1
|
Wei S, Cheng RJ, Li S, Lu C, Zhang Q, Wu Q, Zhao X, Tian X, Zeng X, Liu Y. MSC-microvesicles protect cartilage from degradation in early rheumatoid arthritis via immunoregulation. J Nanobiotechnology 2024; 22:673. [PMID: 39497131 PMCID: PMC11536868 DOI: 10.1186/s12951-024-02922-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 10/10/2024] [Indexed: 11/06/2024] Open
Abstract
OBJECTIVE As research into preclinical rheumatoid arthritis (pre-RA) has advanced, a growing body of evidence suggests that abnormalities in RA-affected joint cartilage precede the onset of arthritis. Thus, early prevention and treatment strategies are imperative. In this study, we aimed to explore the protective effects of mesenchymal stem cell (MSC)-derived microvesicles (MVs) on cartilage degradation in a collagen-induced arthritis (CIA) mouse model. METHODS A CIA mouse model was established to observe early pathological changes in cartilage (days 21-25) through histological and radiological examinations. On day 22, MSCs-MVs were intravenously injected into the mice with CIA. Radiological, histological, and flow cytometric examinations were conducted to observe inflammation and cartilage changes in these mice compared to the mice with CIA and the control mice. In vitro, chondrocytes were cultured with inflammatory factors such as IL-1β and TNFα to simulate inflammatory damage to cartilage. After the addition of MVs, changes in inflammatory levels and collagen expression were measured via Western blotting, immunofluorescence, enzyme-linked immunosorbent assays (ELISAs), and quantitative PCR to determine the role of MVs in maintaining chondrocytes. RESULTS MSC-MVs expressed vesicular membrane proteins (CD63 and Annexin V) and surface markers characteristic of MSCs (CD44, CD73, CD90, and CD105). In the early stages of CIA in mice, a notable decrease in collagen content was observed in the joint cartilage. In mice with CIA, injection of MSCs-MVs resulted in a significant reduction in the peripheral blood levels of IL-1β, TNFα, and IL-6, along with a decrease in the ratio of proinflammatory T and B cells. Additionally, MSC-MVs downregulated the expression of IL-1β, TNFα, MMP-13, and ADAMTS-5 in cartilage while maintaining the stability of type I and type II collagen. These MVs also attenuated the destruction of cartilage, which was evident on imaging. In vitro experiments demonstrated that MSC-MVs effectively suppressed the secretion of the inflammatory factors IL-1β, TNFα, and IL-6 in stimulated peripheral blood mononuclear cells (PBMCs). CONCLUSIONS MSCs-MVs can inhibit the decomposition of the inflammation-induced cartilage matrix by regulating immune cell inflammatory factors to attenuate cartilage destruction. MSC-MVs are promising effective treatments for the early stages of RA.
Collapse
Affiliation(s)
- Shixiong Wei
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College. National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology. State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital. Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Rui-Juan Cheng
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Sujia Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chenyang Lu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qiuping Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qiuhong Wu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xueting Zhao
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xinping Tian
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College. National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology. State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital. Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| | - Xiaofeng Zeng
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College. National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology. State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital. Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
2
|
Woo HE, Cho JY, Lim YH. Propionibacterium freudenreichii MJ2-derived extracellular vesicles inhibit RANKL-induced osteoclastogenesis and improve collagen-induced rheumatoid arthritis. Sci Rep 2024; 14:24973. [PMID: 39443658 PMCID: PMC11500175 DOI: 10.1038/s41598-024-76911-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024] Open
Abstract
Rheumatoid arthritis causes excessive bone loss by stimulating osteoclast differentiation. Extracellular vesicles are valuable disease markers, conveyors of distant cell-to-cell communication, and carriers for drug delivery. The aim of this study was to investigate the anti-osteoclastogenic effects of extracellular vesicles derived from dairy Propionibacterium freudenreichii MJ2 (PFEVs) and the improvement effect of PFEVs on collagen-induced arthritis (CIA) animal model. PFEVs were observed by scanning electron microscopy, transmission electron microscopy, nanoparticle tracking analysis, and LC-MS/MS. The inhibitory activity of PFEVs against receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclast differentiation was investigated in RAW 264.7 cells. PFEVs significantly decreased the expression levels of genes and proteins related to osteoclast differentiation. PFEVs decreased RANK-RANKL binding. In a CIA mouse model, PFEVs treatment significantly reduced arthritis scores and collagen-specific immunoglobulins. PFEVs treatment also reduced pro-inflammatory cytokines and increased anti-inflammatory cytokines. The anti-inflammatory effects were confirmed by H&E staining, and PFEVs treatment inhibited osteoclastogenesis in the CIA mouse model. In conclusion, PFEVs inhibited osteoclast differentiation by inhibiting RANK-RANKL signaling, thereby decreasing the expression of osteoclast differentiation-related genes. PFEVs also improved collagen-induced arthritis by inhibiting inflammation and osteoclastogenesis.
Collapse
Affiliation(s)
- Hee-Eun Woo
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Joo-Young Cho
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Young-Hee Lim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea.
- School of Biosystems and Biomedical Sciences, Korea University, Seoul, 02841, Republic of Korea.
- Department of Laboratory Medicine, Korea University Guro Hospital, Seoul, 08308, Republic of Korea.
| |
Collapse
|
3
|
Kim A, Xie F, Abed OA, Moon JJ. Vaccines for immune tolerance against autoimmune disease. Adv Drug Deliv Rev 2023; 203:115140. [PMID: 37980949 PMCID: PMC10757742 DOI: 10.1016/j.addr.2023.115140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/21/2023]
Abstract
The high prevalence and rising incidence of autoimmune diseases have become a prominent public health issue. Autoimmune disorders result from the immune system erroneously attacking the body's own healthy cells and tissues, causing persistent inflammation, tissue injury, and impaired organ function. Existing treatments primarily rely on broad immunosuppression, leaving patients vulnerable to infections and necessitating lifelong treatments. To address these unmet needs, an emerging frontier of vaccine development aims to restore immune equilibrium by inducing immune tolerance to autoantigens, offering a potential avenue for a cure rather than mere symptom management. We discuss this burgeoning field of vaccine development against inflammation and autoimmune diseases, with a focus on common autoimmune disorders, including multiple sclerosis, type 1 diabetes, rheumatoid arthritis, inflammatory bowel disease, and systemic lupus erythematosus. Vaccine-based strategies provide a new pathway for the future of autoimmune disease therapeutics, heralding a new era in the battle against inflammation and autoimmunity.
Collapse
Affiliation(s)
- April Kim
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Fang Xie
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Omar A Abed
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor 48109, USA.
| |
Collapse
|
4
|
Kang JH, Lee HY, Kim NY, Lee DS, Yim M. Extracellular Prdx1 mediates bacterial infection and inflammatory bone diseases. Life Sci 2023; 333:122140. [PMID: 37797684 DOI: 10.1016/j.lfs.2023.122140] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/30/2023] [Accepted: 09/30/2023] [Indexed: 10/07/2023]
Abstract
AIM We aimed to determine the role of extracellular peroxiredoxin 1 (Prdx1) in the pathogenesis of bacterial infections and inflammatory bone disease. MATERIALS AND METHODS We first investigated the role of Prdx1 using knockout mice. Next, we determined the role of extracellular Prdx1 in bacterial infections by using a neutralizing antibody against Prdx1. We finally investigated whether blockade of extracellular Prdx1 affected high- or low-grade inflammatory bone diseases using calvarial osteolysis, collagen-induced arthritis (CIA), and microgravity-induced bone loss in mouse models. KEY FINDINGS The lack of Prdx1 increased susceptibility to infections by Listeria monocytogenes or Escherichia coli. Prdx1 is released into the serum upon E. coli infection, and blockade of extracellular Prdx1 confers significant protection against bacterial infections. Our data suggested that circulating Prdx1 is increased by the development of osteolytic disease, and that blockade of extracellular Prdx1 exerts therapeutic effects against high- and low-grade inflammatory bone loss. In addition, the release of Prdx1 under inflammatory osteolytic conditions partly depends on non-canonical TIR-domain-containing adapter-inducing interferon-β (TRIF)-caspase-11-gasdemin D (GSDMD) inflammasome pathways. SIGNIFICANCE Extracellular Prdx1 is involved in the development of bacterial infections and inflammatory bone disease. Thus, extracellular Prdx1 may represent a novel therapeutic target for bacterial infections or inflammatory osteolytic diseases.
Collapse
Affiliation(s)
- Ju-Hee Kang
- College of Pharmacy, Sookmyung Women's University, Seoul 140-742, Republic of Korea
| | - Hwa-Yeong Lee
- College of Pharmacy, Sookmyung Women's University, Seoul 140-742, Republic of Korea
| | - Na-Young Kim
- College of Pharmacy, Sookmyung Women's University, Seoul 140-742, Republic of Korea
| | - Dong-Seok Lee
- School of Life Sciences & Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Mijung Yim
- College of Pharmacy, Sookmyung Women's University, Seoul 140-742, Republic of Korea.
| |
Collapse
|
5
|
Wang H. The Potential of Collagen Treatment for Comorbid Diseases. Polymers (Basel) 2023; 15:3999. [PMID: 37836047 PMCID: PMC10574914 DOI: 10.3390/polym15193999] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/27/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Collagen, the most abundant protein in our bodies, plays a crucial role in maintaining the structural integrity of various tissues and organs. Beyond its involvement in skin elasticity and joint health, emerging research suggests that collagen may significantly impact the treatment of complex diseases, particularly those associated with tissue damage and inflammation. The versatile functions of collagen, including skin regeneration, improving joint health, and increasing bone strength, make it potentially useful in treating different diseases. To the best of my knowledge, the strategy of using collagen to treat comorbid diseases has not been widely studied. This paper aims to explore the potential of collagen in treating comorbid diseases, including rheumatoid arthritis, osteoarthritis, osteoporosis, psoriatic arthritis, sarcopenia, gastroesophageal reflux, periodontitis, skin aging, and diabetes mellitus. Collagen-based therapies have shown promise in managing comorbidities due to their versatile properties. The multifaceted nature of collagen positions it as a promising candidate for treating complex diseases and addressing comorbid conditions. Its roles in wound healing, musculoskeletal disorders, cardiovascular health, and gastrointestinal conditions highlight the diverse therapeutic applications of collagen in the context of comorbidity management.
Collapse
Affiliation(s)
- Hsiuying Wang
- Institute of Statistics, National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| |
Collapse
|
6
|
Heger LA, Schommer N, Fukui S, Van Bruggen S, Sheehy CE, Chu L, Rajagopal S, Sivanandhan D, Ewenstein B, Wagner DD. Inhibition of protein arginine deiminase 4 prevents inflammation-mediated heart failure in arthritis. Life Sci Alliance 2023; 6:e202302055. [PMID: 37500179 PMCID: PMC10374941 DOI: 10.26508/lsa.202302055] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
Rheumatoid arthritis is a prototypic inflammatory condition with affected patients being at greater risk of incident heart failure (HF). Targeting innate immune cell function in the pathogenesis of HF bears the potential to guide the development of future therapies. A collagen-induced arthritis (CIA) model in DBA/1 J mice was used to generate arthritis. Mice with CIA developed concentric hypertrophic myocardial remodeling, left ventricular (LV) diastolic dysfunction, and HF with elevated plasma B-type natriuretic peptide levels but preserved LV ejection fraction. Key features of HF in CIA were increased infiltration of activated neutrophils, deposition of neutrophil extracellular traps in the myocardium, and increased tissue levels of the proinflammatory cytokine IL-1β. Specific inhibition of protein arginine deiminase 4 (PAD4) by an orally available inhibitor (JBI-589), administered after the onset of clinical arthritis, prevented HF with reduced neutrophil infiltration. We identify PAD4-mediated neutrophil activation and recruitment as the key thromboinflammatory pathway driving HF development in arthritis. Targeting PAD4 may be a viable therapeutic approach for the prevention of HF secondary to chronic inflammation.
Collapse
Affiliation(s)
- Lukas A Heger
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Departement of Cardiology and Angiology, University Hospital Freiburg Bad Krozingen, Freiburg, Germany
| | - Nicolas Schommer
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Departement of Cardiology and Angiology, University Hospital Freiburg Bad Krozingen, Freiburg, Germany
| | - Shoichi Fukui
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Stijn Van Bruggen
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Center of Molecular and Vascular Biology, Department of Cardiovascular Science, KU Leuven, Leuven, Belgium
| | - Casey E Sheehy
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Long Chu
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | | | | | - Bruce Ewenstein
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Denisa D Wagner
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
7
|
Chen M, Fu W, Xu H, Liu CJ. Tau deficiency inhibits classically activated macrophage polarization and protects against collagen-induced arthritis in mice. Arthritis Res Ther 2023; 25:146. [PMID: 37559125 PMCID: PMC10410869 DOI: 10.1186/s13075-023-03133-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Tau protein serves a pro-inflammatory function in neuroinflammation. However, the role of tau in other inflammatory disorders such as rheumatoid arthritis (RA) is less explored. This study is to investigate the role of endogenous tau and the potential mechanisms in the pathogenesis of inflammatory arthritis. METHODS We established collagen-induced arthritis (CIA) model in wild-type and Tau-/- mice to compare the clinical score and arthritis incidence. Micro-CT analysis was used to evaluate bone erosion of ankle joints. Histological analysis was performed to assess inflammatory cell infiltration, cartilage damage, and osteoclast activity in the ankle joints. Serum levels of pro-inflammatory cytokines were measured by ELISA. The expression levels of macrophage markers were determined by immunohistochemistry staining and quantitative real-time PCR. RESULTS Tau expression was upregulated in joints under inflammatory condition. Tau deletion in mice exhibited milder inflammation and protected against the progression of CIA, evidenced by reduced serum levels of pro-inflammatory cytokines and attenuated bone loss, inflammatory cell infiltration, cartilage damage, and osteoclast activity in the ankle joints. Furthermore, tau deficiency led to the inhibition of classically activated type 1 (M1) macrophage polarization in the synovium. CONCLUSION Tau is a previously unrecognized critical regulator in the pathogenesis of RA and may provide a potential therapeutic target for autoimmune and inflammatory joint diseases.
Collapse
Affiliation(s)
- Meng Chen
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Wenyu Fu
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Huiyun Xu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Chuan-Ju Liu
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY, USA.
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA.
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
8
|
Cui P, Shao T, Liu W, Li M, Yu M, Zhao W, Song Y, Ding Y, Liu J. Advanced review on type II collagen and peptide: preparation, functional activities and food industry application. Crit Rev Food Sci Nutr 2023; 64:11302-11319. [PMID: 37459185 DOI: 10.1080/10408398.2023.2236699] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Type II collagen is a homologous super-helical structure consisting of three identical α1(II) chains. It is a major component of animal cartilage, and is widely used in the food industry. Type II collagen can be extracted by acids, salts, enzymes, and via auxiliary methods and can be further hydrolyzed chemically and enzymatically to produce collagen peptides. Recent studies have shown that type II collagen and its polypeptides have good self-assembly properties and important biological activities, such as maintaining cartilage tissue integrity, inducing immune tolerance, stimulating chondrocyte growth and redifferentiation, and providing antioxidant benefits. This review focuses specifically on type II collagen and describes its structure, extraction, and purification, as well as the preparation of type II collagen peptides. In particular, the self-assembly properties and functional activities of type II collagen and collagen peptides are reviewed. In addition, recent research advances in the application of type II collagen and collagen peptides in functional foods, food additives, food coating materials, edible films, and carriers for the food industry are presented. This paper provides more detailed and comprehensive information on type II collagen and peptide for their application.
Collapse
Affiliation(s)
- Pengbo Cui
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, P. R. China
| | - Tianlun Shao
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, P. R. China
| | - Weilin Liu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, P. R. China
| | - Mengyu Li
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, P. R. China
| | - Mingxiao Yu
- Meitek Technology Company Limited, Qingdao, P. R. China
| | - Weixue Zhao
- Meitek Technology Company Limited, Qingdao, P. R. China
| | - Yanzhuo Song
- Meitek Technology Company Limited, Qingdao, P. R. China
| | - Yuting Ding
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, P. R. China
| | - Jianhua Liu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, P. R. China
| |
Collapse
|
9
|
Zhang Y, Gao Y, Ding Y, Jiang Y, Chen H, Zhan Z, Liu X. Targeting KAT2A inhibits inflammatory macrophage activation and rheumatoid arthritis through epigenetic and metabolic reprogramming. MedComm (Beijing) 2023; 4:e306. [PMID: 37313329 PMCID: PMC10258526 DOI: 10.1002/mco2.306] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 06/15/2023] Open
Abstract
Epigenetic regulation of inflammatory macrophages governs inflammation initiation and resolution in the pathogenesis of rheumatoid arthritis (RA). Nevertheless, the mechanisms underlying macrophage-mediated arthritis injuries remain largely obscure. Here, we found that increased expression of lysine acetyltransferase 2A (KAT2A) in synovial tissues was closely correlated with inflammatory joint immunopathology in both RA patients and experimental arthritis mice. Administration of MB-3, the KAT2A-specific chemical inhibitor, significantly ameliorated the synovitis and bone destruction in collagen-induced arthritis model. Both pharmacological inhibition and siRNA silencing of KAT2A, not only suppressed innate stimuli-triggered proinflammatory gene (such as Il1b and Nlrp3) transcription but also impaired NLR family pyrin domain containing 3 (NLRP3) inflammasome activation in vivo and in vitro. Mechanistically, KAT2A facilitated macrophage glycolysis reprogramming through suppressing nuclear factor-erythroid 2-related factor 2 (NRF2) activity as well as downstream antioxidant molecules, which supported histone 3 lysine 9 acetylation (H3K9ac) and limited NRF2-mediated transcriptional repression of proinflammatory genes. Our study proves that acetyltransferase KAT2A licenses metabolic and epigenetic reprogramming for NLRP3 inflammasome activation in inflammatory macrophages, thereby targeting KAT2A represents a potential therapeutic approach for patients suffering from RA and relevant inflammatory diseases.
Collapse
Affiliation(s)
- Yunkai Zhang
- Department of Pathogen BiologyNaval Medical UniversityShanghaiChina
- National Key Laboratory of Immunity & InflammationNaval Medical UniversityShanghaiChina
| | - Ying Gao
- Department of RheumatologyChanghai Hospital, Naval Medical UniversityShanghaiChina
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaShanghai East Hospital, Tongji University School of MedicineShanghaiChina
| | - Yingying Ding
- Department of Pathogen BiologyNaval Medical UniversityShanghaiChina
| | - Yuyu Jiang
- Department of Pathogen BiologyNaval Medical UniversityShanghaiChina
| | - Huiying Chen
- Department of Pathogen BiologyNaval Medical UniversityShanghaiChina
| | - Zhenzhen Zhan
- Key Laboratory of Arrhythmias of the Ministry of Education of ChinaShanghai East Hospital, Tongji University School of MedicineShanghaiChina
- Department of Liver Surgery, Shanghai Institute of TransplantationRenji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xingguang Liu
- Department of Pathogen BiologyNaval Medical UniversityShanghaiChina
- National Key Laboratory of Immunity & InflammationNaval Medical UniversityShanghaiChina
| |
Collapse
|
10
|
Wang XQ, Cai HH, Deng QW, Chang YZ, Peng YP, Qiu YH. Dopamine D2 receptor on CD4 + T cells is protective against inflammatory responses and signs in a mouse model of rheumatoid arthritis. Arthritis Res Ther 2023; 25:87. [PMID: 37237413 DOI: 10.1186/s13075-023-03071-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 05/20/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Dopamine is a neurotransmitter and has been found to regulate lymphocytes by acting on dopamine receptors (DRs). CD4+ T cells express all the five subtypes of DRs, D1R to D5R. Although CD4+ T cells have been involved in pathogenesis of rheumatoid arthritis (RA), roles of DRs expressed on these cells in RA are poorly understood. This study determined whether D2R expressed on CD4+ T cells regulates inflammatory responses and signs in collagen type II (CII)-induced arthritis (CIA), a mouse model of RA. METHODS DBA/1 mice and C57BL/6 mice with global D1r or D2r deficiency (D1r-/- or D2r-/-) or CD4+ T cell-specific D2r deletion (D2rfl/fl/CD4Cre) were used to prepare CIA model by intradermal injection of CII. D2R agonist sumanirole was intraperitoneally administered in CIA mice. CD4+ T cells obtained from CIA mice were exposed to sumanirole or/and D2R antagonist L-741,626 in vitro. Arthritic symptoms were assessed by clinical arthritis scores. Flow cytometric assay measured frequencies of CD4+ T cell subsets (Th1, Th2, Th17 and Treg cells). Expression of specific transcription factors for the CD4+ T cell subsets was tested by Western blot. Cytokine production was estimated by quantitative PCR and ELISA. RESULTS CIA mice manifested a bias of CD4+ T cells towards Th1 and Th17 cells. D2r-/- CIA mice showed a stronger bias towards Th1 and Th17 phenotypes than CIA mice, while D1r-/- CIA mice did not show the changes. CD4+ T cell-specific D2r deletion exacerbated both the polarization towards Th1 and Th17 cells and the symptoms of arthritis. Sumanirole administration in CIA mice ameliorated the bias of CD4+ T cells towards Th1 and Th17 phenotypes as well as arthritic symptoms. Sumanirole treatment of in vitro CD4+ T cells obtained from CIA mice promoted the shift to Treg cells, and the effect of sumanirole was blocked by L-741,626. CONCLUSIONS D2R expressed on CD4+ T cells is protective against imbalance between pro-inflammatory and anti-inflammatory T cells and arthritic symptoms in CIA.
Collapse
Affiliation(s)
- Xiao-Qin Wang
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, China
| | - Huan-Huan Cai
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, China
| | - Qiao-Wen Deng
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, China
| | - Ya-Zhou Chang
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yu-Ping Peng
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, China.
| | - Yi-Hua Qiu
- Department of Physiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, 226001, China.
| |
Collapse
|
11
|
Wang S, Zhou Y, Huang J, Li H, Pang H, Niu D, Li G, Wang F, Zhou Z, Liu Z. Advances in experimental models of rheumatoid arthritis. Eur J Immunol 2023; 53:e2249962. [PMID: 36330559 DOI: 10.1002/eji.202249962] [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: 04/23/2022] [Revised: 10/16/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by persistent articular inflammation and joint damage. RA was first described over 200 years ago; however, its etiology and pathophysiology remain insufficiently understood. The current treatment of RA is mainly empirical or based on the current understanding of etiology with limited efficacy and/or substantial side effects. Thus, the development of safer and more potent therapeutics, validated and optimized in experimental models, is urgently required. To improve the transition from bench to bedside, researchers must carefully select the appropriate experimental models as well as draw the right conclusions. Here, we summarize the establishment, pathological features, potential mechanisms, advantages, and limitations of the currently available RA models. The aim of the review is to help researchers better understand available RA models; discuss future trends in RA model development, which can help highlight new translational and human-based avenues in RA research.
Collapse
Affiliation(s)
- Siwei Wang
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China.,Honghu Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangtze University, Honghu, Hubei Province, China
| | - Yanhua Zhou
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China.,Honghu Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangtze University, Honghu, Hubei Province, China
| | - Jiangrong Huang
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Huilin Li
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Huidan Pang
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Dandan Niu
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Guangyao Li
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Fei Wang
- Department of Experiment and Training, Hubei College of Chinese Medicine, Hubei Province, China
| | - Zushan Zhou
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China.,Honghu Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangtze University, Honghu, Hubei Province, China
| | - Zhenzhen Liu
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| |
Collapse
|
12
|
Šteigerová M, Šíma M, Slanař O. Pathogenesis of Collagen-Induced Arthritis: Role of Immune Cells with Associated Cytokines and Antibodies, Comparison with Rheumatoid Arthritis. Folia Biol (Praha) 2023; 69:41-49. [PMID: 38063000 DOI: 10.14712/fb2023069020041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Collagen-induced arthritis is the most com-mon in vivo model of rheumatoid arthritis used for investigation of new potential therapies in preclinical research. Rheumatoid arthritis is a systemic inflammatory and autoimmune disease affecting joints, accompanied by significant extra-articular symptoms. The pathogenesis of rheumatoid arthritis and collagen-induced arthritis involves a so far properly unexplored network of immune cells, cytokines, antibodies and other factors. These agents trigger the autoimmune response leading to polyarthritis with cell infiltration, bone and cartilage degeneration and synovial cell proliferation. Our review covers the knowledge about cytokines present in the rat collagen-induced arthritis model and the factors affecting them. In addition, we provide a comparison with rheumatoid arthritis and a description of their important effects on the development of both diseases. We discuss the crucial roles of various immune cells (subtypes of T and B lymphocytes, dendritic cells, monocytes, macrophages), fibroblast-like synoviocy-tes, and their related cytokines (TNF-α, IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12, IL-17, IL-23, GM-CSF, TGF-β). Finally, we also focus on key antibodies (rheu-matoid factor, anti-citrullinated protein antibodies, anti-collagen II antibodies) and tissue-degrading enzymes (matrix metalloproteinases).
Collapse
Affiliation(s)
- Monika Šteigerová
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Martin Šíma
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Ondřej Slanař
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| |
Collapse
|
13
|
Li Z, Bai X, Fan Y, Jia Q, Zhang H, Hou H. Structure of type II collagen from sturgeon cartilage and its effect on adjuvant-induced rheumatoid arthritis in rats. Food Funct 2022; 13:6152-6165. [PMID: 35582851 DOI: 10.1039/d1fo03929f] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The purpose of this paper was to extract and characterize type II collagen of sturgeon cartilage (SC-CII), and to explore the effects of taking SC-CII orally on rheumatoid arthritis (RA) in rats. SC-CII showed a triple-helix structure (RPN = 0.12), with d1 of 11.82 Å and d2 of 4.08 Å, which was analyzed by FT-IR, CD, XRD, and MS. It was constructed of the repeating tripeptide unit Gly-X-Y, where X and Y are generally Pro or Hyp, proved by amino acid composition and peptide mass fingerprinting. Furthermore, the effects of SC-CII on RA were evaluated. Ankle thickness was significantly decreased in SC-CII groups, with changes in lymphocyte proliferation also observed. Compared with the model control group, there was an evident decrease in TNF-α, IL-1β, COX-2, MCP-1, and TLR-4 mRNA levels, but no remarkable differences in APF, MMP-3, and MyD88 mRNA levels in the SC-CII groups. In addition, TNF-α, IL-1β, RF, Anti-CII Ab were significantly reduced in the SC-CII groups, proved by ELISA. Therefore, SC-CII showed alleviating effects on RA through the TLR4/MyD88-NFκB pathway.
Collapse
Affiliation(s)
- Zhaoxia Li
- College of Food Science and Engineering, Ocean University of China, No. 5, YuShan Road, Qingdao, Shandong Province, 266003, P.R. China. .,Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong Province, 266071, P.R. China
| | - Xue Bai
- College of Food Science and Engineering, Ocean University of China, No. 5, YuShan Road, Qingdao, Shandong Province, 266003, P.R. China.
| | - Yan Fan
- College of Food Science and Engineering, Ocean University of China, No. 5, YuShan Road, Qingdao, Shandong Province, 266003, P.R. China.
| | - Qiannan Jia
- College of Food Science and Engineering, Ocean University of China, No. 5, YuShan Road, Qingdao, Shandong Province, 266003, P.R. China.
| | - Hongwei Zhang
- Technology Center of Qingdao Customs, No. 83, Xinyue Road, Qingdao, Shandong Province 266109, P.R. China
| | - Hu Hou
- College of Food Science and Engineering, Ocean University of China, No. 5, YuShan Road, Qingdao, Shandong Province, 266003, P.R. China. .,Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong Province, 266071, P.R. China
| |
Collapse
|
14
|
Yang Y, Shi GN, Wu X, Xu M, Chen CJ, Zhou Y, Wei YZ, Wu L, Cui FF, Sun L, Zhang TT. Quercetin Impedes Th17 Cell Differentiation to Mitigate Arthritis Involving PPARγ-Driven Transactivation of SOCS3 and Redistribution Corepressor SMRT from PPARγ to STAT3. Mol Nutr Food Res 2022; 66:e2100826. [PMID: 35384292 DOI: 10.1002/mnfr.202100826] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 03/03/2022] [Indexed: 01/08/2023]
Abstract
SCOPE Quercetin (QU) is one of the most abundant flavonoids in plants and has attracted the attention of researchers because of its remarkable antirheumatoid arthritis (RA) effects and extremely low adverse reactions. However, the underlying mechanism needs further study. METHODS AND RESULTS Flow cytometry, immunofluorescence, enzyme linked immunosorbent assay (ELISA), and quantitative real-time polymerase chain reaction (qRT-PCR) reveal the obvious inhibitory effects of QU on Th17 cell differentiation in arthritic mice. More importantly, QU markedly limits the development of Th17 cell polarization, which is virtually compromised by the treatment with peroxisome proliferator activated receptor γ (PPARγ) inhibitor GW9662 and knockdown of PPARγ. Additionally, molecular dynamics simulation and immunofluorescence exhibit QU directly binds to PPARγ and increases PPARγ nuclear translocation. Besides, QU confers its moderation effect on suppressor of cytokine signaling protein (SOCS3)/signal transducer and activator of transcription 3 (STAT3) axis partially depending on PPARγ. Furthermore, coimmunoprecipitation shows QU redistributes the corepressor silencing mediator for retinoid and thyroid-hormone receptors (SMRT) from PPARγ to STAT3. Finally, the inhibition of Th17 response and the antiarthritic effect of QU are nullified by GW9662 treatment in arthritic mice. CONCLUSION QU targets PPARγ and consequently inhibits Th17 cell differentiation by dual inhibitory activity of STAT3 to exert antiarthritic effect. The findings facilitate its development and put forth a stage for uncovering the mechanism of other naturally occurring compounds with chemical structures similar to QU.
Collapse
Affiliation(s)
- Yan Yang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Department of Pharmacy, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Gao-Na Shi
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xin Wu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Min Xu
- Department of Pharmacy, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Cheng-Juan Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yu Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ya-Zi Wei
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lei Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fen-Fang Cui
- Department of Pharmacy, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Lan Sun
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tian-Tai Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
15
|
Li W, Ura K, Takagi Y. Industrial application of fish cartilaginous tissues. Curr Res Food Sci 2022; 5:698-709. [PMID: 35479656 PMCID: PMC9035649 DOI: 10.1016/j.crfs.2022.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/22/2022] [Accepted: 04/03/2022] [Indexed: 11/26/2022] Open
Abstract
Cartilage is primarily composed of proteoglycans and collagen. Bioactive compounds derived from animal cartilage, such as chondroitin sulfate and type II collagen, have multiple bioactivities and are incorporated in popular health products. The aging population and increases in degenerative and chronic diseases will stimulate the rapid growth of market demand for cartilage products. Commercial production of bioactive compounds primarily involves the cartilages of mammals and poultry. However, these traditional sources are associated zoonosis concerns; thus, cartilage products from the by-products of fish processing has gained increasing attention because of their high level of safety and other activities. In this review, we summarize the current state of research into fish-derived cartilage products and their application, and discuss future trends and tasks to encourage further expansion and exploitation. At present, shark cartilage is the primary source of marine cartilage. However, the number of shark catches is decreasing worldwide, owing to overfishing. This review considers the potential alternative fish cartilage sources for industrialization. Three keys, the sustainable production of fish, new fish-processing model, and market demand, have been discussed for the future realization of efficient fish cartilage use. The industrialization of fish-derived cartilage products is beneficial for achieving sustainable development of local economies and society. Bioactive compounds derived from fish cartilage are popular as health products. Type II collagen and chondroitin sulfate are the major cartilage bioactive compounds. Cartilaginous fishes, sturgeons, and salmonids are potential fish cartilage sources. Keys for industrialization are fish production, processing model, and market demands. Industrialization of fish cartilage products accords with sustainable development.
Collapse
|
16
|
Li J, Gu P, Tong RS, Wang J, Zhang S, He D, Li JQ. Therapeutic effects of benzoylaconitine and paeoniflorin in rats with collagen-induced arthritis. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e191132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Jie Li
- University of Electronic Science and Technology of China, China; Huadu District People’s Hospital of Guangzhou, China
| | - Ping Gu
- University of Electronic Science and Technology of China, China; Huadu District People’s Hospital of Guangzhou, China
| | - Rong-Sheng Tong
- University of Electronic Science and Technology of China, China; Sichuan Province, China
| | - JiaFeng Wang
- University of Electronic Science and Technology of China, China
| | - ShuHan Zhang
- University of Electronic Science and Technology of China, China
| | - Dan He
- University of Electronic Science and Technology of China, China
| | - Jin-Qi Li
- University of Electronic Science and Technology of China, China; Sichuan Province, China
| |
Collapse
|
17
|
Yeom J, Yim DJ, Ma S, Lim YH. Propionibacterium freudenreichii Inhibits RANKL-Induced Osteoclast Differentiation and Ameliorates Rheumatoid Arthritis in Collagen-Induced Arthritis Mice. Microorganisms 2021; 10:microorganisms10010048. [PMID: 35056497 PMCID: PMC8780394 DOI: 10.3390/microorganisms10010048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 11/16/2022] Open
Abstract
Osteoclast differentiation is crucial for bone absorption, and osteoclasts are involved in bone destruction in rheumatoid arthritis (RA). Dairy Propionibacterium freudenreichii is used as a cheese starter and possesses prebiotic and postbiotic properties. It is known to stimulate the growth of bifidobacteria and produces valuable metabolites, such as vitamin B12 and propionic acid. However, limited information is available on the beneficial effects of P. freudenreichii on human disease. Herein, we aimed to investigate the inhibitory effect of P. freudenreichii MJ2 (MJ2) isolated from raw milk on osteoclast differentiation and evaluate the improvement in RA. The murine macrophage cell line, RAW 264.7, and a collagen-induced arthritis (CIA) mouse model were used to perform in vitro and in vivo studies, respectively. Heat-killed P. freudenreichii MJ2 (hkMJ2)-treated cells significantly inhibited RANKL-induced osteoclast differentiation and TRAP activity. HkMJ2-treated cells exhibited significantly decreased expression of genes and proteins related to RANKL-induced osteoclast differentiation. MJ2 administration decreased the arthritic score in the CIA mouse model. Live and dead MJ2 inhibited bone loss and afforded protection against bone erosion and joint damage in CIA mice. MJ2 decreased the levels of collagen-specific antibodies and inflammatory cytokines and the expression of osteoclast differentiation-related genes and proteins in CIA mice. Interestingly, live and dead MJ2 showed similar RA improvement effects in CIA mice. In conclusion, P. freudenreichii MJ2 inhibited osteoclast differentiation by inhibiting the NF-κB signaling pathway and ameliorated CIA.
Collapse
Affiliation(s)
- Jiah Yeom
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Korea; (J.Y.); (D.J.Y.); (S.M.)
| | - Dong Joon Yim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Korea; (J.Y.); (D.J.Y.); (S.M.)
| | - Seongho Ma
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Korea; (J.Y.); (D.J.Y.); (S.M.)
| | - Young-Hee Lim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Korea; (J.Y.); (D.J.Y.); (S.M.)
- School of Biosystems and Biomedical Sciences, College of Health Science, Korea University, Seoul 02841, Korea
- Department of Laboratory Medicine, Korea University Guro Hospital, Seoul 08308, Korea
- Correspondence: ; Tel.: +82-2-3290-5635
| |
Collapse
|
18
|
Lin L, Gu X, Chen L, Zhang T, Wang C, Wang Z, You Q, Ji L. Study on the alleviation of Fengshi Gutong capsule on rheumatoid arthritis through integrating network pharmacology and experimental exploration. JOURNAL OF ETHNOPHARMACOLOGY 2021; 280:114471. [PMID: 34329717 DOI: 10.1016/j.jep.2021.114471] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/19/2021] [Accepted: 07/26/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fengshi Gutong (FSGT) capsule, a traditional Chinese medicine formula, has effects including warming meridians and dispersing cold, and relieving pain by dredging collaterals. FSGT is generally used for the treatment of rheumatoid arthritis (RA) in clinic in China. AIM OF THE STUDY This study aims to investigate the alleviation provided by FSGT capsule on RA in vivo and the engaged mechanism. MATERIALS AND METHODS The collagen-induced arthritis (CIA) mouse model was used to evaluate the alleviation of FSGT capsule on RA in vivo. Network pharmacology was used to find the potential involved molecular targets. Western-blot, Real-time polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) were conducted. Wound healing assay was performed in human umbilical vein endothelial cells (HUVECs). RESULTS FSGT capsule (300, 900 mg/kg) alleviated RA in CIA mice with no obvious side effects. The results from network pharmacology showed that the top 6 molecular targets involved in the FSGT-provided alleviation on RA were interleukin 6 (IL-6), tumor necrosis factor α (TNFα), C-C motif chemokine 2 (CCL2), vascular endothelial growth factor (VEGF), intercellular cell adhesion molecule-1 (ICAM-1), interleukin 1β (IL-1β), and these results imply the critical participation of inhibiting inflammation or angiogenesis. Next, FSGT capsule decreased the elevated serum contents of rheumatoid factor (RF) and VEGF, and some pro-inflammatory cytokines like TNFα and IL-6. Moreover, FSGT capsule also reduced the elevated protein expression of ICAM1, IL-1β and phosphorylated protein kinase B (Akt) in synovium from CIA mice. Further in vitro results showed that totally 13 compounds from FSGT reduced the enhanced IL-1β and inducible nitric oxide synthase (iNOS) mRNA expression in RAW264.7 macrophages stimulated by lipopolysaccharide (LPS). Meanwhile, 7 compounds from FSGT decreased the VEGF-induced HUVEC migration. Among those compounds, benzoylhypaconine (BHA), pseudoephedrine hydrochloride (PSE), glycyrrhetnic acid (GA), isoliquiritigenin (ISL), quercetin (QUER) and kaempferol (KAE) were found to inhibit both inflammation and angiogenesis in vitro. CONCLUSION FSGT capsule ameliorates RA in CIA mice by reducing inflammation, abrogating angiogenesis and relieving pain. Some compounds in FSGT, including BHA, GA, PSE, ISL, QUER and KAE, reduced both inflammation and angiogenesis in vitro, which suggests that those compounds may contribute to the FSGT capsule-provided alleviation on RA.
Collapse
Affiliation(s)
- Li Lin
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Xinnan Gu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Liangni Chen
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Tianyu Zhang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Changhong Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Zhengtao Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Qingling You
- Department of Orthodontics, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Lili Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and The SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
19
|
Qin J, Huang X, Wang N, Zhou P, Zhang H, Chen Z, Liang K, Gong D, Zeng Q, Niu P, Chen A, Yuan L, Yang Z, Su L, Shen N, Deng J, Yu D. Supranutritional selenium suppresses ROS-induced generation of RANKL-expressing osteoclastogenic CD4 + T cells and ameliorates rheumatoid arthritis. Clin Transl Immunology 2021; 10:e1338. [PMID: 34584694 PMCID: PMC8452973 DOI: 10.1002/cti2.1338] [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: 02/28/2021] [Revised: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE The benefit of Se supplementation in rheumatoid arthritis (RA) has been tested in clinical trials, but results remain inconclusive. The objective of this study was to specifically investigate the potential benefit of supranutritional Se by examining human samples from an area with supranutritional Se intake and testing a mouse model of RA. METHODS Peripheral blood mononuclear cells (PBMCs) from RA patients (N = 57) and healthy controls (HC, N = 71) from an area of supranutritional Se intake (Enshi, Hubei, China) were analysed by flow cytometry. Serum cytokine and Se levels were measured by cytometric beads array (CBA) and inductively coupled plasma mass spectrometry (ICP-MS), respectively. With sufficient or supranutritional selenium intake, mice were induced with collagen-induced arthritis (CIA) and examined for disease activity and immunopathology. The influence of Se supplementation in the generation of RANKL-expressing osteoclastogenic CD4+ T cells was investigated by in vitro assays. RESULTS In Enshi city, HC showed the above-normal concentrations of serum Se concentrations while RA patients were enriched in the normal range (70-150 ng mL-1) or below. RA patients with higher Se levels demonstrated milder disease and lower levels of C-reactive protein, IL-6, RANKL and Th17 cells. In the mouse CIA model, supranutritional Se supplementation delayed disease onset, ameliorated joint pathology and reduced CD4+CD44+RANKL+ T cells. Se supplementation could suppress RANKL expression in cultured mouse Th17 cells. CONCLUSION Supranutritional Se suppresses RANKL-expressing osteoclastogenic CD4+ T cells and could be beneficial to RA, which warrants formal testing in randomised clinical trials.
Collapse
Affiliation(s)
- Jiahuan Qin
- Shanghai Institute of RheumatologyChina‐Australia Centre for Personalized ImmunologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xia Huang
- Department of RheumatologyMinda Hospital of Hubei Minzu UniversityEnshiChina
| | - Naiqi Wang
- The University of Queensland Diamantina InstituteFaculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
| | - Pengcheng Zhou
- The University of Queensland Diamantina InstituteFaculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
| | - Hao Zhang
- Laboratory of Immunology for Environment and HealthSchool of Pharmaceutical ScienceShandong Analysis and Test CenterQilu University of Technology (Shandong Academy of Sciences)JinanChina
| | - Zhian Chen
- The University of Queensland Diamantina InstituteFaculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
| | - Kaili Liang
- Shanghai Institute of RheumatologyChina‐Australia Centre for Personalized ImmunologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Dongcheng Gong
- Shanghai Institute of RheumatologyChina‐Australia Centre for Personalized ImmunologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qunxiong Zeng
- Shanghai Institute of RheumatologyChina‐Australia Centre for Personalized ImmunologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Peng Niu
- Shanghai Institute of RheumatologyChina‐Australia Centre for Personalized ImmunologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Anping Chen
- Department of RheumatologyMinda Hospital of Hubei Minzu UniversityEnshiChina
| | - Lin Yuan
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic DiseasesMinda Hospital of Hubei Minzu UniversityEnshiChina
| | - Zhaohui Yang
- Department of OrthopaedicsMinda Hospital of Hubei Minzu UniversityEnshiChina
| | - Linchong Su
- Department of RheumatologyMinda Hospital of Hubei Minzu UniversityEnshiChina
| | - Nan Shen
- Shanghai Institute of RheumatologyChina‐Australia Centre for Personalized ImmunologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of Medicine (SJTUSM)ShanghaiChina
| | - Jun Deng
- Shanghai Institute of RheumatologyChina‐Australia Centre for Personalized ImmunologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai Jiao Tong University School of Medicine (SJTUSM)ShanghaiChina
| | - Di Yu
- Shanghai Institute of RheumatologyChina‐Australia Centre for Personalized ImmunologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- The University of Queensland Diamantina InstituteFaculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
- Laboratory of Immunology for Environment and HealthSchool of Pharmaceutical ScienceShandong Analysis and Test CenterQilu University of Technology (Shandong Academy of Sciences)JinanChina
| |
Collapse
|
20
|
T-bet represses collagen-induced arthritis by suppressing Th17 lineage commitment through inhibition of RORγt expression and function. Sci Rep 2021; 11:17357. [PMID: 34462459 PMCID: PMC8405656 DOI: 10.1038/s41598-021-96699-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/10/2021] [Indexed: 12/31/2022] Open
Abstract
T-bet is a key transcription factor for the T helper 1 lineage and its expression level is negatively correlated to inflammation in patients with rheumatoid arthritis (RA). Our previous study using T-bet transgenic mice revealed over-expression of T-bet completely suppressed collagen-induced arthritis (CIA), a murine model of RA, indicating a potential suppressive role of T-bet in the pathogenesis of autoimmune arthritis. Here, we show T-bet-deficiency exacerbated CIA. T-bet in CD4 + T cells, but not in CD11c + dendritic cells, was critical for regulating the production of IL-17A, IL-17F, IL-22, and TNFα from CD4 + T cells. T-bet-deficient CD4 + T cells showed higher RORγt expression and increased IL-17A production in RORγt-positive cells after CII immunization. In addition, T-bet-deficient naïve CD4 + T cells showed accelerated Th17 differentiation in vitro. CIA induced in CD4-Cre T-betfl/fl (cKO) mice was more severe and T-bet-deficient CD4 + T cells in the arthritic joints of cKO mice showed higher RORγt expression and increased IL-17A production. Transcriptome analysis of T-bet-deficient CD4 + T cells revealed that expression levels of Th17-related genes were selectively increased. Our results indicate that T-bet in CD4 + T cells repressed RORγt expression and function resulting in suppression of arthritogenic Th17 cells and CIA.
Collapse
|
21
|
Applicability and implementation of the collagen-induced arthritis mouse model, including protocols (Review). Exp Ther Med 2021; 22:939. [PMID: 34335888 PMCID: PMC8290431 DOI: 10.3892/etm.2021.10371] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 03/30/2021] [Indexed: 12/16/2022] Open
Abstract
Animal models of rheumatoid arthritis (RA) are essential for studying the pathogenesis of RA in vivo and determining the efficacy of anti-RA drugs. During the past decades, numerous rodent models of arthritis have been evaluated as potential models and the modeling methods are relatively well-developed. Among these models, the collagen-induced arthritis (CIA) mouse model is the first choice and the most widely used because it may be generated rapidly and inexpensively and is relatively similar in pathogenesis to human RA. To date, there have been numerous classic studies and reviews discussing related pathogeneses and modeling methods. Based on this knowledge, combined with the latest convenient and effective methods for CIA model construction, the present review aims to introduce the model to beginners and clarify important details regarding its use. Information on the origin and pathogenesis of the CIA model, the protocol for establishing it, the rate of successful arthritis induction and the methods used to evaluate the severity of arthritis are briefly summarized. With this information, it is expected that researchers who have recently entered the field or are not familiar with this information will be able to start quickly, avoid unnecessary errors and obtain reliable results.
Collapse
|
22
|
Postlethwaite AE, Tuckey RC, Kim TK, Li W, Bhattacharya SK, Myers LK, Brand DD, Slominski AT. 20 S-Hydroxyvitamin D3, a Secosteroid Produced in Humans, Is Anti-Inflammatory and Inhibits Murine Autoimmune Arthritis. Front Immunol 2021; 12:678487. [PMID: 34276665 PMCID: PMC8278399 DOI: 10.3389/fimmu.2021.678487] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
The ability to use large doses of vitamin D3 (D3) to chronically treat autoimmune diseases such as rheumatoid arthritis (RA) is prohibitive due to its calcemic effect which can damage vital organs. Cytochrome P450scc (CYP11A1) is able to convert D3 into the noncalcemic analog 20S-hydroxyvitamin D3 [20S(OH)D3]. We demonstrate that 20S(OH)D3 markedly suppresses clinical signs of arthritis and joint damage in a mouse model of RA. Furthermore, treatment with 20S(OH)D3 reduces lymphocyte subsets such as CD4+ T cells and CD19+ B cells leading to a significant reduction in inflammatory cytokines. The ratio of T reg cells (CD4+CD25+Foxp3+ T cells) to CD3+CD4+ T cells is increased while there is a decrease in critical complement-fixing anti-CII antibodies. Since pro-inflammatory cytokines and antibodies against type II collagen ordinarily lead to destruction of cartilage and bone, their decline explains why arthritis is attenuated by 20(OH) D3. These results provide a basis for further consideration of 20S(OH)D3 as a potential treatment for RA and other autoimmune disorders.
Collapse
Affiliation(s)
- Arnold E. Postlethwaite
- Research Service, Department of Veterans Affairs Medical Center, Memphis, TN, United States
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Robert C. Tuckey
- School of Molecular Sciences, The University of Western Australia, Perth, WA, Australia
| | - Tae-Kang Kim
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Wei Li
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Syamal K. Bhattacharya
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Linda K. Myers
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - David D. Brand
- Research Service, Department of Veterans Affairs Medical Center, Memphis, TN, United States
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Andrzej T. Slominski
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
- Research Service, Department of Veterans Affairs Medical Center, Birmingham, AL, United States
| |
Collapse
|
23
|
Farooq SM, Ashour HM. Type II Collagen-Specific B Cells Induce Immune Tolerance in Th1-Skewed, Th2-Skewed, and Arthritis-Prone Strains of Mice. Cells 2021; 10:cells10040870. [PMID: 33921248 PMCID: PMC8068989 DOI: 10.3390/cells10040870] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 01/15/2023] Open
Abstract
Antigen-specific regulatory T cells play key immune suppressive roles in autoimmune disease models and regulate the peripheral tolerance achieved via anterior chamber-associated immune deviation (ACAID). Articular cartilage has type II collagen (CII), which is a potent autoantigen protein in arthritis. There has not been much research on the clinical importance of CII-associated diseases. Moreover, the capability of CII to induce immune tolerance has not been previously assessed. We reported that delivery of CII either directly into the eye or via intravenous injection of CII-specific ACAID antigen presenting cells (APCs) can induce ACAID. Here, we hypothesized that peripheral tolerance can be induced following adoptive transfer of in vitro generated CII-specific ACAID B cells to naive mice. Delayed hypersensitivity (DTH) assays were used to assess the suppressive ability of adoptively transferred B cells. Immune responses of ACAID B cell-injected mice were significantly suppressed following challenges with CII as compared to positive controls. This effect was replicated in three different strains of mice (C57BL/6, BALB/c, and DBA/1). Thus, CII-specific ACAID B cells were able to induce immune tolerance in Th1-skewed, Th2-skewed, and arthritis-prone mice. ACAID B cell-mediated tolerance induced by CII could have therapeutic implications for the treatment of CII-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Shukkur M. Farooq
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA;
| | - Hossam M. Ashour
- Department of Integrative Biology, College of Arts and Sciences, University of South Florida, St. Petersburg, FL 33701, USA
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
- Correspondence:
| |
Collapse
|
24
|
Xiong H, Luo M, Ju Y, Zhao Z, Zhang M, Xu R, Ren Y, Yang G, Mei Z. Triterpene saponins from Guo-gang-long attenuate collagen-induced arthritis via regulating A20 and inhibiting MAPK pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 269:113707. [PMID: 33358855 DOI: 10.1016/j.jep.2020.113707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/09/2020] [Accepted: 12/15/2020] [Indexed: 05/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The stems of Entada phaseoloides (L.) Merr commonly named "Guo-gang-long", is a traditional Chinese folk medicine that has been used clinically in China for the treatment of arthritis. Our previous study described that triterpene saponins isolated from "Guo-gang-long" could inhibit the inflammatory response. However, the potential mechanism of "Guo-gang-long" on treatment of arthritis, and whether the triterpene saponins responsible for its anti-arthritic effect are unclear. AIM To investigate the function and mechanisms of the triterpene saponins from E. phaseoloides (ES) in collagen-induced arthritis (CIA) rats. MATERIALS AND METHODS The chemical components of ES were analyzed by HPLC. Anti-arthritic activity of ES was investigated in CIA rats, which was established by immunization with bovine type II collagen. Three doses of ES (25, 50 and 100 mg/kg) were administrated using oral gavage to CIA rats daily for 3 weeks. The anti-arthritic activity of ES was evaluated by clinical arthritis scoring, paw swelling and mechanical sensitivity, as well as histological changes in CIA rats. The impacts of ES on the regulation of the ubiquintin-editing enzyme A20 and MAPK signaling pathway, and production of pro-inflammatory cytokines in CIA rats were examined by Western blot, quantitative real-time PCR, ELISA, and immunohistochemical staining, respectively. RESULTS ES treatment relieved the paw swelling, hyperalgesia and joint destruction, and prevented the progression of arthritis in CIA rats. Meanwhile, ES suppressed the excessive mRNA levels and protein expression of TNF-α and IL-17 in synovial tissues and hind paw joints, and reduced the production of IL-1β, TNF-α and IL-17 in serum. Furthermore, ES up-regulated A20 and suppressed the phosphorylation of p38 and ERK1/2 in hind paw joints, as well as inhibiting the activation of spinal p38 in CIA rats. CONCLUSION ES could relieve rheumatic symptoms and prevent the development of rheumatoid arthritis. The effects of ES may be mediated by reducing proinflammatory cytokine levels, up-regulating A20 expression, reducing p38 and ERK1/2 activation in periphery, and inhibiting of phospho-p38 in spinal cord.
Collapse
Affiliation(s)
- Hui Xiong
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Miao Luo
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Yankun Ju
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Zhongqiu Zhao
- Center for the Study of Itch, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, 63110, USA; Barnes-Jewish Hospital, St. Louis, MO, 63110, USA
| | - Man Zhang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Ran Xu
- College of Biology and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
| | - Yongshen Ren
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Guangzhong Yang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China
| | - Zhinan Mei
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, 430074, China.
| |
Collapse
|
25
|
Berberine Delays Onset of Collagen-Induced Arthritis through T Cell Suppression. Int J Mol Sci 2021; 22:ijms22073522. [PMID: 33805383 PMCID: PMC8037694 DOI: 10.3390/ijms22073522] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/17/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
There is evidence that berberine (BBR), a clinically relevant plant compound, ameliorates clinically apparent collagen-induced arthritis (CIA) in vivo. However, to date, there are no studies involving the use of BBR which explore its prophylactic potential in this model of rheumatoid arthritis (RA). The aim of this study was to determine if prophylactic BBR use during the preclinical phase of collagen-induced arthritis would delay arthritic symptom onset, and to characterize the cellular mechanism underlying such an effect. DBA/1J mice were injected with an emulsion of bovine type II collagen (CII) and complete Freund’s adjuvant (day 0) and a booster injection of CII in incomplete Freund’s adjuvant (day 18) to induce arthritis. Mice were then given i.p. injections of 1 mg/kg/day of BBR or PBS (vehicle with 0.01% DMSO) from days 0 to 28, were left untreated (CIA control), or were in a non-arthritic control group (n = 15 per group). Incidence of arthritis in BBR-treated mice was 50%, compared to 90% in both the CIA and PBS controls. Populations of B and T cells from the spleens and draining lymph nodes of mice were examined on day 14 (n = 5 per group) and day 28 (n = 10 per group). BBR-treated mice had significantly reduced populations of CD4+Th and CD4+CXCR5+ Tfh cells, and an increased proportion of Foxp3+ Treg at days 14 and 28, as well as reduced expression of co-stimulatory molecules CD28 and CD154 at both endpoints. The effect seen on T cell populations and co-stimulatory molecule expression in BBR-treated mice was not mirrored in CD19+ B cells. Additionally, BBR-treated mice experienced reduced anti-CII IgG2a and anti-CII total IgG serum concentrations. These results indicate a potential role for BBR as a prophylactic supplement for RA, and that its effect may be mediated specifically through T cell suppression. However, the cellular effector involved raises concern for BBR prophylactic use in the context of vaccine efficacy and other primary adaptive immune responses.
Collapse
|
26
|
New Studies of Pathogenesis of Rheumatoid Arthritis with Collagen-Induced and Collagen Antibody-Induced Arthritis Models: New Insight Involving Bacteria Flora. Autoimmune Dis 2021; 2021:7385106. [PMID: 33833871 PMCID: PMC8016593 DOI: 10.1155/2021/7385106] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/30/2020] [Accepted: 03/04/2021] [Indexed: 12/23/2022] Open
Abstract
Much public research suggests that autoimmune diseases such as rheumatoid arthritis (RA) are induced by aberrant “self” immune responses attacking autologous tissues and organ components. However, recent studies have reported that autoimmune diseases may be triggered by dysbiotic composition changes of the intestinal bacteria and an imbalance between these bacteria and intestinal immune systems. However, there are a few solid concepts or methods to study the putative involvement and relationship of these inner environmental factors in RA pathogenesis. Fortunately, Collagen-Induced Arthritis (CIA) and Collagen Antibody-Induced Arthritis (CAIA) models have been widely used as animal models for studying the pathogenesis of RA. In addition to RA, these models can be extensively used as animal models for studying complicated hypotheses in many diseases. In this review, we introduce some basic information about the CIA and CAIA models as well as how to apply these models effectively to investigate relationships between the pathogenesis of autoimmune diseases, especially RA, and the dysbiosis of intestinal bacterial flora.
Collapse
|
27
|
Hemshekhar M, Anaparti V, El-Gabalawy H, Mookherjee N. A bioavailable form of curcumin, in combination with vitamin-D- and omega-3-enriched diet, modifies disease onset and outcomes in a murine model of collagen-induced arthritis. Arthritis Res Ther 2021; 23:39. [PMID: 33494792 PMCID: PMC7836561 DOI: 10.1186/s13075-021-02423-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Curcumin (CUR), vitamin D3 (D3), and omega-3-fatty acids (O3FA) individually modulate inflammation and pain in arthritis. Although these supplements are widely used, their combinatorial effects have not been defined. In this study, we examined the effects of a D3 and O3FA (VO)-enriched diet in conjunction with a highly bioavailable form of CUR (Cureit/Acumin™) in a collagen-induced arthritis (CIA) murine model. METHODS Male DBA/1J mice were acclimatized to VO-enriched diet and challenged with bovine collagen II (CII). Bioavailable CUR was administered daily by oral gavage from the onset of CII challenge. Disease severity was determined by monitoring joint thickness and standardized clinical score. Cellular infiltration and cartilage degradation in the joints were assessed by histology, serum cytokines profiled by Meso Scale Discovery multiplex assay, and joint matrix metalloproteinases examined by western blots. RESULTS CUR by itself significantly decreased disease severity by ~ 60%. Administration of CUR in CIA mice taking a VO-enriched diet decreased disease severity by > 80% and maximally delayed disease onset and progression. Some of the disease-modifying effects was mediated by CUR alone, e.g., suppression of serum anti-collagen antibodies and decrease of cellular infiltration and MMP abundance in the joints of CIA mice. Although CUR alone suppressed inflammatory cytokines in serum of CIA mice, the combination of CUR and VO diet significantly enhanced the suppression (> 2-fold compared to CUR) of TNF, IFN-γ, and MCP-1, all known to be associated with RA pathogenesis. CONCLUSION This study provides proof-of-concept that the combination of bioavailable CUR, vitamin D3, and O3FA substantially delays the development and severity of CIA. These findings provide a rationale for systematically evaluating these widely available supplements in individuals at risk for developing future RA.
Collapse
Affiliation(s)
- Mahadevappa Hemshekhar
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB, Canada
| | - Vidyanand Anaparti
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB, Canada
| | - Hani El-Gabalawy
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB, Canada.,Division of Rheumatology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada.,Department of Immunology, University of Manitoba, Winnipeg, MB, R3E3P4, Canada
| | - Neeloffer Mookherjee
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB, Canada. .,Department of Immunology, University of Manitoba, Winnipeg, MB, R3E3P4, Canada.
| |
Collapse
|
28
|
Wei Y, Nie Y, Han Z, Huang H, Liao X, Wang X, Fan Z, Zheng Y. Au@polydopamine nanoparticles/tocilizumab composite as efficient scavengers of oxygen free radicals for improving the treatment of rheumatoid arthritis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111434. [PMID: 33255028 DOI: 10.1016/j.msec.2020.111434] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 08/12/2020] [Accepted: 08/20/2020] [Indexed: 12/18/2022]
Abstract
Rheumatoid arthritis (RA) is the most common chronic autoimmune disorder associated with high-cost, side effects, and low therapeutic effects. To improve the treatment of RA, we originally developed a novel anti-RA Au@polydopamine nanoparticles (PDANPs)/TCZ composite using PDANPs as the binding sites of gold nanoparticles (AuNPs) and the drug carries of tocilizumab (TCZ) through a facile and environmentally-friend method, aiming to effectively scavenge oxygen free radicals (OFR) and inhibit the formation of related inflammatory factors. Characterizations showed that AuNPs with the size of 11.4 ± 2.9 nm randomly distributed onto the surface of PDANPs (145.8 ± 31.9 nm), meanwhile TCZ was chemically cross-linked to PDANPs through Schiff base linkage. The synthesized composite had good biocompatibility that can promote the proliferation and growth of chondrocytes and fibroblasts. More importantly, Au@PDANPs/TCZ composite showed more excellent abilities to scavenge OFR and inhibit the related inflammatory factors in vitro and in vivo than that of AuNPs and PDANPs owing to the synergistic scavenging effect, ensuring its best therapeutic effect in RA therapy. This new composite will have application potential in the treatment of RA related disease.
Collapse
Affiliation(s)
- Yuan Wei
- School of Stomatology, Lanzhou University, Lanzhou 730000, PR China
| | - Yingying Nie
- Institute of Sensing Technology, Gansu Academy of Sciences, Lanzhou 730000, PR China
| | - Zongpu Han
- School of Stomatology, Lanzhou University, Lanzhou 730000, PR China
| | - Haofei Huang
- School of Stomatology, Lanzhou University, Lanzhou 730000, PR China
| | - Xiaozhu Liao
- School of Stomatology, Lanzhou University, Lanzhou 730000, PR China
| | - Xusen Wang
- School of Stomatology, Lanzhou University, Lanzhou 730000, PR China
| | - Zengjie Fan
- School of Stomatology, Lanzhou University, Lanzhou 730000, PR China.
| | - Yan Zheng
- School of Stomatology, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
29
|
Clausen AS, Østergaard DE, Holmberg P, Henriksen JR, Tham J, Damborg PP, Jensen AI, Kjaer A, Hansen AE, Andresen TL. Quantitative determination of 64Cu-liposome accumulation at inflammatory and infectious sites: Potential for future theranostic system. J Control Release 2020; 327:737-746. [PMID: 32920081 DOI: 10.1016/j.jconrel.2020.09.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/14/2020] [Accepted: 09/08/2020] [Indexed: 10/23/2022]
Affiliation(s)
- Anne Skovsbo Clausen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Daniella Elisabet Østergaard
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark
| | - Petter Holmberg
- Department of Translational Medicine, Clinical Infection Medicine, Lund University, Skåne University Hospital, Ruth Lundskogs gata 3, SE-214 28 Malmö, Sweden
| | - Jonas Rosager Henriksen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Anker Engelundsvej 1, DK-2800 Kgs. Lyngby, Denmark
| | - Johan Tham
- Department of Translational Medicine, Clinical Infection Medicine, Lund University, Skåne University Hospital, Ruth Lundskogs gata 3, SE-214 28 Malmö, Sweden
| | - Peter Panduro Damborg
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Bülowsvej 7, DK-1870 Frederiksberg, Denmark
| | - Andreas I Jensen
- Department of Health Technology, The Hevesy Laboratory, Technical University of Denmark, Frederiksborgvej 399, DK-4000 Roskilde, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark.
| | - Anders Elias Hansen
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Denmark; Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Anker Engelundsvej 1, DK-2800 Kgs. Lyngby, Denmark
| | - Thomas Lars Andresen
- Department of Health Technology, Section for Biotherapeutic Engineering and Drug Targeting, Technical University of Denmark, Anker Engelundsvej 1, DK-2800 Kgs. Lyngby, Denmark
| |
Collapse
|
30
|
Buckley BJ, Ali U, Kelso MJ, Ranson M. The Urokinase Plasminogen Activation System in Rheumatoid Arthritis: Pathophysiological Roles and Prospective Therapeutic Targets. Curr Drug Targets 2020; 20:970-981. [PMID: 30516104 PMCID: PMC6700755 DOI: 10.2174/1389450120666181204164140] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 11/12/2018] [Accepted: 11/28/2018] [Indexed: 01/09/2023]
Abstract
Rheumatoid Arthritis (RA) is a chronic and progressive inflammatory disease characterized in its early stages by synovial hyperplasia and inflammatory cell infiltration and later by irreversible joint tissue destruction. The Plasminogen Activation System (PAS) is associated with a wide range of physiological and pathophysiological states involving fibrinolysis, inflammation and tissue remodeling. Various components of the PAS are implicated in the pathophysiology of RA. Urokinase Plasminogen Activator (uPA) in particular is a pro-inflammatory mediator that appears to play an important role in the bone and cartilage destruction associated with RA. Clinical studies have shown that uPA and its receptor uPAR are overexpressed in synovia of patients with rheumatoid arthritis. Further, genetic knockdown and antibody-mediated neutralization of uPA have been shown to be protective against induction or progression of arthritis in animal models. The pro-arthritic role of uPA is differentiated from its haemodynamic counterpart, tissue plasminogen activator (tPA), which appears to play a protective role in RA animal models. This review summarises available evidence supporting the PAS as a critical determinant of RA pathogenesis and highlights opportunities for the development of novel uPAS-targeting therapeutics.
Collapse
Affiliation(s)
- Benjamin J Buckley
- School of Chemistry and Molecular Bioscience, University of Wollongong, NSW 2522, Australia.,Illawarra Health & Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Umar Ali
- School of Chemistry and Molecular Bioscience, University of Wollongong, NSW 2522, Australia.,Illawarra Health & Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Michael J Kelso
- School of Chemistry and Molecular Bioscience, University of Wollongong, NSW 2522, Australia.,Illawarra Health & Medical Research Institute, Wollongong, NSW 2522, Australia
| | - Marie Ranson
- School of Chemistry and Molecular Bioscience, University of Wollongong, NSW 2522, Australia.,Illawarra Health & Medical Research Institute, Wollongong, NSW 2522, Australia
| |
Collapse
|
31
|
De S, Kundu S, Chatterjee M. Generation of a robust model for inducing autoimmune arthritis in Sprague Dawley rats. J Pharmacol Toxicol Methods 2019; 102:106659. [PMID: 31837437 DOI: 10.1016/j.vascn.2019.106659] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 08/23/2019] [Accepted: 11/21/2019] [Indexed: 12/16/2022]
Abstract
INTRODUCTION The prerequisite for any experimental model in animals is similarity with the human disease, uniformity in disease severity and incidence. In antigen-induced arthritis models it is generally recognized that the major limitation is inconsistency in terms of incidence and severity. As access to strains like DBA/1 mice or Lewis rats is difficult for resource restrained laboratories, this study aimed to establish a robust and reproducible animal model of rheumatoid arthritis (RA). METHODS Multiple approaches were undertaken for inducing arthritis in Sprague Dawley (SD) and Wistar rats using complete Freund's adjuvant (CFA), collagen type II (CII) emulsion, or different combinations of CII with low dose CFA along with lipopolysaccharide (LPS). The development of arthritis was evaluated by measuring paw edema, arthritis score, body weight, splenic index, histopathology and radiography of paw tissues. RESULTS The combination of CII with low dose CFA and one injection of LPS resulted in 100% incidence of arthritis with disease severity ranging from moderate to severe and results were corroborated by histopathology and radiography. DISCUSSION In a head-to-head comparison between SD and Wistar rats, the disease profile was better sustained and consistent in SD rats, and the use of CII with low dose CFA and LPS induced features akin to human RA. Taken together, a reproducible model of arthritis was established which can be replicated in any laboratory with limited resources.
Collapse
Affiliation(s)
- Soumita De
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research, 244B Acharya JC Bose Road, Kolkata, India
| | - Sunanda Kundu
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research, 244B Acharya JC Bose Road, Kolkata, India
| | - Mitali Chatterjee
- Department of Pharmacology, Institute of Postgraduate Medical Education and Research, 244B Acharya JC Bose Road, Kolkata, India.
| |
Collapse
|
32
|
Raychaudhuri S, Abria C, Harmany ZT, Smith CM, Kundu‐Raychaudhuri S, Raychaudhuri SP, Chaudhari AJ. Quantitative tracking of inflammatory activity at the peak and trough plasma levels of tofacitinib, a Janus kinase inhibitor, via in vivo
18
F‐FDG PET. Int J Rheum Dis 2019; 22:2165-2169. [DOI: 10.1111/1756-185x.13732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/29/2019] [Accepted: 10/03/2019] [Indexed: 01/11/2023]
Affiliation(s)
- Sanchita Raychaudhuri
- Center for Molecular and Genomic Imaging University of California Davis Davis CA USA
- Icahn School of Medicine at Mount Sinai New York NY USA
| | | | - Zachary T. Harmany
- Center for Molecular and Genomic Imaging University of California Davis Davis CA USA
| | - Charles M. Smith
- Center for Molecular and Genomic Imaging University of California Davis Davis CA USA
| | | | - Siba P. Raychaudhuri
- Veterans Affairs Medical Center Mather CA USA
- Division of Rheumatology, Allergy and Clinical Immunology University of California Davis Sacramento CA USA
| | - Abhijit J. Chaudhari
- Center for Molecular and Genomic Imaging University of California Davis Davis CA USA
- Department of Radiology University of California Davis Sacramento CA USA
| |
Collapse
|
33
|
Wang XS, Cao F, Zhang Y, Pan HF. Therapeutic potential of aryl hydrocarbon receptor in autoimmunity. Inflammopharmacology 2019; 28:63-81. [PMID: 31617124 DOI: 10.1007/s10787-019-00651-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/30/2019] [Indexed: 12/11/2022]
Abstract
Aryl hydrocarbon receptor (AhR), a type of transcriptional factor, is widely expressed in immune cells. The activation of AhR signaling pathway depends on its ligands, which exist in environment and can also be produced by metabolism. Normal expressions of AhR and AhR-mediated signaling may be essential for immune responses, and effects of AhR signaling on the development and function of innate and adaptive immune cells have also been revealed in previous studies. Recent studies also indicate that aberrant AhR signaling may be related to autoimmune diseases, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), multiple sclerosis (MS), autoimmune uveitis (AU), autoimmune diabetes, Behcet's disease (BD) and myasthenia gravis (MG). Moreover, administration of AhR ligands or drugs has been proven effective for improving pathological outcomes in some autoimmune diseases or models. In this review, we summarize the effects of AhR on several innate and adaptive immune cells associated with autoimmunity, and the mechanism on how AhR participates in autoimmune diseases. In addition, we also discuss therapeutic potential and application prospect of AhR in autoimmune diseases, so as to provide valuable information for exploring novel and effective approaches to autoimmune disease treatments.
Collapse
Affiliation(s)
- Xiao-Song Wang
- The First Affiliated Hospital of Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, China.,Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China
| | - Fan Cao
- Department of Clinical Medicine, The Second School of Clinical Medicine, Anhui Medical University, 81 Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Yi Zhang
- Reproductive Medicine Center, Anhui Women and Child Health Care Hospital, 15 Yimin Street, Hefei, Anhui, 230011, China
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, China. .,Anhui Province Key Laboratory of Major Autoimmune Diseases, 81 Meishan Road, Hefei, Anhui, China.
| |
Collapse
|
34
|
Mast cells drive IgE-mediated disease but might be bystanders in many other inflammatory and neoplastic conditions. J Allergy Clin Immunol 2019; 144:S19-S30. [DOI: 10.1016/j.jaci.2019.07.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 06/11/2019] [Accepted: 07/08/2019] [Indexed: 01/05/2023]
|
35
|
Li ZY, Zhou JJ, Luo CL, Zhang LM. Activation of TGR5 alleviates inflammation in rheumatoid arthritis peripheral blood mononuclear cells and in mice with collagen II‑induced arthritis. Mol Med Rep 2019; 20:4540-4550. [PMID: 31702035 PMCID: PMC6797944 DOI: 10.3892/mmr.2019.10711] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 04/02/2019] [Indexed: 02/07/2023] Open
Abstract
Rheumatoid arthritis (RA) is characterized by chronic inflammatory synovitis resulting in progressive joint destruction. Persistent synovial inflammation is induced by activation of various inflammatory cells. G-protein-coupled bile acid receptor 1 (TGR5) is a G-protein-coupled receptor activated by various bile acids, which has been reported to act as a key adaptor in regulating various signaling pathways involved in inflammatory responses and a diverse array of physiological processes, including bile acid synthesis, lipid and carbohydrate metabolism, carcinogenesis, immunity and inflammation. In the present study, TGR5 expression was detected in RA peripheral blood mononuclear cells (PBMCs), and its association with clinical disease activity, histological synovitis severity and radiological joint destruction was analyzed. Subsequently, the role and potential underlying mechanisms of TGR5 in the PBMCs of patients with RA and mice with collagen II-induced arthritis (CIA) were investigated. PBMCs were obtained from 50 patients with RA and 40 healthy controls (HCs). The mRNA and protein expression levels of TGR5 were detected in PBMCs via reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and immunofluorescence staining, respectively. Additionally, the levels of proinflammatory cytokines were analyzed by RT-qPCR and enzyme-linked immunosorbent assay (ELISA). The activation of nuclear factor-κB (NF-κB) and IκB kinase a was determined via western blot analysis. The anti-arthritic and anti-inflammatory effects of LCA on mice with CIA were then investigated. The arthritis score was assessed, and the protein levels of proinflammatory cytokines in the plasma of mice were detected via ELISA. TGR5 mRNA expression was significantly downregulated in the PBMCs of patients with RA compared with in those of the HCs (0.53±0.58 for patients vs. 1.49±0.83 for HCs; P<0.001); similar findings were observed at the protein level. The mRNA expression levels of TGR5 in the PBMCs of patients with RA with a high 28-Joint Disease Activity Score (DAS28) were significantly decreased compared with in patients with a low DAS28 (0.81±0.65 for low score vs. 0.35±0.46 for high score; P=0.002). Furthermore, TGR5 expression was significantly correlated with the levels of C-reactive protein (r=−0.429; P=0.002) and the DAS28 (r=−0.383; P=0.006). RT-qPCR and ELISA analyses indicated that lithocholic acid (LCA, 10 mg/kg/day) attenuated lipopolysaccharide-induced proinflammatory cytokine production via inhibition of NF-κB activity in the PBMCs of patients with RA. In addition, the arthritis score was significantly decreased in LCA-treated CIA mice compared with in non-treated CIA mice. The increased production of tumor necrosis factor-α, interleukin (IL)-1β, IL-6 and IL-8 was significantly reduced in the plasma of LCA-treated CIA mice compared with the control. In conclusion, TGR5 may contribute to the inflammation of PBMCs in patients with RA and mice with CIA.
Collapse
Affiliation(s)
- Zhe-Yong Li
- Department of General Surgery, Sir Run Run Shaw Hospital, Hangzhou, Zhejiang 310017, P.R. China
| | - Jing-Jing Zhou
- Department of Rheumatology, Navy General Hospital, Beijing 100048, P.R. China
| | - Chun-Lei Luo
- Department of Nephrology, Ningbo First Hospital, Ningbo, Zhejiang 3150102, P.R. China
| | - Le-Meng Zhang
- Thoracic Medicine Department 1, Hunan Cancer Hospital, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
36
|
Abstract
Objective: To investigate the potential role of β-galactosidase in altering immunoglobulin G (IgG) galactosylation in serum of rheumatoid arthritis (RA).Methods: The expression level and activity of β-galactosidase in serum and CD 19+ B cells were measured by enzyme-linked immune sorbent assay (ELISA). The effect of β-galactosidase on the N-glycan changes in serum from mice intravenously treated with β-galactosidase was observed by linear ion-trap quadrupole-electrospray ionization mass spectrometry (LTQ-ESI-MS). We established a collagen-induced arthritis (CIA) rat model to explore the biological function of β-galactosidase in RA.Results: The expression level of β-galactosidase in serum of 32 patients was elevated when compared with those of 30 healthy controls. The activity and expression level of β-galactosidase in CD19+ B cells from RA patients was higher than those from healthy controls. The ratio of m/z 1142/937 was reduced in mice treated with β-galactosidase when compared with normal mice. We found that β-galactosidase was implicated in the development of inflammation by affecting body weight and elevating the expression level of interleukin-6, tumor necrosis factor-α, and rheumatoid factor in the serum.Conclusions: Our results suggested the high level of β-galactosidase in B cells and serum of RA patients and revealed that altered β-galactosidase may be implicated in the progression of inflammation.
Collapse
Affiliation(s)
- Zhipeng Su
- Institutes of Biology and Medical Science, Soochow University, Suzhou, PR China
| | - Jingjing Gao
- Institutes of Biology and Medical Science, Soochow University, Suzhou, PR China
| | - Qing Xie
- Institutes of Biology and Medical Science, Soochow University, Suzhou, PR China
| | - Yanping Wang
- Institutes of Biology and Medical Science, Soochow University, Suzhou, PR China
| | - Yunsen Li
- Institutes of Biology and Medical Science, Soochow University, Suzhou, PR China
| |
Collapse
|
37
|
Chuang CH, Cheng YC, Lin SC, Lehman CW, Wang SP, Chen DY, Tsai SW, Lin CC. Atractylodin Suppresses Dendritic Cell Maturation and Ameliorates Collagen-Induced Arthritis in a Mouse Model. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:6773-6784. [PMID: 31154759 DOI: 10.1021/acs.jafc.9b01163] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The aim of this study was to evaluate the immunomodulatory effects of atractylodin, a polyethylene alkyne, on the maturation of bone marrow-derived dendritic cells (BM-DC) as well as its antirheumatic effect on collagen-induced arthritis (CIA) in DBA/1 mice. Our results indicate that atractylodin effectively suppressed the secretion of pro-inflammatory cytokines, expression of costimulatory molecules, and p38 MAPK, ERK, and NF-κBp65 signaling pathways in LPS-incubated dendritic cells (DCs). Additionally, the proliferation and cytokine secretion (IFN-γ and IL-17A) of CD8+ and CD4+ T cells were reduced. In a murine CIA model, intraperitoneal injection of atractylodin significantly alleviated the severity of the disease progression, as indicated by reduced paw swelling, clinical arthritis scores, and pathological changes of joint tissues. In addition, the overall proliferation of T cells stimulated by type II collagen and the abundance of Th1 and Th17 in the spleens were also significantly decreased with atractylodin treatments. Furthermore, atractylodin significantly downregulated the expression levels of CD40, CD80, and CD86 of DCs in the spleens. In conclusion, this study shows for the first time that atractylodin has potential to manipulate the maturation of BM-DCs and should be further explored as a therapeutic agent in the treatment of rheumatoid arthritis (RA).
Collapse
Affiliation(s)
- Cheng Hsuan Chuang
- Institute of Biomedical Science , National Chung-Hsing University , Taichung 402 , Taiwan
| | - Yu-Chieh Cheng
- Institute of Biomedical Science , National Chung-Hsing University , Taichung 402 , Taiwan
- Department of Orthopaedics , Tungs' Taichung Metro Harbor Hospital , Taichung 433 , Taiwan
| | - Shih-Chao Lin
- National Center for Biodefense and Infectious Diseases, School of Systems Biology , George Mason University , Manassas , Virginia 20110 , United States
| | - Caitlin W Lehman
- National Center for Biodefense and Infectious Diseases, School of Systems Biology , George Mason University , Manassas , Virginia 20110 , United States
| | - Shun-Ping Wang
- Department of Orthopaedics , Taichung Veterans General Hospital , Taichung 407 , Taiwan
| | - Der-Yuan Chen
- Division of Immunology and Rheumatology, Department of Internal Medicine , China Medical University Hospital , Taichung 404 , Taiwan
| | - Sen-Wei Tsai
- Department of Physical Medicine and Rehabilitation, Taichung Tzu Chi Hospital , Buddhist Tzu Chi Medical Foundation , Taichung 427 , Taiwan
- Department of Physical Medicine and Rehabilitation, School of Medicine , Tzu Chi University , Hualien 970 , Taiwan
| | - Chi-Chien Lin
- Institute of Biomedical Science , National Chung-Hsing University , Taichung 402 , Taiwan
- Department of Medical Research , China Medical University Hospital , Taichung 404 , Taiwan
- Department of Medical Research and Education , Taichung Veterans General Hospital , Taichung 407 , Taiwan
| |
Collapse
|
38
|
Xiong H, Ding X, Wang H, Jiang H, Wu X, Tu C, Wu C, Pi Y, Yang G, Zhao Z, Mei Z. Tibetan medicine Kuan-Jin-Teng exerts anti-arthritic effects on collagen-induced arthritis rats via inhibition the production of pro-inflammatory cytokines and down-regulation of MAPK signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 57:271-281. [PMID: 30802713 DOI: 10.1016/j.phymed.2018.12.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 12/12/2018] [Accepted: 12/16/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND The stems of Tinospora sinensis (Lour.) Merr commonly named "Kuan-Jin-Teng" in Chinese, have been used to treat rheumatoid arthritis as a Tibetan medicine. PURPOSE The effects of the EtOAc fraction of ethanolic extract from the stems of T. sinensis (KJT) on the pro-inflammatory cytokines and MAPK pathway were studied in collagen-induced arthritis (CIA) model. STUDY DESIGN Anti-arthritic activity of KJT was investigated in CIA model. METHODS The chemical constituents of KJT were analyzed by LC-MS and HPLC. The CIA model was established with injecting the bovine CII emulsified in Freund's adjuvant in Wistar rats. Several doses of KJT (50.0, 100.0 and 200.0 mg/kg) were administrated via oral gavage to CIA rats daily for 4 weeks. The anti-arthritic activity of KJT was investigated by clinical arthritis scoring, paw swelling inspection and hyperalgesia measurement, as well as radiological and histological analysis in CIA rats. The impacts of KJT on the activation of MAPK pathway, production of pro-inflammatory cytokines (TNF-α, IL-1β and IL-17) in ankle joints, serum, and spleen in CIA rats were examined by western blot, immunohistochemical staining, ELISA, and quantitative real-time PCR respectively. Lastly, the effects of KJT on production of the nitric oxide (NO) and pro-inflammatory cytokines as well as the regulation of the phosphorylation of p38 and Erk were detected in lipopolysaccharide (LPS)-stimulated RAW264.7 murine macrophage cells. RESULTS KJT significantly alleviated the paw swelling, hyperalgesia and arthritic severity, and reduced the synovial tissue proliferation and inflammatory cell infiltration in the CIA rats. Moreover, KJT suppressed the production of TNF-α, IL-1β, and IL-17 in ankle joints, serum, and spleen and reversed the up-regulation of the phosphorylation of p38 and Erk in CIA rats. KJT was also demonstrated to inhibit the production of NO and pro-inflammatory cytokines (TNF-α, IL-1β and IL-6), and phosphorylation of p38 and Erk in LPS-stimulated RAW264.7 cells. CONCLUSION These results suggest the mechanisms of KJT performing its anti-arthritis effect may be attributed to inhibiting the production of pro-inflammatory cytokines and down-regulating the MAPK signaling pathway.
Collapse
Affiliation(s)
- Hui Xiong
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Xin Ding
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Hua Wang
- School of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Haiqin Jiang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Xinyan Wu
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Chuyue Tu
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Chaoqun Wu
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Yang Pi
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Guangzhong Yang
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Zhongqiu Zhao
- Center for the Study of Itch, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110, United States; Barnes-Jewish Hospital, St. Louis, MO 63110, United States
| | - Zhinan Mei
- School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan 430074, China.
| |
Collapse
|
39
|
Cao YJ, Xu Y, Liu B, Zheng X, Wu J, Zhang Y, Li XS, Qi Y, Sun YM, Wen WB, Hou L, Wan CP. Dioscin, a Steroidal Saponin Isolated from Dioscorea nipponica, Attenuates Collagen-Induced Arthritis by Inhibiting Th17 Cell Response. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:423-437. [PMID: 30827153 DOI: 10.1142/s0192415x19500216] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Dioscin, a steroidal saponin isolated from Dioscorea nipponica Makino, has previously been shown to possess antiarthritic effects. However, the underlying mechanism is still elusive. Herein, we investigated the therapeutic effects of dioscin on collagen-induced arthritis (CIA) in DBA/1 mice and related mechanism. Cytokine production in CII-specific immune responses were measured by enzyme-linked immunosorbent assay (ELISA); Th17 cell-related gene expression, including IL-17A, ROR[Formula: see text] and IL-23p19, were detected by qPCR analysis; Surface marker, T regulatory (Treg) cells and intracellular cytokines (IL-17A and IFN-[Formula: see text]) were evaluated by flow cytometry. We performed Th17 cell differentiation assay in vitro. Results showed that, in vivo, dioscin treatment significantly reduced the severity of CIA, which was accompanied by decreased Th17 response, but not Th1 and Treg response; dioscin-treated mice also showed lower percentage of CD11b[Formula: see text] Gr-1[Formula: see text] neutrophils; In vitro, dioscin treatment suppressed the differentiation of naive CD4[Formula: see text] T cells into Th17 cell and decreased IL-17A production. Collectively, our results indicate that dioscin exerts antiarthritic effects by inhibiting Th17 cell immune response.
Collapse
Affiliation(s)
- Yong-Jun Cao
- Department of Rheumatology, Nantong Hospital Affiliated to Nanjing, University of Traditional Chinese Medicine, Nantong, Jiangsu 226001, P. R. China
| | - Ying Xu
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Bei Liu
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Xi Zheng
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Jian Wu
- Department of Rheumatology, Nantong Hospital Affiliated to Nanjing, University of Traditional Chinese Medicine, Nantong, Jiangsu 226001, P. R. China
| | - Ying Zhang
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Xiao-Si Li
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Yan Qi
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Yu-Meng Sun
- Department of Rheumatology, Nantong Hospital Affiliated to Nanjing, University of Traditional Chinese Medicine, Nantong, Jiangsu 226001, P. R. China
| | - Wei-Bo Wen
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Lifei Hou
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Chun-Ping Wan
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| |
Collapse
|
40
|
Benabdoun HA, Kulbay M, Rondon EP, Vallières F, Shi Q, Fernandes J, Fahmi H, Benderdour M. In vitro and in vivo assessment of the proresolutive and antiresorptive actions of resolvin D1: relevance to arthritis. Arthritis Res Ther 2019; 21:72. [PMID: 30867044 PMCID: PMC6416871 DOI: 10.1186/s13075-019-1852-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/20/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Resolvin D1 (RvD1), an important member of resolvins, exerts a wide spectrum of biological effects, including resolution of inflammation, tissue repair, and preservation of cell viability. The aim of the present study is to investigate the anti-arthritic potential and clarify the bone protective actions of RvD1 in vitro and in vivo. METHODS RAW264.7 cells were treated with 50 ng/ml LPS for 72 h in the presence or absence of RvD1 (0-500 nM). Primary human monocytes were treated with M-CSF + RANKL for 14 days ± RvD1 (0-500 nM) with or without siRNA against RvD1 receptor FPR2. Expressions of inflammatory mediators, degrading enzymes, osteoclasts (OC) formation, and bone resorption were analyzed. The therapeutic effect of RvD1 (0-1000 ng) was carried out in murine collagen antibody-induced arthritis. Arthritis scoring, joint histology, and inflammatory and bone turnover markers were measured. RESULTS RvD1 is not toxic and inhibits OC differentiation and activation. It decreases bone resorption, as assessed by the inhibition of TRAP and cathepsin K expression, hydroxyapatite matrix resorption, and bone loss. In addition, RvD1 reduces TNF-α, IL-1β, IFN-γ, PGE2, and RANK and concurrently enhances IL-10 in OC. Moreover, in arthritic mice, RvD1 alleviates clinical score, paw inflammation, and bone and joint destructions. Besides, RvD1 reduces inflammatory mediators and markedly decreases serum markers of bone and cartilage turnover. CONCLUSION Our results provide additional evidence that RvD1 plays a key role in preventing bone resorption and other pathophysiological changes associated with arthritis. The study highlights the clinical relevance of RvD1 as a potential compound for the treatment of inflammatory arthritis and related bone disorders.
Collapse
Affiliation(s)
- Houda Abir Benabdoun
- Department of Pharmacology, Université de Montréal, Montreal,, QC, Canada.,Orthopedic Research Laboratory, Hôpital du Sacré-Cœur de Montréal, Room K-3045, 5400 Gouin Blvd. West, Montreal, QC, H4J 1C5, Canada
| | - Merve Kulbay
- Orthopedic Research Laboratory, Hôpital du Sacré-Cœur de Montréal, Room K-3045, 5400 Gouin Blvd. West, Montreal, QC, H4J 1C5, Canada
| | - Elsa-Patricia Rondon
- Orthopedic Research Laboratory, Hôpital du Sacré-Cœur de Montréal, Room K-3045, 5400 Gouin Blvd. West, Montreal, QC, H4J 1C5, Canada
| | - Francis Vallières
- Orthopedic Research Laboratory, Hôpital du Sacré-Cœur de Montréal, Room K-3045, 5400 Gouin Blvd. West, Montreal, QC, H4J 1C5, Canada
| | - Qin Shi
- Orthopedic Research Laboratory, Hôpital du Sacré-Cœur de Montréal, Room K-3045, 5400 Gouin Blvd. West, Montreal, QC, H4J 1C5, Canada
| | - Julio Fernandes
- Orthopedic Research Laboratory, Hôpital du Sacré-Cœur de Montréal, Room K-3045, 5400 Gouin Blvd. West, Montreal, QC, H4J 1C5, Canada.,Department of Surgery, Université de Montréal, Montreal, QC, Canada
| | - Hassan Fahmi
- Osteoarthritis Research Unit, Centre Hospitalier de l'Université de Montréal, Montreal, QC, Canada.,Department of Medicine, University of Montreal, Montreal, QC, Canada
| | - Mohamed Benderdour
- Orthopedic Research Laboratory, Hôpital du Sacré-Cœur de Montréal, Room K-3045, 5400 Gouin Blvd. West, Montreal, QC, H4J 1C5, Canada. .,Department of Surgery, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
41
|
Yimam M, Horm T, Wright L, Jiao P, Hong M, Brownell L, Jia Q. UP1306: A Composition Containing Standardized Extracts of Acacia catechu and Morus alba for Arthritis Management. Nutrients 2019; 11:E272. [PMID: 30691120 PMCID: PMC6413154 DOI: 10.3390/nu11020272] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/23/2019] [Accepted: 01/23/2019] [Indexed: 01/13/2023] Open
Abstract
Osteoarthritis (OA) is characterized by progressive articular cartilage degradation. Although there have been significant advances in OA management, to date, there are no effective treatment options to modify progression of the disease. We believe these unmet needs could be bridged by nutrients from natural products. Collagen induced arthritis in rats was developed and utilized to evaluate anti-inflammatory and cartilage protection activity of orally administered botanical composition, UP1306 (50 mg/kg) and Methotrexate (75 µg/kg) daily for three weeks. Objective arthritis severity markers, urine, synovial lavage, and serum were collected. At necropsy, the hock joint from each rat was collected for histopathology analysis. Urinary cartilage degradation marker (CTX-II), pro-inflammatory cytokines (tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), and IL-6), and proteases (Matrix Metallopeptidase 3 (MMP3) and 13) were measured. Rats treated with UP1306 showed statistically significant improvements in arthritis severity markers, including uCTX-II (91.4% vs. collagen-induced arthritis (CIA)), serum IL-1β, TNF-α, and IL-6 levels as well as synovial MMP-13. The histopathology data were also well aligned with the severity score of arthritis for both UP1306 and Methotrexate. UP1306, a botanical composition that contains a standardized blend of extracts from the heartwood of Acacia catechu and the root bark of Morus alba, could potentially be considered as a dietary supplement product for the management of arthritis.
Collapse
Affiliation(s)
- Mesfin Yimam
- Unigen Inc., 2121 South State Street, Suite 400, Tacoma, WA 98405, USA.
| | - Teresa Horm
- Unigen Inc., 2121 South State Street, Suite 400, Tacoma, WA 98405, USA.
| | - Laura Wright
- Fred Hutch Cancer Research, 1100 Fairview Ave N, Seattle, WA 98109, USA.
| | - Ping Jiao
- Unigen Inc., 2121 South State Street, Suite 400, Tacoma, WA 98405, USA.
| | - Mei Hong
- Unigen Inc., 2121 South State Street, Suite 400, Tacoma, WA 98405, USA.
| | - Lidia Brownell
- Unigen Inc., 2121 South State Street, Suite 400, Tacoma, WA 98405, USA.
| | - Qi Jia
- Unigen Inc., 2121 South State Street, Suite 400, Tacoma, WA 98405, USA.
| |
Collapse
|
42
|
Dai Q, Li J, Yun Y, Wang J. Toll-Like Receptor 4-Myeloid Differentiation Primary Response Gene 88 Pathway Is Involved in the Shikonin Treatment of CIA by Regulating Treg/Th17 Expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2018; 2018:2428546. [PMID: 30643526 PMCID: PMC6311288 DOI: 10.1155/2018/2428546] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/17/2018] [Accepted: 12/04/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigate the effect of shikonin on (CIA) collagen-induced arthritis and its influence and mechanism on the balance between Th17 cells and Treg cells. METHODS Three doses of shikonin were administered orally to mice before the onset of CIA, and celecoxib was used as positive control drug. The arthritis response was monitored visually by macroscopic scoring and hindpaw swelling. Histology of knee was used to assess the occurrence of cartilage destruction and bone erosion. Serum collagen type II (C II) antibody levels associated with CIA were assessed with ELISAs. RT-PCR and quantitative PCR were employed to determine the mRNA expression of cytokines and TLRs in the surface of DCs in the patella with adjacent synovium and spleen in CIA. The expression of cytokines and transcription factors in the peripheral immune organs was tested by Western blotting. RESULTS Shikonin treatment suppressed the macroscopic score and incidence of arthritis. Swelling of hind paws, cartilage destruction, and serum anti-C II concentration were delayed with shikonin when compared to controls. Shikonin treatment suppressed the arthritis in a dose-dependent manner. Moreover, the expression of Th17 cytokines (IL-17A) was greatly inhibited both in the synovium and spleen in treated groups compared with those in control groups. The mRNA and protein levels of IL-10 and TGF-β, however, were upregulated after shikonin treatment. The expression of Foxp3 in the synovium and spleen was upregulated, and the expression of ROR-γt in the synovium and spleen was downregulated after shikonin treatment through RT-PCR, quantitative PCR, and Western blotting. The DCs in the spleen of shikonin-treated mice had lower expression of TLR4 and MyD88, and the expression of TLR2 and TLR9 in the spleen was not different between the two groups. CONCLUSION Shikonin has anti-inflammatory effects on CIA. Shikonin treatment can inhibit Th17 cytokines expression and induce Treg responses through inhibiting the activation of TLR4/MyD88 pathway.
Collapse
Affiliation(s)
- Qiaomei Dai
- Department of Pathology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ji Li
- Department of Chinese Formulae, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yu Yun
- Department of Oncology, Traditional Chinese Medical Hospital of Siyang County, Jiangsu, China
| | - Jianwei Wang
- Department of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
43
|
Wu J, Zhao FT, Fan KJ, Zhang J, Xu BX, Wang QS, Tang TT, Wang TY. Dihydromyricetin Inhibits Inflammation of Fibroblast-Like Synoviocytes through Regulation of Nuclear Factor- κB Signaling in Rats with Collagen-Induced Arthritis. J Pharmacol Exp Ther 2018; 368:218-228. [PMID: 30530730 DOI: 10.1124/jpet.118.253369] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/26/2018] [Indexed: 12/15/2022] Open
Abstract
Dihydromyricetin (DMY), the main flavonoid of Ampelopsis grossedentata, has potent anti-inflammatory activity. However, the effect of DMY on chronic autoimmune arthritis remains undefined. In this study, we investigated the therapeutic effects of DMY on collagen-induced arthritis (CIA). Wistar rats were immunized with bovine type II collagen to establish CIA and were then administered DMY intraperitoneally (5, 25, and 50 mg/kg) every other day for 5 weeks. Paw swelling, clinical scoring, and histologic analysis were assessed to determine the therapeutic effects of DMY on the development of arthritis in CIA rats. The results showed that treatment with DMY significantly reduced erythema and swelling in the paws of CIA rats. Pathologic analysis of the knee joints and peripheral blood cytokine assay results confirmed the antiarthritic effects of DMY on synovitis and inflammation. Fibroblast-like synoviocytes (FLSs) were isolated from the synovium of CIA rats and treated with 10 ng/ml interleukin (IL)-1β DMY significantly inhibited the proliferation, migration, and inflammation of IL-1β-induced FLSs, whereas it significantly increased IL-1β-induced FLS apoptosis in a dose-dependent manner (6.25-25 μM). Moreover, DMY suppressed phosphorylation of IκB kinase (IKK) and inhibitor of NF-κB α and subsequently reduced the IL-1β-induced nucleus translocation of NF-κB in FLSs. Through a molecular docking assay, we demonstrated that DMY could directly bind to the Thr9 and Asp88 residues in IKKα and the Asp95, Asn142, and Gln167 residues in IKKβ These findings demonstrate that DMY could alleviate inflammation in CIA rats and attenuate IL-1β-induced activities in FLSs through suppression of NF-κB signaling.
Collapse
Affiliation(s)
- Jing Wu
- Departments of Pharmacy (J.W., K.-J.F., B.-X.X., Q.-S.W., T.-Y.W.) and Rheumatology and Immunology (F.-T.Z.), and Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery (T.-T.T.), Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; and Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas (J.Z.)
| | - Fu-Tao Zhao
- Departments of Pharmacy (J.W., K.-J.F., B.-X.X., Q.-S.W., T.-Y.W.) and Rheumatology and Immunology (F.-T.Z.), and Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery (T.-T.T.), Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; and Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas (J.Z.)
| | - Kai-Jian Fan
- Departments of Pharmacy (J.W., K.-J.F., B.-X.X., Q.-S.W., T.-Y.W.) and Rheumatology and Immunology (F.-T.Z.), and Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery (T.-T.T.), Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; and Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas (J.Z.)
| | - Jun Zhang
- Departments of Pharmacy (J.W., K.-J.F., B.-X.X., Q.-S.W., T.-Y.W.) and Rheumatology and Immunology (F.-T.Z.), and Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery (T.-T.T.), Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; and Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas (J.Z.)
| | - Bing-Xing Xu
- Departments of Pharmacy (J.W., K.-J.F., B.-X.X., Q.-S.W., T.-Y.W.) and Rheumatology and Immunology (F.-T.Z.), and Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery (T.-T.T.), Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; and Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas (J.Z.)
| | - Qi-Shan Wang
- Departments of Pharmacy (J.W., K.-J.F., B.-X.X., Q.-S.W., T.-Y.W.) and Rheumatology and Immunology (F.-T.Z.), and Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery (T.-T.T.), Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; and Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas (J.Z.)
| | - Ting-Ting Tang
- Departments of Pharmacy (J.W., K.-J.F., B.-X.X., Q.-S.W., T.-Y.W.) and Rheumatology and Immunology (F.-T.Z.), and Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery (T.-T.T.), Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; and Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas (J.Z.)
| | - Ting-Yu Wang
- Departments of Pharmacy (J.W., K.-J.F., B.-X.X., Q.-S.W., T.-Y.W.) and Rheumatology and Immunology (F.-T.Z.), and Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery (T.-T.T.), Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China; and Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas (J.Z.)
| |
Collapse
|
44
|
Liu W, Sun Y, Cheng Z, Guo Y, Liu P, Wen Y. Crocin exerts anti-inflammatory and anti-arthritic effects on type II collagen-induced arthritis in rats. PHARMACEUTICAL BIOLOGY 2018; 56. [PMID: 29540097 PMCID: PMC6168764 DOI: 10.1080/13880209.2018.1448874] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
CONTEXT Rheumatoid arthritis (RA) is a common systemic auto-immune disease, which is characterized by chronic and symmetry synovial inflammation. Crocin has been reported to exhibit anti-inflammatory effects in animal models. OBJECTIVE This study investigates the anti-inflammatory and anti-arthritic effects of crocin on type II collagen-induced arthritis (CIA) in Wistar rats. MATERIALS AND METHODS The CIA rat model was established and randomly divided into five groups with or without crocin treatment (10, 20 or 40 mg/kg), which was started on day 21 after arthritis induction and persisted for 36 days. The symptoms and molecular mechanisms of CIA and crocin-treated CIA rats were compared and investigated. RESULTS CIA rats presented severe RA symptoms, including high arthritis score, paw swelling, joint inflammation, bone erosion, chondrocyte death, cartilage destruction, enhanced expressions of matrix metalloproteinase (MMP) and pro-inflammatory cytokines. However, crocin could mitigate these symptoms. Crocin (40 mg/kg) exhibited the most efficient therapeutic function on CIA rats: the histological scores of joint inflammation, bone erosion, chondrocyte death, cartilage surface erosion, and bone erosion of CIA rats receiving 40 mg/kg crocin treatment were comparable to the normal rats. MMP-1, -3 and -13 protein expression levels of CIA rats with 40 mg/kg crocin treatment were decreased to levels similar to normal rats. Moreover, crocin could also inhibit the expression of TNF-α, IL-17, IL-6 and CXCL8 in serum and ankle tissues of CIA rats. CONCLUSIONS In summary, crocin exhibits therapeutic potential for RA, by mitigating the symptoms and inhibiting the pro-inflammatory factor expression.
Collapse
Affiliation(s)
- Wei Liu
- Department of Orthopaedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, China
| | - Yufeng Sun
- Department of Orthopaedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, China
- CONTACT Yufeng Sun Department of Orthopaedics, The Fifth Hospital of Harbin, No. 27 Jiankang Road, Harbin150010, Heilongjiang, China
| | - Zhenping Cheng
- Department of Orthopaedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, China
| | - Yong Guo
- Department of Orthopaedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, China
| | - Peiming Liu
- Department of Orthopaedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, China
| | - Ying Wen
- Department of Orthopaedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, China
| |
Collapse
|
45
|
Zhao X, Long J, Liang F, Liu N, Sun Y, Xi Y. Vaccination with a Novel Antigen-Specific Tolerizing DNA Vaccine Encoding CCOL2A1 Protects Rats from Experimental Rheumatoid Arthritis. Hum Gene Ther 2018; 30:69-78. [PMID: 29901407 DOI: 10.1089/hum.2018.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Antigen-specific tolerizing DNA vaccines are one of the most promising strategies for rheumatoid arthritis (RA) treatment. They act by inducing potent immune tolerance instead of generalized immunosuppression. Recently, we developed a novel antigen-specific tolerizing DNA vaccine pcDNA-CCOL2A1 coding for chicken type II collagen (CCII) and confirmed its potent therapeutic efficacy in an established rat model of collagen-induced arthritis (CIA). Here we report the prophylactic vaccination efficacy of a single 300 μg/kg dose of pcDNA-CCOL2A1 against CIA incidence, severity, and onset. CCOL2A1 transcripts were detected in the blood of CIA rats 14-42 days after intramuscular injection by 300 μg/kg pcDNA-CCOL2A1. The expression of CCOL2A1 transcripts increased quickly on day 21, peaked at day 28, and then gradually decreased thereafter. Importantly, a single prophylactic vaccination of pcDNA-CCOL2A1 14 days before CIA establishment significantly reduced CIA incidence and severity, deferred its onset, and was as efficacious as the current gold standard drug, methotrexate. The marked effects on CIA incidence and severity closely corresponded to the expression of CCOL2A1. Furthermore, prophylactic vaccination with pcDNA-CCOL2A1 markedly decreased serum content of anti-type II collagen (CII) immunoglobulin G (IgG) antibodies, induced Th1-to-Th2 and Tc1-to-Tc2 shifts, and decreased the percentages of CD4+CD29+ and Th17 T cells. Prophylactic vaccination with pcDNA-CCOL2A1 also downregulated various Th1 cytokines, while upregulating both the Th2-type cytokine interleukin-10 and the Th3-type cytokine transforming growth factor β. Our results indicate that the pcDNA-CCOL2A1 DNA vaccine acts as a highly efficient inducer of specific immunotolerance that could be a promising option for RA treatment in the near future.
Collapse
Affiliation(s)
- Xiao Zhao
- Department of Immunology and National Center for Biomedicine Analysis, Beijing 307 Hospital, Beijing, P.R. China
| | - Juan Long
- Department of Immunology and National Center for Biomedicine Analysis, Beijing 307 Hospital, Beijing, P.R. China
| | - Fei Liang
- Department of Immunology and National Center for Biomedicine Analysis, Beijing 307 Hospital, Beijing, P.R. China
| | - Nan Liu
- Department of Immunology and National Center for Biomedicine Analysis, Beijing 307 Hospital, Beijing, P.R. China
| | - Yuying Sun
- Department of Immunology and National Center for Biomedicine Analysis, Beijing 307 Hospital, Beijing, P.R. China
| | - Yongzhi Xi
- Department of Immunology and National Center for Biomedicine Analysis, Beijing 307 Hospital, Beijing, P.R. China
| |
Collapse
|
46
|
Lee JY, Kim GJ, Choi JK, Choi YA, Jeong NH, Park PH, Choi H, Kim SH. 4-(Hydroxymethyl)catechol Extracted From Fungi in Marine Sponges Attenuates Rheumatoid Arthritis by Inhibiting PI3K/Akt/NF-κB Signaling. Front Pharmacol 2018; 9:726. [PMID: 30079020 PMCID: PMC6062625 DOI: 10.3389/fphar.2018.00726] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 06/15/2018] [Indexed: 12/18/2022] Open
Abstract
Rheumatoid arthritis (RA) is a progressive autoimmune disease specific to synovial joints; it causes joint damage and other systemic abnormalities, thereby leading to physical disability and early mortality. Marine sponge-derived fungi, Pestalotiopsis sp., secrete immunosuppressive compounds in the culture broth. In the present study, we isolated 4-(hydroxymethyl)catechol (4-HMC) from these fungal species, and evaluated its anti-RA effects using a murine collagen-induced arthritis model and tumor necrosis factor-α-stimulated human RA synovial fibroblasts. Oral 4-HMC administration decreased the clinical arthritis score, paw thickness, histologic and radiologic changes, and serum IgG1 and IgG2a levels. It prevented the proliferation of helper T (Th) 1/Th17 CD4+ lymphocytes isolated from inguinal lymph nodes, thereby reducing inflammatory cytokine production in CIA mice. It decreased the expression of inflammatory mediators, including cytokines and matrix metalloproteinases (MMPs), both in vitro and in vivo. We observed that 4-HMC suppresses Th immune responses and MMP expression to inhibit inflammatory cytokine production in human RA synovial fibroblasts by modulating the PI3K/Akt/NF-κB pathway. These results verify the anti-RA potential of 4-HMC.
Collapse
Affiliation(s)
- Jong Y Lee
- Department of Pharmacology, School of Medicine, CMRI, Kyungpook National University, Daegu, South Korea
| | - Geum J Kim
- College of Pharmacy, Yeungnam University, Gyeongsan, South Korea
| | - Jin K Choi
- Department of Pharmacology, School of Medicine, CMRI, Kyungpook National University, Daegu, South Korea.,Molecular Immunology Section, Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD, United States
| | - Young-Ae Choi
- Department of Pharmacology, School of Medicine, CMRI, Kyungpook National University, Daegu, South Korea
| | - Na-Hee Jeong
- Department of Pharmacology, School of Medicine, CMRI, Kyungpook National University, Daegu, South Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, South Korea
| | - Hyukjae Choi
- College of Pharmacy, Yeungnam University, Gyeongsan, South Korea
| | - Sang-Hyun Kim
- Department of Pharmacology, School of Medicine, CMRI, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
47
|
Therapeutic Potential of Sclareol in Experimental Models of Rheumatoid Arthritis. Int J Mol Sci 2018; 19:ijms19051351. [PMID: 29751535 PMCID: PMC5983692 DOI: 10.3390/ijms19051351] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 04/25/2018] [Accepted: 04/26/2018] [Indexed: 12/20/2022] Open
Abstract
Previous studies have shown that the natural diterpene compound, sclareol, potentially inhibits inflammation, but it has not yet been determined whether sclareol can alleviate inflammation associated with rheumatoid arthritis (RA). Here, we utilized human synovial cell line, SW982, and an experimental murine model of rheumatoid arthritis, collagen-induced arthritis (CIA), to evaluate the therapeutic effects of sclareol in RA. Arthritic DBA/1J mice were dosed with 5 and 10 mg/kg sclareol intraperitoneally every other day over 21 days. Arthritic severity was evaluated by levels of anti-collagen II (anti-CII) antibody, inflammatory cytokines, and histopathologic examination of knee joint tissues. Our results reveal that the serum anti-CII antibody, cytokines interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α, and IL-17, as well as Th17 and Th1 cell population in inguinal lymph nodes, were significantly lower in sclareol-treated mice compared to the control group. Also, the sclareol treatment groups showed reduced swelling in the paws and lower histological arthritic scores, indicating that sclareol potentially mitigates collagen-induced arthritis. Furthermore, IL-1β-stimulated SW982 cells secreted less inflammatory cytokines (TNF-α and IL-6), which is associated with the downregulation of p38-mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase (ERK), and NF-κB pathways. Overall, we demonstrate that sclareol could relieve arthritic severities by modulating excessive inflammation and our study merits the pharmaceutical development of sclareol as a therapeutic treatment for inflammation associated with RA.
Collapse
|
48
|
Kanashiro A, Shimizu Bassi G, de Queiróz Cunha F, Ulloa L. From neuroimunomodulation to bioelectronic treatment of rheumatoid arthritis. ACTA ACUST UNITED AC 2018; 1:151-165. [PMID: 30740246 DOI: 10.2217/bem-2018-0001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neuronal stimulation is an emerging field in modern medicine to control organ function and reestablish physiological homeostasis during illness. The nervous system innervates most of the peripheral organs and provides a fine tune to control the immune system. Most of these studies have focused on vagus nerve stimulation and the physiological, cellular and molecular mechanisms regulating the immune system. Here, we review the new results revealing afferent vagal signaling pathways, immunomodulatory brain structures, spinal cord-dependent circuits, neural and non-neural cholinergic/catecholaminergic signals and their respective receptors contributing to neuromodulation of inflammation in rheumatoid arthritis. These new neuromodulatory networks and structures will allow the design of innovative bioelectronic or pharmacological approaches for safer and low-cost treatment of arthritis and related inflammatory disorders.
Collapse
Affiliation(s)
- Alexandre Kanashiro
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.,Department of Physiological Sciences, Federal University of São Carlos (UFSCAR), São Carlos, SP, Brazil
| | - Gabriel Shimizu Bassi
- Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fernando de Queiróz Cunha
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Luis Ulloa
- Department of Surgery, Center of Immunology & Inflammation, Rutgers-New Jersey Medical School, Rutgers University, Newark, NJ 07101, USA
| |
Collapse
|
49
|
Hung LH, Wu CH, Lin BF, Hwang LS. Hyperimmune colostrum alleviates rheumatoid arthritis in a collagen-induced arthritis murine model. J Dairy Sci 2018; 101:3778-3787. [DOI: 10.3168/jds.2017-13572] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 12/28/2017] [Indexed: 11/19/2022]
|
50
|
Jayashree S, Nirekshana K, Guha G, Bhakta-Guha D. Cancer chemotherapeutics in rheumatoid arthritis: A convoluted connection. Biomed Pharmacother 2018; 102:894-911. [PMID: 29710545 DOI: 10.1016/j.biopha.2018.03.123] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 03/20/2018] [Accepted: 03/20/2018] [Indexed: 12/11/2022] Open
Abstract
Chemotherapy is one of the most popular therapeutic strategies to treat cancer. However, cancer chemotherapeutics have often been associated with impairment of the immune system, which might consequently lead to an augmented risk of autoimmune disorders, such as rheumatoid arthritis. Though the accurate mechanistic facets of rheumatoid arthritis induction have not been interpreted yet, a conglomeration of genetic and environmental factors might promote its etiology. What makes the scenario more challenging is that patients with rheumatoid arthritis are at a significantly elevated risk of developing various types of cancer. It is intriguing to note that diverse cancer chemotherapy drugs are also commonly used to treat symptoms of rheumatoid arthritis. However, a colossal multitude of such cancer therapeutics has demonstrated highly varied results in rheumatoid arthritis patients, including both beneficial and adverse effects. Herein, we attempt to present a holistic account of the variegated modalities of this complex tripartite cross-talk between cancer, rheumatoid arthritis and chemotherapy drugs in order to decode the sinuous correlation between these two appalling pathological conditions.
Collapse
Affiliation(s)
- S Jayashree
- Cellular Dyshomeostasis Laboratory (CDHL), Department of Biotechnology, School of Chemical and Bio Technology, SASTRA University, Thanjavur, 613 401, Tamil Nadu, India
| | - K Nirekshana
- Cellular Dyshomeostasis Laboratory (CDHL), Department of Biotechnology, School of Chemical and Bio Technology, SASTRA University, Thanjavur, 613 401, Tamil Nadu, India
| | - Gunjan Guha
- Cellular Dyshomeostasis Laboratory (CDHL), Department of Biotechnology, School of Chemical and Bio Technology, SASTRA University, Thanjavur, 613 401, Tamil Nadu, India.
| | - Dipita Bhakta-Guha
- Cellular Dyshomeostasis Laboratory (CDHL), Department of Biotechnology, School of Chemical and Bio Technology, SASTRA University, Thanjavur, 613 401, Tamil Nadu, India.
| |
Collapse
|