1
|
Walton BL, Shattuck-Brandt R, Hamann CA, Tung VW, Colazo JM, Brand DD, Hasty KA, Duvall CL, Brunger JM. A programmable arthritis-specific receptor for guided articular cartilage regenerative medicine. Osteoarthritis Cartilage 2025; 33:231-240. [PMID: 39706287 PMCID: PMC12019866 DOI: 10.1016/j.joca.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/26/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
OBJECTIVE Investigational cell therapies have been developed as disease-modifying agents for the treatment of osteoarthritis (OA), including those that inducibly respond to inflammatory factors driving OA progression. However, dysregulated inflammatory cascades do not specifically signify the presence of OA. Here, we deploy a synthetic receptor platform that regulates cell behaviors in an arthritis-specific fashion to confine transgene expression to sites of cartilage degeneration. DESIGN A single-chain variable fragment specific for type II collagen (CII) that is exposed in damaged cartilage was used to produce a synthetic Notch (synNotch) receptor that enables "CII-synNotch" mesenchymal stromal cells (MSCs) to recognize degraded cartilage. Artificial signaling induced by both CII-treated culture surfaces and primary tissues was measured via fluorescence and luminescence assays. Separate studies measured the ability of CII-synNotch to govern cartilage anabolic activity of MSCs. Finally, a co-culture with ATDC5 chondrocytes was used to determine whether CII-synNotch MSCs can protect chondrocytes against deleterious effects of pro-inflammatory interleukin-1 in a CII-dependent manner. RESULTS CII-synNotch MSCs are highly and selectively responsive to CII, but not type I collagen, as measured by luminescence assays, fluorescence microscopy, and concentrations of secreted transgene products in culture media. CII-synNotch cells exhibit the capacity to distinguish between healthy and damaged cartilage tissue and constrain transgene expression to regions of exposed CII fibers. Receptor-regulated production of cartilage anabolic and anti-inflammatory transgenes was effective to mediate cartilage regenerative functions. CONCLUSION This work demonstrates proof-of-concept that the synNotch platform guides MSCs for spatially regulated, disease-dependent delivery of OA-relevant biologic drugs.
Collapse
Affiliation(s)
- Bonnie L Walton
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA
| | | | - Catherine A Hamann
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA
| | - Victoria W Tung
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA
| | - Juan M Colazo
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA
| | - David D Brand
- Research Service, Lt. Col. Luke Weathers, Jr. VA Medical Center, Memphis, TN 38105, USA
| | - Karen A Hasty
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis VA Medical Center, Memphis, TN 38105, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA; Center for Bone Biology, Vanderbilt University, Nashville, TN 37212, USA
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212, USA; Center for Bone Biology, Vanderbilt University, Nashville, TN 37212, USA; Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37212, USA.
| |
Collapse
|
2
|
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
|
3
|
Zhang F, Zhang Y, Zhou J, Cai Y, Li Z, Sun J, Xie Z, Hao G. Metabolic effects of quercetin on inflammatory and autoimmune responses in rheumatoid arthritis are mediated through the inhibition of JAK1/STAT3/HIF-1α signaling. Mol Med 2024; 30:170. [PMID: 39390367 PMCID: PMC11468292 DOI: 10.1186/s10020-024-00929-1] [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: 06/06/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Rheumatoid arthritis, a chronic autoimmune disease, is characterized by synovial hyperplasia and cartilage erosion. Here, we investigated the potential mechanism of action of quercetin, the main component of flavonoids, in treating rheumatoid arthritis. OBJECT To examine the anti-arthritic effects of quercetin and elucidate the specific mechanisms that differentiate its metabolic effects on autoimmune and inflammatory responses at the synovial cell level. METHODS We created a collagen-induced arthritis (CIA) model in Wistar rats, which were administered quercetin (50 or 100 mg/kg) continuously for four weeks via stomach perfusion. The arthritis score, histopathological staining, radiological assessment, and serum biochemical parameters were used to study the impact of quercetin on disease improvement. Additionally, immunofluorescence was employed to detect JAK1/STAT3/HIF-1α expression in rat joints. Moreover, the effects of quercetin (20, 40, and 80 µmol/L) on the properties and behavior of synovial fibroblasts were evaluated in an in vitro MH7A cell model using flow cytometry, CCK8, and transwell assays. Further, the mRNA expression levels of inflammatory cytokines IL1β, IL6, IL17, and TNFα were assessed by quantitative real-time PCR. Glucose, lactate, lactate dehydrogenase, pyruvate, pyruvate dehydrogenase, and adenosine triphosphate assay kits were employed to measure the metabolic effects of quercetin on synovial fibroblasts. Finally, immunoblotting was used to examine the impact of quercetin on the JAK1/STAT3/HIF-1α signaling pathway in synovial fibroblasts. RESULTS In vivo experiments confirmed the favorable effects of quercetin in CIA rats, including an improved arthritis score and reduced ankle bone destruction, in addition to a decrease in the pro-inflammatory cytokines IL-1β, IL-6, IL-17, and TNF-α in serum. Immunofluorescence verified that quercetin may ameliorate joint injury in rats with CIA by inhibiting JAK1/STAT3/HIF-1α signaling. Various in vitro experiments demonstrated that quercetin effectively inhibits IL-6-induced proliferation of MH7A cells and reduces their migratory and invasive behavior, while inducing apoptosis and reducing the expression of the pro-inflammatory cytokines IL1β, IL6, IL17, and TNFα at the mRNA level. Quercetin caused inhibition of glucose, lactate, lactate dehydrogenase, pyruvate, and adenosine triphosphate and increased pyruvate dehydrogenase expression in MH7A cells. It was further confirmed that quercetin may inhibit energy metabolism and inflammatory factor secretion in MH7A cells through JAK1/STAT3/HIF-1α signaling. CONCLUSIONS Quercetin's action on multiple target molecules and pathways makes it a promising treatment for cartilage injury in rheumatoid arthritis. By reducing joint inflammation, improving joint metabolic homeostasis, and decreasing immune system activation energy, quercetin inhibits the JAK1/STAT3/HIF-1α signaling pathway to improve disease status.
Collapse
Affiliation(s)
- FengQi Zhang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Binwen Road 548, Binjiang District, Hangzhou, Zhejiang, 310053, China
| | - YiYang Zhang
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Binwen Road 548, Binjiang District, Hangzhou, Zhejiang, 310053, China
| | - JiaWang Zhou
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Binwen Road 548, Binjiang District, Hangzhou, Zhejiang, 310053, China
| | - Ying Cai
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Binwen Road 548, Binjiang District, Hangzhou, Zhejiang, 310053, China
| | - ZhiYu Li
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Jing Sun
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - ZhiJun Xie
- Key Laboratory of Chinese Medicine Rheumatology of Zhejiang Province, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Binwen Road 548, Binjiang District, Hangzhou, Zhejiang, 310053, China.
| | - GuiFeng Hao
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
4
|
Postlethwaite AE, Jiao Y, Yang C, Dong W, Aelion JA, Wang B, Postlethwaite BE, Sigal L, Kang AH, Myers LK, Wheller P, Ingels J, Gu W. Optimizing oral immune tolerance to Type II collagen in patients with rheumatoid arthritis: The importance of dose, interfering medication and genetics. Am J Med Sci 2024; 368:300-310. [PMID: 38897565 DOI: 10.1016/j.amjms.2024.06.008] [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: 01/19/2024] [Revised: 05/17/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVES Oral immune tolerance (OT) is a complex process with unknown genetic regulation. Our aim is to explore possible genetic control of OT in patients with rheumatoid arthritis (RA). METHODS RA patients with increased interferon γ production invitro when their isolated peripheral blood mononuclear cells (PBMC) were cultured with type II bovine collagen α1 chain [α1 (II)] were enrolled in this study and were randomly assigned to the "Low dose" type II collagen (CII) group (30 µg/day for 10 weeks, followed by 50 µg/day for 10 weeks, followed by 70 µg/day for 10 weeks) or "High dose" CII group (90 µg/day for 10 weeks, followed by 110 µg/day for 10 weeks, followed by 130 µg/day for 10 weeks). Heparinized blood was obtained at baseline and after each of the 10 weeks treatment for analysis of the invitro production of IFNγ by their PBMC stimulated by α1(II) . Single nucleotide polymorphism (SNP) analysis of the responders and non-responders to oral CII was conducted using GeneChip Mapping 10 K 2.0 Array. RESULTS The SNP A-15,737 was found to associate with the ability of CII to suppress IFNγ production by α1(CII)-stimulated RA PBMC. The potential for SNP A-15,737 to associate with the OT response for patients with another autoimmune disease [OT induced by oral type I bovine collagen (CI) in patients with diffuse cutaneous systemic sclersodid (dsSSc)] was also explored. CONCLUSIONS The ROT1 region plays a role in the control of IFNγ production after oral dosing of auto-antigens, thereby determining if oral tolerance to that antigen will develop.
Collapse
Affiliation(s)
- Arnold E Postlethwaite
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Department of Veterans Affairs Medical Center, Memphis, Tennessee, 38104, USA.
| | - Yan Jiao
- Departments of Orthopaedic Surgery and BME-Campbell Clinic, and Pathology, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA
| | - Chengyuan Yang
- Departments of Orthopaedic Surgery and BME-Campbell Clinic, and Pathology, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA
| | - Wei Dong
- Departments of Orthopaedic Surgery and BME-Campbell Clinic, and Pathology, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA
| | - Jacob A Aelion
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Benjamin Wang
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | - Leonard Sigal
- Gateway Immunosciences, LLC, Stockbridge MA. 01262, USA
| | - Andrew H Kang
- Department of Veterans Affairs Medical Center, Memphis, Tennessee, 38104, USA
| | - Linda K Myers
- Department of Pediatrics University of Tennessee Health Science Center, Memphis, TN, USA
| | - Patricia Wheller
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jesse Ingels
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Weikuan Gu
- Departments of Orthopaedic Surgery and BME-Campbell Clinic, and Pathology, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA; Department of Veterans Affairs Medical Center, Memphis, Tennessee, 38104, USA
| |
Collapse
|
5
|
Zhou D, Jiao W, Shi W, Wang Q, Chen M. Mendelian randomization identifies causal associations between GWAS-associated bacteria and their metabolites and rheumatoid arthritis. Front Microbiol 2024; 15:1431367. [PMID: 39286352 PMCID: PMC11404690 DOI: 10.3389/fmicb.2024.1431367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 08/15/2024] [Indexed: 09/19/2024] Open
Abstract
Background Accumulating evidence suggests that an imbalance of gut microbiota is commonly observed in patients with rheumatoid arthritis (RA). However, it remains unclear whether gut microbiota dysbiosis is a cause or consequence of RA, and the mechanisms by which gut dysbiosis contributes to RA have not been fully understood. This study aimed to investigate the causal relationship between gut microbiota and metabolites with RA. Methods A two-sample Mendelian randomization analysis was performed to estimate the causality of gut microbiota and metabolites on RA. A genome-wide association study (GWAS) of 211 gut microbiota and 217 metabolites was used as the exposure, whereas RA was treated as the outcome. Inverse variance weighted (IVW) was regarded as the primary approach for calculating causal estimates. MR Egger method, Weighted median method, Simple mode method, and weighted mode method were used for sensitive analysis. Metabolic pathway analysis was performed via the web-based Metaconflict 5.0. Additionally, an animal study was undertaken to evaluate the results inferred by Mendelian randomization. Result This study indicated that six gut microbiota taxa (RuminococcaceaeUCG013, Erysipelotrichia, Erysipelotrichaceae, Erysipelotrichales, Clostridia, and Veillonellaceae) were estimated to exert a positive impact on RA. Conversely, seven gut microbiota taxa (Oxalobacter, Cyanobacteria, RuminococcaceaeUCG002, LachnospiraceaeUCG010, Christensenellaceae, Oxalobacteraceae, Anaerostipes) were estimated to exert a negative impact on RA. Three metabolites, namely indole-3-propionate (IPA), glycine and sphingomyelin (SM 16:1), were found to be linked to lower RA risk, while five metabolites (argininosuccinate, CE 20_4, TAG 58_8, PC 40_6, and LPC 20_4) were linked to higher RA risk. Additionally, four metabolic pathways were identified by metabolic pathway analysis. The collagen-induced arthritis (CIA) rats exhibited a higher relative abundance of Class_Clostridia and a lower abundance of Genus_Lachnospiraceae (p < 0.05) than the healthy controls. Conclusion This study identified causal associations between specific gut microbiota, metabolites, and RA. These findings support the significant role of gut microbiota and metabolites in RA pathogenesis.
Collapse
Affiliation(s)
- Donghai Zhou
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Wenyue Jiao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Weiman Shi
- School of Basic Medicine, Zhejiang Chinese Medicine University, Hangzhou, Zhejiang, China
| | - Qiao Wang
- School of Basic Medicine, Zhejiang Chinese Medicine University, Hangzhou, Zhejiang, China
| | - Muzhi Chen
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Rigi G, Kardar G, Hajizade A, Zamani J, Ahmadian G. The effects of Staphylococcus aureus protein a (SpA) on the expression of inflammatory cytokines in autoimmune patients and their probable immune response modulation mechanisms. Cytokine 2024; 183:156745. [PMID: 39217914 DOI: 10.1016/j.cyto.2024.156745] [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: 04/12/2024] [Revised: 07/20/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
The recombinant Staphylococcal protein A (SpA) is widely used in biotechnology to purify polyclonal and monoclonal IgG antibodies. At very low concentrations, the highly-purified form of the protein A can down-regulate the activation of human B-lymphocytes and macrophages which are the key cells in determining autoimmune diseases. In the present study, the efficiency of three different forms of protein A, including native full-length SpA, the recombinant full-length SpA, and a recombinant truncated form of SpA on the reduction of 4 inflammatory cytokines, including IL-8, IL-1β, TNF-α, and IL-6 by peripheral blood mononuclear cell (PBMCs) were studied and compared to an anti-rheumatoid arthritis commercial drug, Enbrel. The recombinant proteins were expressed in E. coli and the native form of SpA was commercially provided. PBMCs were obtained from adult patients with active rheumatoid arthritis (RA) and healthy control donors. Then, the effect of different doses of the three pure forms of SpA in comparison with Enbrel was investigated by analyzing the expression of selected cytokines using ELISA. The results showed that the truncated form of recombinant SpA significantly reduced the expression of cytokines more effectively than the other full-length formulations as well as the commercial drug Enbrel. In silico analysis shows that in the truncated protein, as the radius of gyration increases, the structure of IgG-binding domains become more open and more exposed to IgG. To summarize, our findings indicate that the truncated form of protein A is the most efficient form of SpA as it significantly decreases the secretion of evaluated cytokines from PBMCs in vitro.
Collapse
Affiliation(s)
- Garshasb Rigi
- Department of Genetics, Faculty of Basic Science, Shahrekord University, P. O. Box 115, Shahrekord 881 863 4141, Iran; Department of Industrial Biotechnology, Research Institute of Biotechnology, Shahrekord University, Shahrekord, Iran
| | - Gholamali Kardar
- Immunology Asthma and Allergy Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Hajizade
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Javad Zamani
- Department of System Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), P. O. BOX: 14155-6343, Tehran 1497716316, Iran
| | - Gholamreza Ahmadian
- Department of System Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), P. O. BOX: 14155-6343, Tehran 1497716316, Iran.
| |
Collapse
|
7
|
Chen JC, Ou LS, Kuo ML, Tseng LY, Chang HL. Autoantigen Exposure in Murine Fetuses Elicited Nonpathogenic Autoimmunity. Arch Med Res 2024; 55:103013. [PMID: 38851050 DOI: 10.1016/j.arcmed.2024.103013] [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: 01/11/2024] [Revised: 04/24/2024] [Accepted: 05/23/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND AND AIM Autoimmunity refers to the presence of autoantibodies and autoreactive lymphocytes against the structural molecules of an individual's cells or tissues, known as self-antigens or autoantigens. It might exist in the absence of autoimmune disease. However, how autoimmunity develops remains a mystery, despite the discovery of autoantibodies in human cord blood. METHODS Murine fetuses on day 14 of gestation were subjected to intraperitoneal injection of murine thyroid peroxidase (TPO) peptides or collagen type II (CII) at graded doses via transuterine approach. Postnatally, the recipients were examined for autoantibodies by ELISA and autoreactive lymphocytes by in vitro incorporation of tritium and for the development of autoimmune thyroiditis or arthritis. RESULTS At one month of age, the recipients did not secrete significant levels of anti-TPO or CII IgG2a in sera until a dose of 0.5 µg TPO or 5.0 µg CII was injected in utero. Serum anti-TPO or CII IgG2a persisted for at least two to four months postnatally. In recipients with elevated autoantibodies, their lymphocytes also showed proliferative responses specifically to TPO or CII. However, the development of autoantibodies and autoreactive lymphocytes was not associated with inflammatory cell infiltration of thyroid glands or paw joints even though anti-TPO or CII IgG2a was enhanced by postnatal TPO or CII challenge. CONCLUSION Fetal exposure to free autoantigens could be immunogenic, shedding new light on the in utero origin of autoantibodies and autoreactive lymphocytes. The development of autoimmunity requires a threshold intensity of autoantigen exposure in the fetus.
Collapse
Affiliation(s)
- Jeng-Chang Chen
- Department of Surgery, Chang Gung Children's Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Liang-Shiou Ou
- Division of Allergy, Asthma and Rheumatology, Department of Pediatrics, Chang Gung Children's Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ming-Ling Kuo
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Li-Yun Tseng
- Pediatric Research Center, Chang Gung Children's Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsueh-Ling Chang
- Pediatric Research Center, Chang Gung Children's Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
8
|
Poutoglidou F, Pourzitaki C, Manthou ME, Samoladas E, Saitis A, Malliou F, Kouvelas D. The inhibitory effect of tocilizumab on systemic bone loss and tendon inflammation in a juvenile Collagen-Induced arthritis rat model. Connect Tissue Res 2022; 63:577-589. [PMID: 35175165 DOI: 10.1080/03008207.2022.2042275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE OF THE STUDY Reduced Bone Mineral Density (BMD) is a prevalent comorbidity in Juvenile Idiopathic Arthritis (JIA). Enthesitis and other tendon abnormalities, such as tenosynovitis, tendinitis and tendon ruptures are, also, common extra-articular manifestations of the disease. The aim of the present study was to investigate the effect of tocilizumab, an antibody that binds the Interleukin-6 (IL-6) Receptor, on inflammation-related bone loss and tendon inflammation in an animal model of JIA. MATERIALS AND METHODS The Collagen-Induced Arthritis (CIA) model was induced in male rats followed by intraperitoneal administration of tocilizumab for 8 weeks. Methotrexate, the most widely used Disease-Modifying Antirheumatic Drug in the management of JIA, was, also, administered, either as a monotherapy or as an add-on therapy to tocilizumab. BMD was evaluated with Micro-Computed Tomography (Micro-CT) and histopathological examination. Tendon damage was, also, assessed histologically. Finally, two pro-inflammatory cytokines, Tumor Necrosis Factor-alpha (TNF-a) and Interleukin-23 (IL-23) were quantified in tendon tissues by ELISA analysis. RESULTS Tocilizumab-treated animals exhibited a significantly improved trabecular microarchitecture on micro-CT analysis and histological examination. Tendon morphology was also improved. Anti-IL-6 treatment led to a significant decrease in TNF-a and IL-23 expression in tendon tissue. CONCLUSIONS The results of the present study provide evidence that tocilizumab reduces inflammation-related bone loss and suppresses tendon inflammation in a juvenile CIA rat model. These findings offer perspectives for the management of osteoporosis and enthesitis in JIA.
Collapse
Affiliation(s)
- Frideriki Poutoglidou
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, University Campus, Thessaloniki, Greece
| | - Chryssa Pourzitaki
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, University Campus, Thessaloniki, Greece
| | - Maria Eleni Manthou
- Laboratory of Histology and Embryology, School of Medicine, Aristotle University of Thessaloniki, University Campus, Thessaloniki, Greece
| | - Efthimios Samoladas
- Orthopaedics Division, "Genimatas" Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Athanasios Saitis
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, University Campus, Thessaloniki, Greece
| | - Foteini Malliou
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, University Campus, Thessaloniki, Greece
| | - Dimitrios Kouvelas
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, University Campus, Thessaloniki, Greece
| |
Collapse
|
9
|
Role of Citrullinated Collagen in Autoimmune Arthritis. Int J Mol Sci 2022; 23:ijms23179833. [PMID: 36077232 PMCID: PMC9456437 DOI: 10.3390/ijms23179833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/09/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Citrullination of proteins plays an important role in protein function and it has recently become clear that citrullinated proteins play a role in immune responses. In this study we examined how citrullinated collagen, an extracellular matrix protein, affects T-cell function during the development of autoimmune arthritis. Using an HLA-DR1 transgenic mouse model of rheumatoid arthritis, mice were treated intraperitoneally with either native type I collagen (CI), citrullinated CI (cit-CI), or phosphate buffered saline (PBS) prior to induction of autoimmune arthritis. While the mice given native CI had significantly less severe arthritis than controls administered PBS, mice receiving cit-CI had no decrease in the severity of autoimmune arthritis. Using Jurkat cells expressing the inhibitory receptor leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1), Western blot analysis indicated that while CI and cit-CI bound to LAIR-1 with similar affinity, only CI induced phosphorylation of the LAIR ITIM tyrosines; cit-CI was ineffective. These data suggest that cit-CI acts as an antagonist of LAIR-1 signaling, and that the severity of autoimmune arthritis can effectively be altered by targeting T cells with citrullinated collagen.
Collapse
|
10
|
Zhu Y, Zhang L, Zhang X, Wu D, Chen L, Hu C, Wen C, Zhou J. Tripterygium wilfordii glycosides ameliorates collagen-induced arthritis and aberrant lipid metabolism in rats. Front Pharmacol 2022; 13:938849. [PMID: 36105231 PMCID: PMC9465305 DOI: 10.3389/fphar.2022.938849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease, and the dysregulation of lipid metabolism has been found to play an important role in the pathogenesis of RA and is related to the severity and prognosis of patients. Tripterygium wilfordii glycosides (TWG) is extracted from the roots of Tripterygium wilfordii Hook F. with anti-inflammatory and immunosuppressive effects, and numerous clinical trials have supported its efficacy in the treatment of RA. Some evidence suggested that TWG can modulate the formation of lipid mediators in various innate immune cells; however whether it can improve RA-related lipid disorders has not been systematically studied. In the study, type Ⅱ collagen-induced arthritis (CIA) model was used to investigate the efficacy of TWG in the treatment of RA and its effect on lipid metabolism. Paw volume, arthritis score, pathological changes of ankle joint, serum autoantibodies and inflammatory cytokines were detected to assess the therapeutic effect on arthritis in CIA rats. Then, shotgun lipidomics based on multi-dimensional mass spectrometry platform was performed to explore the alterations in serum lipidome caused by TWG. The study showed that TWG could effectively ameliorate arthritis in CIA rats, such as reducing paw volume and arthritis score, alleviating the pathological damages of joint, and preventing the production of anti-CII autoantibodies and IL-1β cytokine. Significant increase in ceramide and decrease in lysophosphatidylcholine were observed in CIA rats, and were highly correlated with arthritis score and IL-1β level. After TWG treatment, these lipid abnormalities can be corrected to a great extent. These data demonstrate that TWG exerts a beneficial therapeutic effect on aberrant lipid metabolism which may provide new insights for further exploring the role and mechanism of TWG in the treatment of RA.
Collapse
Affiliation(s)
- Yitian Zhu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, China
| | - Luyun Zhang
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiafeng Zhang
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dehong Wu
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Leiming Chen
- Department of Nephrology, Wenzhou Hospital of Integrated Traditional Chinese and Western Medicine, Wenzhou, China
| | - Changfeng Hu
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chengping Wen
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Chengping Wen, ; Jia Zhou, , orcid.org/0000-0003-2182-8440
| | - Jia Zhou
- Institute of Basic Research in Clinical Medicine, College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Chengping Wen, ; Jia Zhou, , orcid.org/0000-0003-2182-8440
| |
Collapse
|
11
|
Chan WC, Tan L, Liu J, Yang Q, Wang J, Wang M, Yue Y, Hao L, Man Y. Inhibition of Rgs10 aggravates periodontitis with collagen-induced arthritis via the NF-κB pathway. Oral Dis 2022; 29:1802-1811. [PMID: 35122384 DOI: 10.1111/odi.14147] [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: 08/23/2021] [Revised: 01/20/2022] [Accepted: 01/31/2022] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To explore the role of the Rgs10-associated nuclear factor (NF)-κB signalling pathway in periodontitis with rheumatoid arthritis. METHODS Porphyromonas gingivalis and collagen were locally applied to mice to establish in vivo periodontitis and rheumatoid arthritis models, respectively. Both agents were administered together to establish the comorbid group. All models were treated with adeno-associated virus-green fluorescent protein (AAV-GFP) or adeno-associated virus small hairpin Rgs10 (AAV-sh-Rgs10). In vivo expression of Rgs10 and inflammatory cytokines was analysed, along with exploration of the NF-κB signalling pathway in lipopolysaccharide (LPS)-stimulated mouse-derived RAW264.7 cells, with and without treatment of small interfering RNA (siRNA; Rgs10-Mus-MSS245072). RESULTS In the comorbidity mouse group (mice with both periodontitis and rheumatoid arthritis), inhibition of Rgs10 exacerbated periodontitis, along with upregulation of phospho-RelA (pP65), tumour necrosis factor-α (TNF-α), and interleukin-6 (IL-6) expression in the NF-κB signalling pathway. Similarly, treatment of LPS-stimulated RAW264.7 cells with siRNA resulted in the inhibition of Rgs10, along with upregulation of pP65, TNF-α, and IL-6 expression in vitro. CONCLUSION Inhibition of Rgs10 in mice with periodontitis and rheumatoid arthritis can promote the progression of periodontitis, indicating the potential therapeutic role of Rgs10 in this condition.
Collapse
Affiliation(s)
- Wei-Cheng Chan
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Liangyu Tan
- Department of Prosthodontics, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, People's Republic of China
| | - Jie Liu
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Qin Yang
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Jiajia Wang
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Min Wang
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Yuan Yue
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Liang Hao
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| | - Yi Man
- The State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Sichuan, People's Republic of China
| |
Collapse
|
12
|
Folate-Targeted Liposomal Formulations Improve Effects of Methotrexate in Murine Collagen-Induced Arthritis. Biomedicines 2022; 10:biomedicines10020229. [PMID: 35203442 PMCID: PMC8869739 DOI: 10.3390/biomedicines10020229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/13/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023] Open
Abstract
Methotrexate (MTX) is first-line therapy for the treatment of rheumatoid arthritis (RA), however, its use may be limited by side effects notably post-injection malaise. When patients are intolerant or become unresponsive, second-line or antibody therapy may be indicated. A folate-targeted liposomal formulation of MTX (FL-MTX) is tropic to arthritic paws and prevents the onset of collagen-induced arthritis (CIA) in the mouse. We optimized the drug-to-lipid molar ratio to 0.15 and demonstrated the therapeutic efficacy of this form at 2 mg/kg MTX intraperitoneal (i.p.) twice a week. These improved liposomes were present in inflamed joints in proportion to the degree of swelling of the paw and bone remodeling activity. FL-MTX had lower hepatic and renal elimination of MTX than the free substance. FL-MTX provided equivalent results when given i.p. or subcutaneous (s.c.) and FL-MTX 2 mg/kg (drug/lipid 0.15), twice weekly, was similar to or more effective than 35 mg/kg MTX (same route and schedule) in reducing the incidence and swelling in the murine CIA model. These results suggest that FL-MTX is a more potent nanotherapeutic formulation than free MTX treatment. Its potential benefits for patients may include reduced frequency of treatment and lower overall doses for a given response.
Collapse
|
13
|
Whittington KB, Prislovsky A, Beaty J, Albritton L, Radic M, Rosloniec EF. CD8 + T Cells Expressing an HLA-DR1 Chimeric Antigen Receptor Target Autoimmune CD4 + T Cells in an Antigen-Specific Manner and Inhibit the Development of Autoimmune Arthritis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:16-26. [PMID: 34819392 PMCID: PMC8702470 DOI: 10.4049/jimmunol.2100643] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/22/2021] [Indexed: 01/03/2023]
Abstract
Ag-specific immunotherapy is a long-term goal for the treatment of autoimmune diseases; however developing a means of therapeutically targeting autoimmune T cells in an Ag-specific manner has been difficult. Through the engineering of an HLA-DR1 chimeric Ag receptor (CAR), we have produced CD8+ CAR T cells that target CD4+ T cells in an Ag-specific manner and tested their ability to inhibit the development of autoimmune arthritis in a mouse model. The DR1 CAR molecule was engineered to contain CD3ζ activation and CD28 signaling domains and a covalently linked autoantigenic peptide from type II collagen (CII; DR1-CII) to provide specificity for targeting the autoimmune T cells. Stimulation of the DR1-CII CAR T cells by an anti-DR Ab induced cytokine production, indicating that the DR1-CAR functions as a chimeric molecule. In vitro CTL assays using cloned CD4+ T cells as target cells demonstrated that the DR1-CII CAR T cells efficiently recognize and kill CD4+ T cells that are specific for the CII autoantigen. The CTL function was highly specific, as no killing was observed using DR1-restricted CD4+ T cells that recognize other Ags. When B6.DR1 mice, in which autoimmune arthritis had been induced, were treated with the DR1-CII CAR T cells, the CII-specific autoimmune CD4+ T cell response was significantly decreased, autoantibody production was suppressed, and the incidence and severity of the autoimmune arthritis was diminished. These data demonstrate that HLA-DR CAR T cells have the potential to provide a highly specific therapeutic approach for the treatment of autoimmune disease.
Collapse
Affiliation(s)
| | | | - Jacob Beaty
- Department of Medicine, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis TN 38163
| | - Lorraine Albritton
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis TN 38163
| | - Marko Radic
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis TN 38163
| | - Edward F. Rosloniec
- Veterans Affairs Medical Center, Memphis TN 38104,Department of Medicine, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis TN 38163,Department of Pathology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis TN 38163
| |
Collapse
|
14
|
Myers LK, Winstead M, Kee JD, Park JJ, Zhang S, Li W, Yi AK, Stuart JM, Rosloniec EF, Brand DD, Tuckey RC, Slominski AT, Postlethwaite AE, Kang AH. 1,25-Dihydroxyvitamin D3 and 20-Hydroxyvitamin D3 Upregulate LAIR-1 and Attenuate Collagen Induced Arthritis. Int J Mol Sci 2021; 22:ijms222413342. [PMID: 34948139 PMCID: PMC8709360 DOI: 10.3390/ijms222413342] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 12/30/2022] Open
Abstract
Vitamin D plays a crucial role in regulation of the immune response. However, treatment of autoimmune diseases with 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] doses sufficient to be effective is prohibitive due to its calcemic and toxic effects. We use the collagen-induced arthritis (CIA) model to analyze the efficacy of the noncalcemic analog of vitamin D, 20S-hydroxyvitamin D3 [20S(OH)D3], as well as 1,25(OH)2D3, to attenuate arthritis and explore a potential mechanism of action. Mice fed a diet deficient in vitamin D developed a more severe arthritis characterized by enhanced secretion of T cell inflammatory cytokines, compared to mice fed a normal diet. The T cell inflammatory cytokines were effectively suppressed, however, by culture of the cells with 20S(OH)D3. Interestingly, one of the consequences of culture with 1,25(OH)2D3 or 20S(OH)D3, was upregulation of the natural inhibitory receptor leukocyte associated immunoglobulin-like receptor-1 (LAIR-1 or CD305). Polyclonal antibodies which activate LAIR-1 were also capable of attenuating arthritis. Moreover, oral therapy with active forms of vitamin D suppressed arthritis in LAIR-1 sufficient DR1 mice, but were ineffective in LAIR-1−/− deficient mice. Taken together, these data show that the effect of vitamin D on inflammation is at least, in part, mediated by LAIR-1 and that non-calcemic 20S(OH)D3 may be a promising therapeutic agent for the treatment of autoimmune diseases such as Rheumatoid Arthritis.
Collapse
Affiliation(s)
- Linda K. Myers
- Department of Pediatrics, University of Tennessee Health Science Center, 50 N. Dunlap, Rm. 461R, Memphis, TN 38103, USA
- Department of Medicine, University of Tennessee Health Science Center, 956 Court Ave., Memphis, TN 38163, USA; (M.W.); (J.D.K.); (J.J.P.); (J.M.S.); (A.E.P.); (A.H.K.)
- Correspondence: ; Tel.: +1-(901)-448-5774; Fax: +1-(901)-448-7265
| | - Michael Winstead
- Department of Medicine, University of Tennessee Health Science Center, 956 Court Ave., Memphis, TN 38163, USA; (M.W.); (J.D.K.); (J.J.P.); (J.M.S.); (A.E.P.); (A.H.K.)
| | - John D. Kee
- Department of Medicine, University of Tennessee Health Science Center, 956 Court Ave., Memphis, TN 38163, USA; (M.W.); (J.D.K.); (J.J.P.); (J.M.S.); (A.E.P.); (A.H.K.)
| | - Jeoungeun J. Park
- Department of Medicine, University of Tennessee Health Science Center, 956 Court Ave., Memphis, TN 38163, USA; (M.W.); (J.D.K.); (J.J.P.); (J.M.S.); (A.E.P.); (A.H.K.)
| | - Sicheng Zhang
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave, Memphis, TN 38103, USA; (S.Z.); (W.L.)
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave, Memphis, TN 38103, USA; (S.Z.); (W.L.)
| | - Ae-Kyung Yi
- Department of Microbiology-Immunology-Biochemistry, University of Tennessee Health Science Center, 858 Madison Ave., Memphis, TN 38163, USA;
| | - John M. Stuart
- Department of Medicine, University of Tennessee Health Science Center, 956 Court Ave., Memphis, TN 38163, USA; (M.W.); (J.D.K.); (J.J.P.); (J.M.S.); (A.E.P.); (A.H.K.)
- Memphis Veterans Affairs Medical Center, 1030 Jefferson Ave., Memphis, TN 38104, USA; (E.F.R.); (D.D.B.)
| | - Edward F. Rosloniec
- Memphis Veterans Affairs Medical Center, 1030 Jefferson Ave., Memphis, TN 38104, USA; (E.F.R.); (D.D.B.)
| | - David D. Brand
- Memphis Veterans Affairs Medical Center, 1030 Jefferson Ave., Memphis, TN 38104, USA; (E.F.R.); (D.D.B.)
| | - Robert C. Tuckey
- School of Molecular Sciences, University of Western Australia, 35 Stirling Highway, Perth, WA 6009, Australia;
| | - Andrzej T. Slominski
- Department of Dermatology, University of Alabama at Birmingham 500 22nd St. S, Birmingham, AL 35294, USA;
- Comprehensive Cancer Center, University of Alabama at Birmingham 1824 6th Ave., Birmingham, AL 35294, USA
- Birmingham Veterans Affairs Medical Center, 700 19th Street S., Birmingham, AL 35233, USA
| | - Arnold E. Postlethwaite
- Department of Medicine, University of Tennessee Health Science Center, 956 Court Ave., Memphis, TN 38163, USA; (M.W.); (J.D.K.); (J.J.P.); (J.M.S.); (A.E.P.); (A.H.K.)
- Memphis Veterans Affairs Medical Center, 1030 Jefferson Ave., Memphis, TN 38104, USA; (E.F.R.); (D.D.B.)
| | - Andrew H. Kang
- Department of Medicine, University of Tennessee Health Science Center, 956 Court Ave., Memphis, TN 38163, USA; (M.W.); (J.D.K.); (J.J.P.); (J.M.S.); (A.E.P.); (A.H.K.)
- Memphis Veterans Affairs Medical Center, 1030 Jefferson Ave., Memphis, TN 38104, USA; (E.F.R.); (D.D.B.)
| |
Collapse
|
15
|
A Bilayer Osteochondral Scaffold with Self‐Assembled Monomeric Collagen Type‐I, Type‐II, and Polymerized Chondroitin Sulfate Promotes Chondrogenic and Osteogenic Differentiation of Mesenchymal Stem Cells. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
16
|
Poutoglidou F, Pourzitaki C, Manthou ME, Saitis A, Malliou F, Kouvelas D. Infliximab and Tocilizumab Reduce Anxiety-Like Behaviour and Improve Cognitive Performance in a Juvenile Collagen-Induced Arthritis Rat Model. Inflammation 2021; 45:445-459. [PMID: 34515956 DOI: 10.1007/s10753-021-01560-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/03/2021] [Indexed: 12/16/2022]
Abstract
Anxiety disorders and cognitive decline are highly prevalent in rheumatic diseases, including Juvenile Idiopathic Arthritis (JIA). In this study, we investigated the effect of long-term treatment with infliximab and tocilizumab on anxiety-like behaviour and cognitive performance in a juvenile collagen-induced arthritis (CIA) rat model. Forty-nine rats with established moderate arthritis were randomly allocated into 7 equal groups: negative control, vehicle, methotrexate, infliximab, tocilizumab, methotrexate + infliximab and methotrexate + tocilizumab groups. Behavioural tests were performed to evaluate anxiety-like behaviour and cognitive function. Neuropathological changes were investigated by histological examination at the level of the hippocampus, the amygdala and the prefrontal cortex. Also, the expression of Brain-Derived Neurotrophic Factor (BDNF), a biomarker associated with neuronal survival and plasticity, was determined in the hippocampus and the amygdala by RT-qPCR. We found that both infliximab and tocilizumab reduced anxiety-like behaviour in the elevated-plus and elevated-zero maze tests. Tocilizumab, also, improved cognitive function in the olfactory social memory and passive avoidance tests. Anti-cytokine treatment reversed the histopathological changes in the brain induced by CIA. BDNF expression was higher in all treatment groups and especially those receiving monoclonal antibodies combined with methotrexate. Our data provide evidence that chronic infliximab and tocilizumab treatment reduces anxiety-like behaviour, improves cognitive function, reverses neuropathological changes and increases central BDNF expression in a juvenile arthritis rat model. These findings may be translated to humans to address behavioural comorbidities associated with JIA.
Collapse
Affiliation(s)
- Frideriki Poutoglidou
- Department of Clinical Pharmacology, School of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece.
| | - Chryssa Pourzitaki
- Department of Clinical Pharmacology, School of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| | - Maria Eleni Manthou
- Laboratory of Histology and Embryology, Medical School, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| | - Athanasios Saitis
- Department of Clinical Pharmacology, School of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| | - Foteini Malliou
- Department of Clinical Pharmacology, School of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| | - Dimitrios Kouvelas
- Department of Clinical Pharmacology, School of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| |
Collapse
|
17
|
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: 4.0] [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
|
18
|
Pai FT, Lu CY, Lin CH, Wang J, Huang MC, Liu CT, Song YC, Ku CL, Yen HR. Psoralea corylifolia L. Ameliorates Collagen-Induced Arthritis by Reducing Proinflammatory Cytokines and Upregulating Myeloid-Derived Suppressor Cells. Life (Basel) 2021; 11:life11060587. [PMID: 34205531 PMCID: PMC8235662 DOI: 10.3390/life11060587] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/12/2021] [Accepted: 06/15/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Rheumatoid arthritis is an autoimmune disease that may lead to severe complications. The fruit of Psoralea corylifolia L. (PCL) is widely used in traditional Chinese medicine as a well-known herbal treatment for orthopedic diseases. However, there is a lack of studies of its effects on rheumatoid arthritis. The purpose of the study was to investigate the effects and mechanisms of concentrated herbal granules of PCL on rheumatoid arthritis to provide some insights for future development of new drug for the treatment of rheumatoid arthritis. Methods: We used collagen-induced arthritis (CIA) DBA/1J mice as an experimental model to mimic human rheumatoid arthritis. The mice were immunized with collagen on days 0 and 21 and then orally administered 200 mg/kg/day PCL on days 22–49. Starch was used as a control. The mice were sacrificed on day 50. Clinical phenotypes, joint histopathology, and immunological profiles were measured. Results: Compared to the CIA or CIA + Starch group, the CIA + PCL group had significantly ameliorated clinical severity and decreased paw swelling. Histopathological analysis of the hind paws showed that PCL mitigated the erosion of cartilage and the proliferation of synovial tissues. There were significant differences in the levels of TNF-α, IL-6 and IL-17A, as measured by ELISA, and the percentages of CD4 + IL-17A+, CD4 + TNF-α+, CD4 + IFN-γ+ T cells. Furthermore, we also found that in mice treated with CIA + PCL, the percentage and number of bone marrow-derived suppressor cells (MDSCs; Gr1+ CD11b+) increased significantly. Conclusions: We provided evidence for the potential antiarthritic effects of PCL through the inhibition of inflammation and increase of MDSCs. These findings indicate that PCL may be a promising therapeutic herb for the treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- Fu-Tzu Pai
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Research Center for Traditional Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan; (C.-H.L.); (C.-T.L.); (Y.-C.S.)
| | - Cheng-You Lu
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97002, Taiwan;
| | - Chia-Hsin Lin
- Research Center for Traditional Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan; (C.-H.L.); (C.-T.L.); (Y.-C.S.)
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan;
| | - John Wang
- Department of Pathology, China Medical University Hospital, Taichung 40447, Taiwan;
| | - Ming-Cheng Huang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan;
- Department of Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan
| | - Chuan-Teng Liu
- Research Center for Traditional Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan; (C.-H.L.); (C.-T.L.); (Y.-C.S.)
| | - Ying-Chyi Song
- Research Center for Traditional Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan; (C.-H.L.); (C.-T.L.); (Y.-C.S.)
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| | - Cheng-Lung Ku
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Correspondence: (C.-L.K.); (H.-R.Y.); Tel.: +886-3-211-8800 (ext. 3496) (C.-L.K.); +886-4-2205-3366 (ext. 3313) (H.-R.Y.)
| | - Hung-Rong Yen
- Research Center for Traditional Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan; (C.-H.L.); (C.-T.L.); (Y.-C.S.)
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan;
- Department of Chinese Medicine, China Medical University Hospital, Taichung 40447, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan
- Department of Biotechnology, Asia University, Taichung 41354, Taiwan
- Correspondence: (C.-L.K.); (H.-R.Y.); Tel.: +886-3-211-8800 (ext. 3496) (C.-L.K.); +886-4-2205-3366 (ext. 3313) (H.-R.Y.)
| |
Collapse
|
19
|
Poutoglidou F, Pourzitaki C, Manthou ME, Samoladas E, Saitis A, Malliou F, Kouvelas D. Infliximab prevents systemic bone loss and suppresses tendon inflammation in a collagen-induced arthritis rat model. Inflammopharmacology 2021; 29:661-672. [PMID: 33982199 DOI: 10.1007/s10787-021-00815-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/24/2021] [Indexed: 11/24/2022]
Abstract
Reduced Bone Mineral Density (BMD) and tendon abnormalities, such as tenosynovitis and enthesitis, are prevalent comorbidities in patients with rheumatoid arthritis (RA). The aim of the present study was to investigate the effect of chronic treatment with infliximab on BMD and tendon inflammation in an animal model of inflammatory arthritis. Collagen-Induced Arthritis (CIA) was induced in rats, followed by long-term intraperitoneal administration of infliximab. Two additional groups of animals received methotrexate either as a monotherapy or as a co-treatment to infliximab. BMD was evaluated by Micro-Computed Tomography (Micro-CT) and bone histological examination. Tendon inflammation was assessed histologically and by quantitative ELISA analysis of pro-inflammatory cytokines in tendon tissues. Both methotrexate and infliximab treatment alleviated joint inflammation and reduced paw edema. Infliximab-treated animals exhibited an improved trabecular microarchitecture on micro-CT and histological analysis compared to both non-treated and methotrexate-treated animals. Infliximab almost reversed the pathological changes in tendons induced by CIA. Finally, we observed statistically significant declines in tendon TNF-a and IL-23 levels after infliximab treatment. Our study provides evidence that infliximab prevents arthritis-related osteoporosis and suppresses tendon inflammation in an animal model of inflammatory arthritis, in addition to controlling disease activity. These findings offer perspectives for the management of osteoporosis and enthesitis in RA.
Collapse
Affiliation(s)
- Frideriki Poutoglidou
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece.
| | - Chryssa Pourzitaki
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| | - Maria Eleni Manthou
- Laboratory of Histology and Embryology, School of Medicine, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| | - Efthimios Samoladas
- Orthopeadics Division of Gennimatas Hospital, School of Medicine, Aristotle University of Thessaloniki, 54124, Thessaloniki, Greece
| | - Athanasios Saitis
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| | - Foteini Malliou
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| | - Dimitrios Kouvelas
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, University Campus, 54124, Thessaloniki, Greece
| |
Collapse
|
20
|
Si Miao San Attenuates Inflammation and Oxidative Stress in Rats with CIA via the Modulation of the Nrf2/ARE/PTEN Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:2843623. [PMID: 33628297 PMCID: PMC7892228 DOI: 10.1155/2021/2843623] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/08/2020] [Accepted: 02/02/2021] [Indexed: 12/24/2022]
Abstract
Objective Si Miao San (SMS) is a traditional Chinese formula used in China to treat rheumatic diseases. To date, its mechanism in rheumatoid arthritis (RA) treatment is uncertain. Our study aims to assess the antiarthritic effects of SMS in experimental arthritic rats. Materials and Methods SMS (8.63, 4.31, and 2.16 g/kg/day) was orally administered after the first immunization from day 14 to day 53. The effects of SMS on rats with collagen-induced arthritis (CIA) were evaluated by arthritis score and histological assessment. The levels of cytokines and anti-CII antibodies in rat serum were measured by ELISAs. The expression of oxidative stress parameters was detected by biochemical assay kits. The levels of Nrf2, HO-1, NQO1, and PTEN were determined by western blotting. Results Medium- and high-dose SMS treatment significantly decreased arthritis scores and alleviated ankle joint histopathology in the rats with CIA. It inhibited the production of IL-6, TNF-α, COX-2, and PGE2 in rat serum. SMS also suppressed the expression of anti-CII antibodies IgG1 and IgG2a. Moreover, SMS significantly suppressed the levels of MDA and MPO in the synovial tissues while increasing the levels of SOD and CAT in the rats with CIA. The levels of Nrf2, HO-1, NQO1, and PTEN were upregulated by SMS in rat synovial tissues. Conclusions This study demonstrated that SMS effectively alleviated the disease progression of CIA by decreasing the levels of proinflammatory cytokines and reducing oxidative stress damage, as indicated by IL-6, TNF-α, COX-2, and PGE2 levels; inhibiting the overproduction of MDA and MPO; and enhancing antioxidant enzymes by upregulating the Nrf2/ARE/PTEN signalling pathway.
Collapse
|
21
|
Becart S, Whittington KB, Prislovsky A, Rao NL, Rosloniec EF. The role of posttranslational modifications in generating neo-epitopes that bind to rheumatoid arthritis-associated HLA-DR alleles and promote autoimmune T cell responses. PLoS One 2021; 16:e0245541. [PMID: 33465118 PMCID: PMC7815092 DOI: 10.1371/journal.pone.0245541] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022] Open
Abstract
While antibodies to citrullinated proteins have become a diagnostic hallmark in rheumatoid arthritis (RA), we still do not understand how the autoimmune T cell response is influenced by these citrullinated proteins. To investigate the role of citrullinated antigens in HLA-DR1- and DR4-restricted T cell responses, we utilized mouse models that express these MHC-II alleles to determine the relationship between citrullinated peptide affinity for these DR molecules and the ability of these peptides to induce a T cell response. Using a set of peptides from proteins thought to be targeted by the autoimmune T cell responses in RA, aggrecan, vimentin, fibrinogen, and type II collagen, we found that while citrullination can enhance the binding affinity for these DR alleles, it does not always do so, even when in the critical P4 position. Moreover, if peptide citrullination does enhance HLA-DR binding affinity, it does not necessarily predict the generation of a T cell response. Conversely, citrullinated peptides can stimulate T cells without changing the peptide binding affinity for HLA-DR1 or DR4. Furthermore, citrullination of an autoantigen, type II collagen, which enhances binding affinity to HLA-DR1 did not enhance the severity of autoimmune arthritis in HLA-DR1 transgenic mice. Additional analysis of clonal T cell populations stimulated by these peptides indicated cross recognition of citrullinated and wild type peptides can occur in some instances, while in others cases the citrullination generates a novel T cell epitope. Finally, cytokine profiles of the wild type and citrullinated peptide stimulated T cells unveiled a significant disconnect between proliferation and cytokine production. Altogether, these data demonstrate the lack of support for a simplified model with universal correlation between affinity for HLA-DR alleles, immunogenicity and arthritogenicity of citrullinated peptides. Additionally they highlight the complexity of both T cell receptor recognition of citrulline as well as its potential conformational effects on the peptide:HLA-DR complex as recognized by a self-reactive cell receptor.
Collapse
Affiliation(s)
- Stephane Becart
- Immunology Discovery, Janssen Research and Development LLC, San Diego, California, United States of America
| | | | - Amanda Prislovsky
- Veterans Affairs Medical Center, Memphis, Tennessee, United States of America
| | - Navin L. Rao
- Immunology Discovery, Janssen Research and Development LLC, Spring House, Pennsylvania, United States of America
| | - Edward F. Rosloniec
- Veterans Affairs Medical Center, Memphis, Tennessee, United States of America
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Department of Pathology, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
22
|
Han Y, Wang J, Jin M, Jia L, Yan C, Wang Y. Shentong Zhuyu Decoction Inhibits Inflammatory Response, Migration, and Invasion and Promotes Apoptosis of Rheumatoid Arthritis Fibroblast-like Synoviocytes via the MAPK p38/PPAR γ/CTGF Pathway. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6187695. [PMID: 33511203 PMCID: PMC7826240 DOI: 10.1155/2021/6187695] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/23/2020] [Accepted: 12/29/2020] [Indexed: 11/18/2022]
Abstract
INTRODUCTION The current study is aimed at exploring the effect of Shentong Zhuyu Decoction on the proliferation, migration, invasion, and apoptosis of rheumatoid arthritis fibroblast-like synoviocytes (RA-FLS) and its underlying molecular mechanism. MATERIALS AND METHODS The type II collagen-induced arthritis (CIA) model was established. Subsequently, the RA-FLS were isolated from the CIA rat model and identified by immunohistochemistry. The viability, apoptosis, cell cycle, migration, and invasion of RA-FLS were detected by the cell counting kit 8 (CCK-8) assay, flow cytometry, wound-healing assay, and transwell invasion assay, respectively. The levels of MAPK p38, PPARγ, CTGF, Bcl-2, Bax, caspase-3, IL-1β, MMP-3, CDK4, and cyclin D1 were determined by qRT-PCR and western blotting, respectively. RESULTS After treatment with Shentong Zhuyu Decoction medicated serum, the OD570 value, migrative and invasive abilities, and the secretion of IL-1β, MMP-3 were remarkably decreased in RA-FLS, while the apoptosis rate was increased. Further, results showed that Shentong Zhuyu Decoction inhibited the transition from the G1 phase to S phase. Additionally, Shentong Zhuyu Decoction significantly inhibited the expression of Bcl-2, CDK4, cyclin D1, MAPK p-p38, and CTGF, whereas elevated the levels of Bax, caspase-3, and PPARγ. Importantly, the effects of Shentong Zhuyu Decoction were consistent with the trends of MAPK P38 inhibitor (SB203580) and PPARγ agonist (GW1929). CONCLUSIONS Shentong Zhuyu Decoction inhibited viability, inflammatory response, migration, invasion, and transition from the G1 phase to S phase and promoted apoptosis of RA-FLS via the MAPK p38/PPARγ/CTGF pathway.
Collapse
Affiliation(s)
- Ying Han
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
- Department of Chinese Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Wang
- Department of Chinese Medicine Diagnostics, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Meng Jin
- Department of Chinese Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lin Jia
- Department II of Respiratory, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, China
| | - Cuihuan Yan
- Institute of Integrated Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yali Wang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
23
|
Park JE, Brand DD, Rosloniec EF, Yi AK, Stuart JM, Kang AH, Myers LK. Leukocyte-associated immunoglobulin-like receptor 1 inhibits T-cell signaling by decreasing protein phosphorylation in the T-cell signaling pathway. J Biol Chem 2020; 295:2239-2247. [PMID: 31932281 DOI: 10.1074/jbc.ra119.011150] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/09/2020] [Indexed: 12/23/2022] Open
Abstract
Multiple observations implicate T-cell dysregulation as a central event in the pathogenesis of rheumatoid arthritis. Here, we investigated mechanisms for suppressing T-cell activation via the inhibitory receptor leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1). To determine how LAIR-1 affects T-cell receptor (TCR) signaling, we compared 1) T cells from LAIR-1-sufficient and -deficient mice, 2) Jurkat cells expressing either LAIR-1 mutants or C-terminal Src kinase (CSK) mutants, and 3) T cells from mice that contain a CSK transgene susceptible to chemical inhibition. Our results indicated that LAIR-1 engagement by collagen or by complement C1q (C1Q, which contains a collagen-like domain) inhibits TCR signaling by decreasing the phosphorylation of key components in the canonical T-cell signaling pathway, including LCK proto-oncogene SRC family tyrosine kinase (LCK), LYN proto-oncogene SRC family tyrosine kinase (LYN), ζ chain of T-cell receptor-associated protein kinase 70 (ZAP-70), and three mitogen-activated protein kinases (extracellular signal-regulated kinase, c-Jun N-terminal kinase 1/2, and p38). The intracellular region of LAIR-1 contains two immunoreceptor tyrosine-based inhibition motifs that are both phosphorylated by LAIR-1 activation, and immunoprecipitation experiments revealed that Tyr-251 in LAIR-1 binds CSK. Using CRISPR/Cas9-mediated genome editing, we demonstrate that CSK is essential for the LAIR-1-induced inhibition of the human TCR signal transduction. T cells from mice that expressed a PP1 analog-sensitive form of CSK (CskAS) corroborated these findings, and we also found that Tyr-251 is critical for LAIR-1's inhibitory function. We propose that LAIR-1 activation may be a strategy for controlling inflammation and may offer a potential therapeutic approach for managing autoimmune diseases.
Collapse
Affiliation(s)
- Jeoung-Eun Park
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - David D Brand
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163; Department of MicrobiologyγÇô-ImmunologyγÇô-Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee 38163; Research Service, Veterans Affairs Medical Center, Memphis, Tennessee 38104
| | - Edward F Rosloniec
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163; Research Service, Veterans Affairs Medical Center, Memphis, Tennessee 38104
| | - Ae-Kyung Yi
- Department of MicrobiologyγÇô-ImmunologyγÇô-Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - John M Stuart
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163; Research Service, Veterans Affairs Medical Center, Memphis, Tennessee 38104
| | - Andrew H Kang
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163; Research Service, Veterans Affairs Medical Center, Memphis, Tennessee 38104
| | - Linda K Myers
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163.
| |
Collapse
|
24
|
Yoon TW, Kim YI, Cho H, Brand DD, Rosloniec EF, Myers LK, Postlethwaite AE, Hasty KA, Stuart JM, Yi AK. Ameliorating effects of Gö6976, a pharmacological agent that inhibits protein kinase D, on collagen-induced arthritis. PLoS One 2019; 14:e0226145. [PMID: 31809526 PMCID: PMC6897462 DOI: 10.1371/journal.pone.0226145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/20/2019] [Indexed: 12/16/2022] Open
Abstract
Toll-like receptor (TLR) signaling can contribute to the pathogenesis of arthritis. Disruption of TLR signaling at early stages of arthritis might thereby provide an opportunity to halt the disease progression and ameliorate outcomes. We previously found that Gö6976 inhibits TLR-mediated cytokine production in human and mouse macrophages by inhibiting TLR-dependent activation of protein kinase D1 (PKD1), and that PKD1 is essential for proinflammatory responses mediated by MyD88-dependent TLRs. In this study, we investigated whether PKD1 contributes to TLR-mediated proinflammatory responses in human synovial cells, and whether Gö6976 treatment can suppress the development and progression of type II collagen (CII)-induced arthritis (CIA) in mouse. We found that TLR/IL-1R ligands induced activation of PKD1 in human fibroblast-like synoviocytes (HFLS). TLR/IL-1R-induced expression of cytokines/chemokines was substantially inhibited in Gö6976-treated HFLS and PKD1-knockdown HFLS. In addition, serum levels of anti-CII IgG antibodies, and the incidence and severity of arthritis after CII immunization were significantly reduced in mice treated daily with Gö6976. Synergistic effects of T-cell receptor and TLR, as well as TLR alone, on spleen cell proliferation and cytokine production were significantly inhibited in the presence of Gö6976. Our results suggest a possibility that ameliorating effects of Gö6976 on CIA may be due to its ability to inhibit TLR/IL-1R-activated PKD1, which might play an important role in proinflammatory responses in arthritis, and that PKD1 could be a therapeutic target for inflammatory arthritis.
Collapse
Affiliation(s)
- Tae Won Yoon
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Young-In Kim
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Hongsik Cho
- Department of Orthopedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - David D. Brand
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Edward F. Rosloniec
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Linda K. Myers
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Arnold E. Postlethwaite
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Karen A. Hasty
- Department of Orthopedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - John M. Stuart
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Ae-Kyung Yi
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
25
|
|
26
|
Chlorogenic Acid Inhibits BAFF Expression in Collagen-Induced Arthritis and Human Synoviocyte MH7A Cells by Modulating the Activation of the NF- κB Signaling Pathway. J Immunol Res 2019; 2019:8042097. [PMID: 31240234 PMCID: PMC6556285 DOI: 10.1155/2019/8042097] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/01/2019] [Accepted: 04/10/2019] [Indexed: 11/22/2022] Open
Abstract
B cell activating factor (BAFF), a member of the tumor necrosis factor (TNF) family, plays a critical role in the pathogenesis and progression of rheumatoid arthritis (RA). Chlorogenic acid (CGA) is a phenolic compound and exerts antiarthritic activities in arthritis. However, it is not clear whether the anti-inflammatory property of CGA is associated with the regulation of BAFF expression. In this study, we found that treatment of the collagen-induced arthritis (CIA) mice with CGA significantly attenuated arthritis progression and markedly inhibited BAFF production in serum as well as the production of serum TNF-α. Furthermore, CGA inhibits TNF-α-induced BAFF expression in a dose-dependent manner and apoptosis in MH7A cells. Mechanistically, we found the DNA-binding site for the transcription factor NF-κB in the BAFF promoter region is required for this regulation. Moreover, CGA reduces the DNA-binding activity of NF-κB to the BAFF promoter region and suppresses BAFF expression through the NF-κB pathway in TNF-α-stimulated MH7A cells. These results suggest that CGA may serve as a novel therapeutic agent for the treatment of RA by targeting BAFF.
Collapse
|
27
|
Langan D, Kim EY, Moudgil KD. Modulation of autoimmune arthritis by environmental 'hygiene' and commensal microbiota. Cell Immunol 2019; 339:59-67. [PMID: 30638679 PMCID: PMC8056395 DOI: 10.1016/j.cellimm.2018.12.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/09/2018] [Accepted: 12/09/2018] [Indexed: 12/20/2022]
Abstract
Observations in patients with autoimmune diseases and studies in animal models of autoimmunity have revealed that external environmental factors including exposure to microbes and the state of the host gut microbiota can influence susceptibility to autoimmunity and subsequent disease development. Mechanisms underlying these outcomes continue to be elucidated. These include deviation of the cytokine response and imbalance between pathogenic versus regulatory T cell subsets. Furthermore, specific commensal organisms are associated with enhanced severity of arthritis in susceptible individuals, while exposure to certain microbes or helminths can afford protection against this disease. In addition, the role of metabolites (e.g., short-chain fatty acids, tryptophan catabolites), produced either by the microbes themselves or from their action on dietary products, in modulation of arthritis is increasingly being realized. In this context, re-setting of the microbial dysbiosis in RA using prebiotics, probiotics, or fecal microbial transplant is emerging as a promising approach for the prevention and treatment of arthritis. It is hoped that advances in defining the interplay between gut microbiota, dietary products, and bioactive metabolites would help in the development of therapeutic regimen customized for the needs of individual patients in the near future.
Collapse
Affiliation(s)
- David Langan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Baltimore VA Medical Center, Baltimore, MD 21201, United States
| | - Eugene Y Kim
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Department of Biomedical Sciences, Washington State University, Spokane, WA 99224, United States
| | - Kamal D Moudgil
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Department of Medicine, Division of Rheumatology, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Baltimore VA Medical Center, Baltimore, MD 21201, United States.
| |
Collapse
|
28
|
Pan YJ, Wang WH, Huang TY, Weng WH, Fang CK, Chen YC, Hwang JJ. Quetiapine ameliorates collagen-induced arthritis in mice via the suppression of the AKT and ERK signaling pathways. Inflamm Res 2018; 67:847-861. [PMID: 30109356 DOI: 10.1007/s00011-018-1176-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 06/12/2018] [Accepted: 08/01/2018] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE AND DESIGN To investigate the amelioration effects of quetiapine on rheumatoid arthritis with RAW 264.7 macrophage and collagen-induced arthritis (CIA) DBA/1J mouse model. SUBJECTS RAW 264.7 macrophage and DBA/1J mice. TREATMENT Lipopolysaccharide and collagen. METHODS RAW 264.7 macrophages stimulated by lipopolysaccharide (LPS) followed by quetiapine treatments were investigated. Activations of CD80 and CD86 were analyzed by flow cytometry. Pro-inflammatory cytokines such as IL-6, TNF-α and IL-1β were analyzed by ELISA. Proteins involved in signaling pathways related to the formation of rheumatoid arthritis were assayed by Western blotting. Therapeutic efficacy of quetiapine in CIA mouse model was also assayed. 18F-FDG/micro-PET was used to monitor the inflammation status in the joints, and the severity of bone erosion was evaluated with micro-CT and H&E staining. RESULTS The inhibition of pro-inflammatory cytokines by quetiapine was found through the ERK and AKT phosphorylation and subsequent NF-κB and CREB signaling pathways. Pro-inflammatory cytokines such as IL-17, IL-6 and IL-1β were decreased, while immunosuppressive factors such as TGF-β and IL-10 were increased in CIA mice treated with quetiapine. Notably, no uptake of 18F-FDG and bone erosion was found with micro-PET images on days 32 and 43 in the quetiapine-treated and normal control groups. However, significant uptake of 18F-FDG could be observed in the CIA group during the same time course. Similar results were further verified with ex vivo autoradiography. CONCLUSION Taken together, these results suggest that quetiapine is a potential anti-inflammatory drug, and may be used as an adjuvant for the treatment of rheumatoid arthritis.
Collapse
Affiliation(s)
- Yi-Ju Pan
- Department of Psychiatry, Far Eastern Memorial Hospital, Banciao, New Taipei City, 220, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wei-Hsun Wang
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua, Taiwan.,Department of Medical Imaging and Radiology, Shu-Zen Junior College of Medicine and Management, Kao-hsiung, Taiwan
| | - Tzu-Yao Huang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Wei-Hsiang Weng
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Chun-Kai Fang
- Department of Psychiatry and Suicide Prevention Center, Mackay Memorial Hospital, No. 155, Sec.2, Li-Nong Street, Bei-tou, Taipei, 112, Taiwan
| | - Yu-Chan Chen
- Department of Radiation Oncology, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | - Jeng-Jong Hwang
- Department of Biomedical Imaging and Radiological Sciences, National Yang-Ming University, Taipei, Taiwan. .,Biophotonics and Molecular Imaging Research Center, National Yang-Ming University, Taipei, Taiwan.
| |
Collapse
|
29
|
Alabarse PV, Lora PS, Silva JM, Santo RC, Freitas EC, de Oliveira MS, Almeida AS, Immig M, Teixeira VO, Filippin LI, Xavier RM. Collagen-induced arthritis as an animal model of rheumatoid cachexia. J Cachexia Sarcopenia Muscle 2018; 9:603-612. [PMID: 29575818 PMCID: PMC5989855 DOI: 10.1002/jcsm.12280] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/21/2017] [Accepted: 12/07/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Rheumatoid arthritis is characterized by chronic polyarticular synovitis and presents systemic changes that impact quality of life, such as impaired muscle function, seen in up to 66% of the patients. This can progress to severely debilitating state known as rheumatoid cachexia-without loss of fat mass and body weight-for which there is little consensus in terms of diagnosis or treatment. This study aims to evaluate whether the collagen-induced arthritis (CIA) animal model also develops clinical and functional features characteristic of rheumatoid cachexia. METHODS Male DBA1/J mice were randomly divided into 2 groups: healthy animals (CO, n = 11) and CIA animals (n = 13). The clinical score and edema size, animal weight and food intake, free exploratory locomotion, grip strength, and endurance exercise performance were tested 0, 18, 35, 45, 55, and 65 days after disease induction. After euthanasia, several organs, visceral and brown fat, and muscles were dissected and weighed. Muscles were used to assess myofiber diameter. Ankle joint was used to assess arthritis severity by histological score. Statistical analysis were performed using one-way and two-way analyses of variance followed by Tukey's and Bonferroni's test or t-test of Pearson and statistical difference were assumed for a P value under 0.05. RESULTS The CIA had significantly higher arthritis scores and larger hind paw edema volumes than CO. The CIA had decreased endurance exercise performance total time (fatigue; 23, 22, 24, and 21% at 35, 45, 55, and 65 days, respectively), grip strength (27, 55, 63, 60, and 66% at 25, 35, 45, 55, and 65 days, respectively), free locomotion (43, 57, 59, and 66% at 35, 45, 55, and 65 days, respectively), and tibialis anterior and gastrocnemius muscle weight (25 and 24%, respectively) compared with CO. Sarcoplasmic ratios were also reduced in CIA (TA: 23 and GA: 22% less sarcoplasmic ratio), confirming the atrophy of skeletal muscle mass in these animals than in CO. Myofiber diameter was also reduced 45% in TA and 41% in GA in CIA when compared with the CO. Visceral and brown fat were lighter in CIA (54 and 39%, respectively) than CO group. CONCLUSIONS The CIA model is a valid experimental model for rheumatoid cachexia given that the clinical changes observed were similar to those described in patients with rheumatoid arthritis.
Collapse
Affiliation(s)
- Paulo V.G. Alabarse
- Laboratório de Doenças AutoimunesHospital de Clínicas de Porto AlegrePorto AlegreBrazil
- Faculdade de MedicinaUniversidade Federal do Rio Grande do Sul, R. Ramiro Barcelos, 2350Porto Alegre90035‐003Brazil
| | - Priscila S. Lora
- Laboratório de Doenças AutoimunesHospital de Clínicas de Porto AlegrePorto AlegreBrazil
- Universidade do Vale do Rio dos SinosSão LeopoldoBrazil
| | - Jordana M.S. Silva
- Laboratório de Doenças AutoimunesHospital de Clínicas de Porto AlegrePorto AlegreBrazil
- Faculdade de MedicinaUniversidade Federal do Rio Grande do Sul, R. Ramiro Barcelos, 2350Porto Alegre90035‐003Brazil
| | - Rafaela C.E. Santo
- Laboratório de Doenças AutoimunesHospital de Clínicas de Porto AlegrePorto AlegreBrazil
- Faculdade de MedicinaUniversidade Federal do Rio Grande do Sul, R. Ramiro Barcelos, 2350Porto Alegre90035‐003Brazil
| | - Eduarda C. Freitas
- Laboratório de Doenças AutoimunesHospital de Clínicas de Porto AlegrePorto AlegreBrazil
- Faculdade de MedicinaUniversidade Federal do Rio Grande do Sul, R. Ramiro Barcelos, 2350Porto Alegre90035‐003Brazil
| | - Mayara S. de Oliveira
- Laboratório de Doenças AutoimunesHospital de Clínicas de Porto AlegrePorto AlegreBrazil
- Faculdade de MedicinaUniversidade Federal do Rio Grande do Sul, R. Ramiro Barcelos, 2350Porto Alegre90035‐003Brazil
| | - Andrelise S. Almeida
- Laboratório de Doenças AutoimunesHospital de Clínicas de Porto AlegrePorto AlegreBrazil
- Faculdade de BiomedicinaUniversidade do Vale do Rio dos SinosSão LeopoldoBrazil
| | - Mônica Immig
- Laboratório de Doenças AutoimunesHospital de Clínicas de Porto AlegrePorto AlegreBrazil
- Faculdade de BiomedicinaUniversidade do Vale do Rio dos SinosSão LeopoldoBrazil
| | - Vivian O.N. Teixeira
- Laboratório de Doenças AutoimunesHospital de Clínicas de Porto AlegrePorto AlegreBrazil
- Faculdade de MedicinaUniversidade Federal do Rio Grande do Sul, R. Ramiro Barcelos, 2350Porto Alegre90035‐003Brazil
| | - Lidiane I. Filippin
- Laboratório de Doenças AutoimunesHospital de Clínicas de Porto AlegrePorto AlegreBrazil
- Universidade La SalleCanoasBrazil
| | - Ricardo M. Xavier
- Laboratório de Doenças AutoimunesHospital de Clínicas de Porto AlegrePorto AlegreBrazil
- Faculdade de MedicinaUniversidade Federal do Rio Grande do Sul, R. Ramiro Barcelos, 2350Porto Alegre90035‐003Brazil
| |
Collapse
|
30
|
Mesenchymal Stem Cells Promote the Osteogenesis in Collagen-Induced Arthritic Mice through the Inhibition of TNF- α. Stem Cells Int 2018; 2018:4069032. [PMID: 29853911 PMCID: PMC5964571 DOI: 10.1155/2018/4069032] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 04/17/2018] [Indexed: 01/01/2023] Open
Abstract
Objective To investigate the effects of umbilical cord mesenchymal stem cell (UC-MSC) transplantation on joint damage and osteoporosis in collagen-induced arthritis (CIA) mice and to explore the mechanisms by which UC-MSCs modulate the osteogenic differentiation. Methods CIA mice were divided into the following treated groups: UC-MSC transplantation group, antitumor necrosis factor- (TNF-) α group, and zoledronic acid (ZA) group. Microcomputed tomography (micro-CT) was used to analyze the bone morphology parameters. Osteogenic differentiation of treated CIA mice was determined. Bone marrow mesenchymal stem cells (BM-MSCs) from CIA mice were treated with TNF-α in vitro to explore their effects on osteogenesis. Results The arthritis score was significantly reduced in the UC-MSC transplantation and anti-TNF-α-treated CIA groups, compared with control mice (P < 0.001). Micro-CT showed that CIA mice developed osteoporosis at 12 weeks after immunization. The bone morphology parameters were partially improved in UC-MSC-treated CIA mice. Impaired osteogenic differentiation functions were indicated by decreased ALP activity (P < 0.001) and reduced mRNA and protein levels of osteogenic marker genes (P < 0.05) in CIA mice compared with DBA/1 mice. UC-MSC treatment significantly upregulated the impaired osteogenic differentiation ability in CIA mice. Meanwhile, the serum TNF-α level was decreased significantly in the UC-MSC group. The osteogenesis was reduced with the addition of TNF-α in vitro. Conclusion This study demonstrated that UC-MSC transplantation not only significantly improved the joint damage but also played a beneficial role in osteoporosis in CIA mice. Mechanistically, the improved osteogenic differentiation of CIA under UC-MSC treatment may be achieved by inhibition of TNF-α.
Collapse
|
31
|
Fu J, Nogueira SV, Drongelen VV, Coit P, Ling S, Rosloniec EF, Sawalha AH, Holoshitz J. Shared epitope-aryl hydrocarbon receptor crosstalk underlies the mechanism of gene-environment interaction in autoimmune arthritis. Proc Natl Acad Sci U S A 2018; 115:4755-4760. [PMID: 29666259 PMCID: PMC5939100 DOI: 10.1073/pnas.1722124115] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The susceptibility to autoimmune diseases is affected by genetic and environmental factors. In rheumatoid arthritis (RA), the shared epitope (SE), a five-amino acid sequence motif encoded by RA-associated HLA-DRB1 alleles, is the single most significant genetic risk factor. The risk conferred by the SE is increased in a multiplicative way by exposure to various environmental pollutants, such as cigarette smoke. The mechanism of this synergistic interaction is unknown. It is worth noting that the SE has recently been found to act as a signal transduction ligand that facilitates differentiation of Th17 cells and osteoclasts in vitro and in vivo. Intriguingly, the aryl hydrocarbon receptor (AhR), a transcription factor that mediates the xenobiotic effects of many pollutants, including tobacco combustion products, has been found to activate similar biologic effects. Prompted by these similarities, we sought to determine whether the SE and AhR signaling pathways interact in autoimmune arthritis. Here we uncovered a nuclear factor kappa B-mediated synergistic interaction between the SE and AhR pathways that leads to markedly enhanced osteoclast differentiation and Th17 polarization in vitro. Administration of AhR pathway agonists to transgenic mice carrying human SE-coding alleles resulted in a robust increase in arthritis severity, bone destruction, overabundance of osteoclasts, and IL17-expressing cells in the inflamed joints and draining lymph nodes of arthritic mice. Thus, this study identifies a previously unrecognized mechanism of gene-environment interaction that could provide insights into the well-described but poorly understood amplification of the genetic risk for RA upon exposure to environmental pollutants.
Collapse
Affiliation(s)
- Jiaqi Fu
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Sarah V Nogueira
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | | | - Patrick Coit
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Song Ling
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Edward F Rosloniec
- Veterans Affairs Medical Center and Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Amr H Sawalha
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Joseph Holoshitz
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109;
| |
Collapse
|
32
|
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: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 12/28/2017] [Indexed: 11/19/2022]
|
33
|
Park JE, Majumdar S, Brand DD, Rosloniec EF, Yi AK, Stuart JM, Kang AH, Myers LK. The role of Syk in peripheral T cells. Clin Immunol 2018; 192:50-57. [PMID: 29673901 DOI: 10.1016/j.clim.2018.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 04/12/2018] [Accepted: 04/12/2018] [Indexed: 12/15/2022]
Abstract
The aim of this study was to understand how Syk affects peripheral T cell function. T cells from Syk-/- chimeric mice and DR1 Sykfl/fl CD4cre conditional mice gave strong CD3-induced Th1, Th2, and Th17 cytokine responses. However, an altered peptide ligand (APL) of human CII (256-276) with two substitutions (F263N, E266D), also called A12, elicited only Th2 cytokine responses from Sykfl/fl T cells but not Sykfl/fl-CD4cre T cells. Western blots revealed a marked increase in the phosphorylation of Syk, JNK and p38 upon A12/DR1 activation in WT or Sykfl/fl T cells but not in Sykfl/flCD4-cre cells. We demonstrate that Syk is required for the APL- induction of suppressive cytokines. Chemical Syk inhibitors blocked activation of GATA-3 by peptide A12/DR1. In conclusion, this study provides novel insights into the role that Syk plays in directing T cell activity, and may shape therapeutic approaches for autoimmune diseases.
Collapse
Affiliation(s)
- Jeoung-Eun Park
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Sirshendu Majumdar
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - David D Brand
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, United States; Microbiology-Immunology-Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, United States; Research Service, Veterans Affairs Medical Center, Memphis, TN 38104, United States
| | - Edward F Rosloniec
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, United States; Research Service, Veterans Affairs Medical Center, Memphis, TN 38104, United States
| | - Ae-Kyung Yi
- Microbiology-Immunology-Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - John M Stuart
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, United States; Research Service, Veterans Affairs Medical Center, Memphis, TN 38104, United States
| | - Andrew H Kang
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, United States; Research Service, Veterans Affairs Medical Center, Memphis, TN 38104, United States
| | - Linda K Myers
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163, United States.
| |
Collapse
|
34
|
Kaempferol targeting on the fibroblast growth factor receptor 3-ribosomal S6 kinase 2 signaling axis prevents the development of rheumatoid arthritis. Cell Death Dis 2018. [PMID: 29540697 PMCID: PMC5851988 DOI: 10.1038/s41419-018-0433-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Rheumatoid arthritis (RA) is a systemic inflammatory disease that mainly affects the synovial joints. Although involvement of the fibroblast growth factor (FGF) signaling pathway has been suggested as an important modulator in RA development, no clear evidence has been provided. In this study, we found that synovial fluid basic FGF (bFGF) concentration was significantly higher in RA than in osteoarthritis (OA) patients. bFGF stimulates proliferation and migration of human fibroblast-like synoviocytes (FLSs) by activation of the bFGF-FGF receptor 3 (FGFR3)-ribosomal S6 kinase 2 (RSK2) signaling axis. Moreover, a molecular docking study revealed that kaempferol inhibited FGFR3 activity by binding to the active pocket of the FGFR3 kinase domain. Kaempferol forms hydrogen bonds with the FGFR3 backbone oxygen of Glu555 and Ala558 and the side chain of Lys508. Notably, the inhibition of bFGF-FGFR3-RSK2 signaling by kaempferol suppresses the proliferation and migration of RA FLSs and the release of activated T-cell-mediated inflammatory cytokines, such as IL-17, IL-21, and TNF-α. We further found that activated phospho-FGFR3 and -RSK2 were more highly observed in RA than in OA synovium. The hyperplastic lining and sublining lymphoid aggregate layers of RA synovium showed p-RSK2-expressing CD68+ macrophages with high frequency, while pRSK2-expressing CD4+ T-cells was observed at a lower frequency. Notably, kaempferol administration in collagen-induced arthritis mice relieved the frequency and severity of arthritis. Kaempferol reduced osteoclast differentiation in vitro and in vivo relative to the controls and was associated with the inhibition of osteoclast markers, such as tartrate-resistant acid phosphatase, integrin β3, and MMP9. Conclusively, our data suggest that bFGF-induced FGFR3-RSK2 signaling may play a critical role during the initiation and progression of RA in terms of FLS proliferation and enhanced osteoclastogenesis, and that kaempferol may be effective as a new treatment for RA.
Collapse
|
35
|
Choi S, Park H, Jung S, Kim EK, Cho ML, Min JK, Moon SJ, Lee SM, Cho JH, Lee DH, Nam JH. Therapeutic Effect of Exogenous Truncated IK Protein in Inflammatory Arthritis. Int J Mol Sci 2017; 18:E1976. [PMID: 28906466 PMCID: PMC5618625 DOI: 10.3390/ijms18091976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 09/10/2017] [Accepted: 09/11/2017] [Indexed: 12/18/2022] Open
Abstract
Inhibitor K562 (IK) protein was first isolated from the culture medium of K562, a leukemia cell line. It is known to be an inhibitory regulator of interferon-γ-induced major histocompatibility complex class (MHC) II expression. Previously, we found that transgenic (Tg) mice constitutively expressing truncated IK (tIK) showed reduced numbers of pathogenic Th1 and Th17 cells, which are known to be involved in the development of rheumatoid arthritis (RA). Here, we investigated whether exogenous tIK protein has a therapeutic effect in arthritis in disease models and analyzed its mechanism. Exogenous tIK protein was produced in an insect expression system and applied to the collagen antibody-induced arthritis (CAIA) mouse disease model. Injection of tIK protein alleviated the symptoms of arthritis in the CAIA model and reduced Th1 and Th17 cell populations. In addition, treatment of cultured T cells with tIK protein induced expression of A20, a negative regulator of nuclear factor-κB (NFκB)-induced inflammation, and reduced expression of several transcription factors related to T cell activation. We conclude that exogenous tIK protein has the potential to act as a new therapeutic agent for RA patients, because it has a different mode of action to biopharmaceutical agents, such as tumor necrosis factor antagonists, that are currently used to treat RA.
Collapse
Affiliation(s)
- Seulgi Choi
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - HyeLim Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - SeoYeon Jung
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| | - Eun-Kyung Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Jun-Ki Min
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Su-Jin Moon
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea.
| | - Sang-Myeong Lee
- Department of Biotechnology, Chonbuk National University, Iksan 54596, Korea.
| | - Jang-Hee Cho
- Biomaterials Research Center, Cellinbio, Suwon 16680, Korea.
| | - Dong-Hee Lee
- Biomaterials Research Center, Cellinbio, Suwon 16680, Korea.
| | - Jae-Hwan Nam
- Department of Biotechnology, The Catholic University of Korea, Bucheon 14662, Korea.
| |
Collapse
|
36
|
Kim S, Easterling ER, Price LC, Smith SL, Coligan JE, Park JE, Brand DD, Rosloniec EF, Stuart JM, Kang AH, Myers LK. The Role of Leukocyte-Associated Ig-like Receptor-1 in Suppressing Collagen-Induced Arthritis. THE JOURNAL OF IMMUNOLOGY 2017; 199:2692-2700. [PMID: 28887430 DOI: 10.4049/jimmunol.1700271] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 08/10/2017] [Indexed: 11/19/2022]
Abstract
Several observations implicate a critical role for T cell dysregulation as a central problem in rheumatoid arthritis. We investigated a mechanism for suppressing T cell activation by stimulating a natural inhibitory receptor called leukocyte-associated Ig-like receptor-1 (LAIR-1). The collagen-induced arthritis (CIA) model and DR-1 transgenic mice were used to study the importance of LAIR-1 in autoimmune arthritis. Splenocytes from wild-type or LAIR-1-/- mice were stimulated with soluble anti-CD3 Ab in the presence or absence of α1(II) and supernatants were collected for cytokine analysis. B6.DR1 mice were immunized with type II collagen/CFA to induce arthritis and were treated with either the stimulatory mAb to LAIR-1 or a hamster IgG control. Finally, B6.DR1/LAIR-1-/- and B6.DR1/LAIR-1+/+ mice were challenged for CIA and mean severity scores were recorded thrice weekly. Using splenocytes or purified CD4+ cells that were sufficient in LAIR-1, CD3-induced cytokine secretion was significantly suppressed in the presence of collagen, whereas LAIR-1-deficient splenocytes had no attenuation. Treatment with a stimulatory mAb to LAIR-1 also significantly attenuated CIA in the LAIR+/+ mice. When B6.DR1/LAIR-1-/- mice were immunized with type II collagen they developed more severe arthritis and had a greater percentage of affected limbs than the wild-type mice. These data demonstrate that collagen can suppress the T cell cytokine response through the action of LAIR-1. Treatment with stimulating LAIR-1 Abs suppresses CIA whereas B6.DR1/LAIR-1-/- mice develop more severe arthritis than wild-type controls. These data suggest that LAIR-1 may be a potential therapeutic target for suppressing rheumatoid arthritis.
Collapse
Affiliation(s)
- Seunghyun Kim
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Ellis R Easterling
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Lauren C Price
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Savannah L Smith
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163
| | - John E Coligan
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852
| | - Jeoung-Eun Park
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163
| | - David D Brand
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163.,Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38163.,Research Service, Veterans Affairs Medical Center, Memphis, TN 38104; and
| | - Edward F Rosloniec
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163.,Research Service, Veterans Affairs Medical Center, Memphis, TN 38104; and
| | - John M Stuart
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163.,Research Service, Veterans Affairs Medical Center, Memphis, TN 38104; and
| | - Andrew H Kang
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163.,Research Service, Veterans Affairs Medical Center, Memphis, TN 38104; and
| | - Linda K Myers
- Division of Rheumatology, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38163
| |
Collapse
|
37
|
Urushima H, Fujimoto M, Mishima T, Ohkawara T, Honda H, Lee H, Kawahata H, Serada S, Naka T. Leucine-rich alpha 2 glycoprotein promotes Th17 differentiation and collagen-induced arthritis in mice through enhancement of TGF-β-Smad2 signaling in naïve helper T cells. Arthritis Res Ther 2017; 19:137. [PMID: 28615031 PMCID: PMC5471956 DOI: 10.1186/s13075-017-1349-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/26/2017] [Indexed: 11/10/2022] Open
Abstract
Background Leucine-rich alpha 2 glycoprotein (LRG) has been identified as a serum protein elevated in patients with active rheumatoid arthritis (RA). Although the function of LRG is ill-defined, LRG binds with transforming growth factor (TGF)-β and enhances Smad2 phosphorylation. Considering that the imbalance between T helper 17 (Th17) cells and regulatory T cells (Treg) plays important roles in the pathogenesis of RA, LRG may affect arthritic pathology by enhancing the TGF-β-Smad2 pathway that is pivotal for both Treg and Th17 differentiation. The purpose of this study was to explore the contribution of LRG to the pathogenesis of arthritis, with a focus on the role of LRG in T cell differentiation. Methods The differentiation of CD4 T cells and the development of collagen-induced arthritis (CIA) were examined in wild-type mice and LRG knockout (KO) mice. To examine the influence of LRG on T cell differentiation, naïve CD4 T cells were isolated from LRG KO mice and cultured under Treg- or Th17-polarization condition in the absence or presence of recombinant LRG. Results In the CIA model, LRG deficiency led to ameliorated arthritis and reduced Th17 differentiation with no influence on Treg differentiation. By addition of recombinant LRG, the expression of IL-6 receptor (IL-6R) was enhanced through TGF-β-Smad2 signaling. In LRG KO mice, the IL-6R expression and IL-6-STAT3 signaling was attenuated in naïve CD4 T cells, compared to wild-type mice. Conclusions Our findings suggest that LRG upregulates IL-6R expression in naïve CD4 T cells by the enhancement of TGF-β-smad2 pathway and promote Th17 differentiation and arthritis development.
Collapse
Affiliation(s)
- Hayato Urushima
- Laboratory of Immune Signal, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Minoru Fujimoto
- Laboratory of Immune Signal, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan. .,Center for Intractable Immune Disease, Kochi Medical School, Kochi University, Kochi, Japan.
| | - Takashi Mishima
- Laboratory of Immune Signal, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Tomoharu Ohkawara
- Laboratory of Immune Signal, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Hiromi Honda
- Laboratory of Immune Signal, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Hyun Lee
- Laboratory of Immune Signal, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan
| | - Hirohisa Kawahata
- Department of Medical Technology, Morinomiya University of medical science, Osaka, Japan
| | - Satoshi Serada
- Laboratory of Immune Signal, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan.,Center for Intractable Immune Disease, Kochi Medical School, Kochi University, Kochi, Japan
| | - Tetsuji Naka
- Laboratory of Immune Signal, National Institutes of Biomedical Innovation, Health and Nutrition, 7-6-8 Saito-Asagi, Ibaraki, Osaka, 567-0085, Japan.,Center for Intractable Immune Disease, Kochi Medical School, Kochi University, Kochi, Japan
| |
Collapse
|
38
|
Xu J, Feng Z, Chen S, Zhu J, Wu X, Chen X, Li J. Taxol alleviates collagen-induced arthritis in mice by inhibiting the formation of microvessels. Clin Rheumatol 2017; 38:19-27. [PMID: 28455825 PMCID: PMC6342865 DOI: 10.1007/s10067-017-3646-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/05/2017] [Accepted: 04/18/2017] [Indexed: 11/28/2022]
Abstract
The objective of the present study is to evaluate the inhibitory effects of taxol (PTX) on angiogenesis in a collagen-induced arthritis (CIA) mouse model. Collagen II (C II) and complete Freund’s adjuvant (CFA) were used in C57BL/6 (H-2b) mice to generate the CIA model. Random grouping was performed in the normal control group, CIA model group, PTX 1.5 mg/kg group, PTX 1.0 mg/kg group, and PTX 0.5 mg/kg group. Arthritis index scores, tissue pathology scores, and synovium microvessel density (MVD) analysis were performed. Immunohistochemistry and enzyme-linked immunosorbent assay were used to detect the expression of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-α (HIF-1α). The correlation between MVD and pathological scores and between MVD and the expression of VEGF as well as HIF-1α in the synovium were also evaluated. After PTX treatment, the three intervention group arthritis index scores were reduced when compared with the CIA group. The total histological scores in the three PTX treatment groups were lower than those in the CIA group. Similarly, PTX significantly alleviated the scores for synovitis, pannus formation, and bone destruction. Compared with the CIA group, the MVD of the three intervention groups decreased in a dose-dependent manner. The expression of VEGF and HIF-1α in synovial tissues and serum also significantly decreased after PTX treatment. Further analysis showed that MVD and pathological scores and MVD and expression of VEGF as well as HIF-1α in the synovium were positively correlated. PTX may alleviate CIA by suppressing angiogenesis, providing new insights into the treatment of rheumatoid arthritis (RA). VEGF and HIF-1α may be targets for PTX suppression of microvessel formation.
Collapse
Affiliation(s)
- Juan Xu
- Department of Internal Medicine of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhitao Feng
- Department of Internal Medicine of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.,Department of Traditional Chinese Medicine, Medical College of China Three Gorges University, Yichang, Hubei, 443002, China
| | - Shixian Chen
- Department of Internal Medicine of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Junqing Zhu
- Department of Rheumatology, Nanfang Hospital, Southern Medical University, No. 1838, North of Guangzhou Avenue, Guangzhou, Guangdong, 510515, China
| | - Xianghui Wu
- Laboratory Animal Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Xiaoguang Chen
- Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Juan Li
- Department of Internal Medicine of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China. .,Department of Rheumatology, Nanfang Hospital, Southern Medical University, No. 1838, North of Guangzhou Avenue, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
39
|
Niu H, Xu M, Li S, Chen J, Luo J, Zhao X, Gao C, Li X. High-Performance Liquid Chromatography (HPLC) Quantification of Liposome-Delivered Doxorubicin in Arthritic Joints of Collagen-Induced Arthritis Rats. Med Sci Monit Basic Res 2017; 23:150-158. [PMID: 28408733 PMCID: PMC5400028 DOI: 10.12659/msmbr.904103] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Neoangiogenesis occurring in inflamed articular synovium in early rheumatoid arthritis (RA) is characterized by enhanced vascular permeability that allows nanoparticle agents, including liposomes, to deliver encapsulated drugs to arthritic joints and subsequently improve therapeutic efficacy and reduce adverse effects. However, the targeting distribution of liposomes in arthritic joints during RA has not been quantitatively demonstrated. We performed this study to evaluate the targeting distribution of PEGylated doxorubicin liposomes in the arthritic joints of collagen-induced arthritis (CIA) rats by high-performance liquid chromatography (HPLC). Material/Methods Two doxorubicin formulations were administered to CIA rats via tail intravenous injection at a single dose (50 mg/m2). CIA rats were sacrificed and the tissues of the inflamed ankle joints were collected. The content of doxorubicin in the arthritic joints was analyzed by a validated and reproducible HPLC method. A two-way ANOVA for 2×5 factorial design was used for statistical analysis. Results The developed HPLC method was sensitive, precise, and reproducible. The method was successfully applied to quantify doxorubicin content in arthritic tissues. At each time point (6, 12, 24, 48, and 72 h), doxorubicin content in the arthritic joints of the doxorubicin liposome group (DOX-LIP group) was higher than in the free doxorubicin group (DOX group) (P<0.05). In the DOX-LIP group, doxorubicin levels in the arthritic joints increased gradually and significantly in the interval of 6–72 h post-administration. Conclusions PEGylated doxorubicin liposomes were targeted to, accumulated, and retained in the arthritic joints of CIA rats. The present study indicates that liposome encapsulation increases the therapeutic efficacy of antirheumatic drugs, presenting a promising therapeutic strategy for RA.
Collapse
Affiliation(s)
- Hongqing Niu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Menghua Xu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Shuangtian Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Junwei Chen
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Jing Luo
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Xiangcong Zhao
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Chong Gao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| |
Collapse
|
40
|
Tamaddon M, Burrows M, Ferreira SA, Dazzi F, Apperley JF, Bradshaw A, Brand DD, Czernuszka J, Gentleman E. Monomeric, porous type II collagen scaffolds promote chondrogenic differentiation of human bone marrow mesenchymal stem cells in vitro. Sci Rep 2017; 7:43519. [PMID: 28256634 PMCID: PMC5335259 DOI: 10.1038/srep43519] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 01/25/2017] [Indexed: 12/20/2022] Open
Abstract
Osteoarthritis (OA) is a common cause of pain and disability and is often associated with the degeneration of articular cartilage. Lesions to the articular surface, which are thought to progress to OA, have the potential to be repaired using tissue engineering strategies; however, it remains challenging to instruct cell differentiation within a scaffold to produce tissue with appropriate structural, chemical and mechanical properties. We aimed to address this by driving progenitor cells to adopt a chondrogenic phenotype through the tailoring of scaffold composition and physical properties. Monomeric type-I and type-II collagen scaffolds, which avoid potential immunogenicity associated with fibrillar collagens, were fabricated with and without chondroitin sulfate (CS) and their ability to stimulate the chondrogenic differentiation of human bone marrow-derived mesenchymal stem cells was assessed. Immunohistochemical analyses showed that cells produced abundant collagen type-II on type-II scaffolds and collagen type-I on type-I scaffolds. Gene expression analyses indicated that the addition of CS - which was released from scaffolds quickly - significantly upregulated expression of type II collagen, compared to type-I and pure type-II scaffolds. We conclude that collagen type-II and CS can be used to promote a more chondrogenic phenotype in the absence of growth factors, potentially providing an eventual therapy to prevent OA.
Collapse
Affiliation(s)
- M. Tamaddon
- Department of Materials, University of Oxford, Oxford OX1 3PH, UK
- Craniofacial Development and Stem Cell Biology, King’s College London, London SE1 9RT, UK
| | - M. Burrows
- Craniofacial Development and Stem Cell Biology, King’s College London, London SE1 9RT, UK
| | - S. A. Ferreira
- Craniofacial Development and Stem Cell Biology, King’s College London, London SE1 9RT, UK
| | - F. Dazzi
- Division of Cancer Studies, Rayne Institute, King’s College London, London SE5 9NU, UK
| | - J. F. Apperley
- Centre for Haematology, Department of Medicine, Imperial College London, London W12 0NN, UK
- John Goldman Centre for Cellular Therapy, Imperial College Healthcare NHS Trust, London W12 0HS, UK
| | - A. Bradshaw
- John Goldman Centre for Cellular Therapy, Imperial College Healthcare NHS Trust, London W12 0HS, UK
| | - D. D. Brand
- Research Service, Memphis VA Medical Center, Departments of Medicine and Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN 38104, USA
| | - J. Czernuszka
- Department of Materials, University of Oxford, Oxford OX1 3PH, UK
| | - E. Gentleman
- Craniofacial Development and Stem Cell Biology, King’s College London, London SE1 9RT, UK
| |
Collapse
|
41
|
Abdalmula A, Dooley LM, Kaufman C, Washington EA, House JV, Blacklaws BA, Ghosh P, Itescu S, Bailey SR, Kimpton WG. Immunoselected STRO-3 + mesenchymal precursor cells reduce inflammation and improve clinical outcomes in a large animal model of monoarthritis. Stem Cell Res Ther 2017; 8:22. [PMID: 28173831 PMCID: PMC5297153 DOI: 10.1186/s13287-016-0460-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 12/04/2016] [Accepted: 12/16/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The purpose of this study was to investigate the therapeutic efficacy of intravenously administered immunoselected STRO-3 + mesenchymal precursor cells (MPCs) on clinical scores, joint pathology and cytokine production in an ovine model of monoarthritis. METHODS Monoarthritis was established in 16 adult merino sheep by administration of bovine type II collagen into the left hock joint following initial sensitization to this antigen. After 24 h, sheep were administered either 150 million allogeneic ovine MPCs (n = 8) or saline (n = 8) intravenously (IV). Lameness, joint swelling and pain were monitored and blood samples for leukocytes and cytokine levels were collected at intervals following arthritis induction. Animals were necropsied 14 days after arthritis induction and gross and histopathological evaluations were undertaken on tissues from the arthritic (left) and contralateral (right) joints. RESULTS MPC-treated sheep demonstrated significantly reduced clinical signs of lameness, joint pain and swelling compared with saline controls. They also showed decreased cartilage erosions, synovial stromal cell activation and angiogenesis. This was accompanied by decreased infiltration of the synovial tissues by CD4+ lymphocytes and CD14+ monocytes/macrophages. Over the 3 days following joint arthropathy induction, the numbers of neutrophils circulating in the blood and plasma concentrations of activin A were significantly reduced in animals administered MPCs. CONCLUSIONS The results of this study have demonstrated the capacity of IV-administered MPCs to mitigate the clinical signs and some of the inflammatory mediators responsible for joint tissue destruction in a large animal model of monoarthritis.
Collapse
MESH Headings
- Activins/blood
- Animals
- Antigens, Surface/genetics
- Antigens, Surface/immunology
- Arthritis, Experimental/chemically induced
- Arthritis, Experimental/genetics
- Arthritis, Experimental/pathology
- Arthritis, Experimental/therapy
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/pathology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/pathology
- Cell Differentiation
- Cell Movement
- Collagen Type II/administration & dosage
- Disease Models, Animal
- Female
- Gene Expression
- Interferon-gamma/biosynthesis
- Interferon-gamma/immunology
- Interleukin-10/biosynthesis
- Interleukin-10/immunology
- Interleukin-17/biosynthesis
- Interleukin-17/immunology
- Joints/immunology
- Joints/pathology
- Macrophages/immunology
- Macrophages/pathology
- Mesenchymal Stem Cell Transplantation
- Mesenchymal Stem Cells/cytology
- Mesenchymal Stem Cells/immunology
- Monocytes/immunology
- Monocytes/pathology
- Neutrophils/immunology
- Neutrophils/pathology
- Sheep, Domestic
- Synovial Fluid/chemistry
- Synovial Fluid/cytology
- Synovial Fluid/immunology
- Treatment Outcome
Collapse
Affiliation(s)
- Anwar Abdalmula
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC 5010 Australia
| | - Laura M. Dooley
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC 5010 Australia
| | - Claire Kaufman
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC 5010 Australia
| | - Elizabeth A. Washington
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC 5010 Australia
| | - Jacqueline V. House
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3010 Australia
| | - Barbara A. Blacklaws
- Department of Veterinary Medicine, University of Cambridge, Cambridge, CB3 0ES UK
| | - Peter Ghosh
- Mesoblast Ltd, 55 Collins Street, Melbourne, VIC 3000 Australia
| | - Silviu Itescu
- Mesoblast Ltd, 55 Collins Street, Melbourne, VIC 3000 Australia
| | - Simon R. Bailey
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC 5010 Australia
| | - Wayne G. Kimpton
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC 5010 Australia
| |
Collapse
|
42
|
Xiang Y, Zhang T, Yin C, Zhou J, Huang R, Gao S, Zheng L, Wang X, Manyande A, Tian X, Chen J, Fang J. Effects of the stem extracts of Schisandra glaucescens Diels on collagen-induced arthritis in Balb/c mice. JOURNAL OF ETHNOPHARMACOLOGY 2016; 194:1078-1086. [PMID: 27840082 DOI: 10.1016/j.jep.2016.11.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 08/01/2016] [Accepted: 11/04/2016] [Indexed: 05/20/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Schisandra glaucescens Diels (SGD) is used in a subclass of traditional Chinese medicine known as "Tujia drugs". It has been long used for the treatment of rheumatoid arthritis (RA), cough with dyspnea, spontaneous sweating, night sweating, chronic diarrhea, and neurasthenia. As a woody liana growing in mountain jungles at the altitudes of 750-1800m, it is mainly distributed in Sichuan and Hubei Provinces of China. AIM OF THE STUDY To evaluate the antiarthritic activity of acetate (EA) and n-butanol (Bu) fractions of SGD extract on a collagen-induced arthritis mice model. MATERIALS AND METHODS Acute toxicity of EA and Bu fractions of SGD extract was evaluated by gavage on normal mice. Pharmacological investigations were conducted on arthritis male Balb/c mice. The animal model was induced by immunization with type II bovine collagen (CII) on the 1st and the 14th day of the experimental schedule. EA fraction (104, 312, 936mg/kg), Bu fraction (156, 469, 1407mg/kg) of SGD extract was orally administered every two days since the 15th day for 3 weeks. Progression of edema in the paws was measured using a vernier caliper every 3 days since the 10th day. At the end of the experiment, the spleen index and histological changes of the hind knee joints were investigated. Additionally, to explore the possible antirheumatic mechanisms of the EA and Bu fractions, ELISA was carried out to analyze TNF-α, IL-10, IL-6 and IL-1β in the serum. RESULTS The half lethal doses of both EA and Bu fractions were much higher than the dose administered in the pharmacological investigations. Oral administration of EA fraction and Bu fraction of SGD extract significantly and does-dependently inhibited type ІІ collagen induced arthritis (CIA) in mice, as indicated by the effects on paws swelling and spleen index. Histopathological examinations demonstrated that SGD effectively protected the bones and cartilages of knee joints from erosion, lesion and deformation. Besides, the serum concentrations of cytokines TNF-α, IL-1β and IL-6 were significantly lower than the ones from the vehicle control group. Respectively, while cytokine IL-10 was remarkably higher compare with the vehicle control group. CONCLUSIONS SGD might be a safe and effective candidate for the treatment of RA, and deserves further investigation on the chemical components in both EA and Bu fractions of SGD extract.
Collapse
Affiliation(s)
- Yan Xiang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Tiantian Zhang
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chunpin Yin
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiewen Zhou
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rong Huang
- Department of Ophthalmology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430061, Hubei, China
| | - Shasha Gao
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lamei Zheng
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaogang Wang
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Anne Manyande
- School of Psychology, Social Work and Human Sciences, University of West London, London, UK
| | - Xuebi Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Jiachun Chen
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jinbo Fang
- School of Pharmacy, Hubei Key Laboratory of Natural Medicinal Chemistry and Resource Evaluation, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
43
|
Park JE, Rotondo JA, Cullins DL, Brand DD, Yi AK, Stuart JM, Kang AH, Myers LK. Characterization of the Syk-Dependent T Cell Signaling Response to an Altered Peptide. THE JOURNAL OF IMMUNOLOGY 2016; 197:4569-4575. [PMID: 27837109 DOI: 10.4049/jimmunol.1600771] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 10/12/2016] [Indexed: 11/19/2022]
Abstract
Rheumatoid arthritis is an autoimmune disorder characterized by T cell dysregulation. We have shown that an altered peptide ligand (A9) activates T cells to use an alternate signaling pathway that is dependent on FcRγ and spleen tyrosine kinase, resulting in downregulation of inflammation. In the experiments described in this study, we have attempted to determine the molecular basis of this paradox. Three major Src family kinases found in T cells (Lck, Fyn, and Lyn) were tested for activation following stimulation by A9/I-Aq Unexpectedly we found they are not required for T cell functions induced by A9/I-Aq, nor are they required for APL stimulation of cytokines. On the other hand, the induction of the second messenger inositol trisphosphate and the mobilization of calcium are clearly triggered by the APL A9/I-Aq stimulation and are required for cytokine production, albeit the cytokines induced are different from those produced after activation of the canonical pathway. DBA/1 mice doubly deficient in IL-4 and IL-10 were used to confirm that these two cytokines are important for the APL-induced attenuation of arthritis. These studies provide a basis for exploring the effectiveness of analog peptides and the inhibitory T cells they induce as therapeutic tools for autoimmune arthritis.
Collapse
Affiliation(s)
- Jeoung-Eun Park
- Department of Medicine, University of Tennessee Health Science Center, Memphis TN 38163
| | - Jeffrey A Rotondo
- Department of Medicine, University of Tennessee Health Science Center, Memphis TN 38163
| | - David L Cullins
- Department of Medicine, University of Tennessee Health Science Center, Memphis TN 38163
| | - David D Brand
- Department of Medicine, University of Tennessee Health Science Center, Memphis TN 38163.,Research Service, Veterans Affairs Medical Center, Memphis TN 38104
| | - Ae-Kyung Yi
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis TN 38163; and
| | - John M Stuart
- Department of Medicine, University of Tennessee Health Science Center, Memphis TN 38163.,Research Service, Veterans Affairs Medical Center, Memphis TN 38104
| | - Andrew H Kang
- Department of Medicine, University of Tennessee Health Science Center, Memphis TN 38163.,Research Service, Veterans Affairs Medical Center, Memphis TN 38104
| | - Linda K Myers
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis TN 38163
| |
Collapse
|
44
|
Chen R, Schwander M, Barbe MF, Chan MM. Ossicular Bone Damage and Hearing Loss in Rheumatoid Arthritis: A Correlated Functional and High Resolution Morphometric Study in Collagen-Induced Arthritic Mice. PLoS One 2016; 11:e0164078. [PMID: 27690307 PMCID: PMC5045188 DOI: 10.1371/journal.pone.0164078] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/19/2016] [Indexed: 01/13/2023] Open
Abstract
Globally, a body of comparative case-control studies suggests that rheumatoid arthritis (RA) patients are more prone to developing hearing loss (HL). However, experimental evidence that supports this hypothesis is still lacking because the human auditory organ is not readily accessible. The aim of this study was to determine the association between bone damage to the ossicles of the middle ear and HL, using a widely accepted murine model of collagen-induced arthritis (RA mice). Diarthrodial joints in the middle ear were examined with microcomputer tomography (microCT), and hearing function was assessed by auditory brainstem response (ABR). RA mice exhibited significantly decreased hearing sensitivity compared to age-matched controls. Additionally, a significant narrowing of the incudostapedial joint space and an increase in the porosity of the stapes were observed. The absolute latencies of all ABR waves were prolonged, but mean interpeak latencies were not statistically different. The observed bone defects in the middle ear that were accompanied by changes in ABR responses were consistent with conductive HL. This combination suggests that conductive impairment is at least part of the etiology of RA-induced HL in a murine model. Whether the inner ear sustains bone erosion or other pathology, and whether the cochlear nerve sustains pathology await subsequent studies. Considering the fact that certain anti-inflammatories are ototoxic in high doses, monitoring RA patients’ auditory function is advisable as part of the effort to ensure their well-being.
Collapse
Affiliation(s)
- Rensa Chen
- Department of Microbiology and Immunology, Lewis Katz School of Medicine-Temple University, Philadelphia, PA, 19140, United States of America
| | - Martin Schwander
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, United States of America
| | - Mary F. Barbe
- Department of Anatomy, Lewis Katz School of Medicine-Temple University, Philadelphia, PA, 19140, United States of America
| | - Marion M. Chan
- Department of Microbiology and Immunology, Lewis Katz School of Medicine-Temple University, Philadelphia, PA, 19140, United States of America
- * E-mail:
| |
Collapse
|
45
|
MicroRNA-21 Promotes Proliferation of Fibroblast-Like Synoviocytes through Mediation of NF-κB Nuclear Translocation in a Rat Model of Collagen-Induced Rheumatoid Arthritis. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9279078. [PMID: 27429986 PMCID: PMC4939181 DOI: 10.1155/2016/9279078] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 05/03/2016] [Accepted: 05/17/2016] [Indexed: 11/17/2022]
Abstract
MicroRNA-21 (miR-21) is overexpressed in patients with rheumatoid arthritis (RA). This study was designed to investigate the effect and mechanism of miR-21 on cell proliferation in fibroblast-like synoviocytes (FLS) of RA. FLS were primary-cultured from a rat RA model. RA-FLS and normal FLS were infected with lentivirus (anti-miR-21 or pro-miR-21) for overexpression or downregulation of miR-21, respectively. The effects of miR-21 overexpression or inhibition on nucleoprotein NF-κB levels and FLS cell proliferation were evaluated by western blotting and MTT assays. The effects of an inhibitor of NF-κB nuclear translocation (BAY 11-7082) were also evaluated. The results showed that the levels of miR-21 and nucleoprotein NF-κB were increased in FLS of RA model rats compared to the control group. Downregulation of miR-21 in RA FLS led to a significant decrease in nucleoprotein NF-κB levels and cell proliferation rates compared to the antinegative control (NC) group. However, miR-21 overexpression in normal FLS resulted in a significant increase of nucleoprotein NF-κB levels and cell proliferation rates compared to the pro-NC group. The effects of miR-21 overexpression were reversed by BAY 11-7082. We concluded that upregulated miR-21 in FLS in RA model rats may promote cell proliferation by facilitating NF-κB nuclear translocation, thus affecting the NF-κB pathway.
Collapse
|
46
|
Christensen AD, Haase C, Cook AD, Hamilton JA. K/BxN Serum-Transfer Arthritis as a Model for Human Inflammatory Arthritis. Front Immunol 2016; 7:213. [PMID: 27313578 PMCID: PMC4889615 DOI: 10.3389/fimmu.2016.00213] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/17/2016] [Indexed: 12/29/2022] Open
Abstract
The K/BxN serum-transfer arthritis (STA) model is a murine model in which the immunological mechanisms occurring in rheumatoid arthritis (RA) and other arthritides can be studied. To induce K/BxN STA, serum from arthritic transgenic K/BxN mice is transferred to naive mice and manifestations of arthritis occur a few days later. The inflammatory response in the model is driven by autoantibodies against the ubiquitously expressed self-antigen, glucose-6-phosphate isomerase (G6PI), leading to the formation of immune complexes that drive the activation of different innate immune cells such as neutrophils, macrophages, and possibly mast cells. The pathogenesis further involves a range of immune mediators including cytokines, chemokines, complement factors, Toll-like receptors, Fc receptors, and integrins, as well as factors involved in pain and bone erosion. Hence, even though the K/BxN STA model mimics only the effector phase of RA, it still involves a wide range of relevant disease mediators. Additionally, as a murine model for arthritis, the K/BxN STA model has some obvious advantages. First, it has a rapid and robust onset of arthritis with 100% incidence in genetically identical animals. Second, it can be induced in a wide range of strain backgrounds and can therefore also be induced in gene-deficient strains to study the specific importance of disease mediators. Even though G6PI might not be an essential autoantigen, for example, in RA, the K/BxN STA model is a useful tool to understand how autoantibodies, in general, drive the progression of arthritis by interacting with downstream components of the innate immune system. Finally, the model has also proven useful as a model wherein arthritic pain can be studied. Taken together, these features make the K/BxN STA model a relevant one for RA, and it is a potentially valuable tool, especially for the preclinical screening of new therapeutic targets for RA and perhaps other forms of inflammatory arthritis. Here, we describe the molecular and cellular pathways in the development of K/BxN STA focusing on the recent advances in the understanding of the important mechanisms. Additionally, this review provides a comparison of the K/BxN STA model to some other arthritis models.
Collapse
Affiliation(s)
- Anne D Christensen
- Department of Medicine, University of Melbourne, Parkville, VIC, Australia; Novo Nordisk A/S, Måløv, Denmark
| | | | - Andrew D Cook
- Department of Medicine, University of Melbourne , Parkville, VIC , Australia
| | - John A Hamilton
- Department of Medicine, University of Melbourne , Parkville, VIC , Australia
| |
Collapse
|
47
|
Effects of Wutou Decoction on DNA Methylation and Histone Modifications in Rats with Collagen-Induced Arthritis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:5836879. [PMID: 27042192 PMCID: PMC4799822 DOI: 10.1155/2016/5836879] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 02/11/2016] [Indexed: 01/12/2023]
Abstract
Background. Wutou decoction (WTD) has been wildly applied in the treatment of rheumatoid arthritis and experimental arthritis in rats for many years. Epigenetic deregulation is associated with the aetiology of rheumatoid arthritis; however, the effects of WTD on epigenetic changes are unclear. This study is set to explore the effects of WTD on DNA methylation and histone modifications in rats with collagen-induced arthritis (CIA). Methods. The CIA model was established by the stimulation of collagen and adjuvant. The knee synovium was stained with hematoxylin and eosin. The DNA methyltransferase 1 (DNMT1) and methylated CpG binding domain 2 (MBD2) expression of peripheral blood mononuclear cells (PBMCs) were determined by Real-Time PCR. The global DNA histone H3-K4/H3-K27 methylation and total histones H3 and H4 acetylation of PBMCs were detected. Results. Our data demonstrated that the DNMT1 mRNA expression was significantly lowered in group WTD compared to that in group CIA (P < 0.05). The DNA methylation level was significantly reduced in group WTD compared to that in group CIA (P < 0.05). Moreover, H3 acetylation of PBMCs was overexpressed in WTD compared with CIA (P < 0.05). Conclusions. WTD may modulate DNA methylation and histone modifications, functioning as anti-inflammatory potential.
Collapse
|
48
|
EAF2 mediates germinal centre B-cell apoptosis to suppress excessive immune responses and prevent autoimmunity. Nat Commun 2016; 7:10836. [PMID: 26935903 PMCID: PMC4782062 DOI: 10.1038/ncomms10836] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 01/25/2016] [Indexed: 01/03/2023] Open
Abstract
Regulated apoptosis of germinal centre (GC) B cells is critical for normal humoral immune responses. ELL-associated factor 2 (EAF2) regulates transcription elongation and has been shown to be an androgen-responsive potential tumour suppressor in prostate by inducing apoptosis. Here we show that EAF2 is selectively upregulated in GC B cells among various immune cell types and promotes apoptosis of GC B cells both in vitro and in vivo. EAF2 deficiency results in enlarged GCs and elevated antibody production during a T-dependent immune response. After immunization with type II collagen, mice lacking EAF2 produce high levels of collagen-specific autoantibodies and rapidly develop severe arthritis. Moreover, the mutant mice spontaneously produce anti-dsDNA, rheumatoid factor and anti-nuclear antibodies as they age. These results demonstrate that EAF2-mediated apoptosis in GC B cells limits excessive humoral immune responses and is important for maintaining self-tolerance.
Collapse
|
49
|
Miller DC, Whittington KB, Brand DD, Hasty KA, Rosloniec EF. The CII-specific autoimmune T-cell response develops in the presence of FTY720 but is regulated by enhanced Treg cells that inhibit the development of autoimmune arthritis. Arthritis Res Ther 2016; 18:8. [PMID: 26757712 PMCID: PMC4718028 DOI: 10.1186/s13075-015-0909-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 12/29/2015] [Indexed: 12/24/2022] Open
Abstract
Background Fingolimod (FTY720) is an immunomodulating drug that inhibits sphingosine-1-phosphate binding and blocks T-cell egress from lymph nodes. We analyzed the effect of FTY720 on the autoimmune T- and B-cell response in autoimmune arthritis and studied the mechanisms by which it alters the function of T cells. Methods Human leukocyte antigen (HLA)-DR1 humanized mice were immunized with type II collagen (CII) and treated with FTY720 three times per week for 3 weeks. Arthritis was evaluated and autoimmune T- and B-cell responses were measured using proliferation assays, enzyme-linked immunosorbent assays, HLA-DR tetramers, and flow cytometry. The functional capacity of regulatory T (Treg) cells from FTY720-treated mice was measured using an in vitro suppression assay, and the role of Treg cells in inhibiting arthritis in FTY720-treated mice was evaluated using mice treated with anti-CD25 to deplete Treg cells. Results Treatment with FTY720 delayed the onset of arthritis and significantly reduced disease incidence. FTY720 did not prevent the generation of a CII-specific autoimmune T-cell response in vivo. However, as the treatment continued, these T cells became unresponsive to restimulation with antigen in vitro, and this anergic state was reversed by addition of interleukin 2. Measurements of CD4+CD25+Foxp3+ cells in the lymph nodes revealed that the ratio of Treg to helper T (Th) cells increased twofold in the FTY720-treated mice, and in vitro assays indicated that the regulatory function of these cells was enhanced. That FTY720 stimulation of Treg cells played a major role in arthritis inhibition was demonstrated by a loss of disease inhibition and restitution of the T-cell proliferative function after in vivo depletion of the Treg cells. Conclusions While FTY720 affects the recirculation of lymphocytes, its ability to inhibit the development of autoimmune arthritis involves several mechanisms, including the enhancement of Treg cell function by increasing the Treg/Th ratio and increased regulatory function on a per-cell basis. FTY720 did not inhibit the development of the autoimmune T-cell response, but disease inhibition appeared to be mediated by Treg cell–mediated suppression of the CII-specific T cells. These data suggest that specific targeting of Treg cells with FTY720 may be a novel therapy for autoimmunity. Electronic supplementary material The online version of this article (doi:10.1186/s13075-015-0909-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David C Miller
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA. .,Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| | | | - David D Brand
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA. .,Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA. .,Memphis VA Medical Center, 1030 Jefferson Avenue, Memphis, TN, 38104, USA.
| | - Karen A Hasty
- Memphis VA Medical Center, 1030 Jefferson Avenue, Memphis, TN, 38104, USA. .,Department of Orthopaedic Surgery, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| | - Edward F Rosloniec
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA. .,Memphis VA Medical Center, 1030 Jefferson Avenue, Memphis, TN, 38104, USA. .,Department of Pathology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
50
|
Greven DEA, Cohen ES, Gerlag DM, Campbell J, Woods J, Davis N, van Nieuwenhuijze A, Lewis A, Heasmen S, McCourt M, Corkill D, Dodd A, Elvin J, Statache G, Wicks IP, Anderson IK, Nash A, Sleeman MA, Tak PP. Preclinical characterisation of the GM-CSF receptor as a therapeutic target in rheumatoid arthritis. Ann Rheum Dis 2015; 74:1924-30. [PMID: 24936585 PMCID: PMC4602263 DOI: 10.1136/annrheumdis-2014-205234] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 04/28/2014] [Accepted: 05/01/2014] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Previous work has suggested that the granulocyte macrophage colony stimulating factor (GM-CSF)-GM-CSF receptor α axis (GM-CSFRα) may provide a new therapeutic target for the treatment of rheumatoid arthritis (RA). Therefore, we investigated the cellular expression of GM-CSFRα in RA synovial tissue and investigated the effects of anti-GM-CSFRα antibody treatment in vitro and in vivo in a preclinical model of RA. METHODS We compared GM-CSFRα expression on macrophages positive for CD68 or CD163 on synovial biopsy samples from patients with RA or psoriatic arthritis (PsA) to disease controls. In addition, we studied the effects of CAM-3003, an anti-GM-CSFR antibody in a collagen induced arthritis model of RA in DBA/1 mice. The pharmacokinetic profile of CAM-3003 was studied in naïve CD1(ICR) mice (see online supplement) and used to interpret the results of the pharmacodynamic studies in BALB/c mice. RESULTS GM-CSFRα was expressed by CD68 positive and CD163 positive macrophages in the synovium, and there was a significant increase in GM-CSFRα positive cells in patients in patients with RA as well as patients with PsA compared with patients with osteoarthritis and healthy controls. In the collagen induced arthritis model there was a dose dependent reduction of clinical arthritis scores and the number of F4/80 positive macrophages in the inflamed synovium after CAM-3003 treatment. In BALB/c mice CAM-3003 inhibited recombinant GM-CSF mediated margination of peripheral blood monocytes and neutrophils. CONCLUSIONS The findings support the ongoing development of therapies aimed at interfering with GM-CSF or its receptor in various forms of arthritis, such as RA and PsA.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/blood
- Antibodies, Monoclonal/therapeutic use
- Antirheumatic Agents/administration & dosage
- Antirheumatic Agents/blood
- Antirheumatic Agents/therapeutic use
- Arthritis, Experimental/blood
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/immunology
- Arthritis, Psoriatic/immunology
- Arthritis, Rheumatoid/immunology
- Case-Control Studies
- Dose-Response Relationship, Immunologic
- Drug Evaluation, Preclinical/methods
- Female
- Humans
- Male
- Mice, Inbred BALB C
- Mice, Inbred DBA
- Middle Aged
- Molecular Targeted Therapy/methods
- Osteoarthritis/immunology
- Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors
- Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Synovial Membrane/immunology
Collapse
Affiliation(s)
- D E A Greven
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands
| | - E S Cohen
- Department of Respiratory, Inflammation and AutoImmunity Research, MedImmune Limited, Cambridge, UK
| | - D M Gerlag
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands GlaxoSmithKline, Cambridge, UK
| | - J Campbell
- Department of Respiratory, Inflammation and AutoImmunity Research, MedImmune Limited, Cambridge, UK
| | - J Woods
- Department of Respiratory, Inflammation and AutoImmunity Research, MedImmune Limited, Cambridge, UK
| | - N Davis
- Department of Respiratory, Inflammation and AutoImmunity Research, MedImmune Limited, Cambridge, UK
| | - A van Nieuwenhuijze
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - A Lewis
- Department of Respiratory, Inflammation and AutoImmunity Research, MedImmune Limited, Cambridge, UK
| | - S Heasmen
- Department of Respiratory, Inflammation and AutoImmunity Research, MedImmune Limited, Cambridge, UK
| | - M McCourt
- Department of Respiratory, Inflammation and AutoImmunity Research, MedImmune Limited, Cambridge, UK
| | - D Corkill
- Department of Respiratory, Inflammation and AutoImmunity Research, MedImmune Limited, Cambridge, UK
| | - A Dodd
- Department of Respiratory, Inflammation and AutoImmunity Research, MedImmune Limited, Cambridge, UK
| | - J Elvin
- Department of Respiratory, Inflammation and AutoImmunity Research, MedImmune Limited, Cambridge, UK
| | - G Statache
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands
| | - I P Wicks
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - I K Anderson
- Department of Respiratory, Inflammation and AutoImmunity Research, MedImmune Limited, Cambridge, UK
| | - A Nash
- Department of Research and Development, CSL Limited, Parkville, Victoria, Australia
| | - M A Sleeman
- Department of Respiratory, Inflammation and AutoImmunity Research, MedImmune Limited, Cambridge, UK
| | - P P Tak
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/ University of Amsterdam, Amsterdam, The Netherlands GlaxoSmithKline, Stevenage, UK University of Cambridge, Cambridge, UK
| |
Collapse
|