1
|
Dedemadi AG, Sevdali E, Georgiadou D, Valanti EK, Neofotistou-Themeli E, Chanis T, Goutakoli P, Thymiakou E, Drakos E, Christopoulou G, Bournazos S, Constantoulakis P, Verginis P, Kardassis D, Stratikos E, Sidiropoulos P, Chroni A. Dantrolene is an HDL-associated paraoxonase-1 activator with immunosuppressive and atheroprotective properties. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159596. [PMID: 39842506 DOI: 10.1016/j.bbalip.2025.159596] [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: 10/16/2024] [Revised: 01/16/2025] [Accepted: 01/19/2025] [Indexed: 01/24/2025]
Abstract
Human paraoxonase 1 (PON1), an enzyme bound to high-density lipoprotein (HDL), hydrolyzes oxidized lipids and contributes to HDL atheroprotective functions. Decreased serum paraoxonase and arylesterase activities of PON1 have been reported in patients at increased atherosclerosis risk, such as rheumatoid arthritis patients, and associated with arthritis severity and cardiovascular risk. Agents that can modulate PON1 activity and HDL-mediated effects have not been discovered. Aiming to discover chemical tools that enhance PON1 activity, we screened a library of marketed drugs (956 compounds) to identify small molecules that can increase HDL-associated PON1 activity. Screening was performed by a kinetic absorbance assay using human HDL as a source of PON1, and paraoxon and phenyl acetate as substrates to measure paraoxonase and arylesterase activities, respectively. Screening identified the drug dantrolene as a potential PON1 activator, which was confirmed by enzymatic kinetic assays using recombinant wild-type PON1, as well as the PON1[L55M] variant displaying decreased enzyme activity in humans. Furthermore, we used the collagen-induced arthritis (CIA) mouse model to examine the effect of dantrolene on HDL properties and arthritis in vivo. Administration of dantrolene in CIA mice increased paraoxonase and arylesterase activities of PON1, as well as the antioxidant capacity of HDL, and reduced arthritis severity by inhibition of naïve CD4+ T cell differentiation to effector memory cells and generation of Th1 cells. Collectively, our in vitro and in vivo findings indicate using small molecules to enhance HDL-associated PON1 activity is a tractable approach that could lead to novel therapeutics targeting immune responses and atherosclerosis.
Collapse
Affiliation(s)
- Anastasia-Georgia Dedemadi
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece; Department of Chemistry, National and Kapodistrian University of Athens, Zografou, Athens, Greece
| | - Eirini Sevdali
- Laboratory of Rheumatology, Autoimmunity and Inflammation, Medical School, University of Crete, Heraklion, Greece
| | - Daphne Georgiadou
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| | - Eftaxia-Konstantina Valanti
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece
| | - Elpida Neofotistou-Themeli
- Laboratory of Rheumatology, Autoimmunity and Inflammation, Medical School, University of Crete, Heraklion, Greece
| | - Theodoros Chanis
- Laboratory of Rheumatology, Autoimmunity and Inflammation, Medical School, University of Crete, Heraklion, Greece
| | - Panagiota Goutakoli
- Laboratory of Rheumatology, Autoimmunity and Inflammation, Medical School, University of Crete, Heraklion, Greece
| | - Efstathia Thymiakou
- Laboratory of Biochemistry, Medical School, University of Crete, Heraklion, Greece
| | - Elias Drakos
- Department of Pathology, Medical School, University of Crete, Heraklion, Greece
| | | | | | | | - Panayotis Verginis
- Laboratory of Immune Regulation and Tolerance, Division of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
| | - Dimitris Kardassis
- Laboratory of Biochemistry, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
| | - Efstratios Stratikos
- Department of Chemistry, National and Kapodistrian University of Athens, Zografou, Athens, Greece
| | - Prodromos Sidiropoulos
- Laboratory of Rheumatology, Autoimmunity and Inflammation, Medical School, University of Crete, Heraklion, Greece; Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece.
| | - Angeliki Chroni
- Institute of Biosciences and Applications, National Center for Scientific Research "Demokritos", Agia Paraskevi, Athens, Greece.
| |
Collapse
|
2
|
Sen K, Kumar Das S, Ghosh N, Sinha K, Sil PC. Lupeol: A dietary and medicinal triterpene with therapeutic potential. Biochem Pharmacol 2024; 229:116545. [PMID: 39293501 DOI: 10.1016/j.bcp.2024.116545] [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: 06/03/2024] [Revised: 09/04/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024]
Abstract
Lupeol, a triterpene derived from various plants, has emerged as a potent dietary supplement with extensive therapeutic potential. This review offers a comprehensive examination of lupeol's applications across diverse health conditions. By meticulously analyzing current scientific literature, we have synthesized findings that underscore lupeol's impact on cancer, diabetes, gastrointestinal disorders, neurological diseases, dermatological conditions, nephrological issues, and cardiovascular health. The review delves into molecular studies that reveal lupeol's ability to modulate disease pathways and alleviate symptoms, positioning it as a promising therapeutic agent. Moreover, we discuss the potential role of lupeol in clinical practice and public health strategies, emphasizing its substantial benefits as a natural compound. This thorough analysis serves as a critical resource for researchers, providing insights into the multifaceted therapeutic properties of lupeol and its potential to significantly enhance health outcomes.
Collapse
Affiliation(s)
- Koushik Sen
- Jhargram Raj College, Jhargram 721507, India
| | | | | | | | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, Kolkata 700054, India.
| |
Collapse
|
3
|
Nan Y, Chen M, Wu W, Huang R, Sun W, Lu Q, Gu Z, Mao X, Xu H, Wang Y. IGF2BP2 regulates the inflammation of fibroblast-like synoviocytes via GSTM5 in rheumatoid arthritis. Cell Death Discov 2024; 10:215. [PMID: 38702323 PMCID: PMC11068746 DOI: 10.1038/s41420-024-01988-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/06/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease with an unknown etiology. RA cannot be fully cured and requires lengthy treatment, imposing a significant burden on both individuals and society. Due to the lack of specific drugs available for treating RA, exploring a key new therapeutic target for RA is currently an important task. Activated fibroblast-like synoviocytes (FLSs) play a crucial role in the progression of RA, which release interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α resulting in abnormal inflammatory reaction in the synovium. A previous study has highlighted the correlation of m6A reader insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2) with inflammation-related diseases in human. However, the role of IGF2BP2 in the inflammatory reaction of FLSs during RA progression has not been assessed. In this study, IGF2BP2 expression was decreased in the synovial tissues of RA patients and collagen-induced arthritis (CIA) rats. Intra-articular injection of an adeno-associated virus (AAV) vector overexpressing IGF2BP2 relieved paw swelling, synovial hyperplasia and cartilage destruction in CIA rats. IGF2BP2 overexpression also inhibited lipopolysaccharide (LPS)-mediated RA fibroblast-like synoviocytes (RA-FLSs) migration and invasion accompanied by a decreased level of inflammatory factors in vitro. Conversely, IGF2BP2 suppression promoted RA-FLSs migration and invasion with an elevated level of inflammatory factors in vitro. The sequencing result showed that glutathione S-transferase Mu 5 (GSTM5), a key antioxidant gene, was the target mRNA of IGF2BP2. Further experiments demonstrated that IGF2BP2 strengthened the stability of GSTM5 mRNA, leading to weakened inflammatory reaction and reduced expression of matrix metalloproteinase 9 and 13 (MMP9, MMP13). Therefore, IGF2BP2-GSTM5 axis may represent a potential therapeutic target for RA treatment.
Collapse
Affiliation(s)
- Yunyi Nan
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, 226001, Nantong, China
| | - Minhao Chen
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, 226001, Nantong, China
| | - Weijie Wu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, 226001, Nantong, China
- Department of Orthopaedics, Affiliated Nantong Hospital of Shanghai University, The Sixth People's Hospital of Nantong, 226001, Nantong, China
| | - Rongrong Huang
- Department of Pharmacy, Affiliated Hospital of Nantong University, 226001, Nantong, China
| | - Weiwei Sun
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, 226001, Nantong, China
| | - Qian Lu
- Department of Rheumatology, Affiliated Hospital of Nantong University, 226001, Nantong, China
| | - Zhifeng Gu
- Department of Rheumatology, Affiliated Hospital of Nantong University, 226001, Nantong, China
| | - Xingxing Mao
- Department of Orthopaedics, Affiliated Nantong Hospital of Shanghai University, The Sixth People's Hospital of Nantong, 226001, Nantong, China.
| | - Hua Xu
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, 226001, Nantong, China.
| | - Youhua Wang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, 226001, Nantong, China.
| |
Collapse
|
4
|
Sherri N, Assaf R, Bitar ER, Znait S, Borghol AH, Kassem A, Rahal EA. Epstein-Barr Virus DNA Exacerbates Arthritis in a Mouse Model via Toll-like Receptor 9. Int J Mol Sci 2024; 25:4661. [PMID: 38731877 PMCID: PMC11083462 DOI: 10.3390/ijms25094661] [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: 03/28/2024] [Revised: 04/20/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Epstein-Barr virus (EBV) DNA is known to be shed upon reactivation of latent EBV. Based on our previous findings linking Toll-like receptor-9 (TLR9) to an EBV DNA-driven surge in IL-17A production, we aimed to examine the therapeutic potential of TLR9 inhibition in EBV DNA-exacerbated arthritis in a collagen-induced arthritis (CIA) mouse model. C57BL/6J mice were administered either collagen, EBV DNA + collagen, EBV DNA + collagen + TLR9 inhibitor, or only the TLR9 inhibitor. After 70 days, paw thicknesses, clinical scores, and gripping strength were recorded. Moreover, affected joints, footpads, and colons were histologically scored. Furthermore, the number of cells co-expressing IL-17A, IFN-γ, and FOXP3 in joint sections was determined by immunofluorescence assays. Significantly decreased paw thicknesses, clinical scores, and histological scores with a significantly increased gripping strength were observed in the group receiving EBV DNA + collagen + TLR9 inhibitor, compared to those receiving EBV DNA + collagen. Similarly, this group showed decreased IL-17A+ IFN-γ+, IL-17A+ FOXP3+, and IL-17A+ IFN-γ+ FOXP3+ foci counts in joints. We show that inhibiting TLR9 limits the exacerbation of arthritis induced by EBV DNA in a CIA mouse model, suggesting that TLR9 could be a potential therapeutic target for rheumatoid arthritis management in EBV-infected individuals.
Collapse
MESH Headings
- Animals
- Mice
- Arthritis, Experimental/virology
- Arthritis, Experimental/pathology
- Arthritis, Experimental/metabolism
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Arthritis, Rheumatoid/virology
- Disease Models, Animal
- DNA, Viral/genetics
- Epstein-Barr Virus Infections/virology
- Epstein-Barr Virus Infections/complications
- Epstein-Barr Virus Infections/pathology
- Herpesvirus 4, Human/physiology
- Interleukin-17/metabolism
- Mice, Inbred C57BL
- Toll-Like Receptor 9/metabolism
Collapse
Affiliation(s)
- Nour Sherri
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107, Lebanon; (N.S.); (R.A.); (E.R.B.); (S.Z.); (A.H.B.); (A.K.)
| | - Rayan Assaf
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107, Lebanon; (N.S.); (R.A.); (E.R.B.); (S.Z.); (A.H.B.); (A.K.)
| | - Elio R. Bitar
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107, Lebanon; (N.S.); (R.A.); (E.R.B.); (S.Z.); (A.H.B.); (A.K.)
| | - Sabah Znait
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107, Lebanon; (N.S.); (R.A.); (E.R.B.); (S.Z.); (A.H.B.); (A.K.)
| | - Abdul Hamid Borghol
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107, Lebanon; (N.S.); (R.A.); (E.R.B.); (S.Z.); (A.H.B.); (A.K.)
| | - Aya Kassem
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107, Lebanon; (N.S.); (R.A.); (E.R.B.); (S.Z.); (A.H.B.); (A.K.)
| | - Elias A. Rahal
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut 1107, Lebanon; (N.S.); (R.A.); (E.R.B.); (S.Z.); (A.H.B.); (A.K.)
- Center for Infectious Diseases Research (CIDR), American University of Beirut, Beirut 1107, Lebanon
| |
Collapse
|
5
|
da Silva PR, Apolinário NDM, da Silva SÂS, Araruna MEC, Costa TB, e Silva YMSDM, da Silva TG, de Moura RO, dos Santos VL. Anti-Inflammatory Activity of N'-(3-(1H-indol-3-yl)benzylidene)-2-cyanoacetohydrazide Derivative via sGC-NO/Cytokine Pathway. Pharmaceuticals (Basel) 2023; 16:1415. [PMID: 37895886 PMCID: PMC10610422 DOI: 10.3390/ph16101415] [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: 08/17/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
The N-acylhydrazone function has been reported as a pharmacophore group of molecules with diverse pharmacological activities, including anti-inflammatory effects. Therefore, this study was designed to evaluate the anti-inflammatory potential of the compound N'-(3-(1H-indol-3-yl)benzylidene)-2-cyanoacetohydrazide (JR19) in vivo. The study started with the carrageenan-induced peritonitis model, followed by an investigation of leukocyte migration using the subcutaneous air pouch test and an assessment of the antinociceptive profile using formalin-induced pain. A preliminary molecular docking study focusing on the crystallographic structures of NFκB, iNOS, and sGC was performed to determine the likely mechanism of action. The computational study revealed satisfactory interaction energies with the selected targets, and the same peritonitis model was used to validate the involvement of the nitric oxide pathway and cytokine expression in the peritoneal exudate of mice pretreated with L-NAME or methylene blue. In the peritonitis assay, JR19 (10 and 20 mg/kg) reduced leukocyte migration by 59% and 52%, respectively, compared to the vehicle group, with the 10 mg/kg dose used in subsequent assays. In the subcutaneous air pouch assay, the reduction in cell migration was 66%, and the response to intraplantar formalin was reduced by 39%, particularly during the inflammatory phase, suggesting that the compound lacks central analgesic activity. In addition, a reversal of the anti-inflammatory effect was observed in mice pretreated with L-NAME or methylene blue, indicating the involvement of iNOS and sGC in the anti-inflammatory response of JR19. The compound effectively and significantly decreased the levels of IL-6, TNF-α, IL-17, and IFN-γ, and this effect was reversed in animals pretreated with L-NAME, supporting a NO-dependent anti-inflammatory effect. In contrast, pretreatment with methylene blue only reversed the reduction in TNF-α levels. Therefore, these results demonstrate the pharmacological potential of the novel N-acylhydrazone derivative, which acts through the nitric oxide pathway and cytokine signaling, making it a strong candidate as an anti-inflammatory and immunomodulatory agent.
Collapse
Affiliation(s)
- Pablo Rayff da Silva
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
- Laboratório de Desenvolvimento e Síntese de Fármacos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| | - Nadjaele de Melo Apolinário
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| | - Simone Ângela Soares da Silva
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| | - Maria Elaine Cristina Araruna
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| | - Thássia Borges Costa
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| | - Yvnni M. S. de Medeiros e Silva
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Desenvolvimento e Síntese de Fármacos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| | - Teresinha Gonçalves da Silva
- Departamento de Antibióticos, Centro de Biociências, Universidade Federal de Pernambuco, Recife 50740-520, PE, Brazil;
| | - Ricardo Olímpio de Moura
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Desenvolvimento e Síntese de Fármacos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| | - Vanda Lucia dos Santos
- Programa de Pós Graduação em Ciências Farmacêuticas, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil; (P.R.d.S.); (N.d.M.A.); (S.Â.S.d.S.); (M.E.C.A.); (T.B.C.); (Y.M.S.d.M.e.S.); (V.L.d.S.)
- Laboratório de Ensaios Farmacológicos, Departamento de Farmácia, Universidade Estadual da Paraíba, Campina Grande 58429-500, PB, Brazil
| |
Collapse
|
6
|
Reyes JM, Gutierrez MV, Madariaga H, Otero W, Guzman R, Izquierdo J, Abello M, Velez P, Castillo D, Ponce de Leon D, Lukic T, Amador L. Patient-reported outcomes in RA patients treated with tofacitinib or bDMARDs in real-life conditions in two Latin American countries. REUMATOLOGIA CLINICA 2023; 19:319-327. [PMID: 37286268 DOI: 10.1016/j.reumae.2023.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 02/03/2023] [Indexed: 06/09/2023]
Abstract
OBJECTIVE To describe efficacy, safety, and patient-reported outcomes (PROs) in patients with rheumatoid arthritis (RA) with an inadequate response to conventional synthetic disease-modifying antirheumatic drugs (csDMARDs) treated with tofacitinib or biological DMARDs (bDMARDs) in real-life conditions. METHODS A noninterventional study was performed between March 2017 and September 2019 at 13 sites in Colombia and Peru. Outcomes measured at baseline and at the 6-month follow-up were disease activity (RAPID3 [Routine Assessment of Patients Index Data] score), functional status (HAQ-DI [Health Assessment Questionnaire] score), and quality of life (EQ-5D-3L [EuroQol Questionnaire]). The Disease Activity Score-28 (DAS28-ESR) and frequency of adverse events (AEs) were also reported. Unadjusted and adjusted differences from baseline were estimated and expressed as the least squares mean difference (LSMD). RESULTS Data from 100 patients treated with tofacitinib and 70 patients with bDMARDs were collected. At baseline, the patients' mean age was 53.53 years (SD 13.77), the mean disease duration was 6.31 years (SD 7.01). The change from baseline at month 6 was not statistically significant different in the adjusted LSMD [SD] for tofacitinib vs. bDMARDs for RAPID3 score (-2.55[.30] vs. -2.52[.26]), HAQ-DI score (-.56[.07] vs. -.50[.08]), EQ-5D-3L score (.39[.04] vs. .37[.04]) and DAS28-ESR (-2.37[.22] vs. -2.77[.20]). Patients from both groups presented similar proportions of nonserious and serious AEs. No deaths were reported. CONCLUSION Changes from baseline were not statistically significantly different between tofacitinib and bDMARDs in terms of RAPID3 scores and secondary outcomes. Patients from both groups presented similar proportions of nonserious and serious AEs. CLINICAL TRIAL NUMBER NCT03073109.
Collapse
Affiliation(s)
| | | | - H Madariaga
- Centro Especializado de enfermedades neoplásicas (CEEN), Arequipa, Peru
| | - W Otero
- Centro Servimed, Bucaramanga, Colombia
| | - R Guzman
- Instituto de Enfermedades Autoinmunes Renato Guzmán (IDEARG), Bogota, Colombia
| | | | - M Abello
- Centro Integral de Reumatología Circaribe, Barranquilla, Colombia
| | - P Velez
- Centro de Investigación en Reumatología y Especialidades Médicas (CIREEM), Bogota, Colombia
| | | | | | | | | |
Collapse
|
7
|
Zhang J, Zhang J, Lai R, Peng C, Guo Z, Wang C. Risk-associated single nucleotide polymorphisms of mitochondrial D-loop mediate imbalance of cytokines and redox in rheumatoid arthritis. Int J Rheum Dis 2023; 26:124-131. [PMID: 36253082 DOI: 10.1111/1756-185x.14465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/28/2022] [Accepted: 10/01/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND We have identified rheumatoid arthritis (RA) risk-associated single nucleotide polymorphisms (SNPs) in the mitochondrial displacement loop (D-loop) including the major alleles of nucleotides 195T/C, 16260C/T, and 16519C/T as well as the minor alleles of nucleotides 146T/C and 150C/T previously. OBJECTIVE We evaluated the potential relationships of these SNPs with status for oxidative stress and inflammation cytokines. METHODS The DNA was extracted from blood samples of RA patients, and the SNPs of DNA D-loop were verified by polymerase chain reaction amplification and sequence analysis. Serum levels of inflammatory cytokines including interferon-γ (IFN-γ), interleukin-2 (IL-2), IL-6, IL-10, and tumor necrosis factor-α (TNF-α) were determined by cytometric bead array. Plasma reactive oxygen species (ROS) levels were measured by fluorescent probe technology. RESULTS The RA risk-related allele 16519C was significantly associated with high IFN-γ levels (100.576 ± 11.769 vs 64.268 ± 8.199, 95% confidence interval [CI] -66.317 to -6.299, P = 0.018). This allele also associated with ROS at borderline statistics level (619.295 ± 36.687 vs 526.979 ± 25.896, 95% CI -186.145 to -1.513, P = 0.054). The subsequent analysis also showed that the ROS levels were positively correlated with IFN-γ levels (R = 0.291, P = 0.002). Further analysis showed that RA patients with high C-reactive protein levels displayed a higher ROS level (P = 0.001). CONCLUSION Our results imply that the 16519C allele of the mtDNA D-loop might promote ROS and IFN-γ levels by altering the replication and transcription of mtDNA, thereby modifying RA development. REMARK The potential relationships of RA-associated SNPs in the mitochondrial D-loop with status for oxidative stress and inflammation were evaluated. The 16519C allele of the mtDNA D-loop might promote ROS and IFN-γ levels by altering the replication and transcription of mtDNA to modify RA development.
Collapse
Affiliation(s)
- Jingnan Zhang
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jingjing Zhang
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ruixue Lai
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chenxing Peng
- Department of Immunology and Rheumatology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhanjun Guo
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Cuiju Wang
- Department of Gynecology Ultrasound, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
8
|
Wang S, Zhou Y, Huang J, Li H, Pang H, Niu D, Li G, Wang F, Zhou Z, Liu Z. Advances in experimental models of rheumatoid arthritis. Eur J Immunol 2023; 53:e2249962. [PMID: 36330559 DOI: 10.1002/eji.202249962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 10/16/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by persistent articular inflammation and joint damage. RA was first described over 200 years ago; however, its etiology and pathophysiology remain insufficiently understood. The current treatment of RA is mainly empirical or based on the current understanding of etiology with limited efficacy and/or substantial side effects. Thus, the development of safer and more potent therapeutics, validated and optimized in experimental models, is urgently required. To improve the transition from bench to bedside, researchers must carefully select the appropriate experimental models as well as draw the right conclusions. Here, we summarize the establishment, pathological features, potential mechanisms, advantages, and limitations of the currently available RA models. The aim of the review is to help researchers better understand available RA models; discuss future trends in RA model development, which can help highlight new translational and human-based avenues in RA research.
Collapse
Affiliation(s)
- Siwei Wang
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China.,Honghu Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangtze University, Honghu, Hubei Province, China
| | - Yanhua Zhou
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China.,Honghu Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangtze University, Honghu, Hubei Province, China
| | - Jiangrong Huang
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Huilin Li
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Huidan Pang
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Dandan Niu
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Guangyao Li
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Fei Wang
- Department of Experiment and Training, Hubei College of Chinese Medicine, Hubei Province, China
| | - Zushan Zhou
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China.,Honghu Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangtze University, Honghu, Hubei Province, China
| | - Zhenzhen Liu
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| |
Collapse
|
9
|
Šteigerová M, Šíma M, Slanař O. Pathogenesis of Collagen-Induced Arthritis: Role of Immune Cells with Associated Cytokines and Antibodies, Comparison with Rheumatoid Arthritis. Folia Biol (Praha) 2023; 69:41-49. [PMID: 38063000 DOI: 10.14712/fb2023069020041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Collagen-induced arthritis is the most com-mon in vivo model of rheumatoid arthritis used for investigation of new potential therapies in preclinical research. Rheumatoid arthritis is a systemic inflammatory and autoimmune disease affecting joints, accompanied by significant extra-articular symptoms. The pathogenesis of rheumatoid arthritis and collagen-induced arthritis involves a so far properly unexplored network of immune cells, cytokines, antibodies and other factors. These agents trigger the autoimmune response leading to polyarthritis with cell infiltration, bone and cartilage degeneration and synovial cell proliferation. Our review covers the knowledge about cytokines present in the rat collagen-induced arthritis model and the factors affecting them. In addition, we provide a comparison with rheumatoid arthritis and a description of their important effects on the development of both diseases. We discuss the crucial roles of various immune cells (subtypes of T and B lymphocytes, dendritic cells, monocytes, macrophages), fibroblast-like synoviocy-tes, and their related cytokines (TNF-α, IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12, IL-17, IL-23, GM-CSF, TGF-β). Finally, we also focus on key antibodies (rheu-matoid factor, anti-citrullinated protein antibodies, anti-collagen II antibodies) and tissue-degrading enzymes (matrix metalloproteinases).
Collapse
Affiliation(s)
- Monika Šteigerová
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic.
| | - Martin Šíma
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - Ondřej Slanař
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| |
Collapse
|
10
|
Sakyi SA, Owusu‐Yeboah M, Obirikorang C, Dadzie Ephraim RK, Kwarteng A, Opoku S, Afranie BO, Senu E, Boateng AO, Boakye DK, Buckman TA, Amoani B. Profiling vitamin D, its mediators and proinflammatory cytokines in rheumatoid arthritis: A case-control study. Immun Inflamm Dis 2022; 10:e676. [PMID: 35894711 PMCID: PMC9274797 DOI: 10.1002/iid3.676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION The active form of vitamin D has immunomodulatory and anti-inflammatory effect. Vitamin D is implicated in pathogenesis of rheumatoid arthritis (RA) and its deficiency leads to increased inflammation. Moreover, its production is dependent on concentration of calcium, phosphorus, and parathyroid hormone (PTH). Cytokines mediates inflammation in RA synovium. This study evaluated vitamin D, its mediators and proinflammatory cytokines among RA patients. METHODS In a case-control study, 78 RA patients from Komfo Anokye Teaching Hospital rheumatology clinic and 60 healthy blood donors were recruited. Chemistry analyzer and enzyme-linked immunosorbent assay kits were used to measure biochemical parameters and cytokines. RESULTS We found significantly higher levels of interleukin (IL)-1β, interferon gamma (IFN-γ), and tumor necrosis factor-α (TNF-α) in RA patients compared with controls (p < .05). There was a significant positive correlation between intact parathyroid hormone (iPTH) and IL-10 (r = .30, p < .05) and a negative correlation between IL-6 (r = -0.28, p > .05), IL-1β (r = -0.25, p > .05), TNF-α (r = -0.26, p > .05), IFN-γ (r = -0.24, p > .05), and iPTH. There was a significant negative correlation between IL-1β (r = -0.33, p < .05), IFN- γ (r = -0.29, p < .05), and calcium. CONCLUSION Reduced PTH, calcium, and phosphorus is associated with higher levels of proinflammatory cytokines which may worsen RA disease condition. Vitamin D is therefore not an independent regulator of proinflammatory cytokines in RA.
Collapse
Affiliation(s)
- Samuel A. Sakyi
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Mavis Owusu‐Yeboah
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Christian Obirikorang
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Richard K. Dadzie Ephraim
- Department of Medical Laboratory Sciences, Faculty of Allied HealthUniversity of Cape CoastCape CoastGhana
| | - Alexander Kwarteng
- Department of Biochemistry and BiotechnologyKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Stephen Opoku
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, College of Health SciencesKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Bright O. Afranie
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Ebenezer Senu
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Andy O. Boateng
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Derrick K. Boakye
- Department of Medical Diagnostics, Faculty of Allied Health Sciences, College of Health SciencesKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Tonnies A. Buckman
- Department of Molecular Medicine, School of Medicine and DentistryKwame Nkrumah University of Science and TechnologyKumasiGhana
| | - Benjamin Amoani
- Department of Biomedical Science, School of Allied Health SciencesUniversity of Cape CoastCape CoastGhana
| |
Collapse
|
11
|
Poole JA, Mikuls TR, Thiele GM, Gaurav R, Nelson AJ, Duryee MJ, Mitra A, Hunter C, Wyatt TA, England BR, Ascherman DP. Increased susceptibility to organic dust exposure-induced inflammatory lung disease with enhanced rheumatoid arthritis-associated autoantigen expression in HLA-DR4 transgenic mice. Respir Res 2022; 23:160. [PMID: 35717175 PMCID: PMC9206339 DOI: 10.1186/s12931-022-02085-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 06/13/2022] [Indexed: 11/10/2022] Open
Abstract
Immunogenetic as well as environmental and occupational exposures have been linked to the development of rheumatoid arthritis (RA), RA-associated lung disease, and other primary lung disorders. Importantly, various inhalants can trigger post-translational protein modifications, resulting in lung autoantigen expression capable of stimulating pro-inflammatory and/or pro-fibrotic immune responses. To further elucidate gene-environment interactions contributing to pathologic lung inflammation, we exploited an established model of organic dust extract (ODE) exposure with and without collagen-induced arthritis (CIA) in C57BL/6 wild type (WT) versus HLA-DR4 transgenic mice. ODE-induced airway infiltration driven by neutrophils was significantly increased in DR4 versus WT mice, with corresponding increases in bronchoalveolar lavage fluid (BALF) levels of TNF-⍺, IL-6, and IL-33. Lung histopathology demonstrated increased number of ectopic lymphoid aggregates comprised of T and B cells following ODE exposure in DR4 mice. ODE also induced citrullination, malondialdehyde acetaldehyde (MAA) modification, and vimentin expression that co-localized with MAA and was enhanced in DR4 mice. Serum and BALF anti-MAA antibodies were strikingly increased in ODE-treated DR4 mice. Coupling ODE exposure with Type II collagen immunization (CIA) resulted in similarly augmented pro-inflammatory lung profiles in DR4 mice (relative to WT mice) that was accompanied by a profound increase in infiltrating lung CD4+ and CD8+ T cells as well as CD19+CD11b+ autoimmune B cells. Neither modeling strategy induced significant arthritis. These findings support a model in which environmental insults trigger enhanced post-translational protein modification and lung inflammation sharing immunopathological features with RA-associated lung disease in the selected immunogenetic background of HLA-DR4 mice.
Collapse
Affiliation(s)
- Jill A Poole
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Ted R Mikuls
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Research Service, Omaha, NE, USA
| | - Geoffrey M Thiele
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Research Service, Omaha, NE, USA
| | - Rohit Gaurav
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Amy J Nelson
- Veterans Affairs Nebraska-Western Iowa Health Care System, Research Service, Omaha, NE, USA
| | - Michael J Duryee
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Research Service, Omaha, NE, USA
| | - Ananya Mitra
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Carlos Hunter
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Todd A Wyatt
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Research Service, Omaha, NE, USA
- Department of Environmental, Agricultural & Occupational Health, College of Public Health, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bryant R England
- Department of Internal Medicine, College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Veterans Affairs Nebraska-Western Iowa Health Care System, Research Service, Omaha, NE, USA
| | - Dana P Ascherman
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
12
|
Jonsson AH, Zhang F, Dunlap G, Gomez-Rivas E, Watts GFM, Faust HJ, Rupani KV, Mears JR, Meednu N, Wang R, Keras G, Coblyn JS, Massarotti EM, Todd DJ, Anolik JH, McDavid A, Wei K, Rao DA, Raychaudhuri S, Brenner MB. Granzyme K + CD8 T cells form a core population in inflamed human tissue. Sci Transl Med 2022; 14:eabo0686. [PMID: 35704599 PMCID: PMC9972878 DOI: 10.1126/scitranslmed.abo0686] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
T cell-derived pro-inflammatory cytokines are a major driver of rheumatoid arthritis (RA) pathogenesis. Although these cytokines have traditionally been attributed to CD4 T cells, we have found that CD8 T cells are notably abundant in synovium and make more interferon (IFN)-γ and nearly as much tumor necrosis factor (TNF) as their CD4 T cell counterparts. Furthermore, using unbiased high-dimensional single-cell RNA-seq and flow cytometric data, we found that the vast majority of synovial tissue and synovial fluid CD8 T cells belong to an effector CD8 T cell population characterized by high expression of granzyme K (GzmK) and low expression of granzyme B (GzmB) and perforin. Functional experiments demonstrate that these GzmK+ GzmB+ CD8 T cells are major cytokine producers with low cytotoxic potential. Using T cell receptor repertoire data, we found that CD8 GzmK+ GzmB+ T cells are clonally expanded in synovial tissues and maintain their granzyme expression and overall cell state in blood, suggesting that they are enriched in tissue but also circulate. Using GzmK and GzmB signatures, we found that GzmK-expressing CD8 T cells were also the major CD8 T cell population in the gut, kidney, and coronavirus disease 2019 (COVID-19) bronchoalveolar lavage fluid, suggesting that they form a core population of tissue-associated T cells across diseases and human tissues. We term this population tissue-enriched expressing GzmK or TteK CD8 cells. Armed to produce cytokines in response to both antigen-dependent and antigen-independent stimuli, CD8 TteK cells have the potential to drive inflammation.
Collapse
Affiliation(s)
- A. Helena Jonsson
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Fan Zhang
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
- Center for Data Sciences, Brigham and Women’s Hospital; Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital; Boston, MA 02115, USA
- Department of Biomedical Informatics, Harvard Medical School; Boston, MA 02115, USA
- Broad Institute of MIT and Harvard; Cambridge, MA 02142, USA
- Division of Rheumatology and the Center for Health Artificial Intelligence, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Garrett Dunlap
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Emma Gomez-Rivas
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Gerald F. M. Watts
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Heather J. Faust
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Karishma Vijay Rupani
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Joseph R. Mears
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
- Center for Data Sciences, Brigham and Women’s Hospital; Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital; Boston, MA 02115, USA
- Department of Biomedical Informatics, Harvard Medical School; Boston, MA 02115, USA
- Broad Institute of MIT and Harvard; Cambridge, MA 02142, USA
| | - Nida Meednu
- Division of Rheumatology and the Center for Health Artificial Intelligence, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Runci Wang
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Gregory Keras
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Jonathan S. Coblyn
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Elena M. Massarotti
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Derrick J. Todd
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Jennifer H. Anolik
- Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center; Rochester, NY 14642, USA
| | - Andrew McDavid
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry; Rochester, NY 14642, USA
| | | | - Kevin Wei
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Deepak A. Rao
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| | - Soumya Raychaudhuri
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
- Center for Data Sciences, Brigham and Women’s Hospital; Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital; Boston, MA 02115, USA
- Department of Biomedical Informatics, Harvard Medical School; Boston, MA 02115, USA
- Broad Institute of MIT and Harvard; Cambridge, MA 02142, USA
- Arthritis Research UK Centre for Genetics and Genomics, Centre for Musculoskeletal Research, The University of Manchester; Manchester M13 9PT, UK
| | - Michael B. Brenner
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women’s Hospital and Harvard Medical School; Boston, MA 02115, USA
| |
Collapse
|
13
|
Kim YJ, Lee JY, Yang MJ, Cho HJ, Kim MY, Kim L, Hwang JH. Therapeutic effect of intra-articular injected 3'-sialyllactose on a minipig model of rheumatoid arthritis induced by collagen. Lab Anim Res 2022; 38:8. [PMID: 35314005 PMCID: PMC8939226 DOI: 10.1186/s42826-022-00119-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/16/2022] [Indexed: 12/03/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic inflammatory disease of joint, but there is no known cure. 3′-sialyllactose (3′-SL) is an oligosaccharide that is abundant in breast milk of mammals, and has anti-inflammatory properties. However, the efficacy of 3′-SL on RA remains unclear. The objective of the present study was to evaluate the therapeutic effect of 3′-SL after it was directly injected into the knee joint cavity of a RA minipig model.
Results Minipig RA model was induced by intra-articular injection of bovine type II collagen emulsified with complete or incomplete Freund’s adjuvant into left knee joint. In clinical assessment, lameness and swelling of the hindlimb and increased knee joint width were observed in all animals. After the onset of arthritis, 3′-SL (0, 2, 10, and 50 mg/kg) was directly administered to the left knee joint cavity once a week for 4 weeks. Compared to the vehicle control group, no significant difference in macroscopic observation of the synovial pathology or the expression of inflammation-related genes (IL-1β, TNF-α, and COX2) in the synovial membrane of the knee joint was found. In microscopic observation, cell cloning of the articular cartilage was significantly reduced in proportion to the concentration of 3′-SL administered.
Conclusions Our results suggest that intra-articular injected 3′-SL had a therapeutic effect on collagen-induced arthritis at the cellular level with potential as a medication for RA. Supplementary Information The online version contains supplementary material available at 10.1186/s42826-022-00119-2.
Collapse
Affiliation(s)
- Young June Kim
- Animal Model Research Group, Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeollabukdo, 56212, Republic of Korea
| | - Ju-Young Lee
- Animal Model Research Group, Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeollabukdo, 56212, Republic of Korea
| | - Mi-Jin Yang
- Pathology Research Group, Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeollabukdo, 56212, Republic of Korea
| | - Hyun Jin Cho
- Animal Model Research Group, Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeollabukdo, 56212, Republic of Korea
| | - Min-Young Kim
- GeneChem Inc. A-501, 187 Techno 2-ro, Yuseong-gu, Daejeon, 34025, Republic of Korea
| | - Lila Kim
- GeneChem Inc. A-501, 187 Techno 2-ro, Yuseong-gu, Daejeon, 34025, Republic of Korea
| | - Jeong Ho Hwang
- Animal Model Research Group, Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeollabukdo, 56212, Republic of Korea.
| |
Collapse
|
14
|
McCall JL, Blair HC, Blethen KE, Hall C, Elliott M, Barnett JB. Prenatal cadmium exposure does not induce greater incidence or earlier onset of autoimmunity in the offspring. PLoS One 2021; 16:e0249442. [PMID: 34478449 PMCID: PMC8415597 DOI: 10.1371/journal.pone.0249442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/23/2021] [Indexed: 11/30/2022] Open
Abstract
We previously demonstrated that exposure of adult mice to environmental levels of cadmium (Cd) alters immune cell development and function with increases in anti-streptococcal antibody levels, as well as decreases in splenic natural regulatory T cells (nTreg) in the adult female offspring. Based on these data, we hypothesized that prenatal Cd exposure could predispose an individual to developing autoimmunity as adults. To test this hypothesis, the effects of prenatal Cd on the development of autoimmune diabetes and arthritis were investigated. Non-obese diabetic (NOD) mice were exposed to Cd in a manner identical to our previous studies, and the onset of diabetes was assessed in the offspring. Our results showed a similar time-to-onset and severity of disease to historical data, and there were no statistical differences between Cd-exposed and control offspring. Numerous other immune parameters were measured and none of these parameters showed biologically-relevant differences between Cd-exposed and control animals. To test whether prenatal Cd-exposure affected development of autoimmune arthritis, we used SKG mice. While the levels of arthritis were similar between Cd-exposed and control offspring of both sexes, the pathology of arthritis determined by micro-computed tomography (μCT) between Cd-exposed and control animals, showed some statistically different values, especially in the female offspring. However, the differences were small and thus, the biological significance of these changes is open to speculation. Overall, based on the results from two autoimmune models, we conclude that prenatal exposure to Cd did not lead to a measurable propensity to develop autoimmune disease later in life.
Collapse
Affiliation(s)
- Jamie L. McCall
- Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States of America
| | - Harry C. Blair
- Department of Pathology, Pittsburgh VA Medical Center, Pittsburgh, PA, United States of America
- Department of Cell Biology, the and the University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kathryn E. Blethen
- Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States of America
| | - Casey Hall
- Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States of America
| | - Meenal Elliott
- Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States of America
| | - John B. Barnett
- Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States of America
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, United States of America
| |
Collapse
|
15
|
Abstract
Inflammatory arthritis (IA) is a common disease that affects millions of individuals worldwide. Proinflammatory events during IA pathogenesis are well studied; however, loss of protective immunity remains underexplored. Earlier, we reported that 14-3-3zeta (ζ) has a role in T-cell polarization and interleukin (IL)-17A signal transduction. Here, we demonstrate that 14-3-3ζ knockout (KO) rats develop early-onset severe arthritis in two independent models of IA, pristane-induced arthritis and collagen-induced arthritis. Arthritic 14-3-3ζ KO animals showed an increase in bone loss and immune cell infiltration in synovial joints. Induction of arthritis coincided with the loss of anti-14-3-3ζ antibodies; however, rescue experiments to supplement the 14-3-3ζ antibody by passive immunization did not suppress arthritis. Instead, 14-3-3ζ immunization during the presymptomatic phase resulted in significant suppression of arthritis in both wild-type and 14-3-3ζ KO animals. Mechanistically, 14-3-3ζ KO rats exhibited elevated inflammatory gene signatures at the messenger RNA and protein levels, particularly for IL-1β. Furthermore, the immunization with recombinant 14-3-3ζ protein suppressed IL-1β levels, significantly increased anti-14-3-3ζ antibody levels and collagen production, and preserved bone quality. The 14-3-3ζ protein increased collagen expression in primary rat mesenchymal cells. Together, our findings indicate that 14-3-3ζ causes immune suppression and extracellular remodeling, which lead to a previously unrecognized IA-suppressive function.
Collapse
|
16
|
Applicability and implementation of the collagen-induced arthritis mouse model, including protocols (Review). Exp Ther Med 2021; 22:939. [PMID: 34335888 PMCID: PMC8290431 DOI: 10.3892/etm.2021.10371] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 03/30/2021] [Indexed: 12/16/2022] Open
Abstract
Animal models of rheumatoid arthritis (RA) are essential for studying the pathogenesis of RA in vivo and determining the efficacy of anti-RA drugs. During the past decades, numerous rodent models of arthritis have been evaluated as potential models and the modeling methods are relatively well-developed. Among these models, the collagen-induced arthritis (CIA) mouse model is the first choice and the most widely used because it may be generated rapidly and inexpensively and is relatively similar in pathogenesis to human RA. To date, there have been numerous classic studies and reviews discussing related pathogeneses and modeling methods. Based on this knowledge, combined with the latest convenient and effective methods for CIA model construction, the present review aims to introduce the model to beginners and clarify important details regarding its use. Information on the origin and pathogenesis of the CIA model, the protocol for establishing it, the rate of successful arthritis induction and the methods used to evaluate the severity of arthritis are briefly summarized. With this information, it is expected that researchers who have recently entered the field or are not familiar with this information will be able to start quickly, avoid unnecessary errors and obtain reliable results.
Collapse
|
17
|
Dankers W, den Braanker H, Paulissen SMJ, van Hamburg JP, Davelaar N, Colin EM, Lubberts E. The heterogeneous human memory CCR6+ T helper-17 populations differ in T-bet and cytokine expression but all activate synovial fibroblasts in an IFNγ-independent manner. Arthritis Res Ther 2021; 23:157. [PMID: 34082814 PMCID: PMC8173960 DOI: 10.1186/s13075-021-02532-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/18/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Chronic synovial inflammation is an important hallmark of inflammatory arthritis, but the cells and mechanisms involved are incompletely understood. Previously, we have shown that CCR6+ memory T-helper (memTh) cells and synovial fibroblasts (SF) activate each other in a pro-inflammatory feedforward loop, which potentially drives persistent synovial inflammation in inflammatory arthritis. However, the CCR6+ memTh cells are a heterogeneous population, containing Th17/Th22 and Th17.1 cells. Currently, it is unclear which of these subpopulations drive SF activation and how they should be targeted. In this study, we examined the individual contribution of these CCR6+ memTh subpopulations to SF activation and examined ways to regulate their function. METHODS Th17/Th22 (CXCR3-CCR4+), Th17.1 (CXCR3+CCR4-), DP (CXCR3+CCR4+), and DN (CXCR3-CCR4-) CCR6+ memTh, cells sorted from PBMC of healthy donors or treatment-naïve early rheumatoid arthritis (RA) patients, were cocultured with SF from RA patients with or without anti-IL17A, anti-IFNγ, or 1,25(OH)2D3. Cultures were analyzed by RT-PCR, ELISA, or flow cytometry. RESULTS Th17/Th22, Th17.1, DP, and DN cells equally express RORC but differ in production of TBX21 and cytokines like IL-17A and IFNγ. Despite these differences, all the individual CCR6+ memTh subpopulations, both from healthy individuals and RA patients, were more potent in activating SF than the classical Th1 cells. SF activation was partially inhibited by blocking IL-17A, but not by inhibiting IFNγ or TBX21. However, active vitamin D inhibited the pathogenicity of all subpopulations leading to suppression of SF activation. CONCLUSIONS Human CCR6+ memTh cells contain several subpopulations that equally express RORC but differ in TBX21, IFNγ, and IL-17A expression. All individual Th17 subpopulations are more potent in activating SF than classical Th1 cells in an IFNγ-independent manner. Furthermore, our data suggest that IL-17A is not dominant in this T cell-SF activation loop but that a multiple T cell cytokine inhibitor, such as 1,25(OH)2D3, is able to suppress CCR6+ memTh subpopulation-driven SF activation.
Collapse
Affiliation(s)
- Wendy Dankers
- Department of Rheumatology, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
- Current address: Rheumatology Research Group, Centre for Inflammatory Diseases, School of Clinical Sciences at Monash Health, Monash University, Clayton, Australia
| | - Hannah den Braanker
- Department of Rheumatology, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Sandra M J Paulissen
- Department of Rheumatology, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Jan Piet van Hamburg
- Department of Rheumatology, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
- Current address: Amsterdam Rheumatology and Immunology Center, Department of Clinical Immunology & Rheumatology and Laboratory for Experimental Immunology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | - Nadine Davelaar
- Department of Rheumatology, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Edgar M Colin
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Erik Lubberts
- Department of Rheumatology, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
- Department of Immunology, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
18
|
Malhotra H, Garg V, Singh G. Biomarker Approach Towards Rheumatoid Arthritis Treatment. Curr Rheumatol Rev 2021; 17:162-175. [PMID: 33327920 DOI: 10.2174/1573397116666201216164013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 10/02/2020] [Accepted: 10/19/2020] [Indexed: 01/08/2023]
Abstract
Rheumatoid arthritis is an auto-immune disorder, recognized by cartilage as well as bone destruction, which causes irreversible joint deformities, which further results in functional limitations in the patient. Genes like HLA-DRB1 and PTPN22 are likely implicated in the genetic predisposition of rheumatoid arthritis pathology. The first and foremost clinical manifestation in a person with rheumatoid arthritis is joint destruction followed by cartilage and bone destruction caused by cell-cell interactions. The cell-cell interactions are thought to be initialized through the contact of antigen-presenting cells (APC) with CD4+ cells, leading to the progression of the disease. APC includes a complex of class ІІ major histocompatibility complex molecules along with peptide antigens and binds to the receptors present on the surface of T-cells. Further, the activation of macrophages is followed by the release of various pro-inflammatory cytokines such as IL-1 and TNF-α, which lead to the secretion of enzymes that degrade proteoglycan and collagen, which in turn, increase tissue degradation. Biomarkers like IL-6, IL-12, IL-8 and IL-18, 14-3-3η, RANKL, IFN-γ, IFN-β and TGF-β have been designated as key biomarkers in disease development and progression. The study of these biomarkers is very important as they act as a molecular indicator of pathological processes that aggravate the disease.
Collapse
Affiliation(s)
- Hitesh Malhotra
- Chandigarh College of Pharmacy Landran, Mohali, Punjab, India
| | - Vandana Garg
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| |
Collapse
|
19
|
Fadlallah S, Hussein H, Jallad MA, Shehab M, Jurjus AR, Matar GM, Rahal EA. Effect of Epstein-Barr Virus DNA on the Incidence and Severity of Arthritis in a Rheumatoid Arthritis Mouse Model. Front Immunol 2021; 12:672752. [PMID: 34040613 PMCID: PMC8141727 DOI: 10.3389/fimmu.2021.672752] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/22/2021] [Indexed: 01/02/2023] Open
Abstract
Objective We recently demonstrated that EBV DNA is correlated with proinflammatory responses in mice and in rheumatoid arthritis (RA) patients; hence, we utilized an RA mouse model to examine whether EBV DNA enhances the risk and severity of arthritis and to assess its immunomodulatory effects. Methods C57BL/6J mice were treated with collagen (arthritis-inducing agent), EBV DNA 6 days before collagen, EBV DNA 15 days after collagen, Staphylococcus epidermidis DNA 6 days before collagen, EBV DNA alone, or water. Mice were then monitored for clinical signs and affected joints/footpads were histologically analysed. The relative concentration of IgG anti- chicken collagen antibodies and serum cytokine levels of IL-17A and IFNϒ were determined by ELISA. The number of cells co-expressing IL-17A and IFNϒ in joint histological sections was determined by immunofluorescence. Results The incidence of arthritis was significantly higher in mice that received EBV DNA prior to collagen compared to mice that only received collagen. Similarly, increased clinical scores, histological scores and paw thicknesses with a decreased gripping strength were observed in groups treated with EBV DNA and collagen. The relative concentration of IgG anti-chicken collagen antibodies was significantly increased in the group that received EBV DNA 6 days prior to collagen in comparison to the collagen receiving group. On the other hand, the highest number of cells co-expressing IFNϒ and IL-17A was observed in joints from mice that received both collagen and EBV DNA. Conclusion EBV DNA increases the incidence and severity of arthritis in a RA mouse model. Targeting mediators triggered by viral DNA may hence be a potential therapeutic avenue.
Collapse
Affiliation(s)
- Sukayna Fadlallah
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Hadi Hussein
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Mary-Ann Jallad
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Marwa Shehab
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Abdo R Jurjus
- Department of Anatomy, Cell Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ghassan M Matar
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Elias A Rahal
- Department of Experimental Pathology, Immunology, and Microbiology, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
20
|
Montoya T, Sánchez-Hidalgo M, Castejón ML, Rosillo MÁ, González-Benjumea A, Alarcón-de-la-Lastra C. Dietary Oleocanthal Supplementation Prevents Inflammation and Oxidative Stress in Collagen-Induced Arthritis in Mice. Antioxidants (Basel) 2021; 10:antiox10050650. [PMID: 33922438 PMCID: PMC8145376 DOI: 10.3390/antiox10050650] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/16/2021] [Accepted: 04/21/2021] [Indexed: 02/07/2023] Open
Abstract
Oleocanthal (OLE), a characteristic and exclusive secoiridoid of Oleoaceae family, is mainly found in extra virgin olive oil (EVOO). Previous studies have reported its antioxidant, anti-inflammatory, antimicrobial, anticancer and neuroprotective effects. Since the pathogenesis of rheumatoid arthritis (RA) involves inflammatory and oxidative components, this study was designed to evaluate the preventive role of dietary OLE-supplemented effects in collagen-induced arthritis (CIA) murine model. Animals were fed with a preventive OLE-enriched dietary during 6 weeks previous to CIA induction and until the end of experiment time. At day 43 after first immunization, mice were sacrificed: blood was recollected and paws were histological and biochemically processed. Dietary OLE prevented bone, joint and cartilage rheumatic affections induced by collagen. Levels of circulatory matrix metalloproteinase (MMP)-3 and pro-inflammatory cytokines (IL-6, IL-1β, TNF-α, IL-17, IFN-γ) were significantly decreased in secoiridoid fed animals. Besides, dietary OLE was able to diminish COX-2, mPGES-1 and iNOS protein expressions and, also, PGE2 levels. The mechanisms underlying these protective effects could be related to Nrf-2/HO-1 axis activation and the inhibition of relevant signaling pathways including JAK-STAT, MAPKs and NF-κB, thus controlling the production of inflammatory and oxidative mediators. Overall, our results exhibit preliminary evidences about OLE, as a novel dietary tool for the prevention of autoimmune and inflammatory disorders, such as RA.
Collapse
Affiliation(s)
- Tatiana Montoya
- Department of Pharmacology, School of Pharmacy, Universidad de Sevilla, 41012 Seville, Spain; (T.M.); (M.S.-H.); (M.L.C.); (M.Á.R.)
| | - Marina Sánchez-Hidalgo
- Department of Pharmacology, School of Pharmacy, Universidad de Sevilla, 41012 Seville, Spain; (T.M.); (M.S.-H.); (M.L.C.); (M.Á.R.)
| | - María Luisa Castejón
- Department of Pharmacology, School of Pharmacy, Universidad de Sevilla, 41012 Seville, Spain; (T.M.); (M.S.-H.); (M.L.C.); (M.Á.R.)
| | - María Ángeles Rosillo
- Department of Pharmacology, School of Pharmacy, Universidad de Sevilla, 41012 Seville, Spain; (T.M.); (M.S.-H.); (M.L.C.); (M.Á.R.)
| | | | - Catalina Alarcón-de-la-Lastra
- Department of Pharmacology, School of Pharmacy, Universidad de Sevilla, 41012 Seville, Spain; (T.M.); (M.S.-H.); (M.L.C.); (M.Á.R.)
- Correspondence: ; Tel.: +34-95-455-9877
| |
Collapse
|
21
|
Xu S, Zhang T, Cao Z, Zhong W, Zhang C, Li H, Song J. Integrin-α9β1 as a Novel Therapeutic Target for Refractory Diseases: Recent Progress and Insights. Front Immunol 2021; 12:638400. [PMID: 33790909 PMCID: PMC8005531 DOI: 10.3389/fimmu.2021.638400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 02/26/2021] [Indexed: 12/12/2022] Open
Abstract
Integrins refer to heterodimers consisting of subunits α and β. They serve as receptors on cell membranes and interact with extracellular ligands to mediate intracellular molecular signals. One of the least-studied members of the integrin family is integrin-α9β1, which is widely distributed in various human tissues and organs. Integrin-α9β1 regulates the physiological state of cells through a variety of complex signaling pathways to participate in the specific pathological processes of some intractable diseases. In recent years, an increasing amount of research has focused on the role of α9β1 in the molecular mechanisms of different refractory diseases and its promising potential as a therapeutic target. Accordingly, this review introduces and summarizes recent research related to integrin-α9β1, describes the synergistic functions of α9β1 and its corresponding ligands in cancer, autoimmune diseases, nerve injury and thrombosis and, more importantly, highlights the potential of α9β1 as a distinctive target for the treatment of these intractable diseases.
Collapse
Affiliation(s)
- Shihan Xu
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Tingwei Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Zhengguo Cao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wenjie Zhong
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Chuangwei Zhang
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Han Li
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|
22
|
Hemshekhar M, Anaparti V, El-Gabalawy H, Mookherjee N. A bioavailable form of curcumin, in combination with vitamin-D- and omega-3-enriched diet, modifies disease onset and outcomes in a murine model of collagen-induced arthritis. Arthritis Res Ther 2021; 23:39. [PMID: 33494792 PMCID: PMC7836561 DOI: 10.1186/s13075-021-02423-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Curcumin (CUR), vitamin D3 (D3), and omega-3-fatty acids (O3FA) individually modulate inflammation and pain in arthritis. Although these supplements are widely used, their combinatorial effects have not been defined. In this study, we examined the effects of a D3 and O3FA (VO)-enriched diet in conjunction with a highly bioavailable form of CUR (Cureit/Acumin™) in a collagen-induced arthritis (CIA) murine model. METHODS Male DBA/1J mice were acclimatized to VO-enriched diet and challenged with bovine collagen II (CII). Bioavailable CUR was administered daily by oral gavage from the onset of CII challenge. Disease severity was determined by monitoring joint thickness and standardized clinical score. Cellular infiltration and cartilage degradation in the joints were assessed by histology, serum cytokines profiled by Meso Scale Discovery multiplex assay, and joint matrix metalloproteinases examined by western blots. RESULTS CUR by itself significantly decreased disease severity by ~ 60%. Administration of CUR in CIA mice taking a VO-enriched diet decreased disease severity by > 80% and maximally delayed disease onset and progression. Some of the disease-modifying effects was mediated by CUR alone, e.g., suppression of serum anti-collagen antibodies and decrease of cellular infiltration and MMP abundance in the joints of CIA mice. Although CUR alone suppressed inflammatory cytokines in serum of CIA mice, the combination of CUR and VO diet significantly enhanced the suppression (> 2-fold compared to CUR) of TNF, IFN-γ, and MCP-1, all known to be associated with RA pathogenesis. CONCLUSION This study provides proof-of-concept that the combination of bioavailable CUR, vitamin D3, and O3FA substantially delays the development and severity of CIA. These findings provide a rationale for systematically evaluating these widely available supplements in individuals at risk for developing future RA.
Collapse
Affiliation(s)
- Mahadevappa Hemshekhar
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB, Canada
| | - Vidyanand Anaparti
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB, Canada
| | - Hani El-Gabalawy
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB, Canada.,Division of Rheumatology, Department of Internal Medicine, University of Manitoba, Winnipeg, MB, Canada.,Department of Immunology, University of Manitoba, Winnipeg, MB, R3E3P4, Canada
| | - Neeloffer Mookherjee
- Manitoba Centre for Proteomics and Systems Biology, Department of Internal Medicine, University of Manitoba, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB, Canada. .,Department of Immunology, University of Manitoba, Winnipeg, MB, R3E3P4, Canada.
| |
Collapse
|
23
|
A New Peracetylated Oleuropein Derivative Ameliorates Joint Inflammation and Destruction in a Murine Collagen-Induced Arthritis Model via Activation of the Nrf-2/Ho-1 Antioxidant Pathway and Suppression of MAPKs and NF-κB Activation. Nutrients 2021; 13:nu13020311. [PMID: 33499113 PMCID: PMC7911327 DOI: 10.3390/nu13020311] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/14/2021] [Accepted: 01/15/2021] [Indexed: 02/06/2023] Open
Abstract
Oleuropein (OL), an olive tree secoiridoid and its peracetylated derivate (Per-OL) have exhibited several beneficial effects on LPS-stimulated macrophages and murine experimental systemic lupus erythematosus (SLE). This study was designed to evaluate dietary Per-OL in comparison with OL supplementation effects on collagen-induced arthritis (CIA) murine model. Three-weeks-old DBA-1/J male mice were fed from weaning with a standard commercial diet or experimental enriched-diets in 0.05 % (w/w) OL, 0.05% and 0.025% Per-OL. After six weeks of pre-treatment, arthritis was induced by bovine collagen type II by tail base injection (day 0) and on day 21, mice received a booster injection. Mice were sacrificed 42 days after the first immunization. Both Per-OL and OL diets significantly prevented histological damage and arthritic score development, although no statistically significant differences were observed between both compounds. Also, serum collagen oligomeric matrix protein (COMP), metalloprotease (MMP)-3 and pro-inflammatory cytokines levels were ameliorated in paws from secoiridoids fed animals. Mitogen-activated protein kinases (MAPK)s and nuclear transcription factor-kappa-B (NF-κB) activations were drastically down-regulated whereas nuclear factor E2-related factor 2 (Nrf2) and heme-oxygenase-1 (HO-1) protein expressions were up-regulated in those mice fed with OL and Per-OL diets. We conclude that both Per-OL and its parent compound, OL, supplements might provide a basis for developing a new dietary strategy for the prevention of rheumatoid arthritis.
Collapse
|
24
|
THH Relieves CIA Inflammation by Reducing Inflammatory-related Cytokines. Cell Biochem Biophys 2020; 78:367-374. [PMID: 32363523 DOI: 10.1007/s12013-020-00911-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/07/2020] [Indexed: 12/31/2022]
Abstract
Tripterygium hypoglaucum hutch (THH) is a plant of the genus tripterygium, which is also known as colquhounia, Gelsemiun elegan, and so on. It is mainly distributed in Yunnan, Guizhou, and Sichuan regions and other places in China. To study the immune mechanism of THH on related inflammatory cytokines in collagen II-induced arthritis (CIA) mice, healthy male C57BL/6 mice were used to model CIA mice. Mice received THH 420 mg/kg/day or the same amount of normal saline (NS) by gavage for 20 days. The thickness of the ankle joint in mice was observed, and the arthritis index was calculated. Related inflammatory cytokines were detected by real-time quantitative polymerase chain reaction and enzyme-linked immunosorbent assay. The results showed that after treatment with THH, the CIA mice had less swelling and destruction of the joints as well as decreased foot size and arthritis index. The mRNA and protein levels of TNF-α, IFN-γ, and IL-17A were lower in the THH-treated group than in the NS group (P < 0.05). In summary, THH has great significance in the treatment of CIA mice, including reduced related inflammatory cytokines expression level in both joint tissue and serum. The mechanism of THH in the treatment of CIA may be through the inhibition of the NF-kB-STAT3-IL-17 pathway, which also requires further experimental investigation.
Collapse
|
25
|
Kirpotina LN, Schepetkin IA, Hammaker D, Kuhs A, Khlebnikov AI, Quinn MT. Therapeutic Effects of Tryptanthrin and Tryptanthrin-6-Oxime in Models of Rheumatoid Arthritis. Front Pharmacol 2020; 11:1145. [PMID: 32792961 PMCID: PMC7394103 DOI: 10.3389/fphar.2020.01145] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/13/2020] [Indexed: 01/01/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease involving joint and bone damage that is mediated in part by proteases and cytokines produced by synovial macrophages and fibroblast-like synoviocytes (FLS). Although current biological therapeutic strategies for RA have been effective in many cases, new classes of therapeutics are needed. We investigated anti-inflammatory properties of the natural alkaloid tryptanthrin (TRYP) and its synthetic derivative tryptanthrin-6-oxime (TRYP-Ox). Both TRYP and TRYP-Ox inhibited matrix metalloproteinase (MMP)-3 gene expression in interleukin (IL)-1β-stimulated primary human FLS, as well as IL-1β–induced secretion of MMP-1/3 by FLS and synovial SW982 cells and IL-6 by FLS, SW982 cells, human umbilical vein endothelial cells (HUVECs), and monocytic THP-1 cells, although TRYP-Ox was generally more effective and had no cytotoxicity in vitro. Evaluation of the therapeutic potential of TRYP and TRYP-Ox in vivo in murine arthritis models showed that both compounds significantly attenuated the development of collagen-induced arthritis (CIA) and collagen-antibody–induced arthritis (CAIA), with comparable efficacy. Collagen II (CII)-specific antibody levels were similarly reduced in TRYP- and TRYP-Ox-treated CIA mice. TRYP and TRYP-Ox also suppressed proinflammatory cytokine production by lymph node cells from CIA mice, with TRYP-Ox being more effective in inhibiting IL-17A, granulocyte-macrophage colony-stimulating factor (GM-CSF), and receptor activator of nuclear factor-κB ligand (RANKL). Thus, even though TRYP-Ox generally had a better in vitro profile, possibly due to its ability to inhibit c-Jun N-terminal kinase (JNK), both TRYP and TRYP-Ox were equally effective in inhibiting the clinical symptoms and damage associated with RA. Overall, TRYP and/or TRYP-Ox may represent potential new directions for the pursuit of novel treatments for RA.
Collapse
Affiliation(s)
- Liliya N Kirpotina
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Igor A Schepetkin
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Deepa Hammaker
- Division of Rheumatology, Allergy, and Immunology, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Amanda Kuhs
- Division of Rheumatology, Allergy, and Immunology, School of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Andrei I Khlebnikov
- Kizhner Research Center, Tomsk Polytechnic University, Tomsk, Russia.,Research Institute of Biological Medicine, Altai State University, Barnaul, Russia
| | - Mark T Quinn
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| |
Collapse
|
26
|
Tansakul M, Thim-Uam A, Saethang T, Makjaroen J, Wongprom B, Pisitkun T, Pisitkun P. Deficiency of STING Promotes Collagen-Specific Antibody Production and B Cell Survival in Collagen-Induced Arthritis. Front Immunol 2020; 11:1101. [PMID: 32582187 PMCID: PMC7283782 DOI: 10.3389/fimmu.2020.01101] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 05/06/2020] [Indexed: 12/15/2022] Open
Abstract
The levels of interferon-alpha are high in the serum and synovial fluid of rheumatoid arthritis (RA) patients. Activation of the stimulator of type I interferon genes (STING) mediates the productions of type I interferon and promotes chronic inflammation. STING plays a significant role in autoimmune lupus mice. However, the function of STING in collagen-induced arthritis (CIA) model has never been described. This study aimed to test the function of STING in CIA. The Sting-deficient mice developed arthritis comparable to WT mice. The levels of anti-collagen antibody from Sting-deficient mice were significantly higher than the WT mice. The B cells derived from Sting-deficient mice showed better survival than WT mice in response to the B cell receptor (BCR) stimulation. Activation of STING also induced B cell death, especially in activated B cells. This study demonstrated that the inhibition of STING promotes anti-collagen antibodies and B cell survival, which suggested that STING acts as a negative regulator of B cell function in the CIA model.
Collapse
Affiliation(s)
- Mookmanee Tansakul
- Section for Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Arthid Thim-Uam
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Thammakorn Saethang
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jiradej Makjaroen
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Benjawan Wongprom
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Trairak Pisitkun
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Prapaporn Pisitkun
- Section for Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand.,Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
27
|
Yaekura A, Yoshida K, Morii K, Oketani Y, Okumura I, Kaneshiro K, Shibanuma N, Sakai Y, Hashiramoto A. Chronotherapy targeting cytokine secretion attenuates collagen-induced arthritis in mice. Int Immunopharmacol 2020; 84:106549. [PMID: 32416449 DOI: 10.1016/j.intimp.2020.106549] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Diurnal variation of symptoms are observed in rheumatoid arthritis, especially in productions of cytokines that show peak concentrations during mid night. In contrast, cytokines of collagen-induced arthritis (CIA) mice increase in daytimes under Mid-light condition. By using chronotherapy, differences in drug efficacies according to administration time of Baricitinib, a wide ranged cytokine blocker, were examined in CIA mice. METHODS CIA mice were administered a dose of 3 mg/kg of Baricitinib once a day at zeitgeber time (ZT) 0 or ZT12 for 21 days. Arthritis scores, histopathology and factors related to joint destruction in sera were examined. Phosphorylation of STAT3 in liver, expressions of cytokines in spleen, and Interleukin (IL)-6 and tumor necrosis factor (TNF)-α in sera were measured. RESULTS In CIA mice, diurnal variations were observed both in expressions of cytokines and phosphorylation of STAT3. Arthritis scores of ZT0/12 group decreased from day3 as compared to untreated mice, and those of ZT0 group significantly decreased as compared to ZT12 group from day12. Pathological findings, immunohistochemistry of cytokines and Receptor activator of nuclear factor kappa-Β ligand (RANKL)/osteoprotegerin ratio in sera well reflected results of arthritis scores. Diurnal variation of STAT3 phosphorylation was suppressed in ZT0 group. At ZT2, expressions of IL-6/Interferon-γ/TNF/granulocyte-macrophage colony-stimulating factor in ZT0 group were significantly decreased as compared to untreated mice, though not in ZT12 group. In ZT0 group, IL-6 and TNF-α in sera were decreased for longer time than that in ZT12 group. CONCLUSION Chronotherapy using Baricitinib targeting cytokine secretions is effective in CIA mice. Clinical applications of chronotherapy can be expected to enhance the drug efficacy.
Collapse
Affiliation(s)
- Arisa Yaekura
- Department of Biophysics, Division of Clinical of Immunology, Kobe University Graduate School of Health Sciences, Tomogaoka 7-10-2, Suma, Kobe 654-0142, Japan
| | - Kohsuke Yoshida
- Department of Biophysics, Division of Clinical of Immunology, Kobe University Graduate School of Health Sciences, Tomogaoka 7-10-2, Suma, Kobe 654-0142, Japan
| | - Kanta Morii
- Department of Biophysics, Division of Clinical of Immunology, Kobe University Graduate School of Health Sciences, Tomogaoka 7-10-2, Suma, Kobe 654-0142, Japan
| | - Yuto Oketani
- Department of Biophysics, Division of Clinical of Immunology, Kobe University Graduate School of Health Sciences, Tomogaoka 7-10-2, Suma, Kobe 654-0142, Japan
| | - Ikumi Okumura
- Department of Biophysics, Division of Clinical of Immunology, Kobe University Graduate School of Health Sciences, Tomogaoka 7-10-2, Suma, Kobe 654-0142, Japan
| | - Kenta Kaneshiro
- Department of Biophysics, Division of Clinical of Immunology, Kobe University Graduate School of Health Sciences, Tomogaoka 7-10-2, Suma, Kobe 654-0142, Japan
| | - Nao Shibanuma
- Department of Orthopaedic Surgery, Kobe Kaisei Hospital, Shinohara-Kita 3-11-15, Nada, Kobe 657-0068, Japan
| | - Yoshitada Sakai
- Department of Rehabilitation Science, Kobe University Graduate School of Medicine, Kusunoki 7-5-1, Cyuoh, Kobe 650-0017, Japan
| | - Akira Hashiramoto
- Department of Biophysics, Division of Clinical of Immunology, Kobe University Graduate School of Health Sciences, Tomogaoka 7-10-2, Suma, Kobe 654-0142, Japan.
| |
Collapse
|
28
|
Zampieri R, Brozzetti A, Pericolini E, Bartoloni E, Gabrielli E, Roselletti E, Lomonosoff G, Meshcheriakova Y, Santi L, Imperatori F, Merlin M, Tinazzi E, Dotta F, Nigi L, Sebastiani G, Pezzotti M, Falorni A, Avesani L. Prevention and treatment of autoimmune diseases with plant virus nanoparticles. SCIENCE ADVANCES 2020; 6:eaaz0295. [PMID: 32494704 PMCID: PMC7202875 DOI: 10.1126/sciadv.aaz0295] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 02/12/2020] [Indexed: 05/15/2023]
Abstract
Plant viruses are natural, self-assembling nanostructures with versatile and genetically programmable shells, making them useful in diverse applications ranging from the development of new materials to diagnostics and therapeutics. Here, we describe the design and synthesis of plant virus nanoparticles displaying peptides associated with two different autoimmune diseases. Using animal models, we show that the recombinant nanoparticles can prevent autoimmune diabetes and ameliorate rheumatoid arthritis. In both cases, this effect is based on a strictly peptide-related mechanism in which the virus nanoparticle acts both as a peptide scaffold and as an adjuvant, showing an overlapping mechanism of action. This successful preclinical testing could pave the way for the development of plant viruses for the clinical treatment of human autoimmune diseases.
Collapse
Affiliation(s)
- Roberta Zampieri
- Department of Biotechnology, University of Verona, Verona, Italy
- Diamante srl, Strada Le Grazie, 15, 37134 Verona, Italy
| | | | - Eva Pericolini
- Department of Surgical, Medical, Dental and Morphological Sciences with interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Bartoloni
- Department of Medicine, University of Perugia, Perugia, Italy
| | - Elena Gabrielli
- Department of Medicine, University of Perugia, Perugia, Italy
| | | | | | | | - Luca Santi
- Department of Agriculture and Forest Sciences, University of La Tuscia, Viterbo, Italy
| | - Francesca Imperatori
- Department of Agriculture and Forest Sciences, University of La Tuscia, Viterbo, Italy
| | - Matilde Merlin
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Elisa Tinazzi
- Department of Medicine, University of Verona, Verona, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Laura Nigi
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Umberto Di Mario Foundation ONLUS, Toscana Life Sciences, Siena, Italy
| | - Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
- Umberto Di Mario Foundation ONLUS, Toscana Life Sciences, Siena, Italy
| | - Mario Pezzotti
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Alberto Falorni
- Department of Medicine, University of Perugia, Perugia, Italy
| | - Linda Avesani
- Department of Biotechnology, University of Verona, Verona, Italy
| |
Collapse
|
29
|
Phenotypic and functional characterization of natural killer cells in rheumatoid arthritis-regulation with interleukin-15. Sci Rep 2020; 10:5858. [PMID: 32246007 PMCID: PMC7125139 DOI: 10.1038/s41598-020-62654-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/13/2020] [Indexed: 11/23/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovial inflammation and joint destruction. Previous studies have shown that natural killer (NK) cells may play an important role in the pathogenesis of RA. Interleukin (IL)-15, a pro-inflammatory cytokine which induces proliferation and differentiation of NK cells, is overexpressed in RA. In this present study, we examine various NKRs and adhesion molecule expression on NK cells from RA patients and their response to IL-15 stimulation. We also sought to study cytokine-induced memory-like (CIML) NK cells in RA patients. We established that 1. RA patients had higher NK cell percentages in peripheral blood and their serum IL-15 levels were higher compared to healthy volunteers; 2. NK cells from RA patients showed lower NKp46 expression and an impaired CD69 response to IL-15; 3. NK cells from RA patients showed higher CD158b and CD158e expression but lower CD62L expression; 4. exogenous IL-15 up-regulated CD69, CD158b, CD158e but down-regulated NKp46 and CD62L expression in RA; 5. As to CIML NK cells, restimulation - induced NK cytotoxicity and IFN-γ production was impaired in RA patients, 6. Reduced NKp46, perforin, and granzyme B expression on NK cells was found in RA patients with bone deformity and erosion, 7. RA disease activity (DAS28) showed inverse correlation with the percentages of CD56+CD3− NK cells, and NKp46 and perforin expression on NK cells, respectively. Taken together, our study demonstrated differential expression of various NK receptors in RA patients. NKp46, CD158e, and perforin expression on NK cells may serve as markers of RA severity.
Collapse
|
30
|
Yu X, Song Z, Rao L, Tu Q, Zhou J, Yin Y, Chen D. Synergistic induction of CCL5, CXCL9 and CXCL10 by IFN-γ and NLRs ligands on human fibroblast-like synoviocytes-A potential immunopathological mechanism for joint inflammation in rheumatoid arthritis. Int Immunopharmacol 2020; 82:106356. [PMID: 32151958 DOI: 10.1016/j.intimp.2020.106356] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/30/2020] [Accepted: 02/25/2020] [Indexed: 12/13/2022]
Abstract
Interferon-γ (IFN-γ) is traditionally regarded as a proinflammatory cytokine by virtue of its strong macrophage activating potential and its association with Th1 driven immune responses. NOD1 and NOD2 are cytoplasmic receptors that can initiate the initial immune response by sensing bacterial components or danger signals. In this study, we investigated the immunopathological roles of IFN-γ and NOD1, 2 ligands iE-DAP/MDP on the activation of fibroblast-like synoviocytes (FLS) in RA. FLS constitutively express functional NOD1 and NOD2, and the gene and protein expression of NOD1 and NOD2 could be enhanced by the treatment with IFN-γ. The synergistic effect was observed in the combined treatment of IFN-γ and NOD1 ligand iE-DAP or NOD2 ligand MDP on the release of CCL5, CXCL9 and CXCL10 from FLS, and its effect was in a dose-dependent manner. The co-stimulation which IFN-γ combined with iE-DAP/MDP could abolish the inhibition of CXCL8 level by IFN-γ alone. Further investigations showed that synergistic effects on the production of CCL5, CXCL9 and CXCL10 in FLS stimulated by IFN-γ and iE-DAP/MDP were differentially regulated by intracellular activation of NF-κB, p38MAPK and ERK pathways. In conclusion, our data confirmed the inflammatory effect of IFN-γ and iE-DAP/MDP on human FLS for the first time and therefore provided a new insight into the IFN-γ combined with NOD1 or NOD2 activated immunopathological mechanisms mediated by distinct intracellular signal transduction in joint inflammation of RA.
Collapse
Affiliation(s)
- Xiaoyan Yu
- Department of Clinical Laboratory; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation base of Child development and Critical Disorders; Children's Hospital of Chongqing Medical University, Chongqing, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Zhixin Song
- Department of Clinical Laboratory; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation base of Child development and Critical Disorders; Children's Hospital of Chongqing Medical University, Chongqing, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Lubei Rao
- Department of Clinical Laboratory; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation base of Child development and Critical Disorders; Children's Hospital of Chongqing Medical University, Chongqing, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Qianqian Tu
- Department of Clinical Laboratory; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation base of Child development and Critical Disorders; Children's Hospital of Chongqing Medical University, Chongqing, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Jie Zhou
- Department of Clinical Laboratory; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation base of Child development and Critical Disorders; Children's Hospital of Chongqing Medical University, Chongqing, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yibing Yin
- Key Laboratory of Diagnostic Medicine Designated by the Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Dapeng Chen
- Department of Clinical Laboratory; Ministry of Education Key Laboratory of Child Development and Disorders; National Clinical Research Center for Child Health and Disorders; China International Science and Technology Cooperation base of Child development and Critical Disorders; Children's Hospital of Chongqing Medical University, Chongqing, PR China; Chongqing Key Laboratory of Pediatrics, Chongqing, China.
| |
Collapse
|
31
|
Patel P, Meghani N, Kansara K, Kumar A. Nanotherapeutics for the Treatment of Cancer and Arthritis. Curr Drug Metab 2020; 20:430-445. [PMID: 30479211 DOI: 10.2174/1389200220666181127102720] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 10/11/2018] [Accepted: 10/11/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Nanotechnology is gaining significant attention worldwide for the treatment of complex diseases such as AIDS (acquired immune deficiency syndrome), cancer and rheumatoid arthritis. Nanomedicine is the application of nanotechnology used for diagnosis and treatment for the disease that includes the preservation and improvement of human health by covering an area such as drug delivery using nanocarriers, nanotheranostics and nanovaccinology. The present article provides an insight into several aspects of nanomedicine such as usages of multiple types of nanocarriers, their status, advantages and disadvantages with reference to cancer and rheumatoid arthritis. METHODS An extensive search was performed on the bibliographic database for research article on nanotechnology and nanomedicine along with looking deeply into the aspects of these diseases, and how all of them are co-related. We further combined all the necessary information from various published articles and briefed to provide the current status. RESULTS Nanomedicine confers a unique technology against complex diseases which includes early diagnosis, prevention, and personalized therapy. The most common nanocarriers used globally are liposomes, polymeric nanoparticles, dendrimers, metallic nanoparticles, magnetic nanoparticles, solid lipid nanoparticles, polymeric micelles and nanotubes among others. CONCLUSION Nanocarriers are used to deliver drugs and biomolecules like proteins, antibody fragments, DNA fragments, and RNA fragments as the base of cancer biomarkers.
Collapse
Affiliation(s)
- Pal Patel
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Nikita Meghani
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Krupa Kansara
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Ashutosh Kumar
- Biological and Life Sciences, School of Arts and Sciences, Ahmedabad University, Central Campus, Navrangpura, Ahmedabad, 380009, Gujarat, India
| |
Collapse
|
32
|
Choi EW, Kim S. Relationships between Cytokine Levels and Disease Parameters during the Development of a Collagen-induced Arthritis Model in Cynomolgus Macaques ( Macaca fascicularis). Comp Med 2019; 69:228-239. [PMID: 31068244 DOI: 10.30802/aalas-cm-18-000058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In rheumatoid arthritis research, NHP models of collagen-induced arthritis are important because these species share many immunologic and pathologic features with humans. In addition, serum levels of various cytokines in patients with rheumatoid arthritis have been studied as immune markers for disease prediction, early diagnosis, and effective therapeutic management. The purpose of this study was to identify changes in cytokine levels that occur during the development of collagen-induced arthritis in female cynomolgus macaques (n = 8) and to assess the relationships between these changes and various disease parameters. Blood samples were collected weekly before (week 0) and after (weeks 1 through 7) immunization with type II collagen; clinicopathologic and cytokine data from those samples and other clinical parameters were used in correlation analysis. Serum levels of IFN γ, chemokine (C-C motif) ligand 2 (CCL2), and IL6 showed significant changes after generation of collagen-induced arthritis. IFNγ levels showed a strong negative correlation with body weight (an indicator of general body condition), and CCL2 and IL6 showed moderate negative correlation with body weight. Serum IL6 levels showed moderate positive correlation with the soft tissue swelling score and strong positive correlation with serum C-reactive protein levels in our NHP model of collagen-induced arthritis. In addition, serum levels of matrix metalloproteinase 3 increased significantly after inoculation with type II collagen and showed a moderate positive correlation with serum levels of C-reactive protein, IL6, and IL15. These results suggest close correlations between various cytokines and disease parameters in NHP models of rheumatoid arthritis. These cytokines therefore potentially could be used as markers for monitoring the efficacy of novel treatments in NHP models of rheumatoid arthritis.
Collapse
Affiliation(s)
- Eun Wha Choi
- Department of Veterinary Clinical Pathology, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Sungjoo Kim
- Department of Surgery, Division of Transplantation, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea, and GenNBio, Seoul, Republic of Korea
| |
Collapse
|
33
|
Heckrodt TJ, Chen Y, Singh R. Synthesis of the isotopically labeled JAK1/3 inhibitor [D]-R545 and its oxidative metabolite [D]-R935: Protecting group-directed regioselective bromination to access 3,4,5-substituted anilines. Tetrahedron 2019. [DOI: 10.1016/j.tet.2019.02.059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
34
|
Cao YJ, Xu Y, Liu B, Zheng X, Wu J, Zhang Y, Li XS, Qi Y, Sun YM, Wen WB, Hou L, Wan CP. Dioscin, a Steroidal Saponin Isolated from Dioscorea nipponica, Attenuates Collagen-Induced Arthritis by Inhibiting Th17 Cell Response. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:423-437. [PMID: 30827153 DOI: 10.1142/s0192415x19500216] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Dioscin, a steroidal saponin isolated from Dioscorea nipponica Makino, has previously been shown to possess antiarthritic effects. However, the underlying mechanism is still elusive. Herein, we investigated the therapeutic effects of dioscin on collagen-induced arthritis (CIA) in DBA/1 mice and related mechanism. Cytokine production in CII-specific immune responses were measured by enzyme-linked immunosorbent assay (ELISA); Th17 cell-related gene expression, including IL-17A, ROR[Formula: see text] and IL-23p19, were detected by qPCR analysis; Surface marker, T regulatory (Treg) cells and intracellular cytokines (IL-17A and IFN-[Formula: see text]) were evaluated by flow cytometry. We performed Th17 cell differentiation assay in vitro. Results showed that, in vivo, dioscin treatment significantly reduced the severity of CIA, which was accompanied by decreased Th17 response, but not Th1 and Treg response; dioscin-treated mice also showed lower percentage of CD11b[Formula: see text] Gr-1[Formula: see text] neutrophils; In vitro, dioscin treatment suppressed the differentiation of naive CD4[Formula: see text] T cells into Th17 cell and decreased IL-17A production. Collectively, our results indicate that dioscin exerts antiarthritic effects by inhibiting Th17 cell immune response.
Collapse
Affiliation(s)
- Yong-Jun Cao
- Department of Rheumatology, Nantong Hospital Affiliated to Nanjing, University of Traditional Chinese Medicine, Nantong, Jiangsu 226001, P. R. China
| | - Ying Xu
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Bei Liu
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Xi Zheng
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Jian Wu
- Department of Rheumatology, Nantong Hospital Affiliated to Nanjing, University of Traditional Chinese Medicine, Nantong, Jiangsu 226001, P. R. China
| | - Ying Zhang
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Xiao-Si Li
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Yan Qi
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Yu-Meng Sun
- Department of Rheumatology, Nantong Hospital Affiliated to Nanjing, University of Traditional Chinese Medicine, Nantong, Jiangsu 226001, P. R. China
| | - Wei-Bo Wen
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Lifei Hou
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Chun-Ping Wan
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| |
Collapse
|
35
|
Dai Q, Li J, Yun Y, Wang J. Toll-Like Receptor 4-Myeloid Differentiation Primary Response Gene 88 Pathway Is Involved in the Shikonin Treatment of CIA by Regulating Treg/Th17 Expression. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2018; 2018:2428546. [PMID: 30643526 PMCID: PMC6311288 DOI: 10.1155/2018/2428546] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/17/2018] [Accepted: 12/04/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To investigate the effect of shikonin on (CIA) collagen-induced arthritis and its influence and mechanism on the balance between Th17 cells and Treg cells. METHODS Three doses of shikonin were administered orally to mice before the onset of CIA, and celecoxib was used as positive control drug. The arthritis response was monitored visually by macroscopic scoring and hindpaw swelling. Histology of knee was used to assess the occurrence of cartilage destruction and bone erosion. Serum collagen type II (C II) antibody levels associated with CIA were assessed with ELISAs. RT-PCR and quantitative PCR were employed to determine the mRNA expression of cytokines and TLRs in the surface of DCs in the patella with adjacent synovium and spleen in CIA. The expression of cytokines and transcription factors in the peripheral immune organs was tested by Western blotting. RESULTS Shikonin treatment suppressed the macroscopic score and incidence of arthritis. Swelling of hind paws, cartilage destruction, and serum anti-C II concentration were delayed with shikonin when compared to controls. Shikonin treatment suppressed the arthritis in a dose-dependent manner. Moreover, the expression of Th17 cytokines (IL-17A) was greatly inhibited both in the synovium and spleen in treated groups compared with those in control groups. The mRNA and protein levels of IL-10 and TGF-β, however, were upregulated after shikonin treatment. The expression of Foxp3 in the synovium and spleen was upregulated, and the expression of ROR-γt in the synovium and spleen was downregulated after shikonin treatment through RT-PCR, quantitative PCR, and Western blotting. The DCs in the spleen of shikonin-treated mice had lower expression of TLR4 and MyD88, and the expression of TLR2 and TLR9 in the spleen was not different between the two groups. CONCLUSION Shikonin has anti-inflammatory effects on CIA. Shikonin treatment can inhibit Th17 cytokines expression and induce Treg responses through inhibiting the activation of TLR4/MyD88 pathway.
Collapse
Affiliation(s)
- Qiaomei Dai
- Department of Pathology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ji Li
- Department of Chinese Formulae, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yu Yun
- Department of Oncology, Traditional Chinese Medical Hospital of Siyang County, Jiangsu, China
| | - Jianwei Wang
- Department of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
36
|
Liu W, Sun Y, Cheng Z, Guo Y, Liu P, Wen Y. Crocin exerts anti-inflammatory and anti-arthritic effects on type II collagen-induced arthritis in rats. PHARMACEUTICAL BIOLOGY 2018; 56. [PMID: 29540097 PMCID: PMC6168764 DOI: 10.1080/13880209.2018.1448874] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
CONTEXT Rheumatoid arthritis (RA) is a common systemic auto-immune disease, which is characterized by chronic and symmetry synovial inflammation. Crocin has been reported to exhibit anti-inflammatory effects in animal models. OBJECTIVE This study investigates the anti-inflammatory and anti-arthritic effects of crocin on type II collagen-induced arthritis (CIA) in Wistar rats. MATERIALS AND METHODS The CIA rat model was established and randomly divided into five groups with or without crocin treatment (10, 20 or 40 mg/kg), which was started on day 21 after arthritis induction and persisted for 36 days. The symptoms and molecular mechanisms of CIA and crocin-treated CIA rats were compared and investigated. RESULTS CIA rats presented severe RA symptoms, including high arthritis score, paw swelling, joint inflammation, bone erosion, chondrocyte death, cartilage destruction, enhanced expressions of matrix metalloproteinase (MMP) and pro-inflammatory cytokines. However, crocin could mitigate these symptoms. Crocin (40 mg/kg) exhibited the most efficient therapeutic function on CIA rats: the histological scores of joint inflammation, bone erosion, chondrocyte death, cartilage surface erosion, and bone erosion of CIA rats receiving 40 mg/kg crocin treatment were comparable to the normal rats. MMP-1, -3 and -13 protein expression levels of CIA rats with 40 mg/kg crocin treatment were decreased to levels similar to normal rats. Moreover, crocin could also inhibit the expression of TNF-α, IL-17, IL-6 and CXCL8 in serum and ankle tissues of CIA rats. CONCLUSIONS In summary, crocin exhibits therapeutic potential for RA, by mitigating the symptoms and inhibiting the pro-inflammatory factor expression.
Collapse
Affiliation(s)
- Wei Liu
- Department of Orthopaedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, China
| | - Yufeng Sun
- Department of Orthopaedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, China
- CONTACT Yufeng Sun Department of Orthopaedics, The Fifth Hospital of Harbin, No. 27 Jiankang Road, Harbin150010, Heilongjiang, China
| | - Zhenping Cheng
- Department of Orthopaedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, China
| | - Yong Guo
- Department of Orthopaedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, China
| | - Peiming Liu
- Department of Orthopaedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, China
| | - Ying Wen
- Department of Orthopaedics, The Fifth Hospital of Harbin, Harbin, Heilongjiang, China
| |
Collapse
|
37
|
Integrin, alpha9 subunit blockade suppresses collagen-induced arthritis with minimal systemic immunomodulation. Eur J Pharmacol 2018; 833:320-327. [PMID: 29932925 DOI: 10.1016/j.ejphar.2018.06.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 06/17/2018] [Accepted: 06/18/2018] [Indexed: 11/21/2022]
Abstract
Integrin, alpha9 subunit (hereinafter, alpha9) has been identified as a novel putative therapeutic target for rheumatoid arthritis (RA). Support for this target comes from the observations that alpha9 is overexpressed both in the joints of RA patients and in animal models of arthritis. In the experimental models, the increase in alpha9 expression precedes the onset of arthritic symptoms. The current study presents data on the pharmacological profile of an anti-alpha9 antibody in a collagen-induced arthritis (CIA) mouse model. Administration of an alpha9-blocking antibody in CIA mice suppressed the development of arthritis and significantly decreased plasma level of activated fibroblast-like synoviocyte (FLS)-derived biomarkers without reducing the formation of anti-type II collagen antibodies. While anti-alpha9 antibody administration significantly suppress the accumulation of immune cells in arthritic joints it had no effect on immune cell number in the spleen. Furthermore, in non-arthritic mice, alpha9 had no inhibitory effect in either a mixed lymphocyte reaction (MLR) or in a delayed type hypersensitivity (DTH) reaction. These results suggest that blocking alpha9 exerts its anti-arthritic effect through suppression of FLS-activation via a non-immune mediated mechanism. Finally, therapeutic administration of anti-alpha9 antibody alleviated established arthritis in CIA mice. Our data provide evidence that alpha9 blockade is a promising therapy for joint inflammation with minimal systemic immunomodulation.
Collapse
|
38
|
Nehmar R, Mariotte A, de Cauwer A, Sibilia J, Bahram S, Georgel P. Therapeutic Perspectives for Interferons and Plasmacytoid Dendritic Cells in Rheumatoid Arthritis. Trends Mol Med 2018. [DOI: 10.1016/j.molmed.2018.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
39
|
Lee SH, Kwon JY, Kim SY, Jung K, Cho ML. Interferon-gamma regulates inflammatory cell death by targeting necroptosis in experimental autoimmune arthritis. Sci Rep 2017; 7:10133. [PMID: 28860618 PMCID: PMC5579272 DOI: 10.1038/s41598-017-09767-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/28/2017] [Indexed: 02/06/2023] Open
Abstract
Interferon γ (IFN-γ) induces an inflammatory response and apoptotic cell death. Rheumatoid arthritis (RA) is a systemic inflammatory disease associated with increased levels of inflammatory mediators, including tumour necrosis factor α (TNF-α) and T helper (Th) 17 cells, and downregulation of apoptosis of inflammatory cells. We hypothesized that IFN-γ would reduce inflammatory cell death in vitro and that loss of IFN-γ would aggravate inflammation in vivo. IFN-γ downregulated necroptosis and the expression of cellular FLICE-like inhibitory protein (cFLIPL) and mixed lineage kinase domain-like (MLKL). However, loss of IFN-γ promoted the production of cFLIPL and MLKL, and necroptosis. IFN-γ deficiency increased Th17 cell number and upregulated the expression of IL-17 and TNF-α. Expression of MLKL, receptor interacting protein kinase (RIPK)1, and RIPK3 was increased in the joints of mice with collagen-induced arthritis (CIA). Compared with wild-type mice with CIA, IFN-γ−/− CIA mice showed exacerbation of cartilage damage and joint inflammation, and acceleration of MLKL, RIPK1, and RIPK3 production in the joints. IFN-γ deficiency induced the activation of signal transducer and activator of transcription 3. These results suggest that IFN-γ regulates inflammatory cell death and may have potential for use in the treatment of RA.
Collapse
Affiliation(s)
- Seung Hoon Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ji Ye Kwon
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Se-Young Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | | | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, South Korea. .,Laboratory of Immune Network, Conversant Research Consortium in Immunologic disease, College of Medicine, The Catholic University of Korea, Seoul, South Korea.
| |
Collapse
|
40
|
Li P, Zheng Y, Chen X. Drugs for Autoimmune Inflammatory Diseases: From Small Molecule Compounds to Anti-TNF Biologics. Front Pharmacol 2017; 8:460. [PMID: 28785220 PMCID: PMC5506195 DOI: 10.3389/fphar.2017.00460] [Citation(s) in RCA: 230] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 06/27/2017] [Indexed: 12/14/2022] Open
Abstract
Although initially described as an anti-tumor mediator, tumor necrosis factor-alpha (TNF) is generally considered as the master pro-inflammatory cytokine. It plays a crucial role in the pathogenesis of inflammatory diseases, such as rheumatoid arthritis (RA), inflammatory bowel disease, ankylosing spondylitis (AS), and psoriasis. Consequently, anti-TNF therapy has become mainstay treatment for autoimmune diseases. Historically, anti-inflammatory agents were developed before the identification of TNF. Salicylates, the active components of Willow spp., were identified in the mid-19th century for the alleviation of pain, fever, and inflammatory responses. Study of this naturally occurring compound led to the discovery of aspirin, which was followed by the development of non-steroidal anti-inflammatory drugs (NSAIDs) due to the chemical advances in the 19th–20th centuries. Initially, the most of NSAIDs were organic acid, but the non-acidic compounds were also identified as NSAIDs. Although effective in the treatment of inflammatory diseases, NSAIDs have some undesirable and adverse effect, such as ulcers, kidney injury, and bleeding in the gastrointestinal tract. In the past two decades, anti-TNF biologics were developed. Drugs belong to this class include soluble TNF receptor 2 fusion protein and anti-TNF antibodies. The introduction of anti-TNF therapeutics has revolutionized the management of autoimmune diseases, such as RA, psoriatic arthritis (PsA), plaque psoriasis (PP), AS, CD and ulcerative colitis (UC). Nevertheless, up to 40% of patients have no response to anti-TNF treatment. Furthermore, this treatment is associated with some adverse effects such as increased risk of infection, and even triggered the de novo development of autoimmune diseases. Such harmful effect of anti-TNF treatment is likely caused by the global inhibition of TNF biological functions. Therefore, specific inhibition of TNF receptor (TNFR1 or TNFR2) may represent a safer and more effective treatment, as proposed by some recent studies. In this review article, the historical development of anti-inflammatory drugs after World War II as briefly described above will be reviewed and analyzed. The future trend in the development of novel TNF receptor-targeting therapeutics will be discussed in the context of latest progress in the research of TNF biology.
Collapse
Affiliation(s)
- Ping Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| |
Collapse
|
41
|
Adoptive Cell Therapy of Induced Regulatory T Cells Expanded by Tolerogenic Dendritic Cells on Murine Autoimmune Arthritis. J Immunol Res 2017; 2017:7573154. [PMID: 28702462 PMCID: PMC5494067 DOI: 10.1155/2017/7573154] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 04/01/2017] [Accepted: 04/27/2017] [Indexed: 12/21/2022] Open
Abstract
Objective Tolerogenic dendritic cells (tDCs) can expand TGF-β-induced regulatory T cells (iTregs); however, the therapeutic utility of these expanded iTregs in autoimmune diseases remains unknown. We sought to determine the properties of iTregs expanded by mature tolerogenic dendritic cells (iTregmtDC) in vitro and explore their potential to ameliorate collagen-induced arthritis (CIA) in a mouse model. Methods After induction by TGF-β and expansion by mature tDCs (mtDCs), the phenotype and proliferation of iTregmtDC were assessed by flow cytometry. The ability of iTregs and iTregmtDC to inhibit CD4+ T cell proliferation and suppress Th17 cell differentiation was compared. Following adoptive transfer of iTregs and iTregmtDC to mice with CIA, the clinical and histopathologic scores, serum levels of IFN-γ, TNF-α, IL-17, IL-6, IL-10, TGF-β and anti-CII antibodies, and the distribution of the CD4+ Th subset were assessed. Results Compared with iTregs, iTregmtDC expressed higher levels of Foxp3 and suppressed CD4+ T cell proliferation and Th17 cell differentiation to a greater extent. In vivo, iTregmtDC reduced the severity and progression of CIA more significantly than iTregs, which was associated with a modulated inflammatory cytokine profile, reduced anti-CII IgG levels, and polarized Treg/Th17 balance. Conclusion This study highlights the potential therapeutic utility of iTregmtDC in autoimmune arthritis and should facilitate the future design of iTreg immunotherapeutic strategies.
Collapse
|
42
|
Ethyl Caffeate Ameliorates Collagen-Induced Arthritis by Suppressing Th1 Immune Response. J Immunol Res 2017; 2017:7416792. [PMID: 28706956 PMCID: PMC5494568 DOI: 10.1155/2017/7416792] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 03/26/2017] [Indexed: 12/25/2022] Open
Abstract
The present study was designed to assess the antiarthritic potential of ECF in collagen-induced arthritis (CIA) and explore its underlying mechanism. Methods. In vitro, lymphocyte proliferation assay was measured by CCK-8 kit. In vivo, the therapeutic potential of ECF on CIA was investigated; surface marker, Treg cell, and intracellular cytokines (IL-17A and IFN-γ) were detected by flow cytometry. Th1 cell differentiation assay was performed, and mRNA expression in interferon-γ-related signaling was examined by q-PCR analysis. Results. In vitro, ECF markedly inhibited the proliferation of splenocytes in response to ConA and anti-CD3. In vivo, ECF treatment reduced the severity of CIA, inhibited IFN-γ and IL-6 secretion, and decreased the proportion of CD11b+Gr-1+ splenic neutrophil. Meanwhile, ECF treatment significantly inhibited the IFN-γ expression in CD4+T cell without obviously influencing the development of Th17 cells and T regulatory cells. In vitro, ECF suppressed the differentiation of naive CD4+ T cells into Th1. Furthermore, ECF intensely blocked the transcriptional expression in interferon-γ-related signaling, including IFN-γ, T-bet, STAT1, and STAT4. Conclusion. Our results indicated that ECF exerted antiarthritic potential in collagen-induced arthritis by suppressing Th1 immune response and interferon-γ-related signaling.
Collapse
|
43
|
Sahu D, Sharma S, Singla RK, Panda AK. Antioxidant activity and protective effect of suramin against oxidative stress in collagen induced arthritis. Eur J Pharm Sci 2017; 101:125-139. [PMID: 28189815 DOI: 10.1016/j.ejps.2017.02.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 12/21/2016] [Accepted: 02/06/2017] [Indexed: 01/14/2023]
|
44
|
Moroni L, Selmi C, Angelini C, Meroni PL. Evaluation of Endothelial Function by Flow-Mediated Dilation: a Comprehensive Review in Rheumatic Disease. Arch Immunol Ther Exp (Warsz) 2017; 65:463-475. [DOI: 10.1007/s00005-017-0465-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 03/03/2017] [Indexed: 12/18/2022]
|
45
|
Sapir-Koren R, Livshits G. Rheumatoid arthritis onset in postmenopausal women: Does the ACPA seropositive subset result from genetic effects, estrogen deficiency, skewed profile of CD4(+) T-cells, and their interactions? Mol Cell Endocrinol 2016; 431:145-63. [PMID: 27178986 DOI: 10.1016/j.mce.2016.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Revised: 05/04/2016] [Accepted: 05/09/2016] [Indexed: 12/28/2022]
Abstract
Rheumatoid arthritis (RA) incidence displays a differentiated age-dependent female-to-male ratio in which women outnumber men. Evidence that the peak incidence of RA in women coincides with menopause age, suggests a potential estrogenic role to disease etiology. Estrogens exert physiologically both stimulatory and inhibitory effects on the immune system. Epidemiologic and animal model studies with estrogen deprivation or supplementation suggested estrogens as to play, mainly, a protective role in RA immunopathology. In this review, we propose that some yet unidentified disturbances associated with estrogen circulating levels, differentiated by the menopausal status, play a major role in women's RA susceptibility. We focus on the interaction between estrogen deprivation and genetic risk alleles for anti-citrullinated protein antibodies (ACPA) seropositive RA, as a major driving force for increased immune reactivity and RA susceptibility, in postmenopausal women. This opens up new fields for research concerning the association among different irregular estrogenic conditions, the cytokine milieu, and age/menopausal status bias in RA.
Collapse
Affiliation(s)
- Rony Sapir-Koren
- Human Population Biology Research Group, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gregory Livshits
- Human Population Biology Research Group, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel; Lilian and Marcel Pollak Chair of Biological Anthropology, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
46
|
Liu X, Jiao Y, Cao Y, Deng N, Ma Y, Hasty KA, Kang A, Chen H, Stuart JM, Gu W. Decreased expression levels of Ifi genes is associated to the increased resistance to spontaneous arthritis disease in mice deficiency of IL-1RA. BMC Immunol 2016; 17:25. [PMID: 27480124 PMCID: PMC4970213 DOI: 10.1186/s12865-016-0163-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/12/2016] [Indexed: 01/21/2023] Open
Abstract
Background The mouse strain BALB/c deficient in IL-1 receptor antagonist protein (Il-1ra) develops spontaneous arthritis disease (SAD) while the strain DBA/1 IL1rn-/- with the same deficiency does not. Previously, we mapped a QTL on chromosome 1 for SAD and then developed a congenic mouse strain BALB.D1-1-/- that contains the QTL genomic fragment associated with resistance from DBA/1-/- on a BALB/c-/- background. The congenic strain was relatively resistant to spontaneous arthritis and had delayed onset and reduced severity of disease. We obtained whole genome expression profiles from the spleen of the congenic strain BALB.D1-1-/- and four other strains, the wild type BALB/c, DBA/1 and the deficient DBA/1 IL1rn-/- and the BALB/c IL1rn-/-. We then compared the similarities and differences between the congenic strain and the four parental strains. Here we report the selected potential causal genes based on differential expression levels as well as function of genes. Results There is a considerable number of genes that are differentially expressed between the congenic strain and the three parental strains, BALB/c, DBA/1, and DBA/1-/-. However there only a few differentially expressed genes were identified by comparing the congenic strain and the BALB/c-/-strain. These differentially expressed genes are mainly from T-cell receptor beta chain (Tcrb) and interferon-activatable protein (Ifi) genes. These genes are also differentially expressed between congenic strain and BALB/c strains. However, their expression levels in the congenic strain are similar to that in DBA/1 and DBA/1-/-. The expression level of Tcrb-j gene is positively associated with two genes of Ifi gene 200 cluster. Conclusions Decreased expression levels of Ifi genes is associated to the increased resistance to spontaneous arthritis disease and with down regulation of expressions of Tcrb genes in the mouse congenic strain. Ifi genes may play an important role in the susceptibility to SAD in mice. Electronic supplementary material The online version of this article (doi:10.1186/s12865-016-0163-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoyun Liu
- Mudanjiang Medical College, Mudanjiang, HeilongJiang, 157001, People's Republic of China.,Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center (UTHSC), Memphis, TN, 38163, USA
| | - Yan Jiao
- Mudanjiang Medical College, Mudanjiang, HeilongJiang, 157001, People's Republic of China. .,Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center (UTHSC), Memphis, TN, 38163, USA.
| | - Yanhong Cao
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center (UTHSC), Memphis, TN, 38163, USA.,Institute of Kaschin-beck Disease, Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, China.,Key Laboratory of Etiologic Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health (23618104), Harbin, 150081, China
| | - Nan Deng
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Yonghui Ma
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center (UTHSC), Memphis, TN, 38163, USA
| | - Karen A Hasty
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center (UTHSC), Memphis, TN, 38163, USA.,Research Service, Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis, TN, 38104, USA
| | - Andrew Kang
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Hong Chen
- Center of integrative research, The first Hospital of Qiqihaer City, 30 Gongyuan Road, Longsha District, Qiqihaer, Heilongjiang, 161005, People's Republic of China
| | - John M Stuart
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Research Service, Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis, TN, 38104, USA
| | - Weikuan Gu
- Department of Orthopaedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center (UTHSC), Memphis, TN, 38163, USA. .,Research Service, Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis, TN, 38104, USA.
| |
Collapse
|
47
|
Abstract
Basement membrane components are targets of autoimmune attack in diverse diseases that destroy kidneys, lungs, skin, mucous membranes, joints, and other organs in man. Epitopes on collagen and laminin, in particular, are targeted by autoantibodies and T cells in anti-glomerular basement membrane glomerulonephritis, Goodpasture's disease, rheumatoid arthritis, post-lung transplant bronchiolitis obliterans syndrome, and multiple autoimmune dermatoses. This review examines major diseases linked to basement membrane autoreactivity, with a focus on investigations in patients and animal models that advance our understanding of disease pathogenesis. Autoimmunity to glomerular basement membrane type IV is discussed in depth as a prototypic organ-specific autoimmune disease yielding novel insights into the complexity of anti-basement membrane immunity and the roles of genetic and environmental susceptibility.
Collapse
|
48
|
Biton J, Khaleghparast Athari S, Thiolat A, Santinon F, Lemeiter D, Hervé R, Delavallée L, Levescot A, Roga S, Decker P, Girard JP, Herbelin A, Boissier MC, Bessis N. In Vivo Expansion of Activated Foxp3+ Regulatory T Cells and Establishment of a Type 2 Immune Response upon IL-33 Treatment Protect against Experimental Arthritis. THE JOURNAL OF IMMUNOLOGY 2016; 197:1708-19. [PMID: 27474075 DOI: 10.4049/jimmunol.1502124] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 06/24/2016] [Indexed: 01/03/2023]
Abstract
IL-33 is strongly involved in several inflammatory and autoimmune disorders with both pro- and anti-inflammatory properties. However, its contribution to chronic autoimmune inflammation, such as rheumatoid arthritis, is ill defined and probably requires tight regulation. In this study, we aimed at deciphering the complex role of IL-33 in a model of rheumatoid arthritis, namely, collagen-induced arthritis (CIA). We report that repeated injections of IL-33 during induction (early) and during development (late) of CIA strongly suppressed clinical and histological signs of arthritis. In contrast, a late IL-33 injection had no effect. The cellular mechanism involved in protection was related to an enhanced type 2 immune response, including the expansion of eosinophils, Th2 cells, and type 2 innate lymphoid cells, associated with an increase in type 2 cytokine levels in the serum of IL-33-treated mice. Moreover, our work strongly highlights the interplay between IL-33 and regulatory T cells (Tregs), demonstrated by the dramatic in vivo increase in Treg frequencies after IL-33 treatment of CIA. More importantly, Tregs from IL-33-treated mice displayed enhanced capacities to suppress IFN-γ production by effector T cells, suggesting that IL-33 not only favors Treg proliferation but also enhances their immunosuppressive properties. In concordance with these observations, we found that IL-33 induced the emergence of a CD39(high) Treg population in a ST2L-dependent manner. Our findings reveal a powerful anti-inflammatory mechanism by which IL-33 administration inhibits arthritis development.
Collapse
Affiliation(s)
- Jérôme Biton
- INSERM, U1125, F-93017 Bobigny, France; Sorbonne Paris Cité Université Paris 13, F-93000 Bobigny, France
| | - Sara Khaleghparast Athari
- INSERM, U1125, F-93017 Bobigny, France; Sorbonne Paris Cité Université Paris 13, F-93000 Bobigny, France
| | - Allan Thiolat
- INSERM, U1125, F-93017 Bobigny, France; Sorbonne Paris Cité Université Paris 13, F-93000 Bobigny, France
| | - François Santinon
- INSERM, U1125, F-93017 Bobigny, France; Sorbonne Paris Cité Université Paris 13, F-93000 Bobigny, France
| | - Delphine Lemeiter
- INSERM, U1125, F-93017 Bobigny, France; Sorbonne Paris Cité Université Paris 13, F-93000 Bobigny, France
| | - Roxane Hervé
- INSERM, U1125, F-93017 Bobigny, France; Sorbonne Paris Cité Université Paris 13, F-93000 Bobigny, France
| | | | - Anais Levescot
- INSERM U1082, Pôle Biologie Santé, Centre Hospitalo-Universitaire Poitiers, BP 633, F-86022 Poitiers, France
| | - Stéphane Roga
- Institut de Pharmacologie et de Biologie Structurale CNRS-Université de Toulouse III, F-31000 Toulouse, France; and
| | - Patrice Decker
- INSERM, U1125, F-93017 Bobigny, France; Sorbonne Paris Cité Université Paris 13, F-93000 Bobigny, France
| | - Jean-Philippe Girard
- Institut de Pharmacologie et de Biologie Structurale CNRS-Université de Toulouse III, F-31000 Toulouse, France; and
| | - André Herbelin
- INSERM U1082, Pôle Biologie Santé, Centre Hospitalo-Universitaire Poitiers, BP 633, F-86022 Poitiers, France
| | - Marie-Christophe Boissier
- INSERM, U1125, F-93017 Bobigny, France; Sorbonne Paris Cité Université Paris 13, F-93000 Bobigny, France; Assistance Publique-Hôpitaux de Paris, Hôpital Avicenne, Service de Rhumatologie, F-93009 Bobigny, France
| | - Natacha Bessis
- INSERM, U1125, F-93017 Bobigny, France; Sorbonne Paris Cité Université Paris 13, F-93000 Bobigny, France;
| |
Collapse
|
49
|
Lee WJ, Kim JY, Wu TP, Park LS. The establishment of a porcine rheumatoid arthritis model: Collagen induced arthritis minipig model. J Pharmacol Sci 2016; 132:41-47. [PMID: 27156927 DOI: 10.1016/j.jphs.2016.04.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 04/03/2016] [Accepted: 04/04/2016] [Indexed: 01/10/2023] Open
Abstract
Rheumatoid arthritis (RA) research has been largely dependent on collagen induced arthritis (CIA) rodent models, however, they may not translate well to humans due to innate differences in the size, physiology and lifespan. The present study aimed to establish a CIA porcine model with the physical, hematological, histopathological and etiological properties closer to their human equivalent in an attempt to better meet the needs of RA research. Three month old minipigs were administered of bovine type II collagen (CII) emulsified with complete Freund's adjuvants on Day 1 and incomplete Freund's adjuvants on Day 22, via an intradermal or intra-articular route. The clinical, radiological and hematological assessments of immunized animals were made periodically until Day 43, during which period the onset and progression of arthritis was recorded and characterized. In addition, the histopathological and micro-tomographic assessments of the cartilage degradation with regard to mononuclear cell infiltration, and joint deformity indicated a higher severity in the intradermal injection group over the intra-articular group. With confirmation of the susceptibility to heterogeneous CII for arthritis induction in minipig, the potential suitability of this test system as a large animal model for RA has been demonstrated.
Collapse
Affiliation(s)
- Won Jae Lee
- PWG Genetics Pte Ltd, 15 Tech Park Crescent, 638117 Singapore
| | - Ji Yeon Kim
- PWG Genetics Pte Ltd, 15 Tech Park Crescent, 638117 Singapore
| | | | | |
Collapse
|
50
|
Utilization of nanoparticle technology in rheumatoid arthritis treatment. Biomed Pharmacother 2016; 80:30-41. [PMID: 27133037 DOI: 10.1016/j.biopha.2016.03.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/03/2016] [Accepted: 03/03/2016] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is one of the common and severe autoimmune diseases related to joints. This chronic autoimmune inflammatory disease, leads to functional limitation and reduced quality of life, since as there is bone and cartilage destruction, joint swelling and pain. Current advances and new treatment approaches have considerably postponed disease progression and improved the quality of life for many patients. In spite of major advances in therapeutic options, restrictions on the routes of administration and the necessity for frequent and long-term dosing often result in systemic adverse effects and patient non-compliance. Unlike usual drugs, nanoparticle systems are planned to deliver therapeutic agents especially to inflamed synovium, so avoiding systemic and unpleasant effects. The present review discusses about some of the most successful drugs in RA therapy and their side effects and also focuses on key design parameters of RA-targeted nanotechnology-based strategies for improving RA therapies.
Collapse
|