151
|
King JD, Rowland G, Villasante Tezanos AG, Warwick J, Kraus VB, Lattermann C, Jacobs CA. Joint Fluid Proteome after Anterior Cruciate Ligament Rupture Reflects an Acute Posttraumatic Inflammatory and Chondrodegenerative State. Cartilage 2020; 11:329-337. [PMID: 30033738 PMCID: PMC7298591 DOI: 10.1177/1947603518790009] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE The purpose of this study was to evaluate changes in the synovial fluid proteome following acute anterior cruciate ligament (ACL) injury. DESIGN This study represents a secondary analysis of synovial fluid samples collected from the placebo group of a previous randomized trial. Arthrocentesis was performed twice on 6 patients with an isolated acute ACL tear at a mean of 6 and 14 days postinjury. Synovial fluid was analyzed by a highly multiplexed assay of 1129 proteins (SOMAscan version 3, SomaLogic, Inc., Boulder, CO). Pathway analysis using DAVID was performed; genes included met 3 criteria: significant change between the 2 study time points using a paired t test, significant change between the 2 study time points using a Mann-Whitney nonparametric test, and significant Benjamini post hoc analysis. RESULTS Fifteen analytes demonstrated significant increases between time points. Five of the 15 have been previously associated with the onset and/or severity of rheumatoid arthritis, including apoliopoprotein E and isoform E3, vascular cell adhesion protein 1, interleukin-34, and cell surface glycoprotein CD200 receptor 1. Chondrodegenerative enzymes and products of cartilage degeneration all increased over time following injury: MMP-1 (P = 0.08, standardized response mean [SRM] = 1.00), MMP-3 (P = 0.05, SRM = 0.90), ADAM12 (P = 0.03, SRM = 1.31), aggrecan (P = 0.08, SRM = 1.13), and CTX-II (P = 0.07, SRM = 0.56). Notable pathways that were differentially expressed following injury were the cytokine-cytokine receptor interaction and osteoclast differentiation pathways. CONCLUSIONS The proteomic results and pathway analysis demonstrated a pattern of cartilage degeneration, not only consistent with previous findings but also changes consistent with an inflammatory arthritogenic process post-ACL injury.
Collapse
Affiliation(s)
- John D. King
- Department of Orthopedic Surgery,
University of Kentucky, Lexington, KY, USA
| | - Grant Rowland
- Central Texas Sports Medicine &
Orthopedics, Bryan, TX, USA
| | | | - James Warwick
- College of Medicine, University of
Kentucky, Lexington, KY, USA
| | - Virginia B. Kraus
- Duke Molecular Physiology Institute,
Department of Medicine, Duke University School of Medicine, Durham, NC, USA,Division of Rheumatology, Department of
Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Christian Lattermann
- Department of Orthopaedic Surgery,
Harvard Medical School and Brigham and Women’s Hosptial, Chestnut Hill, MS,
USA
| | - Cale A. Jacobs
- Department of Orthopedic Surgery,
University of Kentucky, Lexington, KY, USA,Cale A. Jacobs, Department of Orthopedic
Surgery & Sports Medicine, University of Kentucky, 740 South Limestone
Street, Room K426, Lexington, KY 40536-0284, USA.
| |
Collapse
|
152
|
Sebastian-Valverde M, Pasinetti GM. The NLRP3 Inflammasome as a Critical Actor in the Inflammaging Process. Cells 2020; 9:cells9061552. [PMID: 32604771 PMCID: PMC7348816 DOI: 10.3390/cells9061552] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/12/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
As a consequence of the considerable increase in the human lifespan over the last century, we are experiencing the appearance and impact of new age-related diseases. The causal relationships between aging and an enhanced susceptibility of suffering from a broad spectrum of diseases need to be better understood. However, one specific shared feature seems to be of capital relevance for most of these conditions: the low-grade chronic inflammatory state inherently associated with aging, i.e., inflammaging. Here, we review the molecular and cellular mechanisms that link aging and inflammaging, focusing on the role of the innate immunity and more concretely on the nucleotide-binding oligomerization domain (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome, as well as how the chronic activation of this inflammasome has a detrimental effect on different age-related disorders.
Collapse
Affiliation(s)
| | - Giulio M. Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- James J. Peters Veterans Affairs Medical Center, Bronx, New York, NY 10468, USA
- Correspondence: ; Tel.: +1-212-241-1952
| |
Collapse
|
153
|
Therapeutic Functions of Stem Cells from Oral Cavity: An Update. Int J Mol Sci 2020; 21:ijms21124389. [PMID: 32575639 PMCID: PMC7352407 DOI: 10.3390/ijms21124389] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/14/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022] Open
Abstract
Adult stem cells have been developed as therapeutics for tissue regeneration and immune regulation due to their self-renewing, differentiating, and paracrine functions. Recently, a variety of adult stem cells from the oral cavity have been discovered, and these dental stem cells mostly exhibit the characteristics of mesenchymal stem cells (MSCs). Dental MSCs can be applied for the replacement of dental and oral tissues against various tissue-damaging conditions including dental caries, periodontitis, and oral cancers, as well as for systemic regulation of excessive inflammation in immune disorders, such as autoimmune diseases and hypersensitivity. Therefore, in this review, we summarized and updated the types of dental stem cells and their functions to exert therapeutic efficacy against diseases.
Collapse
|
154
|
Wiese MD, Manning-Bennett AT, Abuhelwa AY. Investigational IRAK-4 inhibitors for the treatment of rheumatoid arthritis. Expert Opin Investig Drugs 2020; 29:475-482. [PMID: 32255710 DOI: 10.1080/13543784.2020.1752660] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is a chronic inflammatory auto-immune disease that can lead to permanent disability and deformity. Despite current treatment modalities, many patients are still unable to reach remission. Interleukin-1 receptor-associated kinase 4 (IRAK-4) inhibitors are novel agents designed to suppress immune signaling pathways involved in inflammation and joint destruction in RA. Four IRAK-4 inhibitors have entered clinical trials. AREAS COVERED This review summarizes the current stage of development of IRAK-4 inhibitors in clinical trials, detailing their chemistry, pharmacokinetics, and therapeutic potential in the treatment of RA. PubMed, Embase and restricted Google searches were conducted using the term 'IRAK-4', and publicly accessible clinical trial databases were reviewed. EXPERT OPINION IRAK-4 inhibitors are an exciting therapeutic option in RA management because unlike other targeted disease-modifying agents, they target the innate immune system. The role of IRAK-4 as a key component of Toll/Interleukin-1 receptor signaling and its potential for a low rate of infectious complications is particularly exciting and this may facilitate their use in combination treatment. A key aspect of upcoming clinical trials will be the identification of biomarkers predictive of treatment efficacy, which will help to define if and how they will be used in the clinic.
Collapse
Affiliation(s)
- Michael D Wiese
- Clinical and Health Sciences, University of South Australia , Adelaide, Australia.,Health and Biomedical Innovation Group, University of South Australia , Adelaide, Australia
| | - Arkady T Manning-Bennett
- Clinical and Health Sciences, University of South Australia , Adelaide, Australia.,Health and Biomedical Innovation Group, University of South Australia , Adelaide, Australia
| | - Ahmad Y Abuhelwa
- Clinical and Health Sciences, University of South Australia , Adelaide, Australia.,Australian Centre for Precision Medicine, Cancer Research Institute, University of South Australia , Adelaide, Australia
| |
Collapse
|
155
|
Song X, Zhang Y, Dai E. Therapeutic targets of thunder god vine (Tripterygium wilfordii hook) in rheumatoid arthritis (Review). Mol Med Rep 2020; 21:2303-2310. [PMID: 32323812 DOI: 10.3892/mmr.2020.11052] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 03/02/2020] [Indexed: 11/05/2022] Open
Abstract
Celastrol and triptolide, chemical compounds isolated from Tripterygium wilfordii hook (also known as thunder god vine), are effective against rheumatoid arthritis (RA). Celastrol targets numerous signaling pathways involving NF‑κB, endoplasmic reticulum Ca2+‑ATPase, myeloid differentiation factor 2, toll‑like receptor 4, pro‑inflammatory chemokines, DNA damage, cell cycle arrest and apoptosis. Triptolide, inhibits NF‑κB, the receptor activator of NF‑κB (RANK)/RANK ligand/osteoprotegerin signaling pathway, cyclooxygenase‑2, matrix metalloproteases and cytokines. The present review examined the chemistry and bioavailability of celastrol and triptolide, and their molecular targets in treating RA. Clinical studies have demonstrated that T. wilfordii has several promising bioactivities, but its multi‑target toxicity has restricted its application. Thus, dosage control and structural modification of T. wilfordii are required to reduce the toxicity. In this review, future directions for research into these promising natural products are discussed.
Collapse
Affiliation(s)
- Xinqiang Song
- Department of Biological Sciences, Xinyang Normal University, Xinyang, Henan 464000, P.R. China
| | - Yu Zhang
- Department of Biological Sciences, Xinyang Normal University, Xinyang, Henan 464000, P.R. China
| | - Erqin Dai
- Department of Biological Sciences, Xinyang Normal University, Xinyang, Henan 464000, P.R. China
| |
Collapse
|
156
|
Hosseini A, Gharibi T, Marofi F, Javadian M, Babaloo Z, Baradaran B. Janus kinase inhibitors: A therapeutic strategy for cancer and autoimmune diseases. J Cell Physiol 2020; 235:5903-5924. [DOI: 10.1002/jcp.29593] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 01/08/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Arezoo Hosseini
- Immunology Research CenterTabriz University of Medical SciencesTabriz Iran
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
- Student Research CommitteeTabriz University of Medical SciencesTabriz Iran
- Aging Research InstituteTabriz University of Medical SciencesTabriz Iran
| | - Tohid Gharibi
- Immunology Research CenterTabriz University of Medical SciencesTabriz Iran
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
- Student Research CommitteeTabriz University of Medical SciencesTabriz Iran
- Aging Research InstituteTabriz University of Medical SciencesTabriz Iran
| | - Faroogh Marofi
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
| | - Mahsa Javadian
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
| | - Zohreh Babaloo
- Immunology Research CenterTabriz University of Medical SciencesTabriz Iran
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
| | - Behzad Baradaran
- Immunology Research CenterTabriz University of Medical SciencesTabriz Iran
- Department of Immunology, School of MedicineTabriz University of Medical SciencesTabriz Iran
| |
Collapse
|
157
|
Tseng CC, Chen YJ, Chang WA, Tsai WC, Ou TT, Wu CC, Sung WY, Yen JH, Kuo PL. Dual Role of Chondrocytes in Rheumatoid Arthritis: The Chicken and the Egg. Int J Mol Sci 2020; 21:E1071. [PMID: 32041125 PMCID: PMC7038065 DOI: 10.3390/ijms21031071] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 12/22/2022] Open
Abstract
Rheumatoid arthritis (RA) is one of the inflammatory joint diseases that display features of articular cartilage destruction. The underlying disturbance results from immune dysregulation that directly and indirectly influence chondrocyte physiology. In the last years, significant evidence inferred from studies in vitro and in the animal model offered a more holistic vision of chondrocytes in RA. Chondrocytes, despite being one of injured cells in RA, also undergo molecular alterations to actively participate in inflammation and matrix destruction in the human rheumatoid joint. This review covers current knowledge about the specific cellular and biochemical mechanisms that account for the chondrocyte signatures of RA and its potential applications for diagnosis and prognosis in RA.
Collapse
Affiliation(s)
- Chia-Chun Tseng
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.T.); (Y.-J.C.); (W.-A.C.)
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (W.-C.T.); (T.-T.O.); (C.-C.W.); (W.-Y.S.)
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.T.); (Y.-J.C.); (W.-A.C.)
- Department of Physical Medicine and Rehabilitation, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Wei-An Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.T.); (Y.-J.C.); (W.-A.C.)
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan
| | - Wen-Chan Tsai
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (W.-C.T.); (T.-T.O.); (C.-C.W.); (W.-Y.S.)
| | - Tsan-Teng Ou
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (W.-C.T.); (T.-T.O.); (C.-C.W.); (W.-Y.S.)
| | - Cheng-Chin Wu
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (W.-C.T.); (T.-T.O.); (C.-C.W.); (W.-Y.S.)
| | - Wan-Yu Sung
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (W.-C.T.); (T.-T.O.); (C.-C.W.); (W.-Y.S.)
| | - Jeng-Hsien Yen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.T.); (Y.-J.C.); (W.-A.C.)
- Division of Rheumatology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan; (W.-C.T.); (T.-T.O.); (C.-C.W.); (W.-Y.S.)
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (C.-C.T.); (Y.-J.C.); (W.-A.C.)
- Institute of Biomedical Science, National Sun Yat-Sen University, Kaohsiung 80424, Taiwan
| |
Collapse
|
158
|
Reich K, Warren R, Coates L, Di Comite G. Long‐term efficacy and safety of secukinumab in the treatment of the multiple manifestations of psoriatic disease. J Eur Acad Dermatol Venereol 2020; 34:1161-1173. [DOI: 10.1111/jdv.16124] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 11/19/2019] [Indexed: 12/12/2022]
Affiliation(s)
- K. Reich
- Translational Research in Inflammatory Skin Diseases Institute for Health Services Research in Dermatology and Nursing University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Skinflammation® Center Hamburg Germany
- Dermatologikum Berlin Berlin Germany
| | - R.B. Warren
- Dermatology Centre Salford Royal NHS Foundation Trust Manchester NIHR Biomedical Research Centre Manchester Academic Health Science Centre The University of Manchester Manchester UK
| | - L.C. Coates
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences University of Oxford Oxford UK
| | | |
Collapse
|
159
|
Min HK, Kim KW, Lee SH, Kim HR. Roles of mast cells in rheumatoid arthritis. Korean J Intern Med 2020; 35:12-24. [PMID: 31722515 PMCID: PMC6960056 DOI: 10.3904/kjim.2019.271] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/08/2019] [Indexed: 12/11/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory arthritis, and the complex interaction and activation of innate and adaptive immune cells are involved in RA pathogenesis. Mast cells (MCs) are one of the tissue-resident innate immune cells, and they contribute to RA pathogenesis. In the present review, the evidence of the pathologic role of MC in RA is discussed based on human and animal data. In addition, the potential role of MC in RA pathogenesis and the research area that should be focused on in the future are suggested.
Collapse
Affiliation(s)
- Hong Ki Min
- Division of Rheumatology, Department of Internal Medicine, Konkuk University Medical Center, Seoul, Korea
| | - Kyoung-Woon Kim
- Conversant Research Consortium in Immunologic Disease, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Korea
| | - Sang-Heon Lee
- Division of Rheumatology, Department of Internal Medicine, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea
| | - Hae-Rim Kim
- Division of Rheumatology, Department of Internal Medicine, Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Korea
- Correspondence to Hae-Rim Kim, M.D. Division of Rheumatology, Department of Internal Medicine, Konkuk University Medical Center, 120-1 Neungdong-ro, Gwangjin-gu, Seoul 05030, Korea Tel: +82-2-2030-7542, Fax: +82-2-2030-7728, E-mail:
| |
Collapse
|
160
|
Singh V, Kalliolias GD, Ostaszewski M, Veyssiere M, Pilalis E, Gawron P, Mazein A, Bonnet E, Petit-Teixeira E, Niarakis A. RA-map: building a state-of-the-art interactive knowledge base for rheumatoid arthritis. Database (Oxford) 2020; 2020:baaa017. [PMID: 32311035 PMCID: PMC7170216 DOI: 10.1093/database/baaa017] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/21/2020] [Accepted: 02/13/2020] [Indexed: 02/07/2023]
Abstract
Rheumatoid arthritis (RA) is a progressive, inflammatory autoimmune disease of unknown aetiology. The complex mechanism of aetiopathogenesis, progress and chronicity of the disease involves genetic, epigenetic and environmental factors. To understand the molecular mechanisms underlying disease phenotypes, one has to place implicated factors in their functional context. However, integration and organization of such data in a systematic manner remains a challenging task. Molecular maps are widely used in biology to provide a useful and intuitive way of depicting a variety of biological processes and disease mechanisms. Recent large-scale collaborative efforts such as the Disease Maps Project demonstrate the utility of such maps as versatile tools to organize and formalize disease-specific knowledge in a comprehensive way, both human and machine-readable. We present a systematic effort to construct a fully annotated, expert validated, state-of-the-art knowledge base for RA in the form of a molecular map. The RA map illustrates molecular and signalling pathways implicated in the disease. Signal transduction is depicted from receptors to the nucleus using the Systems Biology Graphical Notation (SBGN) standard representation. High-quality manual curation, use of only human-specific studies and focus on small-scale experiments aim to limit false positives in the map. The state-of-the-art molecular map for RA, using information from 353 peer-reviewed scientific publications, comprises 506 species, 446 reactions and 8 phenotypes. The species in the map are classified to 303 proteins, 61 complexes, 106 genes, 106 RNA entities, 2 ions and 7 simple molecules. The RA map is available online at ramap.elixir-luxembourg.org as an open-access knowledge base allowing for easy navigation and search of molecular pathways implicated in the disease. Furthermore, the RA map can serve as a template for omics data visualization.
Collapse
Affiliation(s)
- Vidisha Singh
- Laboratoire Européen de Recherche pour la Polyarthrite Rhumatoïde - Genhotel, Univ Evry, Université Paris-Saclay, 2, rue Gaston Crémieux, 91057 EVRY-GENOPOLE cedex, Evry, France
| | - George D Kalliolias
- Arthritis and Tissue Degeneration Program, Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA
- Weill Cornell Medical Center, Weill Department of Medicine, 525 East 68th Street, New York, NY 10065, USA
| | - Marek Ostaszewski
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, L-4367 Belvaux, Luxembourg
| | - Maëva Veyssiere
- Laboratoire Européen de Recherche pour la Polyarthrite Rhumatoïde - Genhotel, Univ Evry, Université Paris-Saclay, 2, rue Gaston Crémieux, 91057 EVRY-GENOPOLE cedex, Evry, France
| | - Eleftherios Pilalis
- eNIOS Applications P.C., R&D department, Alexandrou Pantou 25, 17671, Kallithea-Athens, Greece
| | - Piotr Gawron
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, L-4367 Belvaux, Luxembourg
| | - Alexander Mazein
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6 Avenue du Swing, L-4367 Belvaux, Luxembourg
| | - Eric Bonnet
- Centre National de Recherche en Génomique Humaine (CNRGH), CEA, 2 rue Gaston Crémieux, CP5706 91057 EVRY-GENOPOLE cedex, Evry, France
| | - Elisabeth Petit-Teixeira
- Laboratoire Européen de Recherche pour la Polyarthrite Rhumatoïde - Genhotel, Univ Evry, Université Paris-Saclay, 2, rue Gaston Crémieux, 91057 EVRY-GENOPOLE cedex, Evry, France
| | - Anna Niarakis
- Laboratoire Européen de Recherche pour la Polyarthrite Rhumatoïde - Genhotel, Univ Evry, Université Paris-Saclay, 2, rue Gaston Crémieux, 91057 EVRY-GENOPOLE cedex, Evry, France
| |
Collapse
|
161
|
Osteoarthritis-related biomarkers profile in chronic anterior cruciate ligament injured knee. Knee 2020; 27:51-60. [PMID: 31926672 DOI: 10.1016/j.knee.2019.12.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 11/12/2019] [Accepted: 12/17/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Increasing evidence has shown the involvement of the innate immune system and inflammatory response in osteoarthritis (OA) pathogenesis; however, anterior cruciate ligament (ACL) tears are recognized risk factors for development of post-traumatic OA. We investigated (1) whether inflammatory mediators involved in OA pathogenesis are also present at significant concentrations in the knee joint sometime after ACL complete tear and may be considered as prognostic biomarkers of progression to secondary OA; and (2) whether quantification in serum may surrogate synovial fluid (SF) measurements in both cases. METHODS Thirty-seven end-stage OA patients and 33 patients with ACL complete tear that were included on the waiting list for knee surgery were consecutively recruited. Serum and SF samples were taken before surgery, and tumor necrosis factor-alpha, (TNF-α), interleukin-6 (IL-6), interleukin-8 (IL-8), matrix metalloproteinase-1 (MMP1), matrix metalloproteinase-3 (MMP3), tissue inhibitor of metalloproteinase-1 (TIMP1), bone morphogenetic protein-7 (BMP7), regulated upon activation normal t-cell expressed and secreted (RANTES), cytokine interferon-γ-induced protein 10 (IP-10) and heat shock protein family A (Hsp70) member 1A (HSPA1A) were quantified by enzyme-linked immunosorbent assay (ELISA.) Normally distributed data were compared using a one-way analysis of variance (ANOVA) test. Data not normally distributed were analyzed using a nonparametric Mann-Whitney rank sum test. Differences in means were compared using a Student's t-test. Correlations were analyzed using Pearson's coefficient of variation. RESULTS Eighty-seven percent of patients with OA and 86% of those with ACL tear had quantifiable levels of biomarkers in SF. SF levels of IL-6, IL-8, MMP1, MMP3, RANTES, IP-10, BMP7 and HSPA1A were significantly lower in ACL injury knees compared with those with OA, but much higher than those reported in control subjects. Serum levels of IL-6, IP-10, and MMP1 were also lower in patients with ACL tears, who had, however, significantly higher TNF-α, HSPA1A, and TIMP1 levels when compared with OA patients. Levels of biomarkers tested in serum and SF samples were significantly different. CONCLUSIONS Our data propose that cytokines IL-6 and IL-8 and the chemokines RANTES, IP-10, MMP1, MMP3, and HSPA1A may be involved in the inflammatory process leading to synovitis, the central lesion in OA onset and development; persistent high levels of these substances sometime after ACL injury suggest that they could play an etiopathogenic role in the maintenance of the inflammatory environment leading to post-traumatic OA. Serum biomarker levels do not appear to faithfully reflect what occurs inside the joint. Thus, most biomarkers cannot yet be considered as useful inflammatory biomarkers of knee joint diseases.
Collapse
|
162
|
Pedrosa M, Gomes J, Laranjeira P, Duarte C, Pedreiro S, Antunes B, Ribeiro T, Santos F, Martinho A, Fardilha M, Domingues MR, Abecasis M, P da Silva JA, Paiva A. Immunomodulatory effect of human bone marrow-derived mesenchymal stromal/stem cells on peripheral blood T cells from rheumatoid arthritis patients. J Tissue Eng Regen Med 2019; 14:16-28. [PMID: 31502378 DOI: 10.1002/term.2958] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/04/2019] [Accepted: 09/03/2019] [Indexed: 12/18/2022]
Abstract
Rheumatoid arthritis (RA) is a Th1/Th17-mediated autoimmune disease whose current treatment, consisting in the blockage of inflammatory cytokines by disease-modifying antirheumatic drugs, is not effective for all patients. The therapeutic potential of mesenchymal stromal/stem cells' (MSCs) immunomodulatory properties is being explored in RA. Here, we investigate the effect of human bone marrow (BM)-MSCs on the expression of cytokines involved in RA physiopathology by the distinct functional compartments of CD4+ and CD8+ T cells from RA patients. Peripheral blood mononuclear cells from healthy individuals (n = 6) and RA patients (n = 12) were stimulated with phorbol myristate acetate plus ionomycin and cultured in the presence/absence of BM-MSCs. The expression of (interleukin) IL-2, tumor necrosis factor alpha (TNF-α), and interferon-gamma (IFN-γ) was evaluated in naive, central memory, effector memory, and effector CD4+ and CD8+ T cells, whereas IL-6, IL-9, and IL-17 expression was measured in total CD4+ and CD8+ T cells. mRNA expression of IL-4, IL-10, transforming growth factor beta (TGF-β), cytotoxic T-lymphocyte-associated antigen 4, and/or forkhead box P3 was quantified in fluorescence-activated cell sorting-purified CD4+ T cells, CD8+ T cells, and CD4+ Treg. BM-MSCs inhibited the production of TNF-α, IL-17, IL-6, IL-2, IFN-γ, and IL-9 by T cells from RA patients, mainly by reducing the percentage of cells producing cytokines. This inhibitory effect was transversal to all T cell subsets analyzed. At mRNA level, BM-MSCs increased expression of IL-10 and TGF-β by CD4+ and CD8+ T cells. BM-MSCs displayed a striking inhibitory action over T cells from RA patients, reducing the expression of cytokines involved in RA physiopathology. Remarkably, BM-MSC-derived immunomodulation affected either naive, effector, and memory T cells.
Collapse
Affiliation(s)
- Mónia Pedrosa
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal.,Signal Transduction Laboratory, Center of Cellular Biology, SACS and Department of Biology, University of Aveiro, Aveiro, Portugal
| | - Joana Gomes
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal.,Mass Spectrometry Center, QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Paula Laranjeira
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal
| | - Cátia Duarte
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal.,Rheumatology Department, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Susana Pedreiro
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal
| | | | - Tânia Ribeiro
- Cell2B Advanced Therapeutics, SA, Cantanhede, Portugal
| | - Francisco Santos
- Cell2B Advanced Therapeutics, SA, Cantanhede, Portugal.,Stemlab SA, Cantanhede, Portugal
| | - António Martinho
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal
| | - Margarida Fardilha
- Signal Transduction Laboratory, Center of Cellular Biology, SACS and Department of Biology, University of Aveiro, Aveiro, Portugal.,Laboratory of Signal Transduction, Institute of Biomedicine-iBiMED, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - M Rosário Domingues
- Mass Spectrometry Center, QOPNA, Department of Chemistry, University of Aveiro, Aveiro, Portugal.,Departament of Biology & CESAM, University of Aveiro, Aveiro, Portugal
| | - Manuel Abecasis
- Serviço de Transplantação de Progenitores Hematopoiéticos (UTM), Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa, Portugal
| | - José António P da Silva
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal.,Rheumatology Department, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Artur Paiva
- Centro do Sangue e da Transplantação de Coimbra, Instituto Português do Sangue e da Transplantação, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,CNC.IBILI, University of Coimbra, Coimbra, Portugal.,Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Instituto Politecnico de Coimbra, ESTESC-Coimbra Health School, Ciencias Biomedicas Laboratoriais, Coimbra, Portugal
| |
Collapse
|
163
|
Xie X, Li H, Wang Y, Wan Z, Luo S, Zhao Z, Liu J, Wu X, Li X, Li X. Therapeutic effects of gentiopicroside on adjuvant-induced arthritis by inhibiting inflammation and oxidative stress in rats. Int Immunopharmacol 2019; 76:105840. [DOI: 10.1016/j.intimp.2019.105840] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/18/2019] [Accepted: 08/19/2019] [Indexed: 12/19/2022]
|
164
|
Hsu T, Nguyen-Tran HH, Trojanowska M. Active roles of dysfunctional vascular endothelium in fibrosis and cancer. J Biomed Sci 2019; 26:86. [PMID: 31656195 PMCID: PMC6816223 DOI: 10.1186/s12929-019-0580-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/09/2019] [Indexed: 12/15/2022] Open
Abstract
Chronic inflammation is the underlying pathological condition that results in fibrotic diseases. More recently, many forms of cancer have also been linked to chronic tissue inflammation. While stromal immune cells and myofibroblasts have been recognized as major contributors of cytokines and growth factors that foster the formation of fibrotic tissue, the endothelium has traditionally been regarded as a passive player in the pathogenic process, or even as a barrier since it provides a physical divide between the circulating immune cells and the inflamed tissues. Recent findings, however, have indicated that endothelial cells in fact play a crucial role in the inflammatory response. Endothelial cells can be activated by cytokine signaling and express inflammatory markers, which can sustain or exacerbate the inflammatory process. For example, the activated endothelium can recruit and activate leukocytes, thus perpetuating tissue inflammation, while sustained stimulation of endothelial cells may lead to endothelial-to-mesenchymal transition that contributes to fibrosis. Since chronic inflammation has now been recognized as a significant contributing factor to tumorigenesis, it has also emerged that activation of endothelium also occurs in the tumor microenvironment. This review summarizes recent findings characterizing the molecular and cellular changes in the vascular endothelium that contribute to tissue fibrosis, and potentially to cancer formation.
Collapse
Affiliation(s)
- Tien Hsu
- Department of Biomedical Sciences and Engineering, National Central University, 300 Jhongda Rd, Taoyuan City, Taiwan, Republic of China. .,Center for Chronic Disease Research, National Central University, 300 Jhongda Rd, Taoyuan City, Taiwan, Republic of China.
| | - Hieu-Huy Nguyen-Tran
- Department of Biomedical Sciences and Engineering, National Central University, 300 Jhongda Rd, Taoyuan City, Taiwan, Republic of China
| | - Maria Trojanowska
- Arthritis Center, Boston University School of Medicine, 75 E. Newton St. Evans Building, Boston, MA, 02118, USA
| |
Collapse
|
165
|
Kim B, Kim KH, Chang Y, Shin S, Shin EC, Choi S. One-Step Microfluidic Purification of White Blood Cells from Whole Blood for Immunophenotyping. Anal Chem 2019; 91:13230-13236. [DOI: 10.1021/acs.analchem.9b03673] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Byeongyeon Kim
- Department of Biomedical Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Kyung Hwan Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Yunjung Chang
- Department of Biomedical Engineering, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Suyeon Shin
- Department of Biomedical Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Sungyoung Choi
- Department of Biomedical Engineering, Hanyang University, Seoul 04763, Republic of Korea
| |
Collapse
|
166
|
Huang J, Zeng T, Zhang X, Tian Y, Wu Y, Yu J, Pei Z, Liu Y, Hu T, Tan L. Clinical diagnostic significance of 14-3-3η protein, high-mobility group box-1, anti-cyclic citrullinated peptide antibodies, anti-mutated citrullinated vimentin antibodies and rheumatoid factor in rheumatoid arthritis. Br J Biomed Sci 2019; 77:19-23. [PMID: 31433746 DOI: 10.1080/09674845.2019.1658425] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Introduction: Circulating markers of rheumatoid arthritis (RA) include the 14-3-3η protein, high-mobility group box-1 (HMGB1), anti-cyclic citrullinated peptide (anti-CCP) antibodies, anti-mutated citrullinated vimentin (anti-MCV) antibodies and rheumatoid factor (RF). We set out to determine which two markers in combination provided best discriminatory power for this disease.Methods: We recruited 108 RA patients, 102 non-RA patients (SLE, AS, Sjogren's syndrome, MCTD) and 90 healthy controls. Sensitivity, specificity, positive predictive value, negative predictive value, positive likelihood ratio, negative likelihood ratio and the Youden index of each analyte were calculated and binary logistic regression analysis and receiver operating characteristic (ROC) curve were performed to evaluate their diagnostic value for RA alone and in paired combination.Results: As expected, all markers were elevated in RA patients (P < 0.05). Binary logistic regression analysis showed that 14-3-3η had the highest odds ratio (95% CI) at 2.4 (1.9-2.8). Anti-CCP and anti-MCV had the highest areas under the curves [AUC (95% CI)] at 0.85 (0.78-0.90) and 0.85 (0.78-0.91) respectively (both P < 0.001). In serial detection (one marker followed by another), no combination had a Youden index >0.6. In parallel analysis (both considered together) several combinations had a Youden index >0.7, of which the highest (0.78) was anti-CCP with anti-MCV, with a sensitivity of 93.3% and specificity of 84.7%.Conclusions: Despite individual increases in serum 14-3-3η, HMGB1, anti-CCP, anti-MCV and RF, the combination of anti-CCP and anti-MCV might be of great help for diagnostic in RA, and so should be considered as routine tests for this disease.
Collapse
Affiliation(s)
- J Huang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, China
| | - T Zeng
- The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - X Zhang
- The Third Hospital of Nanchang City Jiangxi Province, Nanchang, Jiangxi, China
| | - Y Tian
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, China
| | - Y Wu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, China
| | - J Yu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, China
| | - Z Pei
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, China
| | - Y Liu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, China
| | - T Hu
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, China
| | - L Tan
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Jiangxi Province Key Laboratory of Laboratory Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
167
|
Li Y, Xing Q, Wei Y, Zhao L, Zhang P, Han X, Wang J. Activation of RXR by bexarotene inhibits inflammatory conditions in human rheumatoid arthritis fibroblast‑like synoviocytes. Int J Mol Med 2019; 44:1963-1970. [PMID: 31545398 DOI: 10.3892/ijmm.2019.4336] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 06/26/2019] [Indexed: 11/06/2022] Open
Abstract
Rheumatoid arthritis (RA) is a debilitating joint disease characterized by chronic inflammation, pathologic alteration of fibroblast‑like synoviocytes (FLS), destruction of cartilage and bone, and the formation of an invasive pannus. RA‑FLS exhibit increased proliferation and resistance to apoptosis. The retinoid X receptor (RXR) has a role in regulating cell cycle, differentiation and apoptosis, and agonism of RXR has been investigated as a treatment strategy in several types of cancer. However, there is little research on the effects of RXR agonism in other diseases. Bexarotene is a novel selective RXR ligand used in the treatment of T‑cell lymphoma. In the present study, bexarotene was used to investigate the involvement of RXR in tumor necrosis factor‑α (TNF‑α)‑induced RA conditions in human FLS. To the best of our knowledge, this is the first time that RXR has been demonstrated to be expressed in FLS and to be downregulated in response to TNF‑α stimulation. The present study also demonstrated that bexarotene exerted an anti‑inflammatory effect by downregulating expression of interleukin (IL)‑6, IL‑8, monocyte chemoattractant protein‑1, and high mobility group box‑1. Notably, bexarotene also rescued the TNF‑α‑induced downregulation of the anti‑inflammatory cytokines IL‑4 and transforming growth factor‑β1. Bexarotene treatment exhibited a potential protective effect against cartilage degradation by downregulating the expression of matrix metalloproteinase (MMP)‑1, MMP‑3 and MMP‑13. In addition, the present results demonstrated that the effects of bexarotene were mediated through the p38 mitogen‑activated protein kinase/nuclear factor‑κB pathway, via inhibition of p38 protein and the inhibitor α of κB phosphorylation. Taken together, the present findings demonstrated the potential of RXR agonism using bexarotene as a treatment against the development and progression of RA.
Collapse
Affiliation(s)
- Yu Li
- Ache Department, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Qunzhi Xing
- Department of Anesthesiology, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Yuanzhang Wei
- Ache Department, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Lei Zhao
- Ache Department, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Pei Zhang
- Ache Department, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Xuechang Han
- Department of Anesthesiology, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Jing Wang
- Ache Department, First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| |
Collapse
|
168
|
The RNA-Binding Protein KSRP Modulates Cytokine Expression of CD4 + T Cells. J Immunol Res 2019; 2019:4726532. [PMID: 31511826 PMCID: PMC6714327 DOI: 10.1155/2019/4726532] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 05/13/2019] [Accepted: 06/18/2019] [Indexed: 01/09/2023] Open
Abstract
The KH-type splicing regulatory protein (KSRP) is a RNA-binding protein, which regulates the stability of many mRNAs encoding immune-relevant proteins. As KSRP regulates innate immune responses, for instance by the modulation of type I interferon mRNA stability, we were interested whether knockdown of the protein (KSRP−/−) interferes with T cell activation and polarization. Polyclonally stimulated KSRP−/− CD4+ T cells proliferated at a higher extent and higher frequency and expressed the activation marker CD25 more than wild-type T cells. In supernatants of stimulated KSRP−/− CD4+ T cells, levels of IL-5, IL-9, IL-10, and IL-13 were observed to be increased compared to those of the control group. KSRP−/− CD8+ T cells showed no altered proliferative capacity upon polyclonal stimulation, but supernatants contained lower levels of interferon-γ. Similar changes in the cytokine expression patterns were also detected in T cells derived from KSRP−/− mice undergoing arthritis induction indicative of a pathophysiological role of KSRP-dependent T cell polarization. We demonstrated the direct binding of KSRP to the 3′ untranslated region of IL-13, IL-10, and IFN-γ mRNA in in vitro experiments. Moreover, since IL-4 mRNA decay was reduced in KSRP−/− CD4+ T cells, we identify KSRP as a negative regulator of IL-4 expression. These data indicate that overexpression of IL-4, which constitutes the primary inducer of Th2 polarization, may cause the Th2 bias of polyclonally stimulated KSRP−/− CD4+ T cells. This is the first report demonstrating that KSRP is involved in the regulation of T cell responses. We present strong evidence that T cells derived from KSRP−/− mice favor Th2-driven immune responses.
Collapse
|
169
|
Protective effects of Clematichinenoside AR against inflammation and cytotoxicity induced by human tumor necrosis factor-α. Int Immunopharmacol 2019; 75:105563. [PMID: 31408840 DOI: 10.1016/j.intimp.2019.04.010] [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: 04/14/2018] [Revised: 03/31/2019] [Accepted: 04/04/2019] [Indexed: 12/28/2022]
Abstract
Clematichinenoside AR (AR), a major active ingredient extracted from traditional Chinese herb Clematis chinensis Osbeck, has been demonstrated to possess anti-inflammatory and immune-modulatory activities in the treatment of experimental rheumatoid arthritis (RA). The therapeutic potential of AR was supposed to be closely correlated to its ability against tumor necrosis factor-α (TNF-α). Therefore, we aimed to explore the protective effects of Clematichinenoside AR against inflammation and cytotoxicity induced by human TNF-α. AR treatment significantly decreased IL-6 and IL-8 secretion, and attenuated MMP-1 production in human RA-derived fibroblast-like synoviocyte MH7A cells stimulated by recombinant human TNF-α (rhTNF-α). AR might antagonize rhTNF-α-induced responses in MH7A cells through inhibiting p38 and ERK MAPKs signal activation. In TNF-α-sensitive murine fibroblast L929 cells, AR treatment attenuated the proliferation inhibition ratio induced by rhTNF-α/ActD and antagonized rhTNF-α-induced cytotoxicity. The cellular and nuclear morphological alterations in apoptotic characteristics induced by rhTNF-α/ActD in L929 cells were observed to be attenuated by the pretreatment with AR under a phase-contrast and fluorescence microscopy, respectively. The Annexin V-FITC/PI double-staining assay was performed to confirm that AR pretreatment obviously decreased the cell death. The antagonistic effects of AR against rhTNF-α-induced cytotoxicity might be potentially attributed to the degeneration of reactive oxygen species and the increasing of mitochondrial membrane potential, along with the suppression of durative phosphorylation of c-Jun N-terminal kinase (JNK). Collectively, our results indicated that AR antagonizes the inflammatory and cytotoxic activities induced by human TNF-α effectively in vitro, which provided further evidence for a novel mechanism underlying AR for treating RA correlating with excessive TNF-α production.
Collapse
|
170
|
Ma JD, Jing J, Wang JW, Mo YQ, Li QH, Lin JZ, Chen LF, Shao L, Miossec P, Dai L. Activation of the Peroxisome Proliferator-Activated Receptor γ Coactivator 1β/NFATc1 Pathway in Circulating Osteoclast Precursors Associated With Bone Destruction in Rheumatoid Arthritis. Arthritis Rheumatol 2019; 71:1252-1264. [PMID: 30802366 PMCID: PMC6771785 DOI: 10.1002/art.40868] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 02/21/2019] [Indexed: 12/22/2022]
Abstract
Objective Activation of osteoclastogenesis at the bone site in rheumatoid arthritis (RA) is well established. The mechanisms by which circulating osteoclast precursors contribute are still unclear. Peroxisome proliferator–activated receptor γ coactivator 1β (PGC‐1β) is implicated in transcriptional regulation of osteoclastogenesis in mouse models. This study was undertaken to investigate the contribution of PGC‐1β to circulating osteoclast precursors and its link to bone destruction in RA. Methods PGC‐1β expression in RA peripheral blood CD14+ monocytes was increased and showed correlation with joint destruction shown on radiographs. Cells from RA patients or healthy controls were transfected with a lentivirus vector for PGC‐1β gene silencing or overexpression and cultured with macrophage colony‐stimulating factor and RANKL. Bone resorption activity, bone‐degrading enzymes, and signaling molecules were measured in these mature osteoclasts. Results Increased nuclear accumulation of PGC‐1β was observed in RA peripheral blood CD14+ monocytes, and these cells had stronger osteoclastogenesis than in healthy controls. PGC‐1β protein expression was positively correlated with radiographic joint destruction (r = 0.396–0.413; all P < 0.05). PGC‐1β knockdown suppressed (51–82% reduction) the expression of cathepsin K, tartrate‐resistant acid phosphatase (TRAP), and matrix metalloproteinase 9 (MMP‐9), as well as osteoclast differentiation and bone resorption activity. Conversely, PGC‐1β overexpression increased these markers (by 1.5–1.8‐fold) and osteoclastogenesis. VIVIT, an inhibitor of NFATc1 activation, inhibited the effect of overexpressed PGC‐1β by reducing cathepsin K, TRAP, and MMP‐9 expression. Chromatin immunoprecipitation assay and dual‐luciferase reporter gene assay showed PGC‐1β bound to NFATc1 promoter, leading to transcriptional activation. Conclusion Activation of the PGC‐1β/NFATc1 pathway in circulating osteoclast precursors was associated with bone destruction in RA. This may represent a new treatment target.
Collapse
Affiliation(s)
- Jian-Da Ma
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jun Jing
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jun-Wei Wang
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ying-Qian Mo
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qian-Hua Li
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jian-Zi Lin
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Le-Feng Chen
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lan Shao
- First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Pierre Miossec
- University of Lyon and Hospices Civils de Lyon, Lyon, France
| | - Lie Dai
- Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
171
|
Ma JD, Jing J, Wang JW, Yan T, Li QH, Mo YQ, Zheng DH, Gao JL, Nguyen KA, Dai L. A novel function of artesunate on inhibiting migration and invasion of fibroblast-like synoviocytes from rheumatoid arthritis patients. Arthritis Res Ther 2019; 21:153. [PMID: 31234900 PMCID: PMC6591920 DOI: 10.1186/s13075-019-1935-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 06/06/2019] [Indexed: 01/16/2023] Open
Abstract
Introduction Anti-malarial drug artesunate can suppress inflammation and prevent cartilage and bone destruction in collagen-induced arthritis model in rats—suggesting it may be a potent drug for rheumatoid arthritis (RA) therapy. We aimed to investigate its effect on the invasive property of fibroblast-like synoviocytes (FLS) from patients with RA. Methods Synovial tissues were obtained by closed needle biopsy from active RA patients, and FLS were isolated and cultured in vitro. RA-FLS were treated with artesunate at various concentrations, while methotrexate or hydroxychloroquine was employed as comparator drugs. Cell viability, proliferation, cell cycle, apoptosis, migration, invasion, and pseudopodium formation of RA-FLS were assessed by CCK-8 assays, EdU staining, Annexin V-FITC/PI staining, transwell assays, or F-actin staining, respectively. Further, relative changes of expressed proteases were analyzed by Proteome profiler human protease array and verified by quantitative real-time PCR (qPCR), Western blot, and ELISA. The expression of signaling molecules of MAPK, NF-κB, AP-1, and PI3K/Akt pathways were measured by qPCR and Western blot. PDK-1 knockdown by specific inhibitor AR-12 or siRNA transfection was used to verify the pharmacological mechanism of artesunate on RA-FLS. Results Artesunate significantly inhibited the migration and invasion of RA-FLS in a dose-dependent manner with or without TNF-α stimulation. The effect was mediated through artesunate inhibition of MMP-2 and MMP-9 production, and pre-treatment with exogenous MMP-9 reversed the inhibitory effect of artesunate on RA-FLS invasion. Artesunate had a stronger inhibitory effect on migration and invasion of RA-FLS as well as greater anti-inflammatory effect than those of hydroxychloroquine. Similar inhibitory effect was detected between artesunate and methotrexate, and synergy was observed when combined. Mechanistically, artesunate significantly inhibited PDK-1 expression as well as Akt and RSK2 phosphorylation—in a similar manner to PDK-1-specific inhibitor AR-12 or PDK-1 knockdown by siRNA transfection. This inhibition results in suppression of RA-FLS migration and invasion as well as decreased MMP-2 and MMP-9 expression. Conclusions Our study demonstrates artesunate is capable of inhibiting migration and invasion of RA-FLS through suppression of PDK1-induced activation of Akt and RSK2 phosphorylation—suggesting that artesunate may be a potential disease-modifying anti-rheumatic drug for RA. Electronic supplementary material The online version of this article (10.1186/s13075-019-1935-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jian-Da Ma
- Department of Rheumatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Jun Jing
- Department of Rheumatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Jun-Wei Wang
- Department of Rheumatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Tao Yan
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Qian-Hua Li
- Department of Rheumatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Ying-Qian Mo
- Department of Rheumatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Dong-Hui Zheng
- Department of Rheumatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China
| | - Jin-Long Gao
- Institute of Dental Research, Sydney Dental School, The University of Sydney, Sydney, NSW, Australia
| | - Ky-Anh Nguyen
- Institute of Dental Research, Sydney Dental School, The University of Sydney, Sydney, NSW, Australia
| | - Lie Dai
- Department of Rheumatology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, People's Republic of China.
| |
Collapse
|
172
|
Inoue T, Kohno M, Nagahara H, Murakami K, Sagawa T, Kasahara A, Kaneshita S, Kida T, Fujioka K, Wada M, Nakada H, Hla T, Kawahito Y. Upregulation of sphingosine-1-phosphate receptor 3 on fibroblast-like synoviocytes is associated with the development of collagen-induced arthritis via increased interleukin-6 production. PLoS One 2019; 14:e0218090. [PMID: 31173610 PMCID: PMC6555509 DOI: 10.1371/journal.pone.0218090] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/24/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Sphingosine-1-phosphate receptor 3 (S1P3) is one of five receptors for sphingosine-1-phosphate (S1P). S1P/S1P3 signaling is involved in numerous physiological and pathological processes including bone metabolism, sepsis, cancer, and immunity. In rheumatoid arthritis (RA), fibroblast-like synoviocytes (FLSs) are activated by several factors and promote abundant proinflammatory cytokine production and bone destruction. The aim of this study was to investigate whether S1P3 is associated with the development of autoimmune arthritis and the pathogenic function of FLSs. METHODS Wild-type (WT) and S1P3 knockout (S1P3-KO) collagen-induced arthritis (CIA) mice were evaluated with respect to clinical and histological disease severity, along with the levels of anti-collagen antibodies and expression of tumor necrosis factor-α (TNFα) and interleukin-6 (IL-6). S1P3 expression in the synovium was analyzed by real-time reverse-transcription polymerase chain reaction (RT-PCR) and immunofluorescence staining. FLSs isolated from CIA mice were activated with TNFα and S1P3 expression was analyzed by real-time RT-PCR. The role of S1P/S1P3 signaling in activated and non-activated FLSs was investigated by measuring cell proliferation and cyto/chemokine production by real-time RT-PCR and/or enzyme-linked immunosorbent assay. RESULTS Clinical and histological scores, and synovial IL-6 expression were significantly lower in S1P3-KO mice with CIA than in WT mice. Arthritic synovia had higher S1P3 expression than intact synovia and FLSs in arthritic joints expressed S1P3 in vivo. Primary cultured FLSs produced IL-6 in a time-dependent manner in response to S1P stimulation and exhibited higher levels of S1P3 expression after activation with TNFα. S1P3-induced production of IL-6 and MMP-3 was increased in FLSs pre-activated with TNFα. CONCLUSION In this study, we demonstrated that S1P3 expression is associated with the development of autoimmune arthritis via inflammation-induced increases in S1P/S1P3 signaling that increase production of IL-6 in FLSs. Inhibition of S1P/S1P3 signaling could open the door to the development of new therapies for RA.
Collapse
Affiliation(s)
- Takuya Inoue
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masataka Kohno
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
- * E-mail:
| | - Hidetake Nagahara
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ken Murakami
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tomoya Sagawa
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akiko Kasahara
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shunya Kaneshita
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takashi Kida
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kazuki Fujioka
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Makoto Wada
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroshi Nakada
- Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University, Kyoto, Japan
| | - Timothy Hla
- Department of Surgery, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yutaka Kawahito
- Inflammation and Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
173
|
Bai M, He X, He Y, Yuan D, Jin T, Wang L. IL-7R gene polymorphisms among patients with rheumatoid arthritis: A case-control study. Mol Genet Genomic Med 2019; 7:e00738. [PMID: 31131543 PMCID: PMC6625337 DOI: 10.1002/mgg3.738] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/16/2019] [Accepted: 04/18/2019] [Indexed: 12/29/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is the most common inflammatory disease which refers to bony erosions and joint destruction largely caused by genetic factors. Our study aimed to explore whether interleukin‐7 receptor (IL‐7R) gene polymorphisms influenced RA risk in the Han Chinese population. Methods Five single nucleotide polymorphisms (SNPs) in IL‐7R gene were successfully genotyped using Agena MassARRAY platform. The associations between IL‐7R polymorphisms and RA were evaluated by the Chi‐squared test, T test, genetic model analysis, and haplotype analysis. We calculated odds ratios (ORs) and 95% confidence intervals (95% CIs) using logistic regression analysis. Results Rs969129 and rs6451231 in the IL‐7R gene were associated with an increased risk of RA in the allele model (OR = 1.25, 95% CI = 1.05–1.49, p = 0.013; OR = 1.23, 95% CI = 1.03–1.48, p = 0.023), respectively. In the genetic models, rs969129 and rs6451231 were associated with an increased risk of RA. After stratification analysis by age, rs969129 and rs6451231 were associated with an increased risk of RA in patients (age <54). After stratification analysis by gender, rs6451231 was associated with an increased risk of RA in males, while rs969129 was found to be associated with an elevated risk of RA in females. And there was a strong linkage disequilibrium among the four SNPs (rs969129, rs118137916, rs10053847, and rs6451231). Conclusion These results suggested rs969129 and rs6451231 in the IL‐7R gene were associated with an increased risk of RA in the Han Chinese population.
Collapse
Affiliation(s)
- Mei Bai
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, Xianyang, China.,Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous Region, Xianyang, China.,Key Laboratory for Basic Life Science Research of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Xue He
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, Xianyang, China.,Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous Region, Xianyang, China.,Key Laboratory for Basic Life Science Research of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Yongjun He
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, Xianyang, China.,Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous Region, Xianyang, China.,Key Laboratory for Basic Life Science Research of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Dongya Yuan
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, Xianyang, China.,Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous Region, Xianyang, China.,Key Laboratory for Basic Life Science Research of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Tianbo Jin
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, Xianyang, China.,Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous Region, Xianyang, China.,Key Laboratory for Basic Life Science Research of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China.,Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, School of Life Sciences, Northwest University, Xi'an, Shaanxi, China
| | - Li Wang
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, Xianyang, China.,Key Laboratory of High Altitude Environment and Genes Related to Diseases of Tibet Autonomous Region, Xianyang, China.,Key Laboratory for Basic Life Science Research of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, Shaanxi, China
| |
Collapse
|
174
|
Stock AT, Jama HA, Hansen JA, Wicks IP. TNF and IL-1 Play Essential but Temporally Distinct Roles in Driving Cardiac Inflammation in a Murine Model of Kawasaki Disease. THE JOURNAL OF IMMUNOLOGY 2019; 202:3151-3160. [DOI: 10.4049/jimmunol.1801593] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/27/2019] [Indexed: 11/19/2022]
|
175
|
Cellulase-Assisted Extraction, Characterization, and Bioactivity against Rheumatoid Arthritis of Astragalus Polysaccharides. INT J POLYM SCI 2019. [DOI: 10.1155/2019/8514247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
This study investigated the effect of cellulase on the isolation of crude Astragalus polysaccharide (APS), analyzed the monosaccharide component of deproteinized APS, detected the molecular weights of purified APS, and examined the biological activities and the preliminary mechanism against rheumatoid arthritis (RA). Compared with water extraction method, cellulase-assisted extraction increased the yield of crude APS to 154% and polysaccharide contents to 121%. Crude APS was then purified by ethanol precipitation, Sevag deproteinization, and high-performance liquid chromatography (HPLC) analysis; monosaccharide contents of APS were different after cellulase-assisted method, especially galacturonic acid content which significantly increased. DEAE-52 cellulose column chromatography isolated three polysaccharide fractions, including a neutral polysaccharide (APS-water) and two acidic polysaccharides (APS-NaCl1 and APS-NaCl2). Using high-performance gel permeation chromatography (HPGPC), the molecular weights of APS-water, APS-NaCl1, and APS-NaCl2 were identified as 67.7 kDa, 234.1 kDa, and 189.4 kDa, respectively. Then their therapeutic effects and possible mechanism against RA were explored using type II collagen-induced arthritis (CIA) rat model. APS could significantly reduce paw swelling, serum concentration of IL-1β and TNF-α, and the expression levels of NF-κB-p65 and IκBα in synovial membranes in CIA rats. Our study indicated that cellulase significantly increases the yield and polysaccharide contents of crude APS, improves the product quality, and preserves the biological features against RA in CIA rats.
Collapse
|
176
|
Robert M, Miossec P. IL-17 in Rheumatoid Arthritis and Precision Medicine: From Synovitis Expression to Circulating Bioactive Levels. Front Med (Lausanne) 2019; 5:364. [PMID: 30693283 PMCID: PMC6339915 DOI: 10.3389/fmed.2018.00364] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 12/24/2018] [Indexed: 12/17/2022] Open
Abstract
Interleukin (IL)-17A has a direct contribution in early induction and late chronic stages of various inflammatory diseases. In vitro and in vivo experiments have first characterized its local effects on different cell types and then its systemic effects. For instance, IL-17 axis is now identified as a key driver of psoriasis through its effects on keratinocytes. Similar observations apply for rheumatoid arthritis (RA) where IL-17A triggers changes in the synovium that lead to synovitis and maintain local inflammation. These results have prompted the development of biologics to target this cytokine. However, while convincing studies are reported on the efficacy of IL-17 inhibitors in psoriasis, there are conflicting results in RA. Patient heterogeneity but also the involvement of mediators that regulate IL-17 function may explain these results. Therefore, new tools and concepts are required to identify patients that could benefit from these IL-17 targeted therapies in RA and the development of predictive biomarkers of response has started with the emergence of various bioassays. Current strategies are also focusing on synovial biopsies that may be used to stratify patients. From local to systemic levels, new approaches are developing and move the field of RA management into the era of precision medicine.
Collapse
Affiliation(s)
- Marie Robert
- Department of Clinical Immunology and Rheumatology, Immunogenomics and Inflammation Research Unit EA 4130, University of Lyon 1, Hôpital Edouard Herriot, Lyon, France
| | - Pierre Miossec
- Department of Clinical Immunology and Rheumatology, Immunogenomics and Inflammation Research Unit EA 4130, University of Lyon 1, Hôpital Edouard Herriot, Lyon, France
| |
Collapse
|
177
|
Ashton MP, Eugster A, Dietz S, Loebel D, Lindner A, Kuehn D, Taranko AE, Heschel B, Gavrisan A, Ziegler AG, Aringer M, Bonifacio E. Association of Dendritic Cell Signatures With Autoimmune Inflammation Revealed by Single-Cell Profiling. Arthritis Rheumatol 2018; 71:817-828. [PMID: 30511817 DOI: 10.1002/art.40793] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 11/29/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To identify single-cell transcriptional signatures of dendritic cells (DCs) that are associated with autoimmunity, and determine whether those DC signatures are correlated with the clinical heterogeneity of autoimmune disease. METHODS Blood-derived DCs were single-cell sorted from the peripheral blood of patients with rheumatoid arthritis, systemic lupus erythematosus, or type 1 diabetes as well as healthy individuals. DCs were analyzed using single-cell gene expression assays, performed immediately after isolation or after in vitro stimulation of the cells. In addition, protein expression was measured using fluorescence-activated cell sorting. RESULTS CD1c+ conventional DCs and plasmacytoid DCs from healthy individuals exhibited diverse transcriptional signatures, while the DC transcriptional signatures in patients with autoimmune disease were altered. In particular, distinct DC clusters, characterized by up-regulation of TAP1, IRF7, and IFNAR1, were abundant in patients with systemic autoimmune disease, whereas DCs from patients with type 1 diabetes had decreased expression of the regulatory genes PTPN6, TGFB, and TYROBP. The frequency of CD1c+ conventional DCs that expressed a systemic autoimmune profile directly correlated with the extent of disease activity in patients with rheumatoid arthritis (Spearman's r = 0.60, P = 0.03). CONCLUSION DC transcriptional signatures are altered in patients with autoimmune disease and are associated with the level of disease activity, suggesting that immune cell transcriptional profiling could improve our ability to detect and understand the heterogeneity of these diseases, and could guide treatment choices in patients with a complex autoimmune disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Anette-Gabriele Ziegler
- Forschergruppe Diabetes e.V., Institute of Diabetes Research, Helmholtz Zentrum München, and DZD, Neuherberg, Germany
| | | | - Ezio Bonifacio
- TU Dresden, Dresden, Germany, Forschergruppe Diabetes e.V., and DZD, Neuherberg, Germany
| |
Collapse
|
178
|
Zhang Q, Dehaini D, Zhang Y, Zhou J, Chen X, Zhang L, Fang RH, Gao W, Zhang L. Neutrophil membrane-coated nanoparticles inhibit synovial inflammation and alleviate joint damage in inflammatory arthritis. NATURE NANOTECHNOLOGY 2018; 13:1182-1190. [PMID: 30177807 DOI: 10.1038/s41565-018-0254-4] [Citation(s) in RCA: 556] [Impact Index Per Article: 92.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 08/02/2018] [Indexed: 05/18/2023]
Abstract
Rheumatoid arthritis is a common chronic inflammatory disorder and a major cause of disability. Despite the progress made with recent clinical use of anti-cytokine biologics, the response rate of rheumatoid arthritis treatment remains unsatisfactory, owing largely to the complexity of cytokine interactions and the multiplicity of cytokine targets. Here, we show a nanoparticle-based broad-spectrum anti-inflammatory strategy for rheumatoid arthritis management. By fusing neutrophil membrane onto polymeric cores, we prepare neutrophil membrane-coated nanoparticles that inherit the antigenic exterior and associated membrane functions of the source cells, which makes them ideal decoys of neutrophil-targeted biological molecules. It is shown that these nanoparticles can neutralize proinflammatory cytokines, suppress synovial inflammation, target deep into the cartilage matrix, and provide strong chondroprotection against joint damage. In a mouse model of collagen-induced arthritis and a human transgenic mouse model of arthritis, the neutrophil membrane-coated nanoparticles show significant therapeutic efficacy by ameliorating joint damage and suppressing overall arthritis severity.
Collapse
Affiliation(s)
- Qiangzhe Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Diana Dehaini
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Yue Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Julia Zhou
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Xiangyu Chen
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Lifen Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Ronnie H Fang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Weiwei Gao
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Liangfang Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
179
|
IL-6, IL-17 and Stat3 are required for auto-inflammatory syndrome development in mouse. Sci Rep 2018; 8:15783. [PMID: 30361689 PMCID: PMC6202393 DOI: 10.1038/s41598-018-34173-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 10/12/2018] [Indexed: 12/18/2022] Open
Abstract
Auto-inflammatory syndrome, a condition clinically distinct from rheumatoid arthritis, is characterized by systemic inflammation in tissues such as major joints, skin, and internal organs. Autonomous innate-immune activation is thought to promote this inflammation, but underlying pathological mechanisms have not been clarified nor are treatment strategies established. Here, we newly established a mouse model in which IL-1 signaling is conditionally activated in adult mice (hIL-1 cTg) and observed phenotypes similar to those seen in auto-inflammatory syndrome patients. In serum of hIL-1 cTg mice, IL-6 and IL-17 levels significantly increased, and signal transducer and activator of transcription 3 (Stat3) was activated in joints. When we crossed hIL-1 cTg with either IL-6- or IL-17-deficient mice or with Stat3 conditional knockout mice, phenotypes seen in hIL-1 cTg mice were significantly ameliorated. Thus, IL-6, IL-17 and Stat3 all represent potential therapeutic targets for this syndrome.
Collapse
|
180
|
Abstract
Rheumatoid arthritis (RA) is the most important chronic inflammatory joint disease with a prevalence of 1%. When untreated the disease leads to joint destruction and therefore to functional restrictions of the patients and also to increased rates of cardiovascular and malignant diseases. After the pathophysiology of rheumatoid arthritis was better understood, in the last 20 years biologics could be developed, which are directed against targets involved in the inflammatory process in RA. Since then the remission rates of RA have substantially increased. In 2017 Janus kinase (JAK) inhibitors were additionally approved for the treatment of RA in Germany. They further broaden the therapeutic options and, in contrast to biologics, are administered orally. The response rates to therapy are better the earlier the disease is diagnosed and treated. Patients in whom RA is suspected due to a new onset of polyarthritis, should therefore be promptly referred to a rheumatologist.
Collapse
Affiliation(s)
- Torsten Witte
- Klinik für Immunologie und Rheumatologie, Medizinische Hochschule Hannover, Carl-Neuberg-Str. 1, 30625, Hannover, Deutschland.
| |
Collapse
|
181
|
Liu W, Zhang Y, Zhu W, Ma C, Ruan J, Long H, Wang Y. Sinomenine Inhibits the Progression of Rheumatoid Arthritis by Regulating the Secretion of Inflammatory Cytokines and Monocyte/Macrophage Subsets. Front Immunol 2018; 9:2228. [PMID: 30319663 PMCID: PMC6168735 DOI: 10.3389/fimmu.2018.02228] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/07/2018] [Indexed: 12/26/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory arthropathy associated with articular damage and attendant comorbidities. Even although RA treatment has advanced remarkably over the last decade, a significant proportion of patients still do not achieve sustained remission. The cause of RA is not yet known despite the many potential mechanisms proposed. It has been confirmed that RA is associated with dysregulated immune system and persistent inflammation. Therefore, management of inflammation is always the target of therapy. Sinomenine (SIN) is the prescription drug approved by the Chinese government for RA treatment. A previous study found that SIN was a robust anti-inflammation drug. In this study, we screened the different secretory cytokines using inflammation antibody arrays and qRT-PCR in both LPS-induced and SIN-treated RAW264.7 cells followed by evaluation of the ability of SIN to modulate cytokine secretion in a cell model, collagen-induced arthritis (CIA) mouse model, and RA patients. Several clinical indexes affecting the 28-joint disease activity score (DAS28) were determined before and after SIN treatment. Clinical indexes, inflammatory cytokine secretion, and DAS28 were compared among RA patients treated with either SIN or methotrexate (MTX). To explore the mechanism of SIN anti-inflammatory function, RA-associated monocyte/macrophage subsets were determined using flow cytometry in CIA mouse model and RA patients, both treated with SIN. The results demonstrated that SIN regulated IL-6, GM-CSF, IL-12 p40, IL-1α, TNF-α, IL-1β, KC (CXCL1), Eotaxin-2, IL-10, M-CSF, RANTES, and MCP-1 secretion in vivo and in vitro and reduced RA activity and DAS28 in a clinical setting. Furthermore, SIN attenuated CD11b+F4/80+CD64+ resident macrophages in the synovial tissue, CD11b+Ly6C+CD43+ macrophages in the spleen and draining lymph nodes of CIA mice. The percentage of CD14+CD16+ peripheral blood mononuclear cells was reduced by SIN in RA patients. These data indicated that SIN regulates the secretion of multiple inflammatory cytokines and monocyte/macrophage subsets, thereby suppressing RA progression. Therefore, along with MTX, SIN could be an alternative cost-effective anti-inflammatory agent for treating RA.
Collapse
Affiliation(s)
- Weiwei Liu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Yajie Zhang
- Central Laboratory, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Clinical Biobank of Nanjing Hospital of Chinese Medicine, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Weina Zhu
- Central Laboratory, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Clinical Biobank of Nanjing Hospital of Chinese Medicine, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Chunhua Ma
- Central Laboratory, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Clinical Biobank of Nanjing Hospital of Chinese Medicine, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jie Ruan
- Central Laboratory, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Clinical Biobank of Nanjing Hospital of Chinese Medicine, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongyan Long
- Central Laboratory, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Clinical Biobank of Nanjing Hospital of Chinese Medicine, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.,Department of Pediatrics, Nanjing Hospital of Chinese Medicine, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue Wang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China.,The First Clinical Medical School, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
182
|
Wei JL, Fu W, Hettinghouse A, He WJ, Lipson KE, Liu CJ. Role of ADAMTS-12 in Protecting Against Inflammatory Arthritis in Mice By Interacting With and Inactivating Proinflammatory Connective Tissue Growth Factor. Arthritis Rheumatol 2018; 70:1745-1756. [PMID: 29750395 DOI: 10.1002/art.40552] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 05/03/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE It has been reported that ADAMTS-12 is a susceptibility gene for rheumatoid arthritis (RA) development, and its level is significantly increased in RA patients. In addition, ADAMTS-12 is reported to be required for inflammation in otherwise healthy subjects. This study was undertaken to determine the role of ADAMTS-12 and the underlying mechanisms in the pathogenesis of inflammatory arthritis. METHODS The collagen-induced arthritis (CIA) model was established in ADAMTS-12-deficient mice and their control littermates to determine the role of ADAMTS-12 in vivo. Micro-computed tomography scanning was used to demonstrate the destruction of the ankle joint; histologic analysis illustrated synovitis, pannus formation, and bone and cartilage destruction; enzyme-linked immunosorbent assay was performed to measure serum levels of inflammatory cytokines; and protein-protein interaction assays were performed to detect the interactions of ADAMTS-12 and its various deletion mutants with connective tissue growth factor (CTGF). RESULTS Deficiency of ADAMTS-12 led to accelerated inflammatory arthritis in the CIA mouse model. Loss of ADAMTS-12 caused enhanced osteoclastogenesis. In vitro and in vivo protein-protein interaction assays demonstrated that ADAMTS-12 bound and processed CTGF, a previously unrecognized substrate of ADAMTS-12. In addition, deletion of ADAMTS-12 enhanced, while overexpression of ADMATS-12 reduced, CTGF-mediated inflammation. Furthermore, ADAMTS-12 regulation of inflammation was largely lost in CTGF-deficient macrophages. Importantly, blocking of CTGF attenuated elevated inflammatory arthritis seen in the ADAMTS-12-deficient CIA mouse model. CONCLUSION This study provides evidence that ADAMTS-12 is a critical regulator of inflammatory arthritis and that this is mediated, at least in part, through control of CTGF turnover.
Collapse
Affiliation(s)
- Jian-Lu Wei
- New York University Medical Center, New York, New York, and Shandong University Qilu Hospital, Jinan, China
| | - Wenyu Fu
- New York University Medical Center, New York, New York
| | | | - Wen-Jun He
- New York University Medical Center, New York, New York
| | | | - Chuan-Ju Liu
- New York University Medical Center and New York University School of Medicine, New York, New York
| |
Collapse
|
183
|
Li Y, Pan H, Duan H, Chen J, Zhu Z, Fan J, Li P, Yang X, Pan W. Double-layered osmotic pump controlled release tablets of actarit: In vitro and in vivo evaluation. Asian J Pharm Sci 2018; 14:340-348. [PMID: 32104464 PMCID: PMC7032193 DOI: 10.1016/j.ajps.2018.05.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 04/17/2018] [Accepted: 05/17/2018] [Indexed: 12/29/2022] Open
Abstract
The aim of the study was to develop actarit double-layered osmotic pump tablets to overcome the weak points of actarit common tablets, such as short half-life and large plasma concentration fluctuations. Single factor experiment and orthogonal test were applied to optimize the formulation; the pharmacokinetic study was performed in beagle dogs adopting actarit common tablets as reference tablets. The optimal formulation was as follows: drug layer: 150 mg actarit, 240 mg PEO-N80, 50 mg NaCl; push layer: 140 mg PEO-WSR303, 20 mg NaCl; coating solution: 30 g cellulose acetate and 6 g PEG 4000 in 1000 ml 94% acetone solution, 60 mg coating weight gain. The pharmacokinetic study showed that Tmax was prolonged by the contrast of commercial common tablets with constant drug release rate, but the bioavailability was equivalent. And a good in vivo–in vitro correlation of the actarit osmotic pump tablets was also established. The designed actarit osmotic pump tablets can be applied for rheumatoid arthritis, proposing a promising replacement for the marked common products.
Collapse
Affiliation(s)
- Yuenan Li
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang City, Liaoning Province 110016, China
| | - Hao Pan
- Liao Ning University, No. 66, Chongshan Middle Road, Huanggu District, Shenyang City, Liaoning Province 110036, China
| | - Hongliang Duan
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang City, Liaoning Province 110016, China
| | - Jianting Chen
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang City, Liaoning Province 110016, China
| | - Zhihong Zhu
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang City, Liaoning Province 110016, China
| | - Jingxin Fan
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang City, Liaoning Province 110016, China
| | - Pingfei Li
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang City, Liaoning Province 110016, China
| | - Xinggang Yang
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang City, Liaoning Province 110016, China
| | - Weisan Pan
- Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang City, Liaoning Province 110016, China
| |
Collapse
|
184
|
Liu T, Wang X, He YL, Wang Y, Dong L, Ma X, Zheng L, Liu CH, Wang GC, Zheng J, Lan YY, Li YJ. In Vivo and In Vitro Anti-Arthritic Effects of Cardenolide-Rich and Caffeoylquinic Acid-Rich Fractions of Periploca forrestii. Molecules 2018; 23:molecules23081988. [PMID: 30096961 PMCID: PMC6222661 DOI: 10.3390/molecules23081988] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/05/2018] [Accepted: 08/08/2018] [Indexed: 12/29/2022] Open
Abstract
Periploca forrestii Schltr. (P. forrestii) is a species used in Traditional Chinese Medicine (TCM) known as “Miao medicine”, and has a long history of use in the treatment of rheumatism, rheumatoid arthritis (RA), and joint pain. The present study aimed to evaluate the anti-arthritis effects of the cardenolide-rich and caffeoylquinic acid-rich fractions (CDLFs and CQAFs) of P. forrestii in collagen-induced arthritic (CIA) rats, and defined the mechanisms of therapeutic action in MH7A cells treated with TNF-α. Serum rheumatoid factor (RF), TNF-α, IL-6, IL-1β, PGE2, NO, SOD, and MDA were determined by ELISA or other commercially assay kits. Histopathological changes in ankle joint tissues were examined. The mRNA expressions of IL-1β, IL-6, COX-2, and iNOS in MH7A cells were measured by qRT-PCR assays. In addition, the expressions of iNOS, COX-2, and p65 proteins, and the phosphorylation of IκBα, p38, ERK1/2, and JNK proteins in MH7A cells were analyzed by Western blot. The results showed that CDLF and CQAF could suppress the paw swelling in CIA rats at different doses (125 mg/kg, 250 mg/kg, and 500 mg/kg). Histopathological examination suggests that the CDLF and CQAF significantly relieved the damage of the structure of the ankle joint in CIA rats. In addition, serum RF, TNF-α, IL-6, IL-1β, PGE2, NO, and MDA were decreased, along with increased activity of serum SOD. Furthermore, CDLF and CQAF downregulated the expressions of IL-1β, IL-6, COX-2, iNOS, and p65, and inhibited the phosphorylation of IκBα, p38, ERK1/2, and JNK in MH7A cells treated with TNF-α. These findings demonstrated that both CDLF and CQAF exhibited anti-arthritic activity, which might be associated with their inhibitory effects on the NF-κB and MAPK signaling pathways.
Collapse
Affiliation(s)
- Ting Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang 550004, China.
| | - Xia Wang
- Engineering Research Center for the Development and Applications of Ethnic Medicines and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, China.
- School of Pharmacy, Guizhou Medical University, Guiyang 550004, China.
| | - Yan-Ling He
- Engineering Research Center for the Development and Applications of Ethnic Medicines and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, China.
- School of Pharmacy, Guizhou Medical University, Guiyang 550004, China.
| | - Yang Wang
- Engineering Research Center for the Development and Applications of Ethnic Medicines and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, China.
- School of Pharmacy, Guizhou Medical University, Guiyang 550004, China.
| | - Li Dong
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang 550004, China.
| | - Xue Ma
- Engineering Research Center for the Development and Applications of Ethnic Medicines and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, China.
| | - Lin Zheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang 550004, China.
| | - Chun-Hua Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang 550004, China.
| | - Guang-Cheng Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang 550004, China.
| | - Jiang Zheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Key Laboratory of Pharmaceutics of Guizhou Province, Guizhou Medical University, Guiyang 550004, China.
| | - Yan-Yu Lan
- Engineering Research Center for the Development and Applications of Ethnic Medicines and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, China.
| | - Yong-Jun Li
- Engineering Research Center for the Development and Applications of Ethnic Medicines and TCM (Ministry of Education), Guizhou Medical University, Guiyang 550004, China.
| |
Collapse
|
185
|
Collagen-induced arthritis in Dark Agouti rats as a model for study of immunological sexual dimorphisms in the human disease. Exp Mol Pathol 2018; 105:10-22. [DOI: 10.1016/j.yexmp.2018.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 04/05/2018] [Accepted: 05/18/2018] [Indexed: 12/15/2022]
|
186
|
Atayde SR, Velosa APP, Catanozi S, Del Bianco V, Andrade PC, Rodrigues JEDCM, dos Santos Filho A, Antonangelo L, de Mello SBV, Capelozzi VL, Teodoro WR. Collagen V oral administration decreases inflammation and remodeling of synovial membrane in experimental arthritis. PLoS One 2018; 13:e0201106. [PMID: 30059520 PMCID: PMC6066207 DOI: 10.1371/journal.pone.0201106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 06/10/2018] [Indexed: 12/18/2022] Open
Abstract
Because collagen type V (Col V) can be exposed in tissue injury, we hypothesized that oral administration of this collagen species modulates the inflammation and remodeling of experimental synovitis, avoiding joint destruction, and that the modulation may differ according to the temporal administration. Arthritis (IA, n = 20) was induced in Lewis rats by intraarticular (ia) injection of 500 μg of methylated bovine serum albumin (mBSA) emulsified in complete Freund’s adjuvant (CFA) (10 μl) followed by an intraarticular booster of mBSA (50 μg) in saline (50 μl) administered at 7 and 14 days. The control group received saline (50 μl, ia). After the first intraarticular injection, ten IA animals were supplemented via gavage with Col V (500 μg/300 μl) daily for 30 days (IA/Suppl). The control group received saline (50 μL) and Col V supplement in the same way (Suppl). Col V oral administration in IA/Suppl led to 1) inhibited edema and severe inflammatory cell infiltration, 2) decreased collagen fiber content, 3) decreased collagen type I, 4) inhibited lymphocyte subpopulations and macrophages, 5) inhibited IL-1β, IL-10, IL-17 and TNF-α production and 6) increased expression of caspase-9 in the synovial tissue. In conclusion, Col V supplementation decreased synovial inflammation and the fibrotic response, possibly by increased the apoptosis of inflammatory cells.
Collapse
Affiliation(s)
- Silvana Ramos Atayde
- Department of Pathology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, São Paulo, Brazil
- * E-mail:
| | - Ana Paula Pereira Velosa
- Rheumatology Division, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, São Paulo, Brazil
| | - Sergio Catanozi
- Endocrinology Division (LIM 10), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, São Paulo, Brazil
| | - Vanessa Del Bianco
- Endocrinology Division (LIM 10), Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, São Paulo, Brazil
| | - Priscila Cristina Andrade
- Department of Pathology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, São Paulo, Brazil
| | | | - Antonio dos Santos Filho
- Rheumatology Division, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, São Paulo, Brazil
| | - Leila Antonangelo
- Department of Pathology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, São Paulo, Brazil
| | | | - Vera Luiza Capelozzi
- Department of Pathology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, São Paulo, Brazil
| | - Walcy Rosolia Teodoro
- Rheumatology Division, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, São Paulo, Brazil
| |
Collapse
|
187
|
Zhai W, Ma Z, Wang W, Song L, Yi J. Paeoniflorin inhibits Rho kinase activation in joint synovial tissues of rats with collagen-induced rheumatoid arthritis. Biomed Pharmacother 2018; 106:255-259. [PMID: 29966968 DOI: 10.1016/j.biopha.2018.06.130] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 06/15/2018] [Accepted: 06/25/2018] [Indexed: 02/06/2023] Open
Abstract
Paeoniflorin (PF) has many effects, such as anti-inflammation, immune-regulation, abirritation, and so on. However, the protective mechanisms of PF on rheumatoid arthritis (RA) was not completely known. Thus, we explored deeply the protective mechanisms in a collagen-induced RA (CIA) rat model. CIA was induced in rats by intradermal injection of bovine type II collagen in complete Freund's adjuvant. Later, the CIA rats received oral administration of PF (50 and 100 mg/kg) once a day from the day 21, with the treatment lasting for 14 days. A variety of indicators were measured for evaluation of anti-rheumatism effect, including paw swelling, arthritis scores, and histopathological changes. And the contents of pro-inflammatory cytokines, including tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6) in the serum, as well as p-NF-κB p65 and p-MYPT1 in the joint synovial tissues were detected to explore the possible mechanisms. The results demonstrated that PF treatment significantly ameliorated the symptoms in CIA rats, reduced the levels of pro-inflammatory cytokines and paw swelling, down-regulated the expressions of p-NF-κB p65 and p-MYPT1. The present results revealed that PF could effectively improve collagen-induced RA in rats by inhibiting Rho kinase activation in the joint synovial tissues, in turn down-regulating expression of p-NF-κB p65 and reducing contents of pro-inflammatory cytokines. Moreover, PF may be an effective agent for RA.
Collapse
Affiliation(s)
- Wenjing Zhai
- College of Chemical and Biological Engineering, Yichun University, Yichun 336000, China
| | - Zhihong Ma
- Department of Immunology and Pathobiology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Weijie Wang
- Department of Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Lin Song
- Department of Immunology and Pathobiology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Jianfeng Yi
- Key Laboratory for Research on Active Ingredients in Natural Medicine of Jiangxi Province, Yichun University, Yichun 336000, China; The Center for Translational Medicine, Yichun University, Yichun 336000, China.
| |
Collapse
|
188
|
Heluany CS, Kupa LDVK, Viana MN, Fernandes CM, Farsky SHP. Hydroquinone exposure worsens the symptomatology of rheumatoid arthritis. Chem Biol Interact 2018; 291:120-127. [PMID: 29908986 DOI: 10.1016/j.cbi.2018.06.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/07/2018] [Accepted: 06/13/2018] [Indexed: 12/12/2022]
Abstract
The genesis of rheumatoid arthritis (RA) is complex and dependent on genetic background and exposure to environmental xenobiotic. Indeed, smoking is associated to developing and worsening pre-existing RA. Nevertheless, the mechanisms and cigarette compounds involved in the harmful processes have not been elucidated. Here, we investigated if the exposure to hydroquinone (HQ), an abundant pro-oxidative compound of cigarette and benzene metabolite, could worsen the ongoing RA. Hence, collagen-induced arthritis (CIA) was induced in male Wistar rats by s.c. injection of 400 μg (200 μL) of bovine collagen type II emulsified in complete Freund's adjuvant on day 1, and a booster injection was performed on day 7. Exposures to nebulized HQ (25 ppm), saline solution or HQ vehicle solution (5% ethanol in saline) were carried out for 1 h, once a day, on days 21-27 after CIA induction. On day 27, animals were euthanized and samples were collected for further analyses. Exposure to HQ caused loss of weight, intensified paw edema, enhanced levels of tumor necrosis factor-α (TNF-α) and anti-citrullinated protein antibody (ACPA) in the serum; augmented synoviocyte proliferation and influx of aril hydrocarbon receptor (AhR) positive cells into the synovial membrane, altered collagen fibre rearrangement in the synovia, and synoviocytes isolated from HQ exposed rats secreted higher levels of pro-inflammatory cytokines, TNF-α and interleukin-1β. Associated, we point out HQ as an environmental pollutant that aggravates RA, suggesting its participation on worsening RA in smoking patients.
Collapse
Affiliation(s)
- Cintia Scucuglia Heluany
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of São Paulo, Brazil
| | - Leonard de Vinci Kanda Kupa
- Department of Clinical and Toxicological Analysis, Faculty of Pharmaceutical Sciences, University of São Paulo, Brazil
| | | | | | | |
Collapse
|
189
|
Äyräväinen L, Heikkinen AM, Kuuliala A, Ahola K, Koivuniemi R, Laasonen L, Moilanen E, Hämäläinen M, Tervahartiala T, Meurman JH, Leirisalo-Repo M, Sorsa T. Inflammatory biomarkers in saliva and serum of patients with rheumatoid arthritis with respect to periodontal status. Ann Med 2018; 50:333-344. [PMID: 29683364 DOI: 10.1080/07853890.2018.1468922] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE To study prospectively the association of salivary and serum matrix metalloproteinase (MMP)-8, tissue inhibitor of MMPs (TIMP)-1 and interleukin (IL)-6 with periodontal and systemic inflammation in rheumatoid arthritis (RA). We hypothesized that biomarker concentrations reflect inflammation. METHODS Fifty three early untreated RA (ERA) and 28 chronic RA (CRA) patients, underwent rheumatological and dental examinations at baseline and one year later after starting first conventional or biological disease modifying antirheumatic drug. We included 43 control subjects. Saliva and serum samples were analyzed for MMP-8, TIMP-1 and IL-6. Periodontal health was assessed by bleeding on probing (BOP), pocket depth (PD) and periodontal inflammatory burden index (PIBI); RA disease activity was assessed by disease activity score DAS28. Joint destruction was analyzed by the modified Sharp-van der Heijde (SHS) method. RESULTS Serum MMP-8 (p < .001; p < .001) and IL-6 (p < .001; p = .002) were significantly higher in CRA vs. other study groups during the study. Salivary MMP-8 (p = .010) and IL-6 (p = .010) were significantly higher in ERA vs. other study groups at baseline. Salivary MMP-8 was associated with periodontal parameters. CONCLUSION Elevated serum concentrations of MMP-8 and IL-6 in CRA patients reflected chronic RA, while elevated salivary concentrations of MMP-8 levels in ERA patients reflected increased periodontal inflammation. Key messages Concentrations of inflammatory biomarkers in serum and saliva were different between patients with RA and healthy controls. Concentrations of MMP-8 and of IL-6 in serum were elevated in patients with chronic RA reflecting joint inflammation and the burden of established RA. Concentrations of MMP-8 in saliva was elevated already at the early stage of RA and the level of salivary MMP-8 was associated with poor periodontal health both in patients with early and in those with chronic RA.
Collapse
Affiliation(s)
- Leena Äyräväinen
- a Department of Oral and Maxillofacial Diseases , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Anna Maria Heikkinen
- a Department of Oral and Maxillofacial Diseases , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Antti Kuuliala
- b Department of Bacteriology and Immunology , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Kirsi Ahola
- a Department of Oral and Maxillofacial Diseases , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Riitta Koivuniemi
- c Department of Rheumatology , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Leena Laasonen
- d Department of Radiology , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Eeva Moilanen
- e Immunopharmacology Research Group, University of Tampere School of Medicine and Tampere University Hospital , Tampere , Finland
| | - Mari Hämäläinen
- e Immunopharmacology Research Group, University of Tampere School of Medicine and Tampere University Hospital , Tampere , Finland
| | - Taina Tervahartiala
- a Department of Oral and Maxillofacial Diseases , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Jukka H Meurman
- a Department of Oral and Maxillofacial Diseases , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Marjatta Leirisalo-Repo
- c Department of Rheumatology , University of Helsinki and Helsinki University Hospital , Helsinki , Finland
| | - Timo Sorsa
- a Department of Oral and Maxillofacial Diseases , University of Helsinki and Helsinki University Hospital , Helsinki , Finland.,f Department of Dental Medicine , Karolinska Institute , Huddinge , Sweden
| |
Collapse
|
190
|
Rea IM, Gibson DS, McGilligan V, McNerlan SE, Alexander HD, Ross OA. Age and Age-Related Diseases: Role of Inflammation Triggers and Cytokines. Front Immunol 2018; 9:586. [PMID: 29686666 PMCID: PMC5900450 DOI: 10.3389/fimmu.2018.00586] [Citation(s) in RCA: 722] [Impact Index Per Article: 120.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 03/08/2018] [Indexed: 12/11/2022] Open
Abstract
Cytokine dysregulation is believed to play a key role in the remodeling of the immune system at older age, with evidence pointing to an inability to fine-control systemic inflammation, which seems to be a marker of unsuccessful aging. This reshaping of cytokine expression pattern, with a progressive tendency toward a pro-inflammatory phenotype has been called "inflamm-aging." Despite research there is no clear understanding about the causes of "inflamm-aging" that underpin most major age-related diseases, including atherosclerosis, diabetes, Alzheimer's disease, rheumatoid arthritis, cancer, and aging itself. While inflammation is part of the normal repair response for healing, and essential in keeping us safe from bacterial and viral infections and noxious environmental agents, not all inflammation is good. When inflammation becomes prolonged and persists, it can become damaging and destructive. Several common molecular pathways have been identified that are associated with both aging and low-grade inflammation. The age-related change in redox balance, the increase in age-related senescent cells, the senescence-associated secretory phenotype (SASP) and the decline in effective autophagy that can trigger the inflammasome, suggest that it may be possible to delay age-related diseases and aging itself by suppressing pro-inflammatory molecular mechanisms or improving the timely resolution of inflammation. Conversely there may be learning from molecular or genetic pathways from long-lived cohorts who exemplify good quality aging. Here, we will discuss some of the current ideas and highlight molecular pathways that appear to contribute to the immune imbalance and the cytokine dysregulation, which is associated with "inflammageing" or parainflammation. Evidence of these findings will be drawn from research in cardiovascular disease, cancer, neurological inflammation and rheumatoid arthritis.
Collapse
Affiliation(s)
- Irene Maeve Rea
- School of Medicine, Dentistry and Biomedical Science, Queens University Belfast, Belfast, United Kingdom
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Londonderry, United Kingdom
- Care of Elderly Medicine, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - David S. Gibson
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Londonderry, United Kingdom
| | - Victoria McGilligan
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Londonderry, United Kingdom
| | - Susan E. McNerlan
- Regional Genetics Service, Belfast Health and Social Care Trust, Belfast, United Kingdom
| | - H. Denis Alexander
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, University of Ulster, C-TRIC Building, Altnagelvin Area Hospital, Londonderry, United Kingdom
| | - Owen A. Ross
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
- Department of Clinical Genomics, Mayo Clinic, Jacksonville, FL, United States
- School of Medicine and Medical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
191
|
Cook AD, Lee MC, Saleh R, Khiew HW, Christensen AD, Achuthan A, Fleetwood AJ, Lacey DC, Smith JE, Förster I, Hamilton JA. TNF and granulocyte macrophage-colony stimulating factor interdependence mediates inflammation via CCL17. JCI Insight 2018; 3:99249. [PMID: 29563337 DOI: 10.1172/jci.insight.99249] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 02/20/2018] [Indexed: 12/15/2022] Open
Abstract
TNF and granulocyte macrophage-colony stimulating factor (GM-CSF) have proinflammatory activity and both contribute, for example, to rheumatoid arthritis pathogenesis. We previously identified a new GM-CSF→JMJD3 demethylase→interferon regulatory factor 4 (IRF4)→CCL17 pathway that is active in monocytes/macrophages in vitro and important for inflammatory pain, as well as for arthritic pain and disease. Here we provide evidence for a nexus between TNF and this pathway, and for TNF and GM-CSF interdependency. We report that the initiation of zymosan-induced inflammatory pain and zymosan-induced arthritic pain and disease are TNF dependent. Once arthritic pain and disease are established, blockade of GM-CSF or CCL17, but not of TNF, is still able to ameliorate them. TNF is required for GM-CSF-driven inflammatory pain and for initiation of GM-CSF-driven arthritic pain and disease, but not once they are established. TNF-driven inflammatory pain and TNF-driven arthritic pain and disease are dependent on GM-CSF and mechanistically require the same downstream pathway involving GM-CSF→CCL17 formation via JMJD3-regulated IRF4 production, indicating that GM-CSF and CCL17 can mediate some of the proinflammatory and algesic actions of TNF. Given we found that TNF appears important only early in arthritic pain and disease progression, targeting a downstream mediator, such as CCL17, which appears to act throughout the course of disease, could be effective at ameliorating chronic inflammatory conditions where TNF is implicated.
Collapse
Affiliation(s)
- Andrew D Cook
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Ming-Chin Lee
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Reem Saleh
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Hsu-Wei Khiew
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Anne D Christensen
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Adrian Achuthan
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Andrew J Fleetwood
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Derek C Lacey
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Julia E Smith
- Cytokine, Chemokine and Complement DPU, Immunoinflammation TA, GSK Medicines Research Centre, Stevenage, Hertfordshire, United Kingdom
| | - Irmgard Förster
- Immunology and Environment, Life and Medical Sciences Institute University of Bonn, Bonn, Germany
| | - John A Hamilton
- University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria, Australia
| |
Collapse
|
192
|
Chizzolini C, Dufour AM, Brembilla NC. Is there a role for IL-17 in the pathogenesis of systemic sclerosis? Immunol Lett 2018; 195:61-67. [PMID: 28919455 DOI: 10.1016/j.imlet.2017.09.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 09/13/2017] [Accepted: 09/13/2017] [Indexed: 12/19/2022]
Abstract
In systemic sclerosis (SSc) immuno-inflammatory events are central to disease development. Amongst other mediators of inflammation, interleukin 17 (IL-17) and Th17 cells have been reported to be increased in the peripheral blood and target organs including involved skin in SSc. They participate and amplify inflammatory responses by inducing the production of cytokines such as IL-6, chemokines such as CCL2 and CXCL8 (IL-8), matrix metalloproteinases-1, -2, -9 and the expression of adhesion molecules in stromal cells including fibroblasts and endothelial cells. In this respect, IL-17 and Th17 cells behave paradigmatically as documented in other autoimmune pathological conditions or infectious diseases. In experimental animal models of skin and lung fibrosis, IL-17 indirectly enhances the fibrotic process by favoring further inflammation by recruiting inflammatory cells, by activating and/or stimulating the production of TGF-β and other pro-fibrotic mediators, by inhibiting autophagy. Whether the findings generated in animal models of fibrosis can be translated to human SSc is unproven. Furthermore, it is controversial whether IL-17 directly promotes the transdifferentiation of human fibroblasts into myofibroblasts and enhances collagen production, with most of the available evidence against this possibility. The reductionist approach in which fibroblast in monolayers are cultured in plastic dishes under the influence of IL-17 limits the relevance of these findings. Further in vitro/ex vivo models with human tissues are being developed to investigate the real effect of IL-17 on extracellular matrix deposition, since agents blocking IL-17 are available for the clinic and it will be important to know whether their use in SSc would be beneficial or detrimental.
Collapse
Affiliation(s)
- Carlo Chizzolini
- Immunology & Allergy, University Hospital and School of Medicine, Geneva, Switzerland; Pathology and Immunology, University Hospital and School of Medicine, Geneva, Switzerland.
| | - Aleksandra Maria Dufour
- Immunology & Allergy, University Hospital and School of Medicine, Geneva, Switzerland; Pathology and Immunology, University Hospital and School of Medicine, Geneva, Switzerland.
| | - Nicolò Costantino Brembilla
- Pathology and Immunology, University Hospital and School of Medicine, Geneva, Switzerland; Dermatology, University Hospital and School of Medicine, Geneva, Switzerland.
| |
Collapse
|
193
|
Oikonomopoulou K, Diamandis EP, Hollenberg MD, Chandran V. Proteinases and their receptors in inflammatory arthritis: an overview. Nat Rev Rheumatol 2018; 14:170-180. [PMID: 29416136 DOI: 10.1038/nrrheum.2018.17] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Proteinases are enzymes with established roles in physiological and pathological processes such as digestion and the homeostasis, destruction and repair of tissues. Over the past few years, the hormone-like properties of circulating proteinases have become increasingly appreciated. Some proteolytic enzymes trigger cell signalling via proteinase-activated receptors, a family of G protein-coupled receptors that have been implicated in inflammation and pain in inflammatory arthritis. Proteinases can also regulate ion flux owing to the cross-sensitization of transient receptor potential cation channel subfamily V members 1 and 4, which are associated with mechanosensing and pain. In this Review, the idea that proteinases have the potential to orchestrate inflammatory signals by interacting with receptors on cells within the synovial microenvironment of an inflamed joint is revisited in three arthritic diseases: osteoarthritis, spondyloarthritis and rheumatoid arthritis. Unanswered questions are highlighted and the therapeutic potential of modulating this proteinase-receptor axis for the management of disease in patients with these types of arthritis is also discussed.
Collapse
Affiliation(s)
- Katerina Oikonomopoulou
- Centre for Prognosis Studies in Rheumatic Diseases, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Eleftherios P Diamandis
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.,Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada.,Department of Clinical Biochemistry, University Health Network, Toronto, Ontario, Canada
| | - Morley D Hollenberg
- Department of Physiology & Pharmacology, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada.,Department of Medicine, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - Vinod Chandran
- Centre for Prognosis Studies in Rheumatic Diseases, Krembil Research Institute, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada.,Division of Rheumatology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
194
|
Romano C, Del Mastro A, Sellitto A, Solaro E, Esposito S, Cuomo G. Tocilizumab reduces complement C3 and C4 serum levels in rheumatoid arthritis patients. Clin Rheumatol 2018; 37:1695-1700. [DOI: 10.1007/s10067-018-3992-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/10/2018] [Accepted: 01/15/2018] [Indexed: 12/31/2022]
|
195
|
Livshits G, Kalinkovich A. Hierarchical, imbalanced pro-inflammatory cytokine networks govern the pathogenesis of chronic arthropathies. Osteoarthritis Cartilage 2018; 26:7-17. [PMID: 29074297 DOI: 10.1016/j.joca.2017.10.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/15/2017] [Accepted: 10/10/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Chronic inflammatory arthropathies, such as rheumatoid arthritis (RA), spondyloarthritis, including psoriatic arthritis (PsA), ankylosing spondyloarthritis (AS), osteoarthritis (OA), and intervertebral disc degenerative disease (DDD) constitute major public health problems that are anticipated to grow significantly as the human population ages. However, many aspects concerning the molecular mechanisms underlying their onset and progression remain unclear. DESIGN This narrative review critically analyzes the molecular mechanisms underlying the inflammation-associated pathogenesis of the aforementioned joint diseases. This includes, in particular, the major role played by several key soluble factors (such as cytokines and the associated signaling pathways, designated as "fragile nodes") produced by local cells and recruited to the joints' immune cells, whose elimination by specific drugs has dramatically improved the diseases' symptomatology and outcome in human clinical trials or in rodent arthritis models. HYPOTHESIS AND THE AIM OF THIS REVIEW We hypothesize that the pathogenesis of chronic inflammatory arthropathies is governed by hierarchical, imbalanced pro-inflammatory cytokine networks (HIPICNs) (comprising a combination of fragile nodes) that are created during the development of both autoimmune (RA, PsA, and AS) and non-autoimmune (OA and DDD) disorders. The main aim of this review is to provide evidence that despite substantial pathobiological differences between these arthropathies, the HIPICNs created are quite common, thus justifying the merging of these disorders mechanistically and suggesting that these common mechanisms exist in the onset and progression of different joint diseases.
Collapse
Affiliation(s)
- G Livshits
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, Tel Aviv 69978, Israel.
| | - A Kalinkovich
- Human Population Biology Research Unit, Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| |
Collapse
|
196
|
Antinuclear antibodies in autoimmune and allergic diseases. Reumatologia 2017; 55:298-304. [PMID: 29491538 PMCID: PMC5825968 DOI: 10.5114/reum.2017.72627] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 12/14/2017] [Indexed: 01/25/2023] Open
Abstract
Antinuclear antibodies (ANA) are primarily significant in the diagnosis of systemic connective tissue diseases. The relationship between their occurrence in allergic diseases is poorly documented. However, the mechanism of allergic and autoimmune diseases has a common thread. In both cases, an increased production of IgE antibodies and presence of ANA in selected disease entities is observed. Equally important is the activation of basophils secreting proinflammatory factors and affecting the differentiation of TH17 lymphocytes. Both autoimmune and allergic diseases have complex multi-pathogenesis and often occur in genetically predisposed individuals. The presence of antinuclear antibodies was confirmed in many systemic connective tissue diseases and some allergic diseases. Examples include atopic dermatitis, non-allergic asthma, and pollen allergy. Co-occurring allergic and autoimmune disorders induce further search for mechanisms involved in the aetiopathogenesis of both groups of diseases.
Collapse
|
197
|
Detrimental roles of TNF-alpha in the antiphospholipid syndrome and de novo synthesis of antiphospholipid antibodies induced by biopharmaceuticals against TNF-alpha. J Thromb Thrombolysis 2017; 44:565-570. [DOI: 10.1007/s11239-017-1571-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
198
|
Autoantibodies against interleukin-21 correlate with disease activity in patients with rheumatoid arthritis. Clin Rheumatol 2017; 37:75-80. [DOI: 10.1007/s10067-017-3862-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 08/29/2017] [Accepted: 10/01/2017] [Indexed: 11/27/2022]
|
199
|
Savioli B, Abdulahad WH, Brouwer E, Kallenberg CG, de Souza AWS. Are cytokines and chemokines suitable biomarkers for Takayasu arteritis? Autoimmun Rev 2017; 16:1071-1078. [DOI: 10.1016/j.autrev.2017.07.023] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 12/26/2022]
|
200
|
Yamada T, Steinz MM, Kenne E, Lanner JT. Muscle Weakness in Rheumatoid Arthritis: The Role of Ca 2+ and Free Radical Signaling. EBioMedicine 2017; 23:12-19. [PMID: 28781131 PMCID: PMC5605300 DOI: 10.1016/j.ebiom.2017.07.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/13/2017] [Accepted: 07/24/2017] [Indexed: 02/07/2023] Open
Abstract
In addition to the primary symptoms arising from inflammatory processes in the joints, muscle weakness is commonly reported by patients with rheumatoid arthritis (RA). Muscle weakness not only reduces the quality of life for the affected patients, but also dramatically increases the burden on society since patients' work ability decreases. A 25–70% reduction in muscular strength has been observed in pateints with RA when compared with age-matched healthy controls. The reduction in muscle strength is often larger than what could be explained by the reduction in muscle size in patients with RA, which indicates that intracellular (intrinsic) muscle dysfunction plays an important role in the underlying mechanism of muscle weakness associated with RA. In this review, we highlight the present understanding of RA-associated muscle weakness with special focus on how enhanced Ca2 + release from the ryanodine receptor and free radicals (reactive oxygen/nitrogen species) contributes to muscle weakness, and recent developments of novel therapeutic interventions. Muscle weakness is commonly reported by patients with rheumatoid arthritis (RA). Intrinsic muscle weakness is important in the underlying mechanisms of muscle weakness associated with rheumatoid arthritis. Enhanced Ca2 + release and peroxynitrite-induced stress contributes to RA-induced muscle weakness.
Collapse
Affiliation(s)
- Takashi Yamada
- Graduate School of Health Sciences, Sapporo Medical University, Sapporo, Japan
| | - Maarten M Steinz
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Ellinor Kenne
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|