1
|
Tominari T, Matsumoto C, Tanaka Y, Shimizu K, Takatoya M, Sugasaki M, Karouji K, Kasuga U, Miyaura C, Miyata S, Itoh Y, Hirata M, Inada M. Roles of Toll-like Receptor Signaling in Inflammatory Bone Resorption. BIOLOGY 2024; 13:692. [PMID: 39336119 PMCID: PMC11429252 DOI: 10.3390/biology13090692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024]
Abstract
Toll-like receptors (TLRs) are pattern recognition receptors expressed in immune cells, including neutrophils, macrophages, and dendritic cells. Microbe-associated molecular patterns, including bacterial components, membranes, nucleic acids, and flagella are recognized by TLRs in inflammatory immune responses. Periodontal disease is an inflammatory disease known to cause local infections associated with gingival inflammation, subsequently leading to alveolar bone resorption. Prostaglandin E2 (PGE2) is a key mediator of TLR-induced inflammatory bone resorption. We previously reported that membrane-bound PGE synthase (mPGES-1)-deficient mice failed to induce bone resorption by lipopolysaccharide (LPS), a major pathogenic factor involved in periodontal bone resorption. Further experiments exploring specific pathogen-promoting osteoclast differentiation revealed that various TLR ligands induced osteoclast differentiation in a co-culture model. The ligands for TLR2/1, TLR2/6, TLR3, and TLR5, as well as TLR4, induce osteoclast differentiation associated with the production of PGE2 and the receptor activator of nuclear factor-kappa B ligand (RANKL), an inevitable inducer of osteoclast differentiation in osteoblasts. In vivo, local injection of TLR ligands, including TLR2/1, TLR2/6, and TLR3, resulted in severe alveolar bone resorption. This review summarizes the latest findings on TLR-mediated osteoclast differentiation and bone resorption in inflammatory diseases, such as periodontal diseases.
Collapse
Affiliation(s)
- Tsukasa Tominari
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan; (T.T.); (C.M.); (C.M.); (M.H.)
| | - Chiho Matsumoto
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan; (T.T.); (C.M.); (C.M.); (M.H.)
| | - Yuki Tanaka
- Cooperative Major of Advanced Health Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan (K.K.)
| | - Kensuke Shimizu
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan; (T.T.); (C.M.); (C.M.); (M.H.)
| | - Masaru Takatoya
- Cooperative Major of Advanced Health Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan (K.K.)
| | - Moe Sugasaki
- Cooperative Major of Advanced Health Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan (K.K.)
| | - Kento Karouji
- Cooperative Major of Advanced Health Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan (K.K.)
| | - Urara Kasuga
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan; (T.T.); (C.M.); (C.M.); (M.H.)
| | - Chisato Miyaura
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan; (T.T.); (C.M.); (C.M.); (M.H.)
| | - Shinji Miyata
- Inada Research Unit, Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan; (S.M.); (Y.I.)
| | - Yoshifumi Itoh
- Inada Research Unit, Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan; (S.M.); (Y.I.)
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Michiko Hirata
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan; (T.T.); (C.M.); (C.M.); (M.H.)
| | - Masaki Inada
- Department of Biotechnology and Life Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan; (T.T.); (C.M.); (C.M.); (M.H.)
- Cooperative Major of Advanced Health Science, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan (K.K.)
- Inada Research Unit, Institute of Global Innovation Research, Tokyo University of Agriculture and Technology, 2-24-16 Nakacho, Koganei-shi, Tokyo 184-8588, Japan; (S.M.); (Y.I.)
| |
Collapse
|
2
|
Ren W, Zhao L, Sun Y, Wang X, Shi X. HMGB1 and Toll-like receptors: potential therapeutic targets in autoimmune diseases. Mol Med 2023; 29:117. [PMID: 37667233 PMCID: PMC10478470 DOI: 10.1186/s10020-023-00717-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/14/2023] [Indexed: 09/06/2023] Open
Abstract
HMGB1, a nucleoprotein, is expressed in almost all eukaryotic cells. During cell activation and cell death, HMGB1 can function as an alarm protein (alarmin) or damage-associated molecular pattern (DAMP) and mediate early inflammatory and immune response when it is translocated to the extracellular space. The binding of extracellular HMGB1 to Toll-like receptors (TLRs), such as TLR2 and TLR4 transforms HMGB1 into a pro-inflammatory cytokine, contributing to the occurrence and development of autoimmune diseases. TLRs, which are members of a family of pattern recognition receptors, can bind to endogenous DAMPs and activate the innate immune response. Additionally, TLRs are key signaling molecules mediating the immune response and play a critical role in the host defense against pathogens and the maintenance of immune balance. HMGB1 and TLRs are reported to be upregulated in several autoimmune diseases, such as rheumatoid arthritis, systemic lupus erythematosus, type 1 diabetes mellitus, and autoimmune thyroid disease. The expression levels of HMGB1 and some TLRs are upregulated in tissues of patients with autoimmune diseases and animal models of autoimmune diseases. The suppression of HMGB1 and TLRs inhibits the progression of inflammation in animal models. Thus, HMGB1 and TLRs are indispensable biomarkers and important therapeutic targets for autoimmune diseases. This review provides comprehensive strategies for treating or preventing autoimmune diseases discovered in recent years.
Collapse
Affiliation(s)
- Wenxuan Ren
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Lei Zhao
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Ying Sun
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Xichang Wang
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110001, Liaoning, China
| | - Xiaoguang Shi
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
3
|
Niu Q, Gao J, Wang L, Liu J, Zhang L. Regulation of differentiation and generation of osteoclasts in rheumatoid arthritis. Front Immunol 2022; 13:1034050. [PMID: 36466887 PMCID: PMC9716075 DOI: 10.3389/fimmu.2022.1034050] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/31/2022] [Indexed: 09/25/2023] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA), which affects nearly 1% of the world's population, is a debilitating autoimmune disease. Bone erosion caused by periarticular osteopenia and synovial pannus formation is the most destructive pathological changes of RA, also leads to joint deformity and loss of function,and ultimately affects the quality of life of patients. Osteoclasts (OCs) are the only known bone resorption cells and their abnormal differentiation and production play an important role in the occurrence and development of RA bone destruction; this remains the main culprit behind RA. METHOD Based on the latest published literature and research progress at home and abroad, this paper reviews the abnormal regulation mechanism of OC generation and differentiation in RA and the possible targeted therapy. RESULT OC-mediated bone destruction is achieved through the regulation of a variety of cytokines and cell-to-cell interactions, including gene transcription, epigenetics and environmental factors. At present, most methods for the treatment of RA are based on the regulation of inflammation, the inhibition of bone injury and joint deformities remains unexplored. DISCUSSION This article will review the mechanism of abnormal differentiation of OC in RA, and summarise the current treatment oftargeting cytokines in the process of OC generation and differentiation to reduce bone destruction in patients with RA, which isexpected to become a valuable treatment choice to inhibit bone destruction in RA.
Collapse
Affiliation(s)
- Qing Niu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Jinfang Gao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Lei Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Jiaxi Liu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Liyun Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
4
|
Liu L, Tian F, Li GY, Xu W, Xia R. The effects and significance of gut microbiota and its metabolites on the regulation of osteoarthritis: Close coordination of gut-bone axis. Front Nutr 2022; 9:1012087. [PMID: 36204373 PMCID: PMC9530816 DOI: 10.3389/fnut.2022.1012087] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is a common chronic degenerative disease of articular cartilage in middle-aged and older individuals, which can result in the joint pain and dysfunction, and even cause the joint deformity or disability. With the enhancing process of global aging, OA has gradually become a major public health problem worldwide. Explaining pathogenesis of OA is critical for the development of new preventive and therapeutic interventions. In recent years, gut microbiota (GM) has been generally regarded as a “multifunctional organ,” which is closely relevant with a variety of immune, metabolic and inflammatory functions. Meanwhile, more and more human and animal researches have indicated the existence of gut-bone axis and suggested that GM and its metabolites are closely involved in the pathogenic process of OA, which might become a potential and promising intervention target. Based on the close coordination of gut-bone axis, this review aims to summarize and discuss the mechanisms of GM and its metabolites influencing OA from the aspects of the intestinal mucosal barrier modulation, intestinal metabolites modulation, immune modulation and strategies for the prevention or treatment of OA based on perspectives of GM and its metabolites, thus providing a profound knowledge and recognition of it.
Collapse
|
5
|
Umar S, Palasiewicz K, Meyer A, Kumar P, Prabhakar BS, Volin MV, Rahat R, Al-Awqati M, Chang HJ, Zomorrodi RK, Rehman J, Shahrara S. Inhibition of IRAK4 dysregulates SARS-CoV-2 spike protein-induced macrophage inflammatory and glycolytic reprogramming. Cell Mol Life Sci 2022; 79:301. [PMID: 35588018 PMCID: PMC9118817 DOI: 10.1007/s00018-022-04329-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 12/13/2022]
Abstract
Escalated innate immunity plays a critical role in SARS-CoV-2 pathology; however, the molecular mechanism is incompletely understood. Thus, we aim to characterize the molecular mechanism by which SARS-CoV-2 Spike protein advances human macrophage (Mϴ) inflammatory and glycolytic phenotypes and uncover novel therapeutic strategies. We found that human Mϴs exposed to Spike protein activate IRAK4 phosphorylation. Blockade of IRAK4 in Spike protein-stimulated Mϴs nullifies signaling of IRAK4, AKT, and baseline p38 without affecting ERK and NF-κB activation. Intriguingly, IRAK4 inhibitor (IRAK4i) rescues the SARS-CoV-2-induced cytotoxic effect in ACE2+HEK 293 cells. Moreover, the inflammatory reprogramming of Mϴs by Spike protein was blunted by IRAK4i through IRF5 and IRF7, along with the reduction of monokines, IL-6, IL-8, TNFα, and CCL2. Notably, in Spike protein-stimulated Mϴs, suppression of the inflammatory markers by IRAK4i was coupled with the rebalancing of oxidative phosphorylation over metabolic activity. This metabolic adaptation promoted by IRAK4i in Spike protein-activated Mϴs was shown to be in part through constraining PFKBF3, HIF1α, cMYC, LDHA, lactate expression, and reversal of citrate and succinate buildup. IRAK4 knockdown could comparably impair Spike protein-enhanced inflammatory and metabolic imprints in human Mϴs as those treated with ACE2, TLR2, and TLR7 siRNA. Extending these results, in murine models, where human SARS-CoV-2 Spike protein was not recognized by mouse ACE2, TLRs were responsible for the inflammatory and glycolytic responses instigated by Spike protein and were dysregulated by IRAK4i therapy. In conclusion, IRAK4i may be a promising strategy for severe COVID-19 patients by counter-regulating ACE2 and TLR-mediated Mϴ hyperactivation. IRAK4i therapy counteracts Mϴ inflammatory and glycolytic reprogramming triggered by Spike protein. This study illustrates that SARS-CoV-2 Spike protein activates IRAK4 signaling via ACE2 as well as TLR2 and TLR7 sensing in human Mϴs. Remarkably, IRAK4i treatment can dysregulate both ACE-dependent and independent (via TLR sensing) SARS-CoV-2 Spike protein-activated inflammatory and metabolic imprints.
Collapse
Affiliation(s)
- Sadiq Umar
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA
| | - Karol Palasiewicz
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA
| | - Anja Meyer
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA
| | - Prabhakaran Kumar
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, USA
| | - Michael V Volin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - Rani Rahat
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA
| | - Mina Al-Awqati
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA
| | - Huan J Chang
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA
| | - Ryan K Zomorrodi
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA
| | - Jalees Rehman
- Department of Pharmacology and Regenerative Medicine, The University of Illinois at Chicago, Chicago, IL, USA
- Department of Medicine, Division of Cardiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL, USA.
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA.
| |
Collapse
|
6
|
Umar S, Palasiewicz K, Volin MV, Romay B, Rahat R, Tetali C, Arami S, Guma M, Ascoli C, Sweiss N, Zomorrodi RK, O'Neill LAJ, Shahrara S. Metabolic regulation of RA macrophages is distinct from RA fibroblasts and blockade of glycolysis alleviates inflammatory phenotype in both cell types. Cell Mol Life Sci 2021; 78:7693-7707. [PMID: 34705053 PMCID: PMC8739866 DOI: 10.1007/s00018-021-03978-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 09/10/2021] [Accepted: 10/13/2021] [Indexed: 11/29/2022]
Abstract
Recent studies have shown the significance of metabolic reprogramming in immune and stromal cell function. Yet, the metabolic reconfiguration of RA macrophages (MΦs) is incompletely understood during active disease and in crosstalk with other cell types in experimental arthritis. This study elucidates a distinct regulation of glycolysis and oxidative phosphorylation in RA MΦs compared to fibroblast (FLS), although PPP (Pentose Phosphate pathway) is similarly reconfigured in both cell types. 2-DG treatment showed a more robust impact on impairing the RA M1 MΦ-mediated inflammatory phenotype than IACS-010759 (IACS, complexli), by reversing ERK, AKT and STAT1 signaling, IRF8/3 transcription and CCL2 or CCL5 secretion. This broader inhibitory effect of 2-DG therapy on RA M1 MΦs was linked to dysregulation of glycolysis (GLUT1, PFKFB3, LDHA, lactate) and oxidative PPP (NADP conversion to NADPH), while both compounds were ineffective on oxidative phosphorylation. Distinctly, in RA FLS, 2-DG and IACS therapies constrained LPS/IFNγ-induced AKT and JNK signaling, IRF5/7 and fibrokine expression. Disruption of RA FLS metabolic rewiring by 2-DG or IACS therapy was accompanied by a reduction of glycolysis (HIF1α, PFKFB3) and suppression of citrate or succinate buildup. We found that 2-DG therapy mitigated CIA pathology by intercepting joint F480+iNOS+MΦ, Vimentin+ fibroblast and CD3+T cell trafficking along with downregulation of IRFs and glycolytic intermediates. Surprisingly, IACS treatment was inconsequential on CIA swelling, cell infiltration, M1 and Th1/Th17 cytokines (IFN-γ/IL-17) and joint glycolytic mediators. Collectively, our results indicate that blockade of glycolysis is more effective than inhibition of complex 1 in CIA, in part due to its effectiveness on the MΦ inflammatory phenotype.
Collapse
Affiliation(s)
- Sadiq Umar
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA
| | - Karol Palasiewicz
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA
| | - Michael V Volin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - Bianca Romay
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA
| | - Rani Rahat
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA
| | - Chandana Tetali
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA
| | - Shiva Arami
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA
| | - Monica Guma
- Division of Rheumatology, Allergy and Immunology, San Diego, School of Medicine, University of California, La Jolla, CA, USA
- VA Medical Center, San Diego, CA, USA
| | - Christian Ascoli
- Division of Pulmonary, Critical Care, Sleep, and Allergy, The University of Illinois at Chicago, Chicago, IL, USA
| | - Nadera Sweiss
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA
| | - Ryan K Zomorrodi
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL, USA.
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, 840 S Wood Street, CSB suite 1114, Chicago, IL, 60612, USA.
| |
Collapse
|
7
|
Chen L, Lu Q, Chen J, Feng R, Yang C. Upregulating miR-27a-3p inhibits cell proliferation and inflammation of rheumatoid arthritis synovial fibroblasts through targeting toll-like receptor 5. Exp Ther Med 2021; 22:1227. [PMID: 34539823 PMCID: PMC8438689 DOI: 10.3892/etm.2021.10661] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/11/2021] [Indexed: 12/25/2022] Open
Abstract
Rheumatoid arthritis (RA) is a serious chronic inflammatory disease and synovial fibroblasts (SFs) serve a vital role in the pathogenesis and progression of RA. Current studies have demonstrated that dysregulation of microRNAs is involved in RA etiopathogenesis. The present study aimed to investigate the role of microRNA (miR)-27a-3p in RASFs, as well as its molecular mechanism. RASFs were isolated from synovial tissues from patients with RA. Expression of miR-27a-3p and toll-like receptor 5 (TLR5) was detected using reverse transcription-quantitative polymerase chain reaction and western blotting. Cell proliferation, apoptosis and inflammatory response were measured with MTT assay, flow cytometry and ELISA kits, respectively. The target binding between miR-27a-3p and TLR5 was predicted on DIANA TOOLS software, and confirmed by dual-luciferase reporter assay and Biotin-coupled miRNA pull-down assay. Expression of miR-27a-3p was downregulated and TLR5 was upregulated in synovial tissues and RASFs isolated from patients with RA. Functionally, upregulating miR-27a-3p may promote the apoptosis rate of RASFs and suppress cell proliferation and secretions of interleukin (IL)-1β, IL-6 and tumor necrosis factor-α. TLR5 was validated as a downstream target for miR-27a-3p in RASFs, and its expression was negatively regulated by miR-27a-3p. Silencing TLR5 in RASFs may exert similar effects to miR-27a-3p-overexpression; whereas, restoring TLR5 counteracted the suppression of miR-27a-3p-overexpression on RASF proliferation and inflammation, as well as the promotion on apoptosis. miR-27a-3p upregulation may suppress RA progression by inhibiting RASFs proliferation and inflammation through targeting TLR5.
Collapse
Affiliation(s)
- Lifeng Chen
- Department of Rheumatology and Immunology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Qiping Lu
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Jianhua Chen
- Department of Rheumatology and Immunology, General Hospital of Central Theater Command, Wuhan, Hubei 430070, P.R. China
| | - Ruibing Feng
- Department of Orthopedics, Central People's Liberation Army Central Theater, Wuhan, Hubei 430070, P.R. China
| | - Chenxi Yang
- Department of Orthopedics, Graduate School of Hubei University of Traditional Chinese Medicine, Wuhan, Hubei 430061, P.R. China
| |
Collapse
|
8
|
Umar S, Palasiewicz K, Van Raemdonck K, Volin MV, Romay B, Amin MA, Zomorrodi RK, Arami S, Gonzalez M, Rao V, Zanotti B, Fox DA, Sweiss N, Shahrara S. IRAK4 inhibition: a promising strategy for treating RA joint inflammation and bone erosion. Cell Mol Immunol 2021; 18:2199-2210. [PMID: 32415262 PMCID: PMC8429735 DOI: 10.1038/s41423-020-0433-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/30/2020] [Indexed: 01/22/2023] Open
Abstract
Flares of joint inflammation and resistance to currently available biologic therapeutics in rheumatoid arthritis (RA) patients could reflect activation of innate immune mechanisms. Herein, we show that a TLR7 GU-rich endogenous ligand, miR-Let7b, potentiates synovitis by amplifying RA monocyte and fibroblast (FLS) trafficking. miR-Let7b ligation to TLR7 in macrophages (MΦs) and FLSs expanded the synovial inflammatory response. Moreover, secretion of M1 monokines triggered by miR-Let7b enhanced Th1/Th17 cell differentiation. We showed that IRAK4 inhibitor (i) therapy attenuated RA disease activity by blocking TLR7-induced M1 MΦ or FLS activation, as well as monokine-modulated Th1/Th17 cell polarization. IRAK4i therapy also disrupted RA osteoclastogenesis, which was amplified by miR-Let7b ligation to joint myeloid TLR7. Hence, the effectiveness of IRAK4i was compared with that of a TNF inhibitor (i) or anti-IL-6R treatment in collagen-induced arthritis (CIA) and miR-Let7b-mediated arthritis. We found that TNF or IL-6R blocking therapies mitigated CIA by reducing the infiltration of joint F480+iNOS+ MΦs, the expression of certain monokines, and Th1 cell differentiation. Unexpectedly, these biologic therapies were unable to alleviate miR-Let7b-induced arthritis. The superior efficacy of IRAK4i over anti-TNF or anti-IL-6R therapy in miR-Let7b-induced arthritis or CIA was due to the ability of IRAK4i therapy to restrain the migration of joint F480+iNOS+ MΦs, vimentin+ fibroblasts, and CD3+ T cells, in addition to negating the expression of a wide range of monokines, including IL-12, MIP2, and IRF5 and Th1/Th17 lymphokines. In conclusion, IRAK4i therapy may provide a promising strategy for RA therapy by disconnecting critical links between inflammatory joint cells.
Collapse
Affiliation(s)
- Sadiq Umar
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Karol Palasiewicz
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Katrien Van Raemdonck
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Michael V Volin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA
| | - Bianca Romay
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - M Asif Amin
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI, 481096, USA
| | - Ryan K Zomorrodi
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Shiva Arami
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mark Gonzalez
- Department of Orthopedic Surgery, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Vikram Rao
- Pfizer Research, Cambridge, MA, 02139, USA
| | - Brian Zanotti
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, 60515, USA
| | - David A Fox
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI, 481096, USA
| | - Nadera Sweiss
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL, 60612, USA.
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
9
|
Palasiewicz K, Umar S, Romay B, Zomorrodi RK, Shahrara S. Tofacitinib therapy intercepts macrophage metabolic reprogramming instigated by SARS-CoV-2 Spike protein. Eur J Immunol 2021; 51:2330-2340. [PMID: 34107055 PMCID: PMC8237023 DOI: 10.1002/eji.202049159] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/14/2021] [Accepted: 06/07/2021] [Indexed: 12/27/2022]
Abstract
The molecular mechanism of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Spike protein was characterized to identify novel therapies. The impact of tofacitinib, IL-6R Ab, or TNFi therapy was determined on Spike protein or LPS/IFN-γ-induced signaling, inflammation, and metabolic reprogramming in MΦs and/or rheumatoid arthritis (RA) fibroblast-like synoviocyte (FLS). ACE2 frequency was markedly expanded in MΦs compared to T cells and RA FLS. Tofacitinib suppresses Spike protein potentiated STAT1 signaling, whereas this function was unchanged by TNFi. Tofacitinib impairs IL-6/IFN/LPS-induced STAT1 and STAT3 phosphorylation in RA MΦs and FLS. Interestingly, tofacitinib had a broader inhibitory effect on the monokines, glycolytic regulators, or oxidative metabolites compared to IL-6R Ab and TNFi in Spike-protein-activated MΦs. In contrast, all three therapies disrupted IFN-α and IFN-β secretion in response to Spike protein; nonetheless, the IFN-γ was only curtailed by tofacitinib or IL-6R Ab. While tofacitinib counteracted MΦ metabolic rewiring instigated by Spike protein, it was inconsequential on the glycolysis expansion mediated via HK2 and/or LDHA in the activated RA MΦ and FLS. Nevertheless, the potentiated inflammatory response and the diminished oxidative phosphorylation modulated by Spike protein and/or LPS/IFN-γ stimulation in MΦs or RA FLS were reversed by tofacitinib. In conclusion, tofacitinib suppresses MΦ inflammation and immunometabolism triggered by Spike protein and may provide a promising strategy for COVID-19 patients.
Collapse
Affiliation(s)
- Karol Palasiewicz
- Jesse Brown VA Medical CenterChicagoILUSA
- Division of RheumatologyDepartment of Medicine, The University of Illinois at ChicagoChicagoILUSA
| | - Sadiq Umar
- Jesse Brown VA Medical CenterChicagoILUSA
- Division of RheumatologyDepartment of Medicine, The University of Illinois at ChicagoChicagoILUSA
| | - Bianca Romay
- Division of RheumatologyDepartment of Medicine, The University of Illinois at ChicagoChicagoILUSA
| | - Ryan K. Zomorrodi
- Division of RheumatologyDepartment of Medicine, The University of Illinois at ChicagoChicagoILUSA
| | - Shiva Shahrara
- Jesse Brown VA Medical CenterChicagoILUSA
- Division of RheumatologyDepartment of Medicine, The University of Illinois at ChicagoChicagoILUSA
| |
Collapse
|
10
|
Targeting Toll-like Receptor (TLR) Pathways in Inflammatory Arthritis: Two Better Than One? Biomolecules 2021; 11:biom11091291. [PMID: 34572504 PMCID: PMC8464963 DOI: 10.3390/biom11091291] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/22/2021] [Accepted: 08/24/2021] [Indexed: 12/17/2022] Open
Abstract
Inflammatory arthritis is a cluster of diseases caused by unregulated activity of the immune system. The lost homeostasis is followed by the immune attack of one’s self, what damages healthy cells and tissues and leads to chronic inflammation of various tissues and organs (e.g., joints, lungs, heart, eyes). Different medications to control the excessive immune response are in use, however, drug resistances, flare-reactions and adverse effects to the current therapies are common in the affected patients. Thus, it is essential to broaden the spectrum of alternative treatments and to develop disease-modifying drugs. In the last 20 years, the involvement of the innate immune receptors TLRs in inflammatory arthritis has been widely investigated and targeting either the receptor itself or the proteins in the downstream signalling cascades has emerged as a promising therapeutic strategy. Yet, concerns about the use of pharmacological agents that inhibit TLR activity and may leave the host unprotected against invading pathogens and toxicity issues amid inhibition of downstream kinases crucial in various cellular functions have arisen. This review summarizes the existing knowledge on the role of TLRs in inflammatory arthritis; in addition, the likely druggable related targets and the developed inhibitors, and discusses the pros and cons of their potential clinical use.
Collapse
|
11
|
Unterberger S, Davies KA, Rambhatla SB, Sacre S. Contribution of Toll-Like Receptors and the NLRP3 Inflammasome in Rheumatoid Arthritis Pathophysiology. Immunotargets Ther 2021; 10:285-298. [PMID: 34350135 PMCID: PMC8326786 DOI: 10.2147/itt.s288547] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 07/15/2021] [Indexed: 12/20/2022] Open
Abstract
Rheumatoid arthritis (RA) is a progressive autoimmune disease that is characterized by inflammation of the synovial joints leading to cartilage and bone damage. The pathogenesis is sustained by the production of pro-inflammatory cytokines including tumor necrosis factor (TNF), interleukin (IL)-1 and IL-6, which can be targeted therapeutically to alleviate disease severity. Several innate immune receptors are suggested to contribute to the chronic inflammation in RA, through the production of pro-inflammatory factors in response to endogenous danger signals. Much research has focused on toll-like receptors and more recently the nucleotide-binding domain and leucine-rich repeat pyrin containing protein-3 (NLRP3) inflammasome, which is required for the processing and release of IL-1β. This review summarizes the current understanding of the potential involvement of these receptors in the initiation and maintenance of inflammation and tissue damage in RA and experimental arthritis models.
Collapse
Affiliation(s)
- Sarah Unterberger
- Brighton and Sussex Medical School, University of Sussex, Falmer, Brighton, BN1 9PS, UK
| | - Kevin A Davies
- Brighton and Sussex Medical School, University of Sussex, Falmer, Brighton, BN1 9PS, UK
| | | | - Sandra Sacre
- Brighton and Sussex Medical School, University of Sussex, Falmer, Brighton, BN1 9PS, UK
| |
Collapse
|
12
|
Zhong X, Zhang F, Yin X, Cao H, Wang X, Liu D, Chen J, Chen X. Bone Homeostasis and Gut Microbial-Dependent Signaling Pathways. J Microbiol Biotechnol 2021; 31:765-774. [PMID: 34176870 PMCID: PMC9705830 DOI: 10.4014/jmb.2104.04016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/27/2021] [Accepted: 06/13/2021] [Indexed: 12/15/2022]
Abstract
Although research on the osteal signaling pathway has progressed, understanding of gut microbial-dependent signaling pathways for metabolic and immune bone homeostasis remains elusive. In recent years, the study of gut microbiota has shed light on our understanding of bone homeostasis. Here, we review microbiota-mediated gut-bone crosstalk via bone morphogenetic protein/SMADs, Wnt and OPG/receptor activator of nuclear factor-kappa B ligand signaling pathways in direct (translocation) and indirect (metabolite) manners. The mechanisms underlying gut microbiota involvement in these signaling pathways are relevant in immune responses, secretion of hormones, fate of osteoblasts and osteoclasts and absorption of calcium. Collectively, we propose a signaling network for maintaining a dynamic homeostasis between the skeletal system and the gut ecosystem. Additionally, the role of gut microbial improvement by dietary intervention in osteal signaling pathways has also been elucidated. This review provides unique resources from the gut microbial perspective for the discovery of new strategies for further improving treatment of bone diseases by increasing the abundance of targeted gut microbiota.
Collapse
Affiliation(s)
- Xiaohui Zhong
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, P.R. China
| | - Feng Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, P.R. China,Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R. China,Nutritional Department, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R. China
| | - Xinyao Yin
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, P.R. China
| | - Hong Cao
- Clinical Assessment Center of Functional Food, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R. China,Department of Endocrinology, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R. China,Nutritional Department, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R. China
| | - Xuesong Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, P.R. China,Department of Orthopedics, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R.China
| | - Dongsong Liu
- Department of Orthopedics, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R.China
| | - Jing Chen
- Department of Orthopedics, Affiliated Hospital of Jiangnan University, Wuxi 214125, P.R.China
| | - Xue Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, P.R. China,Corresponding author Phone: +86-15861589177 E-mail:
| |
Collapse
|
13
|
Ghafouri-Fard S, Abak A, Shoorei H, Talebi SF, Mohaqiq M, Sarabi P, Taheri M, Mokhtari M. Interaction between non-coding RNAs and Toll-like receptors. Biomed Pharmacother 2021; 140:111784. [PMID: 34087695 DOI: 10.1016/j.biopha.2021.111784] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022] Open
Abstract
Toll-like receptors (TLRs) are a large group of pattern recognition receptors which are involved in the regulation of innate immune responses. Based on the interplay between TLRs and adapter molecules, two distinctive signaling cascades, namely the MyD88-dependent and TRIF-dependent pathways have been recognized. TLRs are involved in the development of a wide variety of diseases including cancer and autoimmune disorders. A large body of evidence has shown interaction between two classes of non-coding RNAs, namely microRNAs (miRNAs) and long noncoding RNAs (lncRNAs). These interactions have prominent roles in the pathogenesis of several disorders including infectious disorders, autoimmune conditions and neoplastic disorders. This review aims at description of the interaction between these non-coding RNAs and TLRs.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefe Abak
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Mahdi Mohaqiq
- School of Advancement, Centennial College, Ashtonbee Campus, Toronto, ON, Canada
| | - Parisa Sarabi
- Deputy for Research & Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Majid Mokhtari
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Umar S, Palasiewicz K, Van Raemdonck K, Volin MV, Romay B, Ahmad I, Tetali C, Sweiss N, Amin MA, Zomorrodi RK, Shahrara S. CCL25 and CCR9 is a unique pathway that potentiates pannus formation by remodeling RA macrophages into mature osteoclasts. Eur J Immunol 2021; 51:903-914. [PMID: 33347617 PMCID: PMC10041658 DOI: 10.1002/eji.202048681] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 10/26/2020] [Accepted: 12/18/2020] [Indexed: 12/18/2022]
Abstract
This study elucidates the mechanism of CCL25 and CCR9 in rheumatoid arthritis (RA). RA synovial fluid (SF) expresses elevated levels of CCL25 compared to OA SF and plasma from RA and normal. CCL25 was released into RA SF by fibroblasts (FLS) and macrophages (MΦs) stimulated with IL-1β and IL-6. CCR9 is also presented on IL-1β and IL-6 activated RA FLS and differentiated MΦs. Conversely, in RA PBMCs neither CCL25 nor CCR9 are impacted by 3-month longitudinal TNF inhibitor therapy. CCL25 amplifies RA FLS and monocyte infiltration via p38 and ERK phosphorylation. CCL25-stimulated RA FLS secrete potentiated levels of IL-8 which is disrupted by p38 and ERK inhibitors. CCL25 polarizes RA monocytes into nontraditional M1 MΦs that produce IL-8 and CCL2. Activation of p38 and ERK cascades are also responsible for the CCL25-induced M1 MΦ development. Unexpectedly, CCL25 was unable to polarize RA PBMCs into effector Th1/Th17 cells. Consistently, lymphokine like RANKL was uninvolved in CCL25-induced osteoclastogenesis; however, this manifestation was regulated by osteoclastic factors such as RANK, cathepsin K (CTSK), and TNF-α. In short, we reveal that CCL25/CCR9 manipulates RA FLS and MΦ migration and inflammatory phenotype in addition to osteoclast formation via p38 and ERK activation.
Collapse
Affiliation(s)
- Sadiq Umar
- Jesse Brown VA Medical Center, Chicago, IL
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - Karol Palasiewicz
- Jesse Brown VA Medical Center, Chicago, IL
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - Katrien Van Raemdonck
- Jesse Brown VA Medical Center, Chicago, IL
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - Michael V. Volin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL
| | - Bianca Romay
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - Imran Ahmad
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - Chandana Tetali
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - Nadera Sweiss
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - M Asif Amin
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI 481096
| | - Ryan K Zomorrodi
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL
- Department of Medicine, Division of Rheumatology, the University of Illinois at Chicago, IL
| |
Collapse
|
15
|
Thwaites RS, Unterberger S, Chamberlain G, Walker-Bone K, Davies KA, Sacre S. TLR1/2 and 5 induce elevated cytokine levels from rheumatoid arthritis monocytes independent of ACPA or RF autoantibody status. Rheumatology (Oxford) 2021; 59:3533-3539. [PMID: 32594150 PMCID: PMC7590412 DOI: 10.1093/rheumatology/keaa220] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/30/2020] [Indexed: 01/29/2023] Open
Abstract
OBJECTIVE RA is an autoimmune inflammatory joint disease. Both RF and ACPA are associated with more progressive disease and higher levels of systemic inflammation. Monocyte activation of toll-like receptors (TLRs) by endogenous ligands is a potential source of increased production of systemic cytokines. RA monocytes have elevated TLRs, some of which are associated with the disease activity score using 28 joints (DAS28). The aim of this study was to measure TLR-induced cytokine production from monocytes, stratified by autoantibody status, to assess if their capacity to induce cytokines is related to autoantibody status or DAS28. METHODS Peripheral blood monocytes isolated from RA patients and healthy controls were stimulated with TLR1/2, TLR2/6, TLR4, TLR5, TLR7, TLR8 and TLR9 ligands for 18 h before measuring IL-6, TNFα and IL-10. Serum was used to confirm the autoantibody status. Cytokine levels were compared with RF, ACPA and DAS28. RESULTS RA monocytes demonstrated significantly increased IL-6 and TNFα upon TLR1/2 stimulation and IL-6 and IL-10 upon TLR5 activation. TLR7 and TLR9 activation did not induce cytokines and no significant differences were observed between RA and healthy control monocytes upon TLR2/6, TLR4 or TLR8 activation. When stratified by ACPA or RF status there were no correlations between autoantibody status and elevated cytokine levels. However, TLR1/2-induced IL-6 did correlate with DAS28. CONCLUSIONS Elevated TLR-induced cytokines in RA monocytes were not related to ACPA or RF status. However, TLR1/2-induced IL-6 was associated with disease activity.
Collapse
Affiliation(s)
- Ryan S Thwaites
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Sarah Unterberger
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | | | - Karen Walker-Bone
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Kevin A Davies
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Sandra Sacre
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| |
Collapse
|
16
|
Wu C, Tan S, Liu L, Cheng S, Li P, Li W, Liu H, Zhang F, Wang S, Ning Y, Wen Y, Zhang F. Transcriptome-wide association study identifies susceptibility genes for rheumatoid arthritis. Arthritis Res Ther 2021; 23:38. [PMID: 33482886 PMCID: PMC7821659 DOI: 10.1186/s13075-021-02419-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/05/2021] [Indexed: 02/06/2023] Open
Abstract
Objective To identify rheumatoid arthritis (RA)-associated susceptibility genes and pathways through integrating genome-wide association study (GWAS) and gene expression profile data. Methods A transcriptome-wide association study (TWAS) was conducted by the FUSION software for RA considering EBV-transformed lymphocytes (EL), transformed fibroblasts (TF), peripheral blood (NBL), and whole blood (YBL). GWAS summary data was driven from a large-scale GWAS, involving 5539 autoantibody-positive RA patients and 20,169 controls. The TWAS-identified genes were further validated using the mRNA expression profiles and made a functional exploration. Results TWAS identified 692 genes with PTWAS values < 0.05 for RA. CRIPAK (PEL = 0.01293, PTF = 0.00038, PNBL = 0.02839, PYBL = 0.0978), MUT (PEL = 0.00377, PTF = 0.00076, PNBL = 0.00778, PYBL = 0.00096), FOXRED1 (PEL = 0.03834, PTF = 0.01120, PNBL = 0.01280, PYBL = 0.00583), and EBPL (PEL = 0.00806, PTF = 0.03761, PNBL = 0.03540, PYBL = 0.04254) were collectively expressed in all the four tissues/cells. Eighteen genes, including ANXA5, AP4B1, ATIC (PTWAS = 0.0113, downregulated expression), C12orf65, CMAH, PDHB, RUNX3 (PTWAS = 0.0346, downregulated expression), SBF1, SH2B3, STK38, TMEM43, XPNPEP1, KIAA1530, NUFIP2, PPP2R3C, RAB24, STX6, and TLR5 (PTWAS = 0.04665, upregulated expression), were validated with integrative analysis of TWAS and mRNA expression profiles. TWAS-identified genes functionally involved in endoplasmic reticulum organization, regulation of cytokine production, TNF signaling pathway, immune response-regulating signaling pathway, regulation of autophagy, etc. Conclusion We identified multiple candidate genes and pathways, providing novel clues for the genetic mechanism of RA. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02419-9.
Collapse
Affiliation(s)
- Cuiyan Wu
- School of Public Health, Xi'an Jiaotong University Health Science Center; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, No.76, Yan Ta West Road, Xi'an, 710061, People's Republic of China.
| | - Sijian Tan
- School of Public Health, Xi'an Jiaotong University Health Science Center; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, No.76, Yan Ta West Road, Xi'an, 710061, People's Republic of China
| | - Li Liu
- School of Public Health, Xi'an Jiaotong University Health Science Center; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, No.76, Yan Ta West Road, Xi'an, 710061, People's Republic of China
| | - Shiqiang Cheng
- School of Public Health, Xi'an Jiaotong University Health Science Center; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, No.76, Yan Ta West Road, Xi'an, 710061, People's Republic of China
| | - Peilin Li
- School of Public Health, Xi'an Jiaotong University Health Science Center; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, No.76, Yan Ta West Road, Xi'an, 710061, People's Republic of China
| | - Wenyu Li
- School of Public Health, Xi'an Jiaotong University Health Science Center; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, No.76, Yan Ta West Road, Xi'an, 710061, People's Republic of China
| | - Huan Liu
- School of Public Health, Xi'an Jiaotong University Health Science Center; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, No.76, Yan Ta West Road, Xi'an, 710061, People's Republic of China
| | - Feng'e Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, No.76, Yan Ta West Road, Xi'an, 710061, People's Republic of China
| | - Sen Wang
- School of Public Health, Xi'an Jiaotong University Health Science Center; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, No.76, Yan Ta West Road, Xi'an, 710061, People's Republic of China
| | - Yujie Ning
- School of Public Health, Xi'an Jiaotong University Health Science Center; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, No.76, Yan Ta West Road, Xi'an, 710061, People's Republic of China
| | - Yan Wen
- School of Public Health, Xi'an Jiaotong University Health Science Center; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, No.76, Yan Ta West Road, Xi'an, 710061, People's Republic of China
| | - Feng Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center; Key Laboratory of Trace Elements and Endemic Diseases, National Health Commission of the People's Republic of China, No.76, Yan Ta West Road, Xi'an, 710061, People's Republic of China.
| |
Collapse
|
17
|
Comparative analysis on the anti-inflammatory/immune effect of mesenchymal stem cell therapy for the treatment of pulmonary arterial hypertension. Sci Rep 2021; 11:2012. [PMID: 33479312 PMCID: PMC7820276 DOI: 10.1038/s41598-021-81244-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/30/2020] [Indexed: 02/06/2023] Open
Abstract
Despite the advancement of targeted therapy for pulmonary arterial hypertension (PAH), poor prognosis remains a reality. Mesenchymal stem cells (MSCs) are one of the most clinically feasible alternative treatment options. We compared the treatment effects of adipose tissue (AD)-, bone marrow (BD)-, and umbilical cord blood (UCB)-derived MSCs in the rat monocrotaline-induced pulmonary hypertension (PH) model. The greatest improvement in the right ventricular function was observed in the UCB-MSCs treated group. The UCB-MSCs treated group also exhibited the greatest improvement in terms of the largest decrease in the medial wall thickness, perivascular fibrosis, and vascular cell proliferation, as well as the lowest levels of recruitment of innate and adaptive immune cells and associated inflammatory cytokines. Gene expression profiling of lung tissue confirmed that the UCB-MSCs treated group had the most notably attenuated immune and inflammatory profiles. Network analysis further revealed that the UCB-MSCs group had the greatest therapeutic effect in terms of the normalization of all three classical PAH pathways. The intravenous injection of the UCB-MSCs, compared with those of other MSCs, showed superior therapeutic effects in the PH model for the (1) right ventricular function, (2) vascular remodeling, (3) immune/inflammatory profiles, and (4) classical PAH pathways.
Collapse
|
18
|
Abstract
Bone homeostasis is maintained by a balance in the levels of osteoclast and osteoblast activity. Osteoclasts are bone-resorbing cells and have been shown to act as key players in various osteolytic diseases. Osteoclasts differentiate from monocyte/macrophage lineage cells in the presence of receptor activator of nuclear factor-κB ligand and macrophage colony-stimulating factor. Osteoblasts support osteoclastogenesis by producing several osteoclast differentiation factors. Toll-like receptors (TLRs) are members of the pattern recognition receptor family that are involved in recognizing pathogen-associated molecular patterns and damage-associated molecular patterns in response to pathogen infection. TLRs regulate osteoclastogenesis and bone resorption through either the myeloid differentiation primary response 88 or the Toll/interleukin-1 receptor domain-containing adapter-inducing interferon-β signaling pathways. Since osteoclasts play a central role in the progression of osteolytic diseases, extensive research focusing on TLR downstream signaling in these cells should be conducted to advance the development of effective TLR modulators. In this review, we summarize the currently available information on the role of TLRs in osteoclast differentiation and osteolytic diseases.
Collapse
Affiliation(s)
- Mijung Yim
- College of Pharmacy, Sookmyung Women's University, Seoul, Korea
| |
Collapse
|
19
|
Zafari P, Golpour M, Hafezi N, Bashash D, Esmaeili SA, Tavakolinia N, Rafiei A. Tuberculosis comorbidity with rheumatoid arthritis: Gene signatures, associated biomarkers, and screening. IUBMB Life 2020; 73:26-39. [PMID: 33217772 DOI: 10.1002/iub.2413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/01/2020] [Accepted: 11/04/2020] [Indexed: 12/19/2022]
Abstract
Rheumatoid arthritis (RA) is known to be related to an elevated risk of infections because of its pathobiology and the use of immunosuppressive therapies. Reactivation of latent tuberculosis (TB) infection is a serious issue in patients with RA, especially after receiving anti-TNFs therapy. TNF blocking reinforces the TB granuloma formation and maintenance and the growth of Mycobacterium tuberculosis (Mtb). After intercurrent of TB infection, the standard recommendation is that the treatment with TNF inhibitors to be withheld despite its impressive effect on suppression of inflammation until the infection has resolved. Knowing pathways and mechanisms that are common between two diseases might help to find the mechanistic basis of this comorbidity, as well as provide us a new approach to apply them as therapeutic targets or diagnostic biomarkers. Also, screening for latent TB before initiation of an anti-TNF therapy can minimize complications. This review summarizes the shared gene signature between TB and RA and discusses the biomarkers for early detection of this infection, and screening procedures as well.
Collapse
Affiliation(s)
- Parisa Zafari
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.,Student Research Committee, Mazandaran University of Medical Sciences, Sari, Iran
| | - Monireh Golpour
- Molecular and Cellular Biology Research Center, Student Research Committee, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nasim Hafezi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Naeimeh Tavakolinia
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Rafiei
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
20
|
Jung JY, Kim JW, Suh CH, Kim HA. Roles of Interactions Between Toll-Like Receptors and Their Endogenous Ligands in the Pathogenesis of Systemic Juvenile Idiopathic Arthritis and Adult-Onset Still's Disease. Front Immunol 2020; 11:583513. [PMID: 33224145 PMCID: PMC7674197 DOI: 10.3389/fimmu.2020.583513] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022] Open
Abstract
Systemic juvenile idiopathic arthritis (JIA) and adult-onset Still’s disease (AOSD) are systemic inflammatory disorders that manifest as high-spiking fever, joint pain, evanescent skin rash, and organomegaly. Their pathogenesis is unclear, but inflammation is triggered by activation of the innate immune system with aberrant production of proinflammatory cytokines. Along with extrinsic factors, intrinsic pathways can trigger an unexpected immune response. Damage-associated molecular patterns (DAMPs) induce the activation of innate immune cells, leading to sterile inflammation in systemic JIA and AOSD. These endogenous proteins interact with Toll-like receptors (TLRs), which are pattern recognition receptors, and mediate immune signaling following stimulation by pathogen-associated molecular patterns and DAMPs. Several DAMPs, such as S100 proteins, play a role in the development or severity of systemic JIA and AOSD, in which their interactions with TLRs are altered. Also, the expression levels of genes encoding DAMPs contribute to the susceptibility to systemic JIA and AOSD. Herein, we review reports that TLR and DAMP signaling initiates and/or maintains the inflammatory response in systemic JIA and AOSD, and their correlations with the clinical characteristics of those diseases. In addition, we assess their utility as biomarkers or therapeutics for systemic JIA and AOSD.
Collapse
Affiliation(s)
- Ju-Yang Jung
- Department of Rheumatology, Ajou University of Medical School, Suwon, South Korea
| | - Ji-Won Kim
- Department of Rheumatology, Ajou University of Medical School, Suwon, South Korea
| | - Chang-Hee Suh
- Department of Rheumatology, Ajou University of Medical School, Suwon, South Korea
| | - Hyoun-Ah Kim
- Department of Rheumatology, Ajou University of Medical School, Suwon, South Korea
| |
Collapse
|
21
|
Mendez ME, Sebastian A, Murugesh DK, Hum NR, McCool JL, Hsia AW, Christiansen BA, Loots GG. LPS-Induced Inflammation Prior to Injury Exacerbates the Development of Post-Traumatic Osteoarthritis in Mice. J Bone Miner Res 2020; 35:2229-2241. [PMID: 32564401 PMCID: PMC7689775 DOI: 10.1002/jbmr.4117] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 06/08/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022]
Abstract
Osteoarthritis (OA) is a debilitating and painful disease characterized by the progressive loss of articular cartilage. Post-traumatic osteoarthritis (PTOA) is an injury-induced type of OA that persists in an asymptomatic phase for years before it becomes diagnosed in ~50% of injured individuals. Although PTOA is not classified as an inflammatory disease, it has been suggested that inflammation could be a major driver of PTOA development. Here we examined whether a state of systemic inflammation induced by lipopolysaccharide (LPS) administration 5-days before injury would modulate PTOA outcomes. RNA-seq analysis at 1-day post-injury followed by micro-computed tomography (μCT) and histology characterization at 6 weeks post-injury revealed that LPS administration causes more severe PTOA phenotypes. These phenotypes included significantly higher loss of cartilage and subchondral bone volume. Gene expression analysis showed that LPS alone induced a large cohort of inflammatory genes previously shown to be elevated in synovial M1 macrophages of rheumatoid arthritis (RA) patients, suggesting that systemic LPS produces synovitis. This synovitis was sufficient to promote PTOA in MRL/MpJ mice, a strain previously shown to be resistant to PTOA. The synovium of LPS-treated injured joints displayed an increase in cellularity, and immunohistological examination confirmed that this increase was in part attributable to an elevation in type 1 macrophages. LPS induced the expression of Tlr7 and Tlr8 in both injured and uninjured joints, genes known to be elevated in RA. We conclude that inflammation before injury is an important risk factor for the development of PTOA and that correlating patient serum endotoxin levels or their state of systemic inflammation with PTOA progression may help develop new, effective treatments to lower the rate of PTOA in injured individuals. © 2020 The Authors. Journal of Bone and Mineral Research published by American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Melanie E Mendez
- Physical and Life Science Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA.,School of Natural Sciences, University of California Merced, Merced, CA, USA
| | - Aimy Sebastian
- Physical and Life Science Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Deepa K Murugesh
- Physical and Life Science Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Nicholas R Hum
- Physical and Life Science Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA.,School of Natural Sciences, University of California Merced, Merced, CA, USA
| | - Jillian L McCool
- Physical and Life Science Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA.,School of Natural Sciences, University of California Merced, Merced, CA, USA
| | - Allison W Hsia
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, USA
| | - Blaine A Christiansen
- Department of Orthopaedic Surgery, University of California Davis Health, Sacramento, CA, USA
| | - Gabriela G Loots
- Physical and Life Science Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA.,School of Natural Sciences, University of California Merced, Merced, CA, USA
| |
Collapse
|
22
|
Mendez ME, Murugesh DK, Sebastian A, Hum NR, McCloy SA, Kuhn EA, Christiansen BA, Loots GG. Antibiotic Treatment Prior to Injury Improves Post-Traumatic Osteoarthritis Outcomes in Mice. Int J Mol Sci 2020; 21:E6424. [PMID: 32899361 PMCID: PMC7503363 DOI: 10.3390/ijms21176424] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 12/21/2022] Open
Abstract
Osteoarthritis (OA) is a painful and debilitating disease characterized by the chronic and progressive degradation of articular cartilage. Post-traumatic OA (PTOA) is a secondary form of OA that develops in ~50% of cases of severe articular injury. Inflammation and re-occurring injury have been implicated as contributing to the progression of PTOA after the initial injury. However, there is very little known about external factors prior to injury that could affect the risk of PTOA development. To examine how the gut microbiome affects PTOA development we used a chronic antibiotic treatment regimen starting at weaning for six weeks prior to ACL rupture, in mice. A six-weeks post-injury histological examination showed more robust cartilage staining on the antibiotic (AB)-treated mice than the untreated controls (VEH), suggesting slower disease progression in AB cohorts. Injured joints also showed an increase in the presence of anti-inflammatory M2 macrophages in the AB group. Molecularly, the phenotype correlated with a significantly lower expression of inflammatory genes Tlr5, Ccl8, Cxcl13, and Foxo6 in the injured joints of AB-treated animals. Our results indicate that a reduced state of inflammation at the time of injury and a lower expression of Wnt signaling modulatory protein, Rspo1, caused by AB treatment can slow down or improve PTOA outcomes.
Collapse
Affiliation(s)
- Melanie E. Mendez
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Deepa K. Murugesh
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Aimy Sebastian
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Nicholas R. Hum
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
- UC Merced, School of Natural Sciences, Merced, CA 95343, USA
| | - Summer A. McCloy
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | - Edward A. Kuhn
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
| | | | - Gabriela G. Loots
- Lawrence Livermore National Laboratories, Physical and Life Sciences Directorate, Livermore, CA 94550, USA; (M.E.M.); (D.K.M.); (A.S.); (N.R.H.); (S.A.M.); (E.A.K.)
- UC Merced, School of Natural Sciences, Merced, CA 95343, USA
| |
Collapse
|
23
|
Keirns BH, Lucas EA, Smith BJ. Phytochemicals affect T helper 17 and T regulatory cells and gut integrity: implications on the gut-bone axis. Nutr Res 2020; 83:30-48. [PMID: 33010588 DOI: 10.1016/j.nutres.2020.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 08/05/2020] [Accepted: 08/11/2020] [Indexed: 12/16/2022]
Abstract
The pathology of osteoporosis is multifactorial, but a growing body of evidence supports an important role of the gut-bone axis, especially in bone loss associated with menopause, rheumatoid arthritis, and periodontal disease. Aberrant T cell responses favoring an increase in the ratio of T helper 17 cells to T regulatory cells play a critical role in the underlying etiology of this bone loss. Many of the dietary phytochemicals known to have osteoprotective activity such as flavonoids, organosulfur compounds, phenolic acids, as well as the oligosaccharides also improve gut barrier function and affect T cell differentiation and activation within gut-associated lymphoid tissues and at distal sites. Here, we examine the potential of these phytochemicals to act as prebiotics and immunomodulating agents, in part targeting the gut to mediate their effects on bone.
Collapse
Affiliation(s)
- Bryant H Keirns
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078.
| | - Edralin A Lucas
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078.
| | - Brenda J Smith
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078.
| |
Collapse
|
24
|
Kim SJ, Chang HJ, Volin MV, Umar S, Van Raemdonck K, Chevalier A, Palasiewicz K, Christman JW, Volkov S, Arami S, Maz M, Mehta A, Zomorrodi RK, Fox DA, Sweiss N, Shahrara S. Macrophages are the primary effector cells in IL-7-induced arthritis. Cell Mol Immunol 2020; 17:728-740. [PMID: 31197255 PMCID: PMC7331600 DOI: 10.1038/s41423-019-0235-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 04/10/2019] [Indexed: 12/16/2022] Open
Abstract
Synovial macrophages are crucial in the development of joint inflammation and bone damage; however, the pathways that control macrophage remodeling in inflammatory M1 cells or bone-eroding osteoclasts are not fully understood. We determined that elevated IL-7R/CD127 expression is the hallmark of rheumatoid arthritis (RA) M1 macrophages and that these cells are highly responsive to interleukin-7 (IL-7)-driven osteoclastogenesis. We established that lipopolysaccharide (LPS), interferon-γ (IFNγ), and tumor necrosis factor-α (TNFα), the classic M1 macrophage mediators, enhance IL-7R expression in RA and murine macrophages. The local expression of IL-7 provokes arthritis, predominantly through escalating the number of F480+iNOS+ cells rather than CD3+ T cells. Ectopic LPS injection stabilizes IL-7-induced arthritis by increasing myeloid IL-7R expression, in part via IFNγ induction. Hence, in RAG-/- mice, IL-7-mediated arthritis is suppressed because of the reduction in myeloid IL-7R expression due to the lack of IFNγ. Moreover, the amelioration of IL-7-induced arthritis by anti-TNF therapy is due to a decrease in the number of cells in the unique F480+iNOS+IL-7R+CCL5+ subset, with no impact on the F480+Arginase+ cell or CD3+ T cell frequency. Consistent with the preclinical findings, the findings of a phase 4 study performed with RA patients following 6 months of anti-TNF therapy revealed that IL-7R expression was reduced without affecting the levels of IL-7. This study shifts the paradigm by discovering that IL-7-induced arthritis is dependent on F480+iNOS+IL-7R+CCL5+ cell function, which activates TH-1 cells to amplify myeloid IL-7R expression and disease severity.
Collapse
Affiliation(s)
- Seung-Jae Kim
- Division of Rheumatology, Jesse Brown VA, Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Huan J Chang
- Division of Rheumatology, Jesse Brown VA, Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Michael V Volin
- Department of Microbiology and Immunology, Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL, 60515, USA
| | - Sadiq Umar
- Division of Rheumatology, Jesse Brown VA, Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Katrien Van Raemdonck
- Division of Rheumatology, Jesse Brown VA, Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Aimee Chevalier
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Karol Palasiewicz
- Division of Rheumatology, Jesse Brown VA, Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - John W Christman
- Pulmonary, Critical Care and Sleep Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, 43210, USA
| | - Suncica Volkov
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Shiva Arami
- Division of Rheumatology, Jesse Brown VA, Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Mehrdad Maz
- Division of Allergy, Clinical Immunology and Rheumatology, The University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Anjali Mehta
- Division of Rheumatology, Jesse Brown VA, Medical Center, Chicago, IL, 60612, USA
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Ryan K Zomorrodi
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - David A Fox
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI, 481096, USA
| | - Nadera Sweiss
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Shiva Shahrara
- Division of Rheumatology, Jesse Brown VA, Medical Center, Chicago, IL, 60612, USA.
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
25
|
Root-Bernstein R. Synergistic Activation of Toll-Like and NOD Receptors by Complementary Antigens as Facilitators of Autoimmune Disease: Review, Model and Novel Predictions. Int J Mol Sci 2020; 21:ijms21134645. [PMID: 32629865 PMCID: PMC7369971 DOI: 10.3390/ijms21134645] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/26/2020] [Accepted: 06/27/2020] [Indexed: 12/29/2022] Open
Abstract
Persistent activation of toll-like receptors (TLR) and nucleotide-binding oligomerization domain-containing proteins (NOD) in the innate immune system is one necessary driver of autoimmune disease (AD), but its mechanism remains obscure. This study compares and contrasts TLR and NOD activation profiles for four AD (autoimmune myocarditis, myasthenia gravis, multiple sclerosis and rheumatoid arthritis) and their animal models. The failure of current AD theories to explain the disparate TLR/NOD profiles in AD is reviewed and a novel model is presented that explains innate immune support of persistent chronic inflammation in terms of unique combinations of complementary AD-specific antigens stimulating synergistic TLRs and/or NODs. The potential explanatory power of the model is explored through testable, novel predictions concerning TLR- and NOD-related AD animal models and therapies.
Collapse
|
26
|
Van Raemdonck K, Umar S, Palasiewicz K, Volkov S, Volin MV, Arami S, Chang HJ, Zanotti B, Sweiss N, Shahrara S. CCL21/CCR7 signaling in macrophages promotes joint inflammation and Th17-mediated osteoclast formation in rheumatoid arthritis. Cell Mol Life Sci 2020; 77:1387-1399. [PMID: 31342120 PMCID: PMC10040247 DOI: 10.1007/s00018-019-03235-w] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/08/2019] [Accepted: 07/18/2019] [Indexed: 10/26/2022]
Abstract
In rheumatoid arthritis (RA), synovial tissue abundantly expresses CCL21, a chemokine strongly associated with RA susceptibility. In this study, we aimed to characterize the functional significance of CCL21/CCR7 signaling in different phases of RA pathogenesis. We determined that CCR7 is a hallmark of RA M1 synovial fluid (SF) macrophages, and its expression in RA monocytes and in vitro differentiated macrophages is closely associated with disease activity score (DAS28). In early stages of RA, monocytes infiltrate the synovial tissue. However, blockade of SF CCL21 or CCR7 prevents RA SF-mediated monocyte migration. CCR7 expression in the newly migrated macrophages can be accentuated by LPS and IFNγ and suppressed by IL-4 treatment. We also uncovered that CCL21 stimulation increases the number of M1-polarized macrophages (CD14+CD86+), resulting in elevated transcription of IL-6 and IL-23. These CCL21-induced M1 cytokines differentiate naïve T cells to Th17 cells, without affecting Th1 cell polarization. In the erosive stages of disease, CCL21 potentiates RA osteoclastogenesis through M1-driven Th17 polarization. Disruption of this intricate crosstalk, by blocking IL-6, IL-23, or IL-17 function, impairs the osteoclastogenic capacity of CCL21. Consistent with our in vitro findings, we establish that arthritis mediated by CCL21 expands the joint inflammation to bone erosion by connecting the differentiation of M1 macrophages with Th17 cells. Disease progression is further exacerbated by CCL21-induced neovascularization. We conclude that CCL21 is an attractive novel target for RA therapy, as blockade of its function may abrogate erosive arthritis modulated by M1 macrophages and Th17 cell crosstalk.
Collapse
Affiliation(s)
- Katrien Van Raemdonck
- Jesse Brown VA Medical Center, Chicago, IL, USA.,Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA
| | - Sadiq Umar
- Jesse Brown VA Medical Center, Chicago, IL, USA.,Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA
| | - Karol Palasiewicz
- Jesse Brown VA Medical Center, Chicago, IL, USA.,Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA
| | - Suncica Volkov
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA
| | - Michael V Volin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - Shiva Arami
- Jesse Brown VA Medical Center, Chicago, IL, USA.,Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA
| | - Huan J Chang
- Jesse Brown VA Medical Center, Chicago, IL, USA.,Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA
| | - Brian Zanotti
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - Nadera Sweiss
- Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL, USA. .,Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, 840 S Wood Street, CSB Suite 1114, Chicago, IL, 60612, USA.
| |
Collapse
|
27
|
Yu F, Qiu C, Xu C, Tian Q, Zhao LJ, Wu L, Deng HW, Shen H. Mendelian Randomization Identifies CpG Methylation Sites With Mediation Effects for Genetic Influences on BMD in Peripheral Blood Monocytes. Front Genet 2020; 11:60. [PMID: 32180791 PMCID: PMC7059767 DOI: 10.3389/fgene.2020.00060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 01/17/2020] [Indexed: 12/18/2022] Open
Abstract
Osteoporosis is mainly characterized by low bone mineral density (BMD) and is an increasingly serious public health concern. DNA methylation is a major epigenetic mechanism that may contribute to the variation in BMD and may mediate the effects of genetic and environmental factors of osteoporosis. In this study, we performed an epigenome-wide DNA methylation analysis in peripheral blood monocytes of 118 Caucasian women with extreme BMD values. Further, we developed and implemented a novel analytical framework that integrates Mendelian randomization with genetic fine mapping and colocalization to evaluate the causal relationships between DNA methylation and BMD phenotype. We identified 2,188 differentially methylated CpGs (DMCs) between the low and high BMD groups and distinguished 30 DMCs that may mediate the genetic effects on BMD. The causal relationship was further confirmed by eliminating the possibility of horizontal pleiotropy, linkage effect and reverse causality. The fine-mapping analysis determined 25 causal variants that are most likely to affect the methylation levels at these mediator DMCs. The majority of the causal methylation quantitative loci and DMCs reside within cell type-specific histone mark peaks, enhancers, promoters, promoter flanking regions and CTCF binding sites, supporting the regulatory potentials of these loci. The established causal pathways from genetic variant to BMD phenotype mediated by DNA methylation provide a gene list to aid in designing future functional studies and lead to a better understanding of the genetic and epigenetic mechanisms underlying the variation of BMD.
Collapse
Affiliation(s)
- Fangtang Yu
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Chuan Qiu
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Chao Xu
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Qing Tian
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Lan-Juan Zhao
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Li Wu
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Hong-Wen Deng
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
- School of Basic Medical Science, Central South University, Changsha, China
| | - Hui Shen
- Center for Bioinformatics and Genomics, Department of Global Biostatistics and Data Science, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| |
Collapse
|
28
|
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: 3.6] [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
|
29
|
Mühl H, Bachmann M. IL-18/IL-18BP and IL-22/IL-22BP: Two interrelated couples with therapeutic potential. Cell Signal 2019; 63:109388. [PMID: 31401146 DOI: 10.1016/j.cellsig.2019.109388] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 02/06/2023]
Abstract
Interleukin (IL)-18 and IL-22 are key components of cytokine networks that play a decisive role in (pathological) inflammation, host defense, and tissue regeneration. Tight regulation of cytokine-driven signaling, inflammation, and immunoactivation is supposed to enable nullification of a given deleterious trigger without mediating overwhelming collateral tissue damage or even activating a cancerous face of regeneration. In fact, feedback regulation by specific cytokine opponents is regarded as a major means by which the immune system is kept in balance. Herein, we shine a light on the interplay between IL-18 and IL-22 and their opponents IL-18 binding protein (IL-18BP) and IL-22BP in order to provide integrated information on their biology, pathophysiological significance, and prospect as targets and/or instruments of therapeutic intervention.
Collapse
Affiliation(s)
- Heiko Mühl
- pharmazentrum frankfurt/ZAFES, University Hospital Goethe University Frankfurt am Main, Theodor-Stern- Kai 7, 60590 Frankfurt am Main, Germany.
| | - Malte Bachmann
- pharmazentrum frankfurt/ZAFES, University Hospital Goethe University Frankfurt am Main, Theodor-Stern- Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
30
|
Petrackova A, Horak P, Radvansky M, Skacelova M, Fillerova R, Kudelka M, Smrzova A, Mrazek F, Kriegova E. Cross-Disease Innate Gene Signature: Emerging Diversity and Abundance in RA Comparing to SLE and SSc. J Immunol Res 2019; 2019:3575803. [PMID: 31396542 PMCID: PMC6664489 DOI: 10.1155/2019/3575803] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 06/12/2019] [Indexed: 12/19/2022] Open
Abstract
Overactivation of the innate immune system together with the impaired downstream pathway of type I interferon-responding genes is a hallmark of rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and systemic sclerosis (SSc). To date, limited data on the cross-disease innate gene signature exists among those diseases. We compared therefore an innate gene signature of Toll-like receptors (TLRs), seven key members of the interleukin (IL)1/IL1R family, and CXCL8/IL8 in peripheral blood mononuclear cells from well-defined patients with active stages of RA (n = 36, DAS28 ≥ 3.2), SLE (n = 28, SLEDAI > 6), and SSc (n = 22, revised EUSTAR index > 2.25). Emerging diversity and abundance of the innate signature in RA patients were detected: RA was characterized by the upregulation of TLR3, TLR5, IL1RAP/IL1R3, IL18R1, and SIGIRR/IL1R8 when compared to SSc (P corr < 0.02) and of TLR2, TLR5, and SIGIRR/IL1R8 when compared to SLE (P corr < 0.02). Applying the association rule analysis, six rules (combinations and expression of genes describing disease) were identified for RA (most frequently included high TLR3 and/or IL1RAP/IL1R3) and three rules for SLE (low IL1RN and IL18R1) and SSc (low TLR5 and IL18R1). This first cross-disease study identified emerging heterogeneity in the innate signature of RA patients with many upregulated innate genes compared to that of SLE and SSc.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Arthritis, Rheumatoid/blood
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/immunology
- Female
- Humans
- Immunity, Innate/genetics
- Interleukin-1/genetics
- Interleukin-1/metabolism
- Interleukin-8/genetics
- Interleukin-8/metabolism
- Leukocytes, Mononuclear/metabolism
- Lupus Erythematosus, Systemic/blood
- Lupus Erythematosus, Systemic/genetics
- Lupus Erythematosus, Systemic/immunology
- Male
- Middle Aged
- Receptors, Interleukin-1 Type I/genetics
- Receptors, Interleukin-1 Type I/metabolism
- Scleroderma, Systemic/blood
- Scleroderma, Systemic/genetics
- Scleroderma, Systemic/immunology
- Toll-Like Receptors/genetics
- Toll-Like Receptors/metabolism
- Transcriptome
Collapse
Affiliation(s)
- Anna Petrackova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Pavel Horak
- Department of Internal Medicine III-Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Martin Radvansky
- Faculty of Electrical Engineering and Computer Science, Department of Computer Science, VSB-Technical University of Ostrava, Ostrava, Czech Republic
| | - Martina Skacelova
- Department of Internal Medicine III-Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Regina Fillerova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Milos Kudelka
- Faculty of Electrical Engineering and Computer Science, Department of Computer Science, VSB-Technical University of Ostrava, Ostrava, Czech Republic
| | - Andrea Smrzova
- Department of Internal Medicine III-Nephrology, Rheumatology and Endocrinology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Frantisek Mrazek
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| | - Eva Kriegova
- Department of Immunology, Faculty of Medicine and Dentistry, Palacky University Olomouc and University Hospital, Olomouc, Czech Republic
| |
Collapse
|
31
|
Badr MT, Häcker G. Gene expression profiling meta-analysis reveals novel gene signatures and pathways shared between tuberculosis and rheumatoid arthritis. PLoS One 2019; 14:e0213470. [PMID: 30845171 PMCID: PMC6405138 DOI: 10.1371/journal.pone.0213470] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 02/21/2019] [Indexed: 12/16/2022] Open
Abstract
Tuberculosis (TB) is among the leading causes of death by infectious diseases. An epidemiological association between Mycobacterium tuberculosis infection and autoimmune diseases like rheumatoid arthritis (RA) has been reported but it remains unclear if there is a causal relationship, and if so, which molecular pathways and regulatory mechanisms contribute to it. Here we used a computational biology approach by global gene expression meta-analysis to identify candidate genes and pathways that may link TB and RA. Data were collected from public expression databases such as NCBI GEO. Studies were selected that analyzed mRNA-expression in whole blood or blood cell populations in human case control studies at comparable conditions. Six TB and RA datasets (41 active TB patients, 33 RA patients, and 67 healthy controls) were included in the downstream analysis. This approach allowed the identification of deregulated genes that had not been identified in the single analysis of TB or RA patients and that were co-regulated in TB and RA patients compared to healthy subjects. The genes encoding TLR5, TNFSF10/TRAIL, PPP1R16B/TIMAP, SIAH1, PIK3IP1, and IL17RA were among the genes that were most significantly deregulated in TB and RA. Pathway enrichment analysis revealed 'T cell receptor signaling pathway', 'Toll-like receptor signaling pathway,' and 'virus defense related pathways' among the pathways most strongly associated with both diseases. The identification of a common gene signature and pathways substantiates the observation of an epidemiological association of TB and RA and provides clues on the mechanistic basis of this association. Newly identified genes may be a basis for future functional and epidemiological studies.
Collapse
Affiliation(s)
- M. T. Badr
- Institute of Medical Microbiology and Hygiene, Medical Center—University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - G. Häcker
- Institute of Medical Microbiology and Hygiene, Medical Center—University of Freiburg, Faculty of Medicine, Freiburg, Germany
- BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
32
|
Li S, Xiang C, Wei X, Sun X, Li R, Li P, Sun J, Wei D, Chen Y, Zhang Y, Wei L. Early supplemental α2-macroglobulin attenuates cartilage and bone damage by inhibiting inflammation in collagen II-induced arthritis model. Int J Rheum Dis 2019; 22:654-665. [PMID: 30609267 DOI: 10.1111/1756-185x.13457] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 11/08/2018] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To determine if early supplemental intra-articular α2-macroglobulin (A2M) has a chondroprotective effect in a collagen II-induced arthritis (CIA) mice model. METHODS DBA/1 mice were randomized into four groups (n = 15/group): (a) CIA + 1.2 μg of A2M; (b) CIA + 0.8 μg of A2M; (c) CIA + 0.4 μg of A2M; (d) vehicle + phosphate-buffered saline (PBS). A2M was injected into right ankles and PBS was injected into the left ankles simultaneously as internal control at days 36, 43 and 50. The CIA inflammation clinical score and ankle thickness were recorded every other day starting on day 21 until sacrifice. Changes in inflammation were monitored by in vivo fluorescence molecular tomography (FMT). Inflammation, cartilage and bone damage were assessed with X-ray, histology and immunohistochemistry. Cartilage and inflammation-related gene expression was quantified by real-time polymerase chain reaction (PCR). RESULTS All mice showed ankle inflammation on day 33. After day 43, lower clinical scores, ankle thickness and Sharp/van der Heijde method scores in A2M-treated ankles compared with PBS-treated ankles. FMT data indicated that the inflammation markers MMPSense and ProSense were significantly elevated in the PBS-treated ankles than A2M-treated ankles. Histology and X-ray analyses indicated that A2M administration resulted in lower levels of inflammatory infiltration and synovial hyperplasia, as well as more typical cartilage and bone organization with increased COL II and Aggrecan staining when compared with PBS-treated ankles. In addition, real-time PCR showed that,matrix metalloproteinase-3, -9, -13, COL X and Runx2 were significantly less expressed in A2M-treated groups than PBS-treated animals. CONCLUSION Early supplemental intra-articular A2M exerts an anti-inflammatory effect and attenuates cartilage and bone damage in a CIA model.
Collapse
Affiliation(s)
- Shengchun Li
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Chuan Xiang
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaochun Wei
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaojuan Sun
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Ruifang Li
- The Third people's Hospital of Hubei Province, Wuhan, China
| | - Pengcui Li
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jian Sun
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Dinglu Wei
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yong Chen
- The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yanxiang Zhang
- The Third people's Hospital of Hubei Province, Wuhan, China
| | - Lei Wei
- The Second Hospital of Shanxi Medical University, Taiyuan, China.,Department of Orthopedics, Warren Alpert Medical School of Brown University/RIH, Providence, Rhode Island
| |
Collapse
|
33
|
Bone erosions in rheumatoid arthritis: recent developments in pathogenesis and therapeutic implications. JOURNAL OF MUSCULOSKELETAL & NEURONAL INTERACTIONS 2018; 18:304-319. [PMID: 30179207 PMCID: PMC6146189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Bone erosions develop early in the course of rheumatoid arthritis (RA) and deteriorate progressively, causing joint damage and resulting in impaired functional capacity of patients. During the last years, considerable number of studies has increased our understanding of the pathogenetic mechanisms mediating the development of bone erosions in RA. Increased production of RANKL and other cytokines, dysregulation of innate immune mechanisms, autoantibodies specific to RA and alterations of microRNA expression stimulate differentiation and function of osteoclasts, which are responsible for the development of bone erosions. Besides, increased levels of cytokines, overproduction of antagonists of the canonical Wnt signaling pathway and deficient production of bone morphogenetic proteins result in impaired osteoblast differentiation and function, undermining the capacity of bone erosions to repair. Disease-modifying antirheumatic drugs, synthetic or biological, currently used in the treatment of RA, can halt the progression of bone erosions and may even lead to partial repair, although complete repair is unattainable. Targeting pathogenetic mechanisms participating in the erosive process may add to the therapeutic effect of DMARDs and help in the prevention or repair of bone erosions. However, more studies are still needed to confirm whether such therapeutic strategies are effective.
Collapse
|
34
|
Bendickova K, Tidu F, Fric J. Calcineurin-NFAT signalling in myeloid leucocytes: new prospects and pitfalls in immunosuppressive therapy. EMBO Mol Med 2018; 9:990-999. [PMID: 28606994 PMCID: PMC5538425 DOI: 10.15252/emmm.201707698] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Myeloid leucocytes mediate host protection against infection and critically regulate inflammatory responses in body tissues. Pattern recognition receptor signalling is crucial for myeloid cell responses to pathogens, but growing evidence suggests an equally potent role for Calcineurin–NFAT signalling in control of myeloid cell function. All major subsets of myeloid leucocytes employ Calcineurin–NFAT signalling during immune responses to pathogens and/or tissue damage, but the influence this pathway exerts on pathogen clearance and host susceptibility to infection is not fully understood. Recent data from experimental models indicate that Calcineurin‐NFAT signalling is essential for infection control, and calcineurin inhibitors used in transplantation medicine (including cyclosporine A and tacrolimus) are now being tested for efficacy in a diverse range of inflammatory conditions and autoimmune pathologies. Efforts to repurpose calcineurin inhibitor drugs for new therapeutic applications may yield rapid improvements in clinical outcomes, but the potential impact of these compounds on myeloid cell function in treated patients is largely unknown. Here we discuss Calcineurin–NFAT control of myeloid leucocyte function in the context of recent therapeutic developments and ongoing clinical studies.
Collapse
Affiliation(s)
- Kamila Bendickova
- Center for Translational Medicine (CTM), International Clinical Research Center (ICRC), St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Federico Tidu
- Center for Translational Medicine (CTM), International Clinical Research Center (ICRC), St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Jan Fric
- Center for Translational Medicine (CTM), International Clinical Research Center (ICRC), St. Anne's University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
35
|
VLR Recognition of TLR5 Expands the Molecular Characterization of Protein Antigen Binding by Non-Ig-based Antibodies. J Mol Biol 2018; 430:1350-1367. [PMID: 29596914 DOI: 10.1016/j.jmb.2018.03.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 03/20/2018] [Accepted: 03/20/2018] [Indexed: 01/24/2023]
Abstract
Variable lymphocyte receptors (VLRs) are unconventional adaptive immune receptors relatively recently discovered in the phylogenetically ancient jawless vertebrates, lamprey and hagfish. VLRs bind antigens using a leucine-rich repeat fold and are the only known adaptive immune receptors that do not utilize an immunoglobulin fold for antigen recognition. While immunoglobulin antibodies have been studied extensively, there are comparatively few studies on antigen recognition by VLRs, particularly for protein antigens. Here we report isolation, functional and structural characterization of three VLRs that bind the protein toll-like receptor 5 (TLR5) from zebrafish. Two of the VLRs block binding of TLR5 to its cognate ligand flagellin in functional assays using reporter cells. Co-crystal structures revealed that these VLRs bind to two different epitopes on TLR5, both of which include regions involved in flagellin binding. Our work here demonstrates that the lamprey adaptive immune system can be used to generate high-affinity VLR clones that recognize different epitopes and differentially impact natural ligand binding to a protein antigen.
Collapse
|
36
|
Sessile Innate Immune Cells. DAMAGE-ASSOCIATED MOLECULAR PATTERNS IN HUMAN DISEASES 2018. [PMCID: PMC7123606 DOI: 10.1007/978-3-319-78655-1_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this chapter, sessile cells of the innate immune system are briefly introduced. Defined as cells equipped with diverse pattern recognition molecules capable of detecting MAMPs and DAMPs, they encompass cells such as epithelial cells, fibroblasts, vascular cells, chondrocytes, osteoblasts, and adipocytes. Located at the body surfaces, epithelial cells represent the first line of innate immune defense against invading microbial pathogens. They are significant contributors to innate mucosal immunity and generate various antimicrobial defense mechanisms. Also, epithelial cells critically contribute to tissue repair via the phenomenon of re-epithelialization. Fibroblasts operate as classical sentinel cells of the innate immune system dedicated to responding to MAMPs and DAMPs emitted upon any tissue injury. Typically, fibroblasts synthesize most of the extracellular matrix of connective tissues, thereby playing a crucial role in tissue repair processes. Vascular cells of the innate immune system represent an evolutionarily developed first-line defense against any inciting insult hitting the vessel walls from the luminal side including bacteria, viruses, microbial toxins, and chemical noxa such as nicotine. Upon such insults and following recognition of MAMPs and DAMPs, vascular cells react with an innate immune response to create an acute inflammatory milieu in the vessel wall aimed at curing the vascular injury concerned. Chondrocytes, osteoblasts, and osteoclasts represent other vital cells of the skeletal system acting as cells of the innate immune system in its wider sense. These cells mediate injury-promoted DAMP-induced inflammatory and regenerative processes specific for the skeletal systems. Finally, adipocytes are regarded as highly active cells of the innate immune system. As white, brown, and beige adipocytes, they operate as a dynamic metabolic organ that can secrete certain bioactive molecules which have endocrine, paracrine, and autocrine actions.
Collapse
|
37
|
Hernandez CJ. Bone Mechanical Function and the Gut Microbiota. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1033:249-270. [DOI: 10.1007/978-3-319-66653-2_12] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
38
|
Le Rossignol S, Ketheesan N, Haleagrahara N. Redox-sensitive transcription factors play a significant role in the development of rheumatoid arthritis. Int Rev Immunol 2017; 37:129-143. [PMID: 28898138 DOI: 10.1080/08830185.2017.1363198] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease which is associated with significant morbidity. Redox sensitive transcription factors including NF-κB, HIF, AP-1, and Nrf2 are intimately involved in the pathogenesis of RA. The treatment of this disease is limited by the elusive nature of the pathogenesis of RA. NF-κB is crucial for the maturation of immune cells as well as production of TNFα and MMPs, which escalate RA. HIF is essential for activation of inflammatory cells, angiogenesis and pannus formation in RA. AP-1 regulates cytokine and MMP production as well as synovial hyperplasia which are key processes in RA. Nrf2 is involved with chondrogenesis, osteoblastogenesis, prostaglandin secretion and ROS production in RA. Targeting two or more of these transcription factors may result in increased efficacy than either therapy in isolation. This review will highlight the control specific mediators on these transcription factors, the subsequent effect of these transcription factors once activated, and then mesh this with the pathogenesis of RA. The elucidation of key transcription factor regulation in the pathogenesis of RA may highlight the novel therapy interventions which may prove to have a greater efficacy than those therapies currently available.
Collapse
Affiliation(s)
- Scott Le Rossignol
- a College of Medicine and Dentistry , James Cook University Townsville , Queensland , Australia
| | - Natkunam Ketheesan
- b Biomedicine, College of Public Health, Medical and Veterinary Sciences , James Cook University , Townsville , Queensland , Australia.,c Australian Institute of Tropical Health and Medicine , James Cook University , Townsville , Queensland , Australia
| | - Nagaraja Haleagrahara
- b Biomedicine, College of Public Health, Medical and Veterinary Sciences , James Cook University , Townsville , Queensland , Australia.,c Australian Institute of Tropical Health and Medicine , James Cook University , Townsville , Queensland , Australia
| |
Collapse
|
39
|
Vergadi E, Ieronymaki E, Lyroni K, Vaporidi K, Tsatsanis C. Akt Signaling Pathway in Macrophage Activation and M1/M2 Polarization. THE JOURNAL OF IMMUNOLOGY 2017; 198:1006-1014. [PMID: 28115590 DOI: 10.4049/jimmunol.1601515] [Citation(s) in RCA: 718] [Impact Index Per Article: 89.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 09/26/2016] [Indexed: 01/05/2023]
Abstract
Macrophages become activated initiating innate immune responses. Depending on the signals, macrophages obtain an array of activation phenotypes, described by the broad terms of M1 or M2 phenotype. The PI3K/Akt/mTOR pathway mediates signals from multiple receptors including insulin receptors, pathogen-associated molecular pattern receptors, cytokine receptors, adipokine receptors, and hormones. As a result, the Akt pathway converges inflammatory and metabolic signals to regulate macrophage responses modulating their activation phenotype. Akt is a family of three serine-threonine kinases, Akt1, Akt2, and Akt3. Generation of mice lacking individual Akt, PI3K, or mTOR isoforms and utilization of RNA interference technology have revealed that Akt signaling pathway components have distinct and isoform-specific roles in macrophage biology and inflammatory disease regulation, by controlling inflammatory cytokines, miRNAs, and functions including phagocytosis, autophagy, and cell metabolism. Herein, we review the current knowledge on the role of the Akt signaling pathway in macrophages, focusing on M1/M2 polarization and highlighting Akt isoform-specific functions.
Collapse
Affiliation(s)
- Eleni Vergadi
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion 71003, Greece; and.,Laboratory of Intensive Care Medicine, School of Medicine, University of Crete, Heraklion 71003, Greece
| | - Eleftheria Ieronymaki
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion 71003, Greece; and
| | - Konstantina Lyroni
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion 71003, Greece; and
| | - Katerina Vaporidi
- Laboratory of Intensive Care Medicine, School of Medicine, University of Crete, Heraklion 71003, Greece
| | - Christos Tsatsanis
- Laboratory of Clinical Chemistry, School of Medicine, University of Crete, Heraklion 71003, Greece; and
| |
Collapse
|
40
|
Kim SJ, Chen Z, Essani AB, Elshabrawy HA, Volin MV, Volkov S, Swedler W, Arami S, Sweiss N, Shahrara S. Identification of a Novel Toll-like Receptor 7 Endogenous Ligand in Rheumatoid Arthritis Synovial Fluid That Can Provoke Arthritic Joint Inflammation. Arthritis Rheumatol 2017; 68:1099-110. [PMID: 26662519 DOI: 10.1002/art.39544] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 12/03/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Levels of Toll-like receptor 7 (TLR-7) are elevated in rheumatoid arthritis (RA), but the impact on RA is unknown because the endogenous ligand for TLR-7 has not been identified. The aim of this study was to identify a TLR-7 endogenous ligand and to determine its role in the pathogenesis of RA. METHODS The presence of an endogenous TLR-7 ligand, microRNA let-7b (miR-let-7b), was examined by real-time polymerase chain reaction (PCR) analysis. Using RA knockdown cells, TLR-7-knockout mice, or antagonist, the specificity of miR-let-7b as a potential ligand for TLR-7 was tested. The mechanism by which ligation of miR-let-7b to TLR-7 promotes disease was investigated in RA myeloid cells by real-time PCR, enzyme-linked immunosorbent assay, and fluorescence-activated cell sorting. We also established the effect of ectopic miR-let-7b expression on arthritic joint inflammation. RESULTS We found that a TLR-7 endogenous ligand resides mainly in RA synovial fluid macrophages. The GU-rich domain in miR-let-7b was found to be essential for TLR-7 ligation, since miR-147, the positive control for GU, was able to stimulate TLR-7+ myeloid cells, whereas miR-124, the negative, non-GU, control, was not. We demonstrated that miR-let-7b or exosomes containing miR-let-7b could transform the RA and/or mouse naive or antiinflammatory macrophages into inflammatory M1 macrophages via TLR-7 ligation. Consistently, we showed that miR-let-7b provokes arthritis by remodeling naive myeloid cells into M1 macrophages via TLR-7 ligation, since joint swelling and M1 macrophages are absent in TLR-7-deficient mice. CONCLUSION The results of this study underscore the importance of miR-let-7b ligation to TLR-7 in the joint during the effector phase of RA.
Collapse
Affiliation(s)
- Seung-Jae Kim
- Jesse Brown VA Medical Center and University of Illinois at Chicago
| | - Zhenlong Chen
- Jesse Brown VA Medical Center and University of Illinois at Chicago
| | - Abdul B Essani
- Jesse Brown VA Medical Center and University of Illinois at Chicago
| | | | - Michael V Volin
- Midwestern University, Chicago College of Osteopathic Medicine, Downers Grove, Illinois
| | | | - William Swedler
- Jesse Brown VA Medical Center and University of Illinois at Chicago
| | - Shiva Arami
- Jesse Brown VA Medical Center and University of Illinois at Chicago
| | | | - Shiva Shahrara
- Jesse Brown VA Medical Center and University of Illinois at Chicago
| |
Collapse
|
41
|
Kim SJ, Chen Z, Essani AB, Elshabrawy HA, Volin MV, Fantuzzi G, McInnes IB, Baker JF, Finn P, Kondos G, Volkov S, Swedler W, Arami S, Sweiss N, Shahrara S. Differential impact of obesity on the pathogenesis of RA or preclinical models is contingent on the disease status. Ann Rheum Dis 2017; 76:731-739. [PMID: 27797749 PMCID: PMC10026536 DOI: 10.1136/annrheumdis-2016-209206] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 07/15/2016] [Accepted: 09/04/2016] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Studies were performed to uncover the significance of obesity in rheumatoid arthritis (RA) and preclinical models. METHODS Preclinical arthritis models were used to examine the impact of obesity on disease onset and remission. Conditioned media from RA adipose tissues were used to investigate the mechanism contributing to joint neutrophil influx and M1 macrophage differentiation observed in early and remission phases of arthritis. RESULTS We report that mice fed with high fat diet (HFD) have an earlier onset of collagen-induced arthritis (CIA) compared with mice on regular diet. However, the differences in CIA joint swelling between the two diet groups are lost once disease is established. We found that early arthritis triggered by obesity is due to elevated joint MIP2/interleukin-8 levels detected in CIA as well as in the RA and mouse adipose tissues and the effect of this chemokine on neutrophil recruitment. Although active disease progression is similarly affected in both diet groups, arthritis resolution is accelerated in lean mice while joint inflammation is sustained in obese mice. We document that HFD can prolong toll-like receptor (TLR)4-induced arthritis by increasing joint monocyte migration and further remodelling the recruited cells into M1 macrophages. Consistently, we show that adipose condition media can transform RA and wild-type naïve myeloid cells into M1 macrophages; however, this function is impaired by TLR4 blockade or deficiency. CONCLUSIONS We conclude that despite established disease being unaffected by obesity, the early and the resolution phases of RA are impacted by obesity through different mechanisms.
Collapse
Affiliation(s)
- Seung-Jae Kim
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, Illinois, USA
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Zhenlong Chen
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, Illinois, USA
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Abdul B Essani
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, Illinois, USA
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Hatem A Elshabrawy
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, Illinois, USA
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Michael V Volin
- Department of Microbiology & Immunology, Midwestern University, Downers Grove, Illinois, USA
| | - Giamila Fantuzzi
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Iain B McInnes
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Joshua F Baker
- Philadelphia VA Medical Center, Philadelphia, Pennsylvania, USA
- Division of Rheumatology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Patricia Finn
- Division of Pulmonary, Division of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - George Kondos
- Division of Cardiology, Division of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Suncica Volkov
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - William Swedler
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, Illinois, USA
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Shiva Arami
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, Illinois, USA
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Nadera Sweiss
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, Illinois, USA
| | - Shiva Shahrara
- Division of Rheumatology, Jesse Brown VA Medical Center, Chicago, Illinois, USA
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
42
|
Elshabrawy HA, Essani AE, Szekanecz Z, Fox DA, Shahrara S. TLRs, future potential therapeutic targets for RA. Autoimmun Rev 2016; 16:103-113. [PMID: 27988432 DOI: 10.1016/j.autrev.2016.12.003] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 09/07/2016] [Indexed: 01/27/2023]
Abstract
Toll like receptors (TLR)s have a central role in regulating innate immunity and in the last decade studies have begun to reveal their significance in potentiating autoimmune diseases such as rheumatoid arthritis (RA). Earlier investigations have highlighted the importance of TLR2 and TLR4 function in RA pathogenesis. In this review, we discuss the newer data that indicate roles for TLR5 and TLR7 in RA and its preclinical models. We evaluate the pathogenicity of TLRs in RA myeloid cells, synovial tissue fibroblasts, T cells, osteoclast progenitor cells and endothelial cells. These observations establish that ligation of TLRs can transform RA myeloid cells into M1 macrophages and that the inflammatory factors secreted from M1 and RA synovial tissue fibroblasts participate in TH-17 cell development. From the investigations conducted in RA preclinical models, we conclude that TLR-mediated inflammation can result in osteoclastic bone erosion by interconnecting the myeloid and TH-17 cell response to joint vascularization. In light of emerging unique aspects of TLR function, we summarize the novel approaches that are being tested to impair TLR activation in RA patients.
Collapse
Affiliation(s)
- Hatem A Elshabrawy
- Division of Rheumatology, Jesse Brown VA, Medical Center, Chicago, IL 60612, USA; Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612, USA
| | - Abdul E Essani
- Division of Rheumatology, Jesse Brown VA, Medical Center, Chicago, IL 60612, USA; Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612, USA
| | - Zoltán Szekanecz
- Department of Rheumatology, Institute of Medicine, University of Debrecen Faculty of Medicine, Nagyerdei Str 98, Debrecen H-4004, Hungary
| | - David A Fox
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shiva Shahrara
- Division of Rheumatology, Jesse Brown VA, Medical Center, Chicago, IL 60612, USA; Department of Medicine, Division of Rheumatology, University of Illinois at Chicago, IL 60612, USA.
| |
Collapse
|
43
|
Hernandez CJ, Guss JD, Luna M, Goldring SR. Links Between the Microbiome and Bone. J Bone Miner Res 2016; 31:1638-46. [PMID: 27317164 PMCID: PMC5434873 DOI: 10.1002/jbmr.2887] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/14/2016] [Accepted: 06/15/2016] [Indexed: 12/28/2022]
Abstract
The human microbiome has been shown to influence a number of chronic conditions associated with impaired bone mass and bone quality, including obesity, diabetes, and inflammatory bowel disease. The connection between the microbiome and bone health, however, has not been well studied. The few studies available demonstrate that the microbiome can have a large effect on bone remodeling and bone mass. The gut microbiome is the largest reservoir of microbial organisms in the body and consists of more than a thousand different species interacting with one another in a stable, dynamic equilibrium. How the microbiome can affect organs distant from the gut is not well understood but is believed to occur through regulation of nutrition, regulation of the immune system, and/or translocation of bacterial products across the gut endothelial barrier. Here we review each of these mechanisms and discuss their potential effect on bone remodeling and bone mass. We discuss how preclinical studies of bone-microbiome interactions are challenging because the microbiome is sensitive to genetic background, housing environment, and vendor source. Additionally, although the microbiome exhibits a robust response to external stimuli, it rapidly returns to its original steady state after a disturbance, making it difficult to sustain controlled changes in the microbiome over time periods required to detect alterations in bone remodeling, mass, or structure. Despite these challenges, an understanding of the mechanisms by which the gut microbiome affects bone has the potential to provide insights into the dissociation between fracture risk and bone mineral density in patients including those with obesity, diabetes, or inflammatory bowel disease. In addition, alteration of the gut microbiome has the potential to serve as a biomarker of bone metabolic activity as well as a target for therapies to improve bone structure and quality using pharmaceutical agents or pre- or probiotics. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Christopher J Hernandez
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA.,Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.,Hospital for Special Surgery, New York, NY, USA
| | - Jason D Guss
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Marysol Luna
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY, USA
| | | |
Collapse
|
44
|
Toll-like receptors and chronic inflammation in rheumatic diseases: new developments. Nat Rev Rheumatol 2016; 12:344-57. [PMID: 27170508 DOI: 10.1038/nrrheum.2016.61] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In the past few years, new developments have been reported on the role of Toll-like receptors (TLRs) in chronic inflammation in rheumatic diseases. The inhibitory function of TLR10 has been demonstrated. Receptors that enhance the function of TLRs, and several TLR inhibitors, have been identified. In addition, the role of the microbiome and TLRs in the onset of rheumatic diseases has been reported. We review novel insights on the role of TLRs in several inflammatory joint diseases, including rheumatoid arthritis, systemic lupus erythematosus, gout and Lyme arthritis, with a focus on the signalling mechanisms mediated by the Toll-IL-1 receptor (TIR) domain, the exogenous and endogenous ligands of TLRs, and the current and future therapeutic strategies to target TLR signalling in rheumatic diseases.
Collapse
|
45
|
Yan L, Liang J, Yao C, Wu P, Zeng X, Cheng K, Yin H. Pyrimidine Triazole Thioether Derivatives as Toll-Like Receptor 5 (TLR5)/Flagellin Complex Inhibitors. ChemMedChem 2015; 11:822-6. [DOI: 10.1002/cmdc.201500471] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Lei Yan
- Center of Basic Molecular Science, Department of Chemistry; Tsinghua University; Beijing 100082 China
| | - Jiaqi Liang
- Center of Basic Molecular Science, Department of Chemistry; Tsinghua University; Beijing 100082 China
| | - Chengbo Yao
- Center of Basic Molecular Science, Department of Chemistry; Tsinghua University; Beijing 100082 China
| | - Peiyao Wu
- Center of Basic Molecular Science, Department of Chemistry; Tsinghua University; Beijing 100082 China
| | - Xianfeng Zeng
- Center of Basic Molecular Science, Department of Chemistry; Tsinghua University; Beijing 100082 China
| | - Kui Cheng
- Center of Basic Molecular Science, Department of Chemistry; Tsinghua University; Beijing 100082 China
| | - Hang Yin
- Center of Basic Molecular Science, Department of Chemistry; Tsinghua University; Beijing 100082 China
| |
Collapse
|
46
|
Zhou Y, Deng HW, Shen H. Circulating monocytes: an appropriate model for bone-related study. Osteoporos Int 2015; 26:2561-72. [PMID: 26194495 DOI: 10.1007/s00198-015-3250-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 07/10/2015] [Indexed: 10/23/2022]
Abstract
Peripheral blood monocytes (PBMs) are an important source of precursors of osteoclasts, the bone-resorbing cells and the cytokines produced by PBMs that have profound effects on osteoclast differentiation, activation, and apoptosis. So PBMs represent a highly valuable and unique working cell model for bone-related study. Finding an appropriate working cell model for clinical and (epi-)genomic studies of human skeletal disorders is a challenge. Peripheral blood monocytes (PBMs) can give rise to osteoclasts, the bone-resorbing cells. Particularly, PBMs provide the sole source of osteoclast precursors for adult peripheral skeleton where the bone marrow is normally hematopoietically inactive. PBMs can secrete potent pro- and anti-inflammatory cytokines, which are important for osteoclast differentiation, activation, and apoptosis. Reduced production of PBM cytokines represents a major mechanism for the inhibitory effects of sex hormones on osteoclastogenesis and bone resorption. Abnormalities in PBMs have been linked to various skeletal disorders/traits, strongly supporting for the biological relevance of PBMs with bone metabolism and disorders. Here, we briefly review the origin and further differentiation of PBMs. In particular, we discuss the close relationship between PBMs and osteoclasts, and highlight the utility of PBMs in study the pathophysiological mechanisms underlying various skeletal disorders.
Collapse
Affiliation(s)
- Y Zhou
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University, New Orleans, LA, 70112, USA
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA, 70118, USA
| | - H-W Deng
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University, New Orleans, LA, 70112, USA
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA, 70118, USA
| | - H Shen
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, Tulane University, New Orleans, LA, 70112, USA.
- Cell and Molecular Biology Department, Tulane University, New Orleans, LA, 70118, USA.
- Center for Bioinformatics and Genomics, Department of Biostatistics and Bioinformatics, School of Public Health and Tropical Medicine, Tulane University, 1440 Canal St., Suite 2001, New Orleans, LA, 70112, USA.
| |
Collapse
|
47
|
Guillou C, Derambure C, Fréret M, Verdet M, Avenel G, Golinski ML, Sabourin JC, Loarer FL, Adriouch S, Boyer O, Lequerré T, Vittecoq O. Prophylactic Injection of Recombinant Alpha-Enolase Reduces Arthritis Severity in the Collagen-Induced Arthritis Mice Model. PLoS One 2015; 10:e0136359. [PMID: 26302382 PMCID: PMC4547710 DOI: 10.1371/journal.pone.0136359] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 07/31/2015] [Indexed: 01/13/2023] Open
Abstract
Objective To evaluate the ability of the glycolytic enzyme alpha-enolase (ENO1) or its immunodominant peptide (pEP1) to reduce the severity of CIA in DBA/1 mice when injected in a prophylactic way. Methods Mice were treated with mouse ENO1 or pEP1 one day prior to collagen II immunization. Clinical assessment was evaluated using 4 parameters (global and articular scores, ankle thickness and weight). Titers of serum anti-ENO1, anti-cyclic citrullinated peptides (anti-CCP) and anti-CII (total IgG and IgG1/IgG2a isotypes) antibodies were measured by ELISA at different time-points. Disease activity was assessed by histological analysis of both anterior and hind paws at the end of experimentation. Results Prophylactic injection of 100 μg of ENO1 reduced severity of CIA. Serum levels of anti-CII antibodies were reduced in ENO1-treated mice. Concordantly, ENO1-treated mice joints presented less severe histological signs of arthritis. ENO1 did not induce a shift toward a Th2 response since IgG1/IgG2a ratio of anti-CII antibodies remained unchanged and IL-4 serum levels were similar to those measured in the control group. Conclusions Pre-immunization with ENO1 or its immunodominant peptide pEP1 reduces CIA severity at the clinical, immunological and histological levels. Effects of pEP1 were less pronounced. This immunomodulatory effect is associated with a reduction in anti-CII antibodies production but is not due to a Th1/Th2 shift.
Collapse
Affiliation(s)
- Clément Guillou
- INSERM, U905, Rouen, France
- Normandy University, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Céline Derambure
- INSERM, U905, Rouen, France
- Normandy University, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Manuel Fréret
- INSERM, U905, Rouen, France
- Normandy University, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Mathieu Verdet
- Rouen University Hospital, Department of Rheumatology, Rouen, France
| | - Gilles Avenel
- Rouen University Hospital, Department of Rheumatology, Rouen, France
| | - Marie-Laure Golinski
- INSERM, U905, Rouen, France
- Normandy University, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Jean-Christophe Sabourin
- Normandy University, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
- Rouen University Hospital, Department of Pathology, Rouen, France
| | | | - Sahil Adriouch
- INSERM, U905, Rouen, France
- Normandy University, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Olivier Boyer
- INSERM, U905, Rouen, France
- Normandy University, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
- Rouen University Hospital, Department of Immunology, Rouen, France
| | - Thierry Lequerré
- INSERM, U905, Rouen, France
- Normandy University, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
- Rouen University Hospital, Department of Rheumatology, Rouen, France
| | - Olivier Vittecoq
- INSERM, U905, Rouen, France
- Normandy University, Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
- Rouen University Hospital, Department of Rheumatology, Rouen, France
- * E-mail:
| |
Collapse
|
48
|
Rosenthal KS, Mikecz K, Steiner HL, Glant TT, Finnegan A, Carambula RE, Zimmerman DH. Rheumatoid arthritis vaccine therapies: perspectives and lessons from therapeutic ligand epitope antigen presentation system vaccines for models of rheumatoid arthritis. Expert Rev Vaccines 2015; 14:891-908. [PMID: 25787143 DOI: 10.1586/14760584.2015.1026330] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The current status of therapeutic vaccines for autoimmune diseases is reviewed with rheumatoid arthritis as the focus. Therapeutic vaccines for autoimmune diseases must regulate or subdue responses to common self-antigens. Ideally, such a vaccine would initiate an antigen-specific modulation of the T-cell immune response that drives the inflammatory disease. Appropriate animal models and types of T helper cells and signature cytokine responses that drive autoimmune disease are also discussed. Interpretation of these animal models must be done cautiously because the means of initiation, autoantigens, and even the signature cytokine and T helper cell (Th1 or Th17) responses that are involved in the disease may differ significantly from those in humans. We describe ligand epitope antigen presentation system vaccine modulation of T-cell autoimmune responses as a strategy for the design of therapeutic vaccines for rheumatoid arthritis, which may also be effective in other autoimmune conditions.
Collapse
|
49
|
Abstract
Even though activation of immunity is associated with bone destruction, new mechanisms have been described in 2014 through which immunology-associated pathways can cooperate to support osteogenesis. These advances support the view of the immune system as a central mechanism which can regulate bone homeostasis, regeneration and destruction.
Collapse
|