1
|
Yildirim D, Baykul M, Edek YC, Gulengul M, Alp GT, Eroglu FS, Adisen E, Kucuk H, Erden A, Goker B, Nas K, Ozturk MA. Could serum HMGB1 levels be a predictor associated with psoriatic arthritis? Biomark Med 2023; 17:871-880. [PMID: 38117143 DOI: 10.2217/bmm-2023-0490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023] Open
Abstract
Background/aim: Psoriasis is a chronic autoimmune disease that predominantly affects the skin and musculoskeletal system. We hypothesized that HMGB1, an inflammatory nuclear protein, may play a role in the musculoskeletal involvement of psoriasis. Methods: Forty patients with psoriasis and 45 with psoriatic arthritis were involved in the study; the results were compared with 22 healthy controls. Serum HMGB1 levels were evaluated from peripheral blood samples. Results: Serum HMGB1 levels were found to be significantly higher in patients with psoriasis regardless of joint involvement (p < 0.001). Also, HMGB1 levels were correlated with the extent of psoriasis. Conclusion: Serum HMGB1 levels may contribute to the progression of psoriasis to psoriatic arthritis and correlate with the severity of skin involvement.
Collapse
Affiliation(s)
- Derya Yildirim
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Gazi University, 06460, Ankara, Turkey
| | - Merve Baykul
- Division of Rheumatology, Department of Physical Medicine & Rehabilitation, Faculty of Medicine, Sakarya University, 54100, Sakarya, Turkey
| | - Yusuf C Edek
- Department of Dermatology, Faculty of Medicine, Gazi University, 06460, Ankara, Turkey
| | - Mehmet Gulengul
- Department of Dermatology, Faculty of Medicine, Gazi University, 06460, Ankara, Turkey
| | - Gizem T Alp
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Gazi University, 06460, Ankara, Turkey
| | - Fatma S Eroglu
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Gazi University, 06460, Ankara, Turkey
| | - Esra Adisen
- Department of Dermatology, Faculty of Medicine, Gazi University, 06460, Ankara, Turkey
| | - Hamit Kucuk
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Gazi University, 06460, Ankara, Turkey
| | - Abdulsamet Erden
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Gazi University, 06460, Ankara, Turkey
| | - Berna Goker
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Gazi University, 06460, Ankara, Turkey
| | - Kemal Nas
- Division of Rheumatology, Department of Physical Medicine & Rehabilitation, Faculty of Medicine, Sakarya University, 54100, Sakarya, Turkey
| | - Mehmet A Ozturk
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine, Gazi University, 06460, Ankara, Turkey
| |
Collapse
|
2
|
Ma W, Zhu J, Bai L, Zhao P, Li F, Zhang S. The role of neutrophil extracellular traps and proinflammatory damage-associated molecular patterns in idiopathic inflammatory myopathies. Clin Exp Immunol 2023; 213:202-208. [PMID: 37289984 PMCID: PMC10361739 DOI: 10.1093/cei/uxad059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 04/13/2023] [Accepted: 05/25/2023] [Indexed: 06/10/2023] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are a group of systemic autoimmune diseases characterized by immune-mediated muscle injury. Abnormal neutrophil extracellular traps (NETs) can be used as a biomarker of IIM disease activity, but the mechanism of NET involvement in IIMs needs to be elucidated. Important components of NETs, including high-mobility group box 1, DNA, histones, extracellular matrix, serum amyloid A, and S100A8/A9, act as damage-associated molecular patterns (DAMPs) to promote inflammation in IIMs. NETs can act on different cells to release large amounts of cytokines and activate the inflammasome, which can subsequently aggravate the inflammatory response. Based on the idea that NETs may be proinflammatory DAMPs of IIMs, we describe the role of NETs, DAMPs, and their interaction in the pathogenesis of IIMs and discuss the possible targeted treatment strategies in IIMs.
Collapse
Affiliation(s)
- Wenlan Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jiarui Zhu
- Department of Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, China
| | - Ling Bai
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Peipei Zhao
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Feifei Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Sigong Zhang
- Department of Rheumatology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
3
|
Kamiya M, Kimura N, Umezawa N, Hasegawa H, Yasuda S. Muscle fiber necroptosis in pathophysiology of idiopathic inflammatory myopathies and its potential as target of novel treatment strategy. Front Immunol 2023; 14:1191815. [PMID: 37483632 PMCID: PMC10361824 DOI: 10.3389/fimmu.2023.1191815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/22/2023] [Indexed: 07/25/2023] Open
Abstract
Idiopathic inflammatory myopathies (IIMs), which are a group of chronic and diverse inflammatory diseases, are primarily characterized by weakness in the proximal muscles that progressively leads to persistent disability. Current treatments of IIMs depend on nonspecific immunosuppressive agents (including glucocorticoids and immunosuppressants). However, these therapies sometimes fail to regulate muscle inflammation, and some patients suffer from infectious diseases and other adverse effects related to the treatment. Furthermore, even after inflammation has subsided, muscle weakness persists in a significant proportion of the patients. Therefore, the elucidation of pathophysiology of IIMs and development of a better therapeutic strategy that not only alleviates muscle inflammation but also improves muscle weakness without increment of opportunistic infection is awaited. Muscle fiber death, which has been formerly postulated as "necrosis", is a key histological feature of all subtypes of IIMs, however, its detailed mechanisms and contribution to the pathophysiology remained to be elucidated. Recent studies have revealed that muscle fibers of IIMs undergo necroptosis, a newly recognized form of regulated cell death, and promote muscle inflammation and dysfunction through releasing inflammatory mediators such as damage-associated molecular patterns (DAMPs). The research on murine model of polymyositis, a subtype of IIM, revealed that the inhibition of necroptosis or HMGB1, one of major DAMPs released from muscle fibers undergoing necroptosis, ameliorated muscle inflammation and recovered muscle weakness. Furthermore, not only the necroptosis-associated molecules but also PGAM5, a mitochondrial protein, and reactive oxygen species have been shown to be involved in muscle fiber necroptosis, indicating the multiple target candidates for the treatment of IIMs acting through necroptosis regulation. This article overviews the research on muscle injury mechanisms in IIMs focusing on the contribution of necroptosis in their pathophysiology and discusses the potential treatment strategy targeting muscle fiber necroptosis.
Collapse
|
4
|
Danieli MG, Antonelli E, Piga MA, Claudi I, Palmeri D, Tonacci A, Allegra A, Gangemi S. Alarmins in autoimmune diseases. Autoimmun Rev 2022; 21:103142. [PMID: 35853572 DOI: 10.1016/j.autrev.2022.103142] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/10/2022] [Indexed: 12/18/2022]
Abstract
Alarmins are endogenous, constitutively expressed, chemotacting and immune activating proteins or peptides released because of non-programmed cell death (i.e. infections, trauma, etc). They are considered endogenous damage-associated molecular patterns (DAMPs), able to induce a sterile inflammation. In the last years, several studies highlighted a possible role of different alarmins in the pathogenesis of various autoimmune and immune-mediated diseases. We reviewed the relevant literature about this topic, for about 160 articles. Particularly, we focused on systemic autoimmune diseases (systemic lupus erythematosus, rheumatoid arthritis, idiopathic inflammatory myopathies, ANCA-associated vasculitides, Behçet's disease) and cutaneous organ-specific autoimmune diseases (vitiligo, psoriasis, alopecia, pemphigo). Finally, we discussed about future perspectives and potential therapeutic implications of alarmins in autoimmune diseases. In fact, identification of receptors and downstream signal transducers of alarmins may lead to the identification of antagonistic inhibitors and agonists, with the capacity to modulate alarmins-related pathways and potential therapeutic applicability.
Collapse
Affiliation(s)
- Maria Giovanna Danieli
- Clinica Medica, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, via Tronto 10/A, 60126 Torrette di Ancona, Italy; Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy.
| | - Eleonora Antonelli
- PostGraduate School of Internal Medicine, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy.
| | - Mario Andrea Piga
- Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy.
| | - Ilaria Claudi
- Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy.
| | - Davide Palmeri
- Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy.
| | - Alessandro Tonacci
- Institute of Clinical Physiology, National Research Council of Italy (IFC-CNR), Via G. Moruzzi 1, 56124 Pisa, Italy.
| | - Alessandro Allegra
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy.
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy.
| |
Collapse
|
5
|
Dong Y, Ming B, Dong L. The Role of HMGB1 in Rheumatic Diseases. Front Immunol 2022; 13:815257. [PMID: 35250993 PMCID: PMC8892237 DOI: 10.3389/fimmu.2022.815257] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/31/2022] [Indexed: 12/19/2022] Open
Abstract
HMGB1, a highly conserved non-histone nuclear protein, is widely expressed in mammalian cells. HMGB1 in the nucleus binds to the deoxyribonucleic acid (DNA) to regulate the structure of chromosomes and maintain the transcription, replication, DNA repair, and nucleosome assembly. HMGB1 is actively or passively released into the extracellular region during cells activation or necrosis. Extracellular HMGB1 as an alarmin can initiate immune response alone or combined with other substances such as nucleic acid to participate in multiple biological processes. It has been reported that HMGB1 is involved in various inflammatory responses and autoimmunity. This review article summarizes the physiological function of HMGB1, the post-translational modification of HMGB1, its interaction with different receptors, and its recent advances in rheumatic diseases and strategies for targeted therapy.
Collapse
Affiliation(s)
- Yuanji Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingxia Ming
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Glucocorticoids in Myositis: Initiation, Tapering, and Discontinuation. Curr Rheumatol Rep 2022; 24:47-53. [PMID: 35275363 DOI: 10.1007/s11926-022-01060-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF THE REVIEW We discuss the evidence behind the use of glucocorticoids in myositis including a discussion of mechanism of action, dosing, tapering, and duration of therapy as well as glucocorticoid-related adverse events that are particularly important in myositis patients. RECENT FINDINGS Studies showing reduction in mortality rates after the use of glucocorticoids, better outcomes in patients treated with glucocorticoids compared to those who did not, and reduction of inflammation in muscle biopsies provide low level evidence to support use of glucocorticoids in myositis. Early initiation of therapy is associated with better functional outcomes. Use of intravenous methylprednisolone in patients with severe disease may lead to quicker recovery and reduction in long-term glucocorticoid exposure. Steroid-related myopathy and osteoporosis are glucocorticoid side effects that are particularly relevant in myositis. The optimal dose and duration of glucocorticoid therapy in myositis currently remain elusive, and this review emphasizes the need for better quality studies in this area.
Collapse
|
7
|
Peng Q, Zhang Y, Liu Y, Liang L, Li W, Tian X, Zhang L, Yang H, Lu X, Wang G. Necroptosis contributes to myofiber death in idiopathic inflammatory myopathies. Arthritis Rheumatol 2022; 74:1048-1058. [PMID: 35077006 DOI: 10.1002/art.42071] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/22/2021] [Accepted: 01/18/2022] [Indexed: 11/05/2022]
Affiliation(s)
- Qing‐Lin Peng
- Department of Rheumatology, Key Lab of Myositis, China‐Japan Friendship Hospital Beijing 100029 China
| | - Ya‐Mei Zhang
- Department of Rheumatology, Key Lab of Myositis, China‐Japan Friendship Hospital Beijing 100029 China
| | - Yan‐Chun Liu
- NMPA Key Laboratory for Quality Evaluation of In Vitro Diagnostics, Beijing Institute of Medical Device Testing Beijing 101111 China
| | - Lin Liang
- Department of Rheumatology, Key Lab of Myositis, China‐Japan Friendship Hospital Beijing 100029 China
| | - Wen‐Li Li
- Department of Rheumatology, Key Lab of Myositis, China‐Japan Friendship Hospital Beijing 100029 China
| | - Xiao‐Lan Tian
- Department of Rheumatology, Key Lab of Myositis, China‐Japan Friendship Hospital Beijing 100029 China
| | - Lu Zhang
- Department of Rheumatology, Key Lab of Myositis, China‐Japan Friendship Hospital Beijing 100029 China
| | - Hong‐Xia Yang
- Department of Rheumatology, Key Lab of Myositis, China‐Japan Friendship Hospital Beijing 100029 China
| | - Xin Lu
- Department of Rheumatology, Key Lab of Myositis, China‐Japan Friendship Hospital Beijing 100029 China
| | - Guo‐Chun Wang
- Department of Rheumatology, Key Lab of Myositis, China‐Japan Friendship Hospital Beijing 100029 China
| |
Collapse
|
8
|
Targeting necroptosis in muscle fibers ameliorates inflammatory myopathies. Nat Commun 2022; 13:166. [PMID: 35013338 PMCID: PMC8748624 DOI: 10.1038/s41467-021-27875-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Abstract
Muscle cell death in polymyositis is induced by CD8+ cytotoxic T lymphocytes. We hypothesized that the injured muscle fibers release pro-inflammatory molecules, which would further accelerate CD8+ cytotoxic T lymphocytes-induced muscle injury, and inhibition of the cell death of muscle fibers could be a novel therapeutic strategy to suppress both muscle injury and inflammation in polymyositis. Here, we show that the pattern of cell death of muscle fibers in polymyositis is FAS ligand-dependent necroptosis, while that of satellite cells and myoblasts is perforin 1/granzyme B-dependent apoptosis, using human muscle biopsy specimens of polymyositis patients and models of polymyositis in vitro and in vivo. Inhibition of necroptosis suppresses not only CD8+ cytotoxic T lymphocytes-induced cell death of myotubes but also the release of inflammatory molecules including HMGB1. Treatment with a necroptosis inhibitor or anti-HMGB1 antibodies ameliorates myositis-induced muscle weakness as well as muscle cell death and inflammation in the muscles. Thus, targeting necroptosis in muscle cells is a promising strategy for treating polymyositis providing an alternative to current therapies directed at leukocytes.
Collapse
|
9
|
Wu SE, Chiu YL, Kao TW, Chen WL. Elevated level of the soluble receptor for advanced glycation end-products involved in sarcopenia: an observational study. BMC Geriatr 2021; 21:531. [PMID: 34620111 PMCID: PMC8495916 DOI: 10.1186/s12877-021-02487-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 09/17/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The soluble receptor for advanced glycation end products (sRAGE) has been proposed to serve as a marker for disease severity, but its role in sarcopenia, an age-related progressive loss of muscle mass and function, remains elusive. This study examines the association between sRAGE and sarcopenia. METHODS A total of 314 community-dwelling elderly adults who had their health examination at Tri-Service General Hospital from 2017 to 2019 underwent protein analysis with enzyme-linked immunosorbent assay. The relationship with sarcopenia and its detailed information, including components and diagnosis status, were examined using linear and logistic regressions. RESULTS As for sarcopenia components, low muscle mass (β = 162.8, p = 0.012) and strength (β = 181.31, p = 0.011) were significantly correlated with sRAGE, but not low gait speed (p = 0.066). With regard to disease status, confirmed sarcopenia (β = 436.93, p < 0.001), but not probable (p = 0.448) or severe sarcopenia (p = 0.488), was significantly correlated with sRAGE. In addition, females revealed a stronger association with sRAGE level by showing significant correlations with low muscle mass (β = 221.72, p = 0.014) and low muscle strength (β = 208.68, p = 0.043). CONCLUSIONS sRAGE level showed a positive association with sarcopenia, illustrating its involvement in the evolution of sarcopenia. This association is more evident in female groups, which may be attributed to the loss of protection from estrogen in postmenopausal women. Utilizing sRAGE level as a prospective marker for sarcopenia deserves further investigation in future studies.
Collapse
Affiliation(s)
- Shou-En Wu
- Department of General Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Department of Family and Community Medicine, Division of Geriatric Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Number 325, Section 2, Chang-gong Rd, Nei-Hu District, 114, Taipei, Taiwan, Republic of China
| | - Yi-Lin Chiu
- Department of Biochemistry , National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Tung-Wei Kao
- Department of General Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China.,Department of Family and Community Medicine, Division of Geriatric Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Number 325, Section 2, Chang-gong Rd, Nei-Hu District, 114, Taipei, Taiwan, Republic of China
| | - Wei-Liang Chen
- Department of General Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China. .,Department of Family and Community Medicine, Division of Geriatric Medicine, Tri-Service General Hospital, and School of Medicine, National Defense Medical Center, Number 325, Section 2, Chang-gong Rd, Nei-Hu District, 114, Taipei, Taiwan, Republic of China. .,Department of Biochemistry , National Defense Medical Center, Taipei, Taiwan, Republic of China.
| |
Collapse
|
10
|
Güler DD, Keleşoğlu Dinçer AB, Karahan ZC, Güler HS, Yayla ME, Sezer S, Aydemir Gülöksüz EG, Okatan İE, Torgutalp M, Eroğlu DŞ, Yüksel ML, Turgay TM, Kınıklı G, Ateş A. High mobility group box-1 levels may be associated with disease activity of Behcet's disease. Turk J Med Sci 2021; 51:2690-2697. [PMID: 34333900 PMCID: PMC8742499 DOI: 10.3906/sag-2101-116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 07/31/2021] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND High mobility group box- 1 (HMGB- 1) is a nuclear protein acting as a proinflammatory molecule. The serum HMGB- 1 levels were found elevated in chronic inflammatory diseases. In this cross-sectional study, serum HMGB- 1 levels in Behcet's disease (BD) patients and healthy controls (HC) were studied. Also, its association with disease activity scores and clinical findings were evaluated. METHODS Ninety BD patients and 50 age-sex matched HC were included in the study. Disease activity scores were assessed by Behcet Disease Current Activity Form (BDCAF) and Behcet Syndrome Activity Score (BSAS). Serum HMGB- 1 levels were measured using a commercial ELISA kit. A p value of < 0.05 was considered to be statistically significant. RESULTS Serum HMGB- 1 levels were significantly higher in BD than in HC (43.26 pg/mL and 16.73 pg/mL; p < 0.001, respectively). Serum HMGB- 1 levels were statistically significantly associated with presence of erythema nodosum (EN) and genital ulcers in the last one month prior to recruitment (p = 0.041 and p < 0.001, respectively). BDCAF and BSAS scores were positively correlated with serum HMGB- 1 level ( p = 0.03 and p = 0.02, respectively). DISCUSSION HMGB - 1 may play a role in the development of BD. Also, due to its positive correlation with disease activity indices, it can be used as a novel disease activity parameter in BD.
Collapse
Affiliation(s)
- Dilara Dönmez Güler
- Department of Internal Medical Sciences, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Ayşe Bahar Keleşoğlu Dinçer
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,Ankara University, Ankara, Turkey
| | - Zeynep Ceren Karahan
- Department of Medical Microbiology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Hasan Selim Güler
- Department of Internal Medical Sciences, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Müçteba Enes Yayla
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,Ankara University, Ankara, Turkey
| | - Serdar Sezer
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,Ankara University, Ankara, Turkey
| | - Emine Gözde Aydemir Gülöksüz
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,Ankara University, Ankara, Turkey
| | - İlyas Ercan Okatan
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,Ankara University, Ankara, Turkey
| | - Murat Torgutalp
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,Ankara University, Ankara, Turkey
| | - Didem Şahin Eroğlu
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,Ankara University, Ankara, Turkey
| | - Mehmet Levent Yüksel
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,Ankara University, Ankara, Turkey
| | - Tahsin Murat Turgay
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,Ankara University, Ankara, Turkey
| | - Gülay Kınıklı
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,Ankara University, Ankara, Turkey
| | - Aşkın Ateş
- Division of Rheumatology, Department of Internal Medicine, Faculty of Medicine,Ankara University, Ankara, Turkey
| |
Collapse
|
11
|
High mobility group box 1 is involved in the pathogenesis of passive transfer myasthenia gravis model. Neuroreport 2021; 32:803-807. [PMID: 33994526 DOI: 10.1097/wnr.0000000000001665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Myasthenia gravis (MG) is an autoimmune disease with autoantibodies against the mainly nicotinic acetylcholine receptor (AChR). High mobility group box1 (HMGB1) acts as a danger signal and drives the pathogenesis of autoimmune-mediated diseases. However, the role of HMGB1 in the pathogenesis of MG is not fully understood. Therefore, in this study, we analyzed serum levels of HMGB1 and immunohistochemical HMGB1 staining of muscle tissues in the passive transfer MG model to investigate the role of HMGB1 in MG. As a result, serum HMGB1 levels tended to be higher and the quantitative score of muscle pathology showed greater HMGB1 deposition (P = 0.02) along with sparser AChR staining and more severe inflammation in the passive transfer MG rats (n = 6) than those in control rats (n = 6). These findings indicate that HMGB1 is an important mediator and biomarker for inflammation in the pathogenesis of MG and can be a therapeutic target in MG.
Collapse
|
12
|
Cytokines and inflammatory mediators as promising markers of polymyositis/dermatomyositis. Curr Opin Rheumatol 2021; 32:534-541. [PMID: 32941247 DOI: 10.1097/bor.0000000000000744] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Idiopathic inflammatory myopathies (IIMs), known also as myositis, represent challenging group of heterogeneous muscle disorders characterized by symmetric proximal muscle weakness and evidence of muscle inflammation. The purpose of this review is to provide important updates on cytokines and inflammatory mediators related to myositis. RECENT FINDINGS In the past 5 years, multiple studies brought a fresh insight into the pathogenesis of myositis by introducing new factors or further characterizing the role of the well established mediators in myositis. Among the mediators reviewed in this article, special attention was paid to interferons, C-X-C motif chemokine ligand 10, interleukin-18 and the IL23/Th17 axis. Some of the recent work has also focused on the nontraditional cytokines, such as adipokines, myokines, S100 proteins, High Mobility Group Box 1 or B-cell activating factor and on several anti-inflammatory mediators. Moreover, microRNAs and their potential to reflect the disease activity or to regulate the inflammatory processes in myositis have recently been subject of intensive investigation. Some of the above-mentioned mediators have been proposed as promising clinical biomarkers or therapeutic targets for myositis. SUMMARY Several recent studies contributed to a better understanding of the pathogenesis of myositis and highlighted the clinical significance of certain inflammatory mediators. Application of these new findings may help to develop innovative approaches for patients' phenotyping, disease activity monitoring and potentially novel therapies.
Collapse
|
13
|
Coskun Benlidayi I, Gupta L. The pathophysiological effects of exercise in the management of idiopathic inflammatory myopathies: A scoping review. Int J Rheum Dis 2021; 24:896-903. [PMID: 33793075 DOI: 10.1111/1756-185x.14104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 11/30/2022]
Abstract
Idiopathic inflammatory myopathy (IIM) is a term used for a heterogeneous group of diseases characterized by severe muscle weakness. In addition to pharmacological treatment options, non-pharmacological methods such as exercising are essential for proper management of myositis. The present article aimed to provide an insight into the potential pathophysiological mechanisms underlying exercise-related benefits in myositis. A systematic search was performed on PubMed/MEDLINE, Scopus, Web of Science, and Google Scholar using the following keywords and their combinations: "idiopathic inflammatory myopathy", "inflammatory myopathy", "myositis", "polymyositis", "dermatomyositis", "inclusion body myositis", and "exercise". Current literature indicates that exercising has impact on both immune and non-immune pathways in patients with IIM. Exercise-related benefits include (a) increased mitochondrial biogenesis/enzyme activity, (b) reconditioning of immune/inflammatory pathways, (c) decreased endoplasmic reticulum stress, (d) modulation of gene expression, (e) increased protein synthesis and cytoskeletal remodeling, and (f) decreased muscle fibrosis and non-muscle area infiltrates. With its certain benefits, exercise stands as a precious non-pharmacological treatment option for patients with IIM.
Collapse
Affiliation(s)
- Ilke Coskun Benlidayi
- Department of Physical Medicine and Rehabilitation, Cukurova University Faculty of Medicine, Adana, Turkey
| | - Latika Gupta
- Department of Clinical Immunology and Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
14
|
Innate immune response in systemic autoimmune diseases: a potential target of therapy. Inflammopharmacology 2020; 28:1421-1438. [DOI: 10.1007/s10787-020-00762-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 09/18/2020] [Indexed: 02/06/2023]
|
15
|
Vijayakumar EC, Bhatt LK, Prabhavalkar KS. High Mobility Group Box-1 (HMGB1): A Potential Target in Therapeutics. Curr Drug Targets 2020; 20:1474-1485. [PMID: 31215389 DOI: 10.2174/1389450120666190618125100] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 02/06/2023]
Abstract
High mobility group box-1 (HMGB1) mainly belongs to the non-histone DNA-binding protein. It has been studied as a nuclear protein that is present in eukaryotic cells. From the HMG family, HMGB1 protein has been focused particularly for its pivotal role in several pathologies. HMGB-1 is considered as an essential facilitator in diseases such as sepsis, collagen disease, atherosclerosis, cancers, arthritis, acute lung injury, epilepsy, myocardial infarction, and local and systemic inflammation. Modulation of HMGB1 levels in the human body provides a way in the management of these diseases. Various strategies, such as HMGB1-receptor antagonists, inhibitors of its signalling pathway, antibodies, RNA inhibitors, vagus nerve stimulation etc. have been used to inhibit expression, release or activity of HMGB1. This review encompasses the role of HMGB1 in various pathologies and discusses its therapeutic potential in these pathologies.
Collapse
Affiliation(s)
- Eyaldeva C Vijayakumar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Kedar S Prabhavalkar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| |
Collapse
|
16
|
De Paepe B. Progressive Skeletal Muscle Atrophy in Muscular Dystrophies: A Role for Toll-like Receptor-Signaling in Disease Pathogenesis. Int J Mol Sci 2020; 21:ijms21124440. [PMID: 32580419 PMCID: PMC7352931 DOI: 10.3390/ijms21124440] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/18/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022] Open
Abstract
Muscle atrophy is an active process controlled by specific transcriptional programs, in which muscle mass is lost by increased protein degradation and/or decreased protein synthesis. This review explores the involvement of Toll-like receptors (TLRs) in the muscle atrophy as it is observed in muscular dystrophies, disorders characterized by successive bouts of muscle fiber degeneration and regeneration in an attempt to repair contraction-induced damage. TLRs are defense receptors that detect infection and recognize self-molecules released from damaged cells. In muscular dystrophies, these receptors become over-active, and are firmly involved in the sustained chronic inflammation exhibited by the muscle tissue, via their induction of pro-inflammatory cytokine expression. Taming the exaggerated activation of TLR2/4 and TLR7/8/9, and their downstream effectors in particular, comes forward as a therapeutic strategy with potential to slow down disease progression.
Collapse
|
17
|
Day J, Otto S, Cash K, Eldi P, Hissaria P, Proudman S, Limaye V, Hayball JD. Aberrant Expression of High Mobility Group Box Protein 1 in the Idiopathic Inflammatory Myopathies. Front Cell Dev Biol 2020; 8:226. [PMID: 32363191 PMCID: PMC7180187 DOI: 10.3389/fcell.2020.00226] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 03/17/2020] [Indexed: 12/31/2022] Open
Abstract
Introduction High Mobility Group Box Protein 1 (HMGB1) is a DNA-binding protein that exerts inflammatory or pro-repair effects upon translocation from the nucleus. We postulate aberrant HMGB1 expression in immune-mediated necrotising myopathy (IMNM). Methods Herein, we compare HMGB1 expression (serological and sarcoplasmic) in patients with IMNM with that of other myositis subtypes using immunohistochemistry and ELISA. Results IMNM (n = 62) and inclusion body myositis (IBM, n = 14) patients had increased sarcoplasmic HMGB1 compared with other myositis patients (n = 46). Sarcoplasmic HMGB1 expression correlated with muscle weakness and histological myonecrosis, inflammation, regeneration and autophagy. Serum HMGB1 levels were elevated in patients with IMNM, dermatomyositis and polymositis, and those myositis patients with extramuscular inflammatory features. Discussion Aberrant HMGB1 expression occurs in myositis patients and correlates with weakness. A unique expression profile of elevated sarcoplasmic and serum HMGB1 was detected in IMNM.
Collapse
Affiliation(s)
- Jessica Day
- Experimental Therapeutics Laboratory, University of South Australia Cancer Research Institute, Adelaide, SA, Australia.,School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia.,Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Sophia Otto
- Royal Adelaide Hospital, Adelaide, SA, Australia.,SA Pathology, Adelaide, SA, Australia
| | | | - Preethi Eldi
- Experimental Therapeutics Laboratory, University of South Australia Cancer Research Institute, Adelaide, SA, Australia.,School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Pravin Hissaria
- Royal Adelaide Hospital, Adelaide, SA, Australia.,SA Pathology, Adelaide, SA, Australia
| | - Susanna Proudman
- Royal Adelaide Hospital, Adelaide, SA, Australia.,Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Vidya Limaye
- Royal Adelaide Hospital, Adelaide, SA, Australia.,Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - John D Hayball
- Experimental Therapeutics Laboratory, University of South Australia Cancer Research Institute, Adelaide, SA, Australia.,School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia.,Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
18
|
Miller FW, Lamb JA, Schmidt J, Nagaraju K. Risk factors and disease mechanisms in myositis. Nat Rev Rheumatol 2019; 14:255-268. [PMID: 29674613 DOI: 10.1038/nrrheum.2018.48] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Autoimmune diseases develop as a result of chronic inflammation owing to interactions between genes and the environment. However, the mechanisms by which autoimmune diseases evolve remain poorly understood. Newly discovered risk factors and pathogenic processes in the various idiopathic inflammatory myopathy (IIM) phenotypes (known collectively as myositis) have illuminated innovative approaches for understanding these diseases. The HLA 8.1 ancestral haplotype is a key risk factor for major IIM phenotypes in some populations, and several genetic variants associated with other autoimmune diseases have been identified as IIM risk factors. Environmental risk factors are less well studied than genetic factors but might include viruses, bacteria, ultraviolet radiation, smoking, occupational and perinatal exposures and a growing list of drugs (including biologic agents) and dietary supplements. Disease mechanisms vary by phenotype, with evidence of shared innate and adaptive immune and metabolic pathways in some phenotypes but unique pathways in others. The heterogeneity and rarity of the IIMs make advancements in diagnosis and treatment cumbersome. Novel approaches, better-defined phenotypes, and international, multidisciplinary consensus have contributed to progress, and it is hoped that these methods will eventually enable therapeutic intervention before the onset or major progression of disease. In the future, preemptive strategies for IIM management might be possible.
Collapse
Affiliation(s)
- Frederick W Miller
- Environmental Autoimmunity Group, Clinical Research Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Janine A Lamb
- Centre for Epidemiology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, UK
| | - Jens Schmidt
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Kanneboyina Nagaraju
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY, USA
| |
Collapse
|
19
|
Kobayashi T, Noguchi M, Nakayama H, Fukano R, Ohga S. Adjuvant recombinant thrombomodulin therapy for hepatopathy induced by vincristine, actinomycin D, and cyclophosphamide in pediatric rhabdomyosarcoma: A case report. Mol Clin Oncol 2019; 11:208-212. [DOI: 10.3892/mco.2019.1864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 04/30/2019] [Indexed: 01/20/2023] Open
Affiliation(s)
- Tetsuko Kobayashi
- Division of Pediatrics, National Hospital Organization Kyushu Cancer Center, Fukuoka 811‑1395, Japan
| | - Maiko Noguchi
- Division of Pediatrics, National Hospital Organization Kyushu Cancer Center, Fukuoka 811‑1395, Japan
| | - Hideki Nakayama
- Division of Pediatrics, National Hospital Organization Kyushu Cancer Center, Fukuoka 811‑1395, Japan
| | - Reiji Fukano
- Division of Pediatrics, National Hospital Organization Kyushu Cancer Center, Fukuoka 811‑1395, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Kyushu University Hospital, Fukuoka 812‑8582, Japan
| |
Collapse
|
20
|
Riuzzi F, Sorci G, Sagheddu R, Chiappalupi S, Salvadori L, Donato R. RAGE in the pathophysiology of skeletal muscle. J Cachexia Sarcopenia Muscle 2018; 9:1213-1234. [PMID: 30334619 PMCID: PMC6351676 DOI: 10.1002/jcsm.12350] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/20/2018] [Accepted: 08/24/2018] [Indexed: 12/14/2022] Open
Abstract
Emerging evidence suggests that the signalling of the Receptor for Advanced Glycation End products (RAGE) is critical for skeletal muscle physiology controlling both the activity of muscle precursors during skeletal muscle development and the correct time of muscle regeneration after acute injury. On the other hand, the aberrant re-expression/activity of RAGE in adult skeletal muscle is a hallmark of muscle wasting that occurs in response to ageing, genetic disorders, inflammatory conditions, cancer, and metabolic alterations. In this review, we discuss the mechanisms of action and the ligands of RAGE involved in myoblast differentiation, muscle regeneration, and muscle pathological conditions. We highlight potential therapeutic strategies for targeting RAGE to improve skeletal muscle function.
Collapse
Affiliation(s)
- Francesca Riuzzi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology
| | - Guglielmo Sorci
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology
| | - Roberta Sagheddu
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology
| | - Sara Chiappalupi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology
| | - Laura Salvadori
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology
| | - Rosario Donato
- Department of Experimental Medicine, University of Perugia, Perugia, Italy.,Interuniversity Institute of Myology.,Centro Universitario di Ricerca sulla Genomica Funzionale, University of Perugia, Perugia, Italy
| |
Collapse
|
21
|
Keller CW, Schmidt J, Lünemann JD. Immune and myodegenerative pathomechanisms in inclusion body myositis. Ann Clin Transl Neurol 2017; 4:422-445. [PMID: 28589170 PMCID: PMC5454400 DOI: 10.1002/acn3.419] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/09/2017] [Accepted: 04/10/2017] [Indexed: 12/17/2022] Open
Abstract
Inclusion Body Myositis (IBM) is a relatively common acquired inflammatory myopathy in patients above 50 years of age. Pathological hallmarks of IBM are intramyofiber protein inclusions and endomysial inflammation, indicating that both myodegenerative and inflammatory mechanisms contribute to its pathogenesis. Impaired protein degradation by the autophagic machinery, which regulates innate and adaptive immune responses, in skeletal muscle fibers has recently been identified as a potential key pathomechanism in IBM. Immunotherapies, which are successfully used for treating other inflammatory myopathies lack efficacy in IBM and so far no effective treatment is available. Thus, a better understanding of the mechanistic pathways underlying progressive muscle weakness and atrophy in IBM is crucial in identifying novel promising targets for therapeutic intervention. Here, we discuss recent insights into the pathomechanistic network of mutually dependent inflammatory and degenerative events during IBM.
Collapse
Affiliation(s)
- Christian W. Keller
- Institute of Experimental ImmunologyLaboratory of NeuroinflammationUniversity of ZürichZürichSwitzerland
| | - Jens Schmidt
- Department of NeurologyUniversity Medical Center GöttingenGöttingenGermany
| | - Jan D. Lünemann
- Institute of Experimental ImmunologyLaboratory of NeuroinflammationUniversity of ZürichZürichSwitzerland
- Department of NeurologyUniversity Hospital ZürichZürichSwitzerland
| |
Collapse
|
22
|
Thompson C, Piguet V, Choy E. The pathogenesis of dermatomyositis. Br J Dermatol 2017; 179:1256-1262. [DOI: 10.1111/bjd.15607] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2016] [Indexed: 11/29/2022]
Affiliation(s)
- C. Thompson
- Department of Infection and Immunity; Cardiff University; Cardiff U.K
- Department of Rheumatology; University Hospital of Wales; Cardiff U.K
| | - V. Piguet
- Department of Infection and Immunity; Cardiff University; Cardiff U.K
- Department of Dermatology; University Hospital of Wales; Cardiff U.K
| | - E. Choy
- Department of Rheumatology; University Hospital of Wales; Cardiff U.K
- Department of Dermatology; University Hospital of Wales; Cardiff U.K
| |
Collapse
|
23
|
Liu L, Deng J, Ji Q, Peng B. High-mobility Group Box 1 Is Associated with the Inflammatory Infiltration and Alveolar Bone Destruction in Rats Experimental Periapical Lesions. J Endod 2017; 43:964-969. [PMID: 28389071 DOI: 10.1016/j.joen.2016.11.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 11/07/2016] [Accepted: 11/17/2016] [Indexed: 10/19/2022]
Abstract
INTRODUCTION This study was conducted to observe the immunohistochemical localization of high-mobility group box 1 (HMGB1) and its receptor, Toll-like receptor 4 (TRL4), in the development of periapical lesions induced in rats. The possible role of these molecules in the pathogenesis of periapical lesions was also explored. METHODS Periapical lesions developed within 35 days after mandibular first molar pulp exposure in Wistar rats. The animals were randomly killed at 0, 7, 14, 21, 28, and 35 days after pulp exposure. The jaws that contained the first molar were obtained and prepared for histologic analysis, enzyme histochemistry, immunohistochemistry, and double immunofluorescence staining. RESULTS From day 0 to 35, the areas of periapical bone loss increased and appeared to be stabilized on day 35. A few HMGB1-positive, TLR4-positive cells and osteoclasts could be observed on day 7. From day 7 to 28, the HMGB1 and TLR4 protein expression increased and subsequently remained stable. The number of osteoclasts multiplied from day 0 to 14 and then gradually decreased from day 14 to 35. Double immunofluorescence staining results showed HMGB1-positive, TLR4-positive cells around periapical lesions surrounding the apical foramen. CONCLUSIONS Thus, HMGB1 and TLR4 may be associated with the pathogenesis of the periapical lesions.
Collapse
Affiliation(s)
- Lingshuang Liu
- Department of Endodontics, The Affiliated Hospital of Qingdao University, Qingdao Shi, Shandong Sheng, China
| | - Jing Deng
- Department of Endodontics, The Affiliated Hospital of Qingdao University, Qingdao Shi, Shandong Sheng, China
| | - Qiuxia Ji
- Department of Periodontology, The Affiliated Hospital of Qingdao University, Qingdao Shi, Shandong Sheng, China
| | - Bin Peng
- Department of Operative Dentistry and Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
24
|
The host defense peptide LL-37 a possible inducer of the type I interferon system in patients with polymyositis and dermatomyositis. J Autoimmun 2017; 78:46-56. [DOI: 10.1016/j.jaut.2016.12.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 12/01/2016] [Accepted: 12/06/2016] [Indexed: 11/22/2022]
|
25
|
Ceribelli A, De Santis M, Isailovic N, Gershwin ME, Selmi C. The Immune Response and the Pathogenesis of Idiopathic Inflammatory Myositis: a Critical Review. Clin Rev Allergy Immunol 2017; 52:58-70. [PMID: 26780034 DOI: 10.1007/s12016-016-8527-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The pathogenesis of idiopathic inflammatory myositis (IIMs, including polymyositis and dermatomyositis) remains largely enigmatic, despite advances in the study of the role played by innate immunity, adaptive immunity, genetic predisposition, and environmental factors in an orchestrated response. Several factors are involved in the inflammatory state that characterizes the different forms of IIMs which share features and mechanisms but are clearly different with respect to the involved sites and characteristics of the inflammation. Cellular and non-cellular mechanisms of both the immune and non-immune systems have been identified as key regulators of inflammation in polymyositis/dermatomyositis, particularly at different stages of disease, leading to the fibrotic state that characterizes the end stage. Among these, a special role is played by an interferon signature and complement cascade with different mechanisms in polymyositis and dermatomyositis; these differences can be identified also histologically in muscle biopsies. Numerous cellular components of the adaptive and innate immune response are present in the site of tissue inflammation, and the complexity of idiopathic inflammatory myositis is further supported by the involvement of non-immune mechanisms such as hypoxia and autophagy. The aim of this comprehensive review is to describe the major pathogenic mechanisms involved in the onset of idiopathic inflammatory myositis and to report on the major working hypothesis with therapeutic implications.
Collapse
Affiliation(s)
- Angela Ceribelli
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital, via A. Manzoni 56, 20089, Rozzano, MI, Italy
- BIOMETRA Department, University of Milan, Milan, Italy
| | - Maria De Santis
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital, via A. Manzoni 56, 20089, Rozzano, MI, Italy
| | - Natasa Isailovic
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital, via A. Manzoni 56, 20089, Rozzano, MI, Italy
| | - M Eric Gershwin
- Division of Rheumatology, Allergy, and Clinical Immunology, University of California Davis, Davis, CA, USA
| | - Carlo Selmi
- Division of Rheumatology and Clinical Immunology, Humanitas Research Hospital, via A. Manzoni 56, 20089, Rozzano, MI, Italy.
- BIOMETRA Department, University of Milan, Milan, Italy.
| |
Collapse
|
26
|
Wan Z, Zhang X, Peng A, He M, Lei Z, Wang Y. TLR4-HMGB1 signaling pathway affects the inflammatory reaction of autoimmune myositis by regulating MHC-I. Int Immunopharmacol 2016; 41:74-81. [PMID: 27816788 DOI: 10.1016/j.intimp.2016.10.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 09/13/2016] [Accepted: 10/12/2016] [Indexed: 10/20/2022]
Abstract
OBJECTIVES To analyze the effects of TLR4 on the expression of the HMGB1, MHC-I and downstream cytokines IL-6 and TNF-α, and to investigate the biological role of the TLR4-HMGB1 signaling pathway in the development of the autoimmune myositis. METHODS We built mice models with experimental autoimmune myositis (EAM) and used the inverted screen experiment to measure their muscle endurance; we also examined inflammatory infiltration of muscle tissues after HE staining; and we assessed the expression of MHC-I using immunohistochemistry. In addition, peripheral blood mononuclear cells (PBMC) were extracted and flow cytometry was utilized to detect the effect of IFN-γ on the expression of MHC-I. Furthermore, PBMCs were treated with IFN-γ, anti-TLR4, anti-HMGB1 and anti-MHC-I. Real-time PCR and western blotting were employed to examine the expressions of TLR4, HMGB1 and MHC-I in different groups. The ELISA method was also utilized to detect the expression of the downstream cytokines TNF-α and IL-6. RESULTS The expressions of TLR4, HMGB1 and MHC-I in muscle tissues from mice with EAM were significantly higher than those in the control group (all P<0.05). After IFN-γ treatment, the expressions of TLR4, HMGB1, MHC-I, TNF-α and IL-6 in PBMCs significantly increased (all P<0.05). The treatment of anti-TLR4, anti-HMGB1 and anti-MHC-I could significantly downregulate the expression of MHC-I (all P<0.05). In addition, anti-TLR4 and anti-HMGB1 significantly reduced the expression of TNF-α and IL-6 (all P<0.05). CONCLUSIONS The TLR4-HMGB1 signaling pathway affects the process of autoimmune myositis inflammation by regulating the expression of MHC-I and other pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Zemin Wan
- Department of Clinical Laboratory, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, Guangdong, China.
| | - Xiujuan Zhang
- Department of Liver Disease, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Anping Peng
- Department of Clinical Laboratory, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Min He
- Department of Clinical Laboratory, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Zhenhua Lei
- Department of Urology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| | - Yunxiu Wang
- Department of Clinical Laboratory, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, Guangdong, China
| |
Collapse
|
27
|
Dysregulated innate immune function in the aetiopathogenesis of idiopathic inflammatory myopathies. Autoimmun Rev 2016; 16:87-95. [PMID: 27666811 DOI: 10.1016/j.autrev.2016.09.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 09/08/2016] [Indexed: 12/20/2022]
Abstract
The idiopathic inflammatory myopathies (IIMs) are a heterogeneous group of systemic muscle conditions that are believed to be autoimmune in nature. They have distinct pathological features, but the aetiopathogenesis of each subtype remains largely unknown. Recently, there has been increased interest in the complex role the innate immune system plays in initiating and perpetuating these conditions, and how this may differ between subtypes. This article summarises the traditional paradigms of IIM pathogenesis and reviews the accumulating evidence for disturbances in innate immune processes in these rare, but debilitating chronic conditions.
Collapse
|
28
|
Pahwa R, Jialal I. The role of the high-mobility group box1 protein-Toll like receptor pathway in diabetic vascular disease. J Diabetes Complications 2016; 30:1186-91. [PMID: 27037040 DOI: 10.1016/j.jdiacomp.2016.03.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 03/07/2016] [Accepted: 03/08/2016] [Indexed: 12/20/2022]
Abstract
OBJECTIVES Increased Toll like receptors (TLRs) especially 2 and 4 have been demonstrated in obesity, metabolic syndrome (MetS) and diabetes resulting in increased cellular inflammation. Since we have shown increased TLR2 and 4 activities in both T1DM and T2DM and MetS, we wanted to elucidate the mechanisms of this sterile inflammation. In T1DM, T2DM and MetS we have shown that high mobility group box 1 protein (HMGB-1), a non-histone DNA binding protein is increased and could be a potential activator of TLRs since it has previously shown to activate TLR2, 4 and 9. We examined the role of HMGB-1 in patients and animal models of diabetes and MetS to determine how important it is as an activator of TLR mediated inflammation and its role in diabetic vascular complications. METHODS A Medline search was conducted using the terms HMGB-1, TLRs and diabetes. RESULTS HMGB-1 levels are increased in patients with diabetes and MetS, and associated with increased biomediators of inflammation. Furthermore data supported a role of HMGB-1 in both diabetic microvascular and macrovascular complications. CONCLUSIONS HMGB-1 interaction with TLRs is implicated in diabetic complications and could be important therapeutic target.
Collapse
Affiliation(s)
- Roma Pahwa
- Laboratory of Atherosclerosis and Metabolic Research, Department of Pathology and InternalMedicine, University of California Davis Medical Center, Sacramento, CA
| | - Ishwarlal Jialal
- Laboratory of Atherosclerosis and Metabolic Research, Department of Pathology and InternalMedicine, University of California Davis Medical Center, Sacramento, CA; Medical Services (Endocrinology and Metabolism), VA Medical Center, Mather, CA.
| |
Collapse
|
29
|
Loell I, Raouf J, Chen YW, Shi R, Nennesmo I, Alexanderson H, Dastmalchi M, Nagaraju K, Korotkova M, Lundberg IE. Effects on muscle tissue remodeling and lipid metabolism in muscle tissue from adult patients with polymyositis or dermatomyositis treated with immunosuppressive agents. Arthritis Res Ther 2016; 18:136. [PMID: 27287443 PMCID: PMC4902919 DOI: 10.1186/s13075-016-1033-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 05/25/2016] [Indexed: 12/24/2022] Open
Abstract
Background Polymyositis (PM) and dermatomyositis (DM) are autoimmune muscle diseases, conventionally treated with high doses of glucocorticoids in combination with immunosuppressive drugs. Treatment is often dissatisfying, with persisting muscle impairment. We aimed to investigate molecular mechanisms that might contribute to the persisting muscle impairment despite immunosuppressive treatment in adult patients with PM or DM using gene expression profiling of repeated muscle biopsies. Methods Paired skeletal muscle biopsies from six newly diagnosed adult patients with DM or PM taken before and after conventional immunosuppressive treatment were examined by gene expression microarray analysis. Selected genes that displayed changes in expression were analyzed by Western blot. Muscle biopsy sections were evaluated for inflammation, T lymphocytes (CD3), macrophages (CD68), major histocompatibility complex (MHC) class I expression and fiber type composition. Results After treatment, genes related to immune response and inflammation, including inflammasome pathways and interferon, were downregulated. This was confirmed at the protein level for AIM-2 and caspase-1 in the inflammasome pathway. Changes in genes involved in muscle tissue remodeling suggested a negative effect on muscle regeneration and growth. Gene markers for fast type II fibers were upregulated and fiber composition was switched towards type II fibers in response to treatment. The expression of genes involved in lipid metabolism was altered, suggesting a potential lipotoxic effect on muscles of the immunosuppressive treatment. Conclusion The anti-inflammatory effect of immunosuppressive treatment was combined with negative effects on genes involved in muscle tissue remodeling and lipid metabolism, suggesting a negative effect on recovery of muscle performance which may contribute to persisting muscle impairment in adult patients with DM and PM.
Collapse
Affiliation(s)
- Ingela Loell
- Karolinska Institutet, Department of Medicine, Rheumatology Unit, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Joan Raouf
- Karolinska Institutet, Department of Medicine, Rheumatology Unit, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Yi-Wen Chen
- Childrens National Medical Center, Research Center for Genetic Medicine, Washington, DC, USA
| | - Rongye Shi
- Center for Human Immunology, Autoimmunity and Inflammation, National Heart/Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Inger Nennesmo
- Karolinska University Hospital Huddinge, Institution for Laboratory Medicine (LABMED), Stockholm, Sweden
| | - Helene Alexanderson
- Karolinska Institutet, Department of NVS, Division of Physical Therapy and Karolinska University Hospital Solna, Physical Therapy Clinic, Stockholm, Sweden
| | - Maryam Dastmalchi
- Karolinska Institutet, Department of Medicine, Rheumatology Unit, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Kanneboyina Nagaraju
- Childrens National Medical Center, Research Center for Genetic Medicine, Washington, DC, USA
| | - Marina Korotkova
- Karolinska Institutet, Department of Medicine, Rheumatology Unit, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Ingrid E Lundberg
- Karolinska Institutet, Department of Medicine, Rheumatology Unit, Karolinska University Hospital Solna, Stockholm, Sweden.
| |
Collapse
|
30
|
Han SJ, Min HJ, Yoon SC, Ko EA, Park SJ, Yoon JH, Shin JS, Seo KY. HMGB1 in the pathogenesis of ultraviolet-induced ocular surface inflammation. Cell Death Dis 2015; 6:e1863. [PMID: 26313914 PMCID: PMC4558494 DOI: 10.1038/cddis.2015.199] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 05/27/2015] [Accepted: 06/16/2015] [Indexed: 01/19/2023]
Abstract
High-mobility group box 1 (HMGB1) functions as a transcription-enhancing nuclear protein as well as a crucial cytokine that regulates inflammation. This study demonstrated that secretion of HMGB1 due to ultraviolet (UV) radiation inducing ocular surface inflammation-mediated reactive oxygen species (ROS) production. After treating conjunctival epithelial cells with UV radiation, HMGB1 was translocated from the nucleus to the cytoplasm and then eventually to the extracellular space. HMGB1 played a crucial role in UV-induced conjunctival neutrophil infiltration, which subsided when mice were pretreated with the HMGB1 inhibitors soluble receptor for advanced glycation endproducts (sRAGEs) and HMGB1 A box protein. In case of using ROS quencher, there was decrease in UV-induced HMGB1 secretion in conjunctival epithelial cells and mice. Considering that UV-induced chronic inflammation causes ocular surface change as pterygium, we have confirmed high HMGB1 translocation and ROS expression in human pterygium. Our findings therefore revealed a previously unknown mechanism of UV-induced ocular inflammation related to ROS and HMGB1 suggesting a new medical therapeutic target.
Collapse
Affiliation(s)
- S J Han
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - H J Min
- Department of Otorhinolaryngology - Head and Neck Surgery, Chung-Ang University College of Medicine, Seoul, Korea.,Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - S C Yoon
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - E A Ko
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea
| | - S J Park
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea
| | - J-H Yoon
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Department of Otorhinolaryngology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute and Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - J-S Shin
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea.,Department of Microbiology, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute and Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Korea
| | - K Y Seo
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
31
|
Findlay AR, Goyal NA, Mozaffar T. An overview of polymyositis and dermatomyositis. Muscle Nerve 2015; 51:638-56. [PMID: 25641317 DOI: 10.1002/mus.24566] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2015] [Indexed: 12/23/2022]
Abstract
Polymyositis and dermatomyositis are inflammatory myopathies that differ in their clinical features, histopathology, response to treatment, and prognosis. Although their clinical pictures differ, they both present with symmetrical, proximal muscle weakness. Treatment relies mainly upon empirical use of corticosteroids and immunosuppressive agents. A deeper understanding of the molecular pathways that drive pathogenesis, careful phenotyping, and accurate disease classification will aid clinical research and development of more efficacious treatments. In this review we address the current knowledge of the epidemiology, clinical characteristics, diagnostic evaluation, classification, pathogenesis, treatment, and prognosis of polymyositis and dermatomyositis.
Collapse
Affiliation(s)
- Andrew R Findlay
- Department of Neurology, University of California, Irvine UC Irvine, MDA ALS and Neuromuscular Center, 200 South Manchester Avenue, Suite 110, Orange, California, 92868, USA
| | | | | |
Collapse
|
32
|
Kurokohchi K, Imataki O, Kubo F. Anti-inflammatory effect of recombinant thrombomodulin for fulminant hepatic failure. World J Gastroenterol 2015; 21:8203-8207. [PMID: 26185395 PMCID: PMC4499366 DOI: 10.3748/wjg.v21.i26.8203] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Revised: 03/01/2015] [Accepted: 03/19/2015] [Indexed: 02/06/2023] Open
Abstract
Fulminant hepatic failure (FHF) is a critical illness that can be comorbid to primary liver damage. FHF shows a high mortality rate, and patients with FHF require intensive therapy, including plasma apheresis. However, intensive care at the present is not enough to restore the severe liver damage or promote hepatocellular reproduction, and a standard therapy for the treatment of FHF has not been established. An 86-year-old female with FHF was admitted to our hospital. Her manifestation demonstrated a clinical situation of systemic inflammatory response syndrome (SIRS) and disseminated intravascular coagulation. A diagnosis of fulminant hepatitis was made according to the definition given in the position paper of the American Association for the Study of Liver Diseases. Her serum hepatocyte growth factor (HGF) level had increased to 11.84 ng/mL. The HGF level indicated massive liver damage as seen in FHF. Recombinant thrombomodulin (rTM) was administered daily from the admission day for 1 wk at 380 U/kg. The patient’s white blood cells and C-reactive protein responded to the rTM treatment within a few days. The HGF level and PT recovered to the normal range. The levels of proinflammatory cytokines (tumor necrosis factor-α and interleukin-1β) were suppressed by the administration of rTM. The patient’s hepatic function (e.g., PT and albumin) completely recovered without plasma exchange. rTM may modulate the over-response of SIRS with the improvement of proinflammatory cytokines. The underlying mechanism is thought to be the inhibitory effect of rTM on high-mobility group box 1 (HMBG1). The pathogenesis of HMBG1 protein in fulminant hepatic failure has been already known. A novel favorable effect of rTM for SIRS would be promising for FHF, and the wide application of rTM for SIRS should be considered.
Collapse
|
33
|
Muth IE, Zschüntzsch J, Kleinschnitz K, Wrede A, Gerhardt E, Balcarek P, Schreiber-Katz O, Zierz S, Dalakas MC, Voll RE, Schmidt J. HMGB1 and RAGE in skeletal muscle inflammation: Implications for protein accumulation in inclusion body myositis. Exp Neurol 2015; 271:189-97. [PMID: 26048613 DOI: 10.1016/j.expneurol.2015.05.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 05/23/2015] [Accepted: 05/30/2015] [Indexed: 01/26/2023]
Abstract
Inflammation is associated with protein accumulation in IBM, but precise mechanisms are elusive. The "alarmin" HMGB1 is upregulated in muscle inflammation. Its receptor RAGE is crucial for β-amyloid-associated neurodegeneration. Relevant signaling via HMGB1/RAGE is expected in IBM pathology. By real-time-PCR, mRNA-expression levels of HMGB1 and RAGE were upregulated in muscle biopsies of patients with IBM and PM, but not in muscular dystrophy or non-myopathic controls. By immunohistochemistry, both molecules displayed the highest signal in IBM, where they distinctly co-localized to intra-fiber accumulations of β-amyloid and neurofilament/tau. In these fibers, identification of phosphorylated Erk suggested that relevant downstream activation is present upon HMGB1 signaling via RAGE. Protein expressions of HMGB1, RAGE, Erk and phosphorylated Erk were confirmed by Western blot. In a well established cell-culture model for pro-inflammatory cell-stress, exposure of human muscle-cells to IL-1β+IFN-γ induced cytoplasmic translocation of HMGB1 and subsequent release as evidenced by ELISA. Upregulation of RAGE on the cell surface was demonstrated by immunocytochemistry and flow-cytometry. Recombinant HMGB1 was equally potent as IL-1β+IFN-γ in causing amyloid-accumulation and cell-death, and both were abrogated by the HMGB1-blocker BoxA. The findings strengthen the concept of unique interactions between degenerative and inflammatory mechanisms and suggest that HMGB1/RAGE signaling is a critical pathway in IBM pathology.
Collapse
Affiliation(s)
- Ingrid E Muth
- Department of Neurology, University Medical Center, Göttingen, Germany
| | - Jana Zschüntzsch
- Department of Neurology, University Medical Center, Göttingen, Germany
| | - Konstanze Kleinschnitz
- Department of Neurology, University Medical Center, Göttingen, Germany; Department of Neuroimmunology, Institute for Multiple Sclerosis Research and Hertie Foundation, University Medical Center, Göttingen, Germany
| | - Arne Wrede
- Department of Neuropathology, University Medical Center, Göttingen, Germany
| | - Ellen Gerhardt
- Department of Neurodegeneration and Restorative Research, Center for Nanoscale Microscopy and Molecular Physiology of the Brain, University Medical Center Göttingen, Germany
| | - Peter Balcarek
- Department of Trauma Surgery, University Medical Center, Göttingen, Germany
| | - Olivia Schreiber-Katz
- Friedrich-Baur-Institute, Department of Neurology, Ludwig-Maximilians-University of München, München, Germany
| | - Stephan Zierz
- Department of Neurology, University Hospital Halle/Saale, Halle/Saale, Germany
| | - Marinos C Dalakas
- Neuroimmunology Unit, Department of Pathophysiology, University of Athens Medical School, Athens, Greece; Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, University Medical Center, Freiburg, Germany
| | - Jens Schmidt
- Department of Neurology, University Medical Center, Göttingen, Germany; Department of Neuroimmunology, Institute for Multiple Sclerosis Research and Hertie Foundation, University Medical Center, Göttingen, Germany.
| |
Collapse
|
34
|
Sciorati C, Monno A, Ascherman DP, Seletti E, Manfredi AA, Rovere-Querini P. Required role of apoptotic myogenic precursors and toll-like receptor stimulation for the establishment of autoimmune myositis in experimental murine models. Arthritis Rheumatol 2015; 67:809-22. [PMID: 25504878 DOI: 10.1002/art.38985] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 12/02/2014] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Muscle regeneration is a hallmark of the idiopathic inflammatory myopathies (IIMs), a group of autoimmune disorders that are characterized by leukocyte infiltration and dysfunction of the skeletal muscle. Despite detailed studies describing the clinical and histopathologic features of IIMs, the immunopathogenesis of these disorders remains undefined. The aim of this study was to investigate the immunopathologic processes of autoimmune myositis in experimental murine models. METHODS Expression of the autoantigen histidyl-transfer RNA synthetase (HisRS) was analyzed in mice with acutely injured or dystrophic muscles, in inflammatory leukocytes, and in purified satellite cells. Anti-HisRS antibodies and myositis induction were assessed in mice after muscle injury and immunization with apoptotic satellite cells or C2C12 myoblasts, in the presence or absence of the Toll-like receptor 7 (TLR-7) agonist R848. RESULTS Muscle necrosis, leukocyte infiltration, and myofiber regeneration induced by toxic agents (cardiotoxin or glycerol) or promoted by genetic disruption of the α-sarcoglycan/dystrophin complex in mice were uniformly associated with up-regulated expression of HisRS. Although regenerating myofibers and purified satellite cells are known to show increased expression of HisRS in these settings, anti-HisRS antibodies were not detectable. However, intramuscular immunization with ultraviolet B-irradiated, HisRS-expressing apoptotic myoblasts in the presence of R848 triggered the production of anti-HisRS IgG antibodies as well as persistent lymphocyte infiltration and prolonged/delayed muscle regeneration. Conversely, intramuscular administration of R848 alone or in combination with living or postapoptotic/necrotic myoblasts failed to generate this myositis phenotype. CONCLUSION In the presence of TLR/adjuvant signals and underlying muscle injury, apoptotic myogenic precursors expressing high levels of autoantigen can provoke autoantibody formation and lymphocytic infiltration of muscle tissue, effectively replicating the features of IIM.
Collapse
|
35
|
HMGB1 expression and muscle regeneration in idiopathic inflammatory myopathies and degenerative joint diseases. J Muscle Res Cell Motil 2015; 36:255-62. [PMID: 25761565 DOI: 10.1007/s10974-015-9411-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 03/05/2015] [Indexed: 01/17/2023]
Abstract
The High-Mobility Group Box 1 protein (HMGB1) is a known nuclear protein which may be released from the nucleus into the cytoplasm and the extracellular space. It is believed that the mobilized HMGB1 plays role in the autoimmune processes as an alarmin, stimulating the immune response. In addition, muscle regeneration and differentiation may also be altered in the inflammatory surroundings. Biopsy specimens derived from patients with idiopathic inflammatory myopathies (IIM) such as polymyositis or dermatomyositis were compared to muscle samples from patients undergoing surgical interventions for coxarthrosis. The biopsy and surgery specimens were used for Western blot analysis, for immunohistochemical detection of HMGB1 in histological preparations and for cell culturing to examine cell proliferation and differentiation. Our data show lower HMGB1 expression, impaired proliferation and slightly altered fusion capacity in the primary cell cultures started from IIM specimens than in cultures of coxarthrotic muscles. The ratio of regenerating muscle fibres with centralised nuclei (myotubes) is lower in the IIM samples than in the coxarthrotic ones but corticosteroid treatment shifts the ratio towards the coxarthrotic value. Our data suggest that the impaired regeneration capacity should also be considered to be behind the muscle weakness in IIM patients. The role of HMGB1 as a pathogenic signal requires further investigation.
Collapse
|
36
|
Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, Huang J, Yu Y, Fan XG, Yan Z, Sun X, Wang H, Wang Q, Tsung A, Billiar TR, Zeh HJ, Lotze MT, Tang D. HMGB1 in health and disease. Mol Aspects Med 2014; 40:1-116. [PMID: 25010388 PMCID: PMC4254084 DOI: 10.1016/j.mam.2014.05.001] [Citation(s) in RCA: 705] [Impact Index Per Article: 70.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/05/2014] [Indexed: 12/22/2022]
Abstract
Complex genetic and physiological variations as well as environmental factors that drive emergence of chromosomal instability, development of unscheduled cell death, skewed differentiation, and altered metabolism are central to the pathogenesis of human diseases and disorders. Understanding the molecular bases for these processes is important for the development of new diagnostic biomarkers, and for identifying new therapeutic targets. In 1973, a group of non-histone nuclear proteins with high electrophoretic mobility was discovered and termed high-mobility group (HMG) proteins. The HMG proteins include three superfamilies termed HMGB, HMGN, and HMGA. High-mobility group box 1 (HMGB1), the most abundant and well-studied HMG protein, senses and coordinates the cellular stress response and plays a critical role not only inside of the cell as a DNA chaperone, chromosome guardian, autophagy sustainer, and protector from apoptotic cell death, but also outside the cell as the prototypic damage associated molecular pattern molecule (DAMP). This DAMP, in conjunction with other factors, thus has cytokine, chemokine, and growth factor activity, orchestrating the inflammatory and immune response. All of these characteristics make HMGB1 a critical molecular target in multiple human diseases including infectious diseases, ischemia, immune disorders, neurodegenerative diseases, metabolic disorders, and cancer. Indeed, a number of emergent strategies have been used to inhibit HMGB1 expression, release, and activity in vitro and in vivo. These include antibodies, peptide inhibitors, RNAi, anti-coagulants, endogenous hormones, various chemical compounds, HMGB1-receptor and signaling pathway inhibition, artificial DNAs, physical strategies including vagus nerve stimulation and other surgical approaches. Future work further investigating the details of HMGB1 localization, structure, post-translational modification, and identification of additional partners will undoubtedly uncover additional secrets regarding HMGB1's multiple functions.
Collapse
Affiliation(s)
- Rui Kang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| | - Ruochan Chen
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Qiuhong Zhang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Wen Hou
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Sha Wu
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Lizhi Cao
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jin Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yan Yu
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xue-Gong Fan
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhengwen Yan
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA; Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Xiaofang Sun
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Experimental Department of Institute of Gynecology and Obstetrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510510, China
| | - Haichao Wang
- Laboratory of Emergency Medicine, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - Qingde Wang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Allan Tsung
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Herbert J Zeh
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Daolin Tang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| |
Collapse
|
37
|
Abstract
High-mobility group box 1 (HMGB1) was originally defined as a ubiquitous nuclear protein, but it was later determined that the protein has different roles both inside and outside of cells. Nuclear HMGB1 regulates chromatin structure and gene transcription, whereas cytosolic HMGB1 is involved in inflammasome activation and autophagy. Extracellular HMGB1 has drawn attention because it can bind to related cell signalling transduction receptors, such as the receptor for advanced glycation end products, Toll-like receptor (TLR)2, TLR4 and TLR9. It also participates in the development and progression of a variety of diseases. HMGB1 is actively secreted by stimulation of the innate immune system, and it is passively released by ischaemia or cell injury. This review focuses on the important role of HMGB1 in the pathogenesis of acute and chronic sterile inflammatory conditions. Strategies that target HMGB1 have been shown to significantly decrease inflammation in several disease models of sterile inflammation, and this may represent a promising clinical approach for treatment of certain conditions associated with sterile inflammation.
Collapse
Affiliation(s)
- A Tsung
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | | |
Collapse
|
38
|
Magna M, Pisetsky DS. The role of HMGB1 in the pathogenesis of inflammatory and autoimmune diseases. Mol Med 2014; 20:138-46. [PMID: 24531836 DOI: 10.2119/molmed.2013.00164] [Citation(s) in RCA: 242] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/05/2014] [Indexed: 12/30/2022] Open
Abstract
High-mobility group box 1 (HMGB1) protein is a highly abundant protein that can promote the pathogenesis of inflammatory and autoimmune diseases once it is in an extracellular location. This translocation can occur with immune cell activation as well as cell death, with the conditions for release associated with the expression of different isoforms. These isoforms result from post-translational modifications, with the redox states of three cysteines at positions 23, 45 and 106 critical for activity. Depending on the redox states of these residues, HMGB1 can induce cytokine production via toll-like receptor 4 (TLR4) or promote chemotaxis by binding the chemokine CXCL12 for stimulation via CXCR4. Fully oxidized HMGB1 is inactive. During the course of inflammatory disease, HMGB1 can therefore play a dynamic role depending on its redox state. As a mechanism to generate alarmins, cell death is an important source of HMGB1, although each major cell death form (necrosis, apoptosis, pyroptosis and NETosis) can lead to different isoforms of HMGB1 and variable levels of association of HMGB1 with nucleosomes. The association of HMGB1 with nucleosomes may contribute to the pathogenesis of systemic lupus erythematosus by producing nuclear material whose immunological properties are enhanced by the presence of an alarmin. Since HMGB1 levels in blood or tissue are elevated in many inflammatory and autoimmune diseases, this molecule can serve as a unique biomarker as well as represent a target of novel therapies to block its various activities.
Collapse
Affiliation(s)
- Melinda Magna
- Duke University Medical Center, Durham, North Carolina, United States of America
| | - David S Pisetsky
- Duke University Medical Center, Durham, North Carolina, United States of America Medical Research Service, Durham Veterans Administration Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
39
|
Johansson L, Snäll J, Sendi P, Linnér A, Thulin P, Linder A, Treutiger CJ, Norrby-Teglund A. HMGB1 in severe soft tissue infections caused by Streptococcus pyogenes. Front Cell Infect Microbiol 2014; 4:4. [PMID: 24524027 PMCID: PMC3906589 DOI: 10.3389/fcimb.2014.00004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 01/09/2014] [Indexed: 12/31/2022] Open
Abstract
Extracellular High Mobility Group Box 1 (HMGB1) has been associated with acute and chronic inflammatory conditions. However, little is known about HMGB1 in necrotizing bacterial infections. We hypothesized that the local HMGB1 response is excessive in severe soft tissue infections (STIs), which are characterized by necrosis and hyperinflammation. To explore this, tissue biopsies were collected from patients with varying severity of Streptococcus pyogenes skin and STIs, including erysipelas, cellulitis, and necrotizing fasciitis. Tissue sections were immunostained for HMGB1, S. pyogenes, and inflammatory cell infiltrates and results quantified by acquired computerized image analysis (ACIA). HMGB1 expression increased in parallel to disease severity and was significantly higher in necrotizing fasciitis than in erysipelas (p = 0.0023). Confocal microscopy of sections co-stained for HMGB1 and cell markers revealed both extracellular and cytoplasmic HMGB1, the latter of which was found predominantly in macrophages. To further verify macrophages as main source of activation triggered HMGB1 release, human macrophages were infected with clinical S. pyogenes isolates. The results demonstrated infection triggered release of HMGB1. Dual staining's visualized HMGB1 in areas close to, but not overlapping, with neutrophils, indicating a potential chemotactic role. In vitro transmigration experiments showed a chemotactic effect of HMGB1 on neutrophils. The data furthermore provided in vivo support that HGMB1 may form immunostimulatory complexes with IL-1β. Taken together, the findings provide the first in vivo evidence that HMGB1 is abundant at the local site of severe bacterial STIs and its levels correlated to severity of infections; hence, indicating its potential value as a biomarker for tissue pathology.
Collapse
Affiliation(s)
- Linda Johansson
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet Stockholm, Sweden
| | - Johanna Snäll
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet Stockholm, Sweden
| | - Parham Sendi
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet Stockholm, Sweden
| | - Anna Linnér
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet Stockholm, Sweden
| | - Pontus Thulin
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet Stockholm, Sweden
| | - Adam Linder
- Division of Infection Medicine, Department of Clinical Sciences, Lund University Hospital Lund, Sweden
| | - Carl-Johan Treutiger
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet Stockholm, Sweden
| | - Anna Norrby-Teglund
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet Stockholm, Sweden
| |
Collapse
|
40
|
Hayashi A, Nagafuchi H, Ito I, Hirota K, Yoshida M, Ozaki S. Lupus antibodies to the HMGB1 chromosomal protein: epitope mapping and association with disease activity. Mod Rheumatol 2014. [DOI: 10.3109/s10165-009-0151-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
41
|
Venalis P, Lundberg IE. Immune mechanisms in polymyositis and dermatomyositis and potential targets for therapy. Rheumatology (Oxford) 2013; 53:397-405. [PMID: 23970542 DOI: 10.1093/rheumatology/ket279] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PM and DM are characterized clinically by weakness and low endurance of skeletal muscle. Other organs are frequently involved, suggesting that idiopathic inflammatory myopathies (IIMs) are systemic inflammatory diseases. Involvement of immune mechanisms in IIMs is supported by the presence of T cells, macrophages and dendritic cells in muscle tissue, by the presence of autoantibodies and by HLA-DR being a strong genetic risk factor. T cells may have direct and indirect toxic effects on muscle fibres, causing muscle fibre necrosis and muscle weakness, but the target of the immune reaction is not known. A newly identified T cell subset, CD28(null) T cells, may have cytotoxic effects in the CD4(+) and CD8(+) T cell phenotype. These cells are apoptosis resistant and may contribute to treatment resistance. Several myositis-specific autoantibodies have been identified, but they are all directed against ubiquitously expressed autoantigens and the specificity of the T cell reactivity is not known. These autoantibodies are associated with distinct clinical phenotypes and some with distinct molecular pathways; e.g. sera from patients with anti-Jo-1 autoantibodies may activate the type I IFN system and these sera also contain high levels of B cell activating factor compared with other IIM subsets. The characterization of patients into subgroups based on autoantibody profiles seems to be a promising way to learn more about the specificities of the immune reactions. Careful phenotyping of infiltrating immune cells in muscle tissue before and after specific therapies and relating the molecular findings to clinical outcome measures may be another way to improve knowledge on specific immune mechanism in IIMs. Such information will be important for the development of new therapies.
Collapse
Affiliation(s)
- Paulius Venalis
- CMM Foundation, Karolinska University Hospital L8:04, 171 76 Stockholm, Sweden.
| | | |
Collapse
|
42
|
Fernandez I, Harlow L, Zang Y, Liu-Bryan R, Ridgway WM, Clemens PR, Ascherman DP. Functional redundancy of MyD88-dependent signaling pathways in a murine model of histidyl-transfer RNA synthetase-induced myositis. THE JOURNAL OF IMMUNOLOGY 2013; 191:1865-72. [PMID: 23842751 DOI: 10.4049/jimmunol.1203070] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
We have previously shown that i.m. administration of bacterially expressed murine histidyl-tRNA synthetase (HRS) triggers florid muscle inflammation (relative to appropriate control proteins) in various congenic strains of mice. Because severe disease develops even in the absence of adaptive immune responses to HRS, we sought to identify innate immune signaling components contributing to our model of HRS-induced myositis. In vitro stimulation assays demonstrated HRS-mediated activation of HEK293 cells transfected with either TLR2 or TLR4, revealing an excitatory capacity exceeding that of other bacterially expressed fusion proteins. Corresponding to this apparent functional redundancy of TLR signaling pathways, HRS immunization of B6.TLR2(-/-) and B6.TLR4(-/-) single-knockout mice yielded significant lymphocytic infiltration of muscle tissue comparable to that produced in C57BL/6 wild-type mice. In contrast, concomitant elimination of TLR2 and TLR4 signaling in B6.TLR2(-/-).TLR4(-/-) double-knockout mice markedly reduced the severity of HRS-induced muscle inflammation. Complementary subfragment analysis demonstrated that aa 60-90 of HRS were absolutely required for in vitro as well as in vivo signaling via these MyD88-dependent TLR pathways--effects mediated, in part, through preferential binding of exogenous ligands capable of activating specific TLRs. Collectively, these experiments indicate that multiple MyD88-dependent signaling cascades contribute to this model of HRS-induced myositis, underscoring the antigenic versatility of HRS and confirming the importance of innate immunity in this system.
Collapse
Affiliation(s)
- Irina Fernandez
- Division of Rheumatology, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Rayavarapu S, Coley W, Kinder TB, Nagaraju K. Idiopathic inflammatory myopathies: pathogenic mechanisms of muscle weakness. Skelet Muscle 2013; 3:13. [PMID: 23758833 PMCID: PMC3681571 DOI: 10.1186/2044-5040-3-13] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 04/22/2013] [Indexed: 11/25/2022] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are a heterogenous group of complex muscle diseases of unknown etiology. These diseases are characterized by progressive muscle weakness and damage, together with involvement of other organ systems. It is generally believed that the autoimmune response (autoreactive lymphocytes and autoantibodies) to skeletal muscle-derived antigens is responsible for the muscle fiber damage and muscle weakness in this group of disorders. Therefore, most of the current therapeutic strategies are directed at either suppressing or modifying immune cell activity. Recent studies have indicated that the underlying mechanisms that mediate muscle damage and dysfunction are multiple and complex. Emerging evidence indicates that not only autoimmune responses but also innate immune and non-immune metabolic pathways contribute to disease pathogenesis. However, the relative contributions of each of these mechanisms to disease pathogenesis are currently unknown. Here we discuss some of these complex pathways, their inter-relationships and their relation to muscle damage in myositis. Understanding the relative contributions of each of these pathways to disease pathogenesis would help us to identify suitable drug targets to alleviate muscle damage and also improve muscle weakness and quality of life for patients suffering from these debilitating muscle diseases.
Collapse
Affiliation(s)
- Sree Rayavarapu
- Research Center for Genetic Medicine, Children's National Medical Center, 111 Michigan Ave NW, Washington DC, USA.
| | | | | | | |
Collapse
|
44
|
Chen Y, Sun W, Gao R, Su Y, Umehara H, Dong L, Gong F. The role of high mobility group box chromosomal protein 1 in rheumatoid arthritis. Rheumatology (Oxford) 2013; 52:1739-47. [PMID: 23584368 DOI: 10.1093/rheumatology/ket134] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
High mobility group box chromosomal protein 1 (HMGB1) is a ubiquitous highly conserved single polypeptide in all mammal eukaryotic cells. HMGB1 exists mainly within the nucleus and acts as a DNA chaperone. When passively released from necrotic cells or actively secreted into the extracellular milieu in response to appropriate signal stimulation, HMGB1 binds to related cell signal transduction receptors, such as RAGE, TLR2, TLR4 and TLR9, and becomes a proinflammatory cytokine that participates in the development and progression of many diseases, such as arthritis, acute lung injury, graft rejection immune response, ischaemia reperfusion injury and autoimmune liver damage. Only a small amount of HMGB1 release occurs during apoptosis, which undergoes oxidative modification on Cys106 and delivers tolerogenic signals to suppress immune activity. This review focuses on the important role of HMGB1 in the pathogenesis of RA, mainly manifested as the aberrant expression of HMGB1 in the serum, SF and synovial tissues; overexpression of signal transduction receptors; abnormal regulation of osteoclastogenesis and bone remodelling leading to the destruction of cartilage and bones. Intervention with HMGB1 may ameliorate the pathogenic conditions and attenuate disease progression of RA. Therefore administration of an HMGB1 inhibitor may represent a promising clinical approach for the treatment of RA.
Collapse
Affiliation(s)
- Yu Chen
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095th Jiefang Avenue, Wuhan, Hubei 430030, China.
| | | | | | | | | | | | | |
Collapse
|
45
|
Wu CX, Guo H, Gong JP, Liu Q, Sun H. The role of high mobility group box chromosomal protein 1 expression in the differential diagnosis of hepatic actinomycosis: a case report. J Med Case Rep 2013; 7:31. [PMID: 23351605 PMCID: PMC3573988 DOI: 10.1186/1752-1947-7-31] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 01/02/2013] [Indexed: 11/24/2022] Open
Abstract
Introduction Primary hepatic actinomycosis is a rare disease, but is important in the differential diagnosis of hepatoma in endemic areas. As high mobility group box chromosomal protein 1 plays an important role in the pathogenesis of both acute and chronic inflammatory conditions, we postulate that high mobility group box chromosomal protein 1 may have a possible pathogenic role in hepatic actinomycosis. To the best of our knowledge, our report is the first to detect an association between highly elevated high mobility group box chromosomal protein 1 expression and hepatic actinomycosis. Case presentation A 67-year-old Chinese man was admitted to our hospital with a three-month history of epigastric pain, anorexia, and subjective weight loss. Ultrasonography and computed tomography of the patient’s abdomen confirmed a hypodense mass measuring seven cm in diameter in the left lateral segment of his liver. A hepatic tumor was suspected and surgical resection was scheduled. Histopathologic examination revealed that the overall features of the hepatic tissues were consistent with hepatic actinomycosis. Whole blood and hepatic tissue samples of the patient, of patients who had hepatocellular carcinoma and of healthy donors were collected. Serum high mobility group box chromosomal protein 1 concentration in actinomycosis was 8.5ng/mL, which was higher than the hepatocellular carcinoma level of 5.2ng/mL and the normal level of <three ng/mL. High mobility group box chromosomal protein 1 messenger ribonucleic acid levels and high mobility group box chromosomal protein 1 protein content in the affected tissues of this patient with hepatic actinomycosis were higher than those of the control and hepatocellular carcinoma tissues. The results of immunohistochemistry showed the following: in the control tissues, high mobility group box chromosomal protein 1 was distributed mainly in the cytoplasm; in the hepatocellular carcinoma tissues, high mobility group box chromosomal protein 1 was distributed primarily in the nucleus; and in the actinomycosis tissues, high mobility group box chromosomal protein 1 was increased in both the cytoplasm and nucleus. Conclusion High mobility group box chromosomal protein 1 may have a potent biological effect on the pathogenesis of hepatic actinomycosis as a novel cytokine and may be a useful marker in the differential diagnosis of hepatic actinomycosis.
Collapse
Affiliation(s)
- Chuan-Xin Wu
- Key Laboratory of Molecular Biology for Infectious Diseases, Ministry of Education, Liver Diseases Research and Treatment Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| | | | | | | | | |
Collapse
|
46
|
Zong M, Bruton JD, Grundtman C, Yang H, Li JH, Alexanderson H, Palmblad K, Andersson U, Harris HE, Lundberg IE, Westerblad H. TLR4 as receptor for HMGB1 induced muscle dysfunction in myositis. Ann Rheum Dis 2012; 72:1390-9. [DOI: 10.1136/annrheumdis-2012-202207] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
47
|
de Souza A, Westra J, Limburg P, Bijl M, Kallenberg C. HMGB1 in vascular diseases: Its role in vascular inflammation and atherosclerosis. Autoimmun Rev 2012; 11:909-17. [DOI: 10.1016/j.autrev.2012.03.007] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 03/22/2012] [Indexed: 11/28/2022]
|
48
|
Harlow L, Fernandez I, Soejima M, Ridgway WM, Ascherman DP. Characterization of TLR4-mediated auto-antibody production in a mouse model of histidyl-tRNA synthetase-induced myositis. Innate Immun 2012; 18:876-85. [PMID: 22582345 DOI: 10.1177/1753425912446714] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We have previously shown that intramuscular immunization with a recombinant fragment of murine histidyl-tRNA synthetase (HRS) in the absence of exogenous adjuvant generates Ag-specific, IgG class switched Abs a murine model of myositis. Markedly diminished IgG anti-HRS auto-Ab responses in TLR4 signaling-deficient C3H/HeJ mice indicate that TLR4 is required for auto-Ab formation and/or class switching in this system. Comparative time course assessment of HRS-immunized C3H/HeOuJ (wild type) and C3H/HeJ (TLR4 mutant) mice shows here that despite significant impairment of class switched IgG anti-HRS responses in TLR4-deficient C3H/HeJ mice, production of IgM anti-HRS auto-Abs is relatively preserved-suggesting that TLR4-mediated signals modulate IgG class switching rather than auto-Ab formation in this genetic background. In C57BL/6-derived knockout mice lacking either MyD88 (B6.MyD88(-/-)) or TRIF (B6.TRIF(-/-)) adaptor molecules, immunization studies indicate that TRIF exerts a dominant role in the generation of HRS-specific IgG auto-Abs. Complementing these analyses, in vitro stimulation of unfractionated, as well as T cell-depleted, C3H/HeOuJ splenocytes with recombinant murine HRS reveals that TLR4-mediated generation of class switched auto-Abs can occur independently of T cell help. Overall, these findings support a broader role for TLR4 in the breakdown of immune tolerance and development of autoimmunity.
Collapse
Affiliation(s)
- Lisa Harlow
- Department of Medicine, Division of Rheumatology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | | | |
Collapse
|
49
|
Abstract
HMGB1 is a non-histone nuclear protein that can serve as an alarmin to drive the pathogenesis of inflammatory and autoimmune disease. Although primarily located in the cell nucleus, HMGB1 can translocate to the cytoplasm, as well as the extracellular space, during cell activation and cell death; during activation, HMGB1 can undergo post-translational modifications. The activity of HMGB1 varies with the redox states of the cysteine residues, which are required for binding to TLR4. In addition to stimulating cells directly, HMGB1 can form immunostimulatory complexes with cytokines and other endogenous and exogenous factors. In the synovia of patients with rheumatoid arthritis, as well as animal models of this disease, extranuclear expression of HMGB1 is increased and blockade of HMGB1 expression attenuates disease in animal models. In systemic lupus erythematosus, HMGB1 can be a component of immune complexes containing anti-DNA because of its interaction with DNA. In myositis, expression of HMGB1 is enhanced in inflamed muscle and can perturb muscle function. Together, these findings indicate that HMGB1 might be an important mediator and biomarker in rheumatic diseases as well as a target of new therapy.
Collapse
|
50
|
|