1
|
Neves A, Viveiros L, Venturelli V, Isenberg DA. Where are we now in biologic drugs for myositis? Rheumatology (Oxford) 2024; 63:2938-2947. [PMID: 38321569 DOI: 10.1093/rheumatology/keae096] [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: 10/03/2023] [Revised: 01/10/2024] [Accepted: 01/30/2024] [Indexed: 02/08/2024] Open
Abstract
Idiopathic inflammatory myopathies (IIMs) are a rare and heterogeneous group of chronic autoimmune disorders. Up to 40% of IIM patients have long-term sequelae and significant functional disability. Its management can be challenging and new therapies are badly needed. The small number of cases with diverse presentations and different diagnostic criteria significantly affect clinical trial results. Only IVIG has been internationally approved for IIM patients. Most clinical trials of new biologic therapies have failed to meet their primary endpoints in IIM, with only one biologic drug recommended for refractory IIM treatment (rituximab), although not approved. We review several new emerging biologic drugs, including B cell depletion therapies, abatacept, Janus kinase inhibitors, and aldesleukin. Encouragingly, some phase II randomized controlled trials have evaluated the efficacy and safety of new biologics in IIM, demonstrating an improvement in clinical and laboratory measures.
Collapse
Affiliation(s)
- Ana Neves
- Internal Medicine Department, Centro Hospitalar Universitário de São João, Oporto, Portugal
| | - Luísa Viveiros
- Internal Medicine Department, Centro Hospitalar Universitário de Santo António, Oporto, Portugal
| | - Veronica Venturelli
- Rheumatology Unit, Department of Medical Sciences, Università degli Studi di Ferrara, Azienda Ospedaliero-Universitaria S. Anna, Cona, Italy
| | - David A Isenberg
- Centre for Rheumatology, Department of Medicine, University College London, London, UK
| |
Collapse
|
2
|
Nishimura A, Nelke C, Huber M, Mensch A, Roth A, Oberwittler C, Zimmerlein B, Krämer HH, Neuen-Jacob E, Stenzel W, Müller-Ladner U, Ruck T, Schänzer A. Differentiating idiopathic inflammatory myopathies by automated morphometric analysis of MHC-1, MHC-2 and ICAM-1 in muscle tissue. Neuropathol Appl Neurobiol 2024; 50:e12998. [PMID: 39030945 DOI: 10.1111/nan.12998] [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: 04/26/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/22/2024]
Abstract
AIMS Diagnosis of idiopathic inflammatory myopathies (IIM) is based on morphological characteristics and the evaluation of disease-related proteins. However, although broadly applied, substantial bias is imposed by the respective methods, observers and individual staining approaches. We aimed to quantify the protein levels of major histocompatibility complex (MHC)-1, (MHC)-2 and intercellular adhesion molecule (ICAM)-1 using an automated morphometric method to mitigate bias. METHODS Double immunofluorescence staining was performed on whole muscle sections to study differences in protein expression in myofibre and endomysial vessels. We analysed all IIM subtypes including dermatomyositis (DM), anti-synthetase syndrome (ASyS), inclusion body myositis (IBM), immune-mediated-necrotising myopathy (IMNM), dysferlinopathy (DYSF), SARS-CoV-2 infection and vaccination-associated myopathy. Biopsies with neurogenic atrophy (NA) and normal morphology served as controls. Bulk RNA-Sequencing (RNA-Seq) was performed on a subset of samples. RESULTS Our study highlights the significance of MHC-1, MHC-2 and ICAM-1 in diagnosing IIM subtypes and reveals distinct immunological profiles. RNASeq confirmed the precision of our method and identified specific gene pathways in the disease subtypes. Notably, ASyS, DM and SARS-CoV-2-associated myopathy showed increased ICAM-1 expression in the endomysial capillaries, indicating ICAM-1-associated vascular activation in these conditions. In addition, ICAM-1 showed high discrimination between different subgroups with high sensitivity and specificity. CONCLUSIONS Automated morphometric analysis provides precise quantitative data on immune-associated proteins that can be integrated into our pathophysiological understanding of IIM. Further, ICAM-1 holds diagnostic value for the detection of IIM pathology.
Collapse
Affiliation(s)
- Anna Nishimura
- Institute of Neuropathology, Justus-Liebig University Giessen, Germany
| | - Christopher Nelke
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Melanie Huber
- Department of Rheumatology and Clinical Immunology, Campus Kerckhoff, Justus-Liebig-University, Giessen, Germany
| | - Alexander Mensch
- Department of Neurology, University Medicine Halle, Halle (Saale), Germany
| | - Angela Roth
- Institute of Neuropathology, Justus-Liebig University Giessen, Germany
| | | | | | - Heidrun H Krämer
- Department of Neurology, Justus-Liebig-University, Giessen, Germany
- Translational Neuroscience Network Giessen (TNNG), Justus Liebig University Giessen, Giessen, Germany
| | - Eva Neuen-Jacob
- Institute of Neuropathology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Werner Stenzel
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ulf Müller-Ladner
- Department of Rheumatology and Clinical Immunology, Campus Kerckhoff, Justus-Liebig-University, Giessen, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Anne Schänzer
- Institute of Neuropathology, Justus-Liebig University Giessen, Germany
- Translational Neuroscience Network Giessen (TNNG), Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
3
|
Robinson TP, Hamidi T, Counts B, Guttridge DC, Ostrowski MC, Zimmers TA, Koniaris LG. The impact of inflammation and acute phase activation in cancer cachexia. Front Immunol 2023; 14:1207746. [PMID: 38022578 PMCID: PMC10644737 DOI: 10.3389/fimmu.2023.1207746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
The development of cachexia in the setting of cancer or other chronic diseases is a significant detriment for patients. Cachexia is associated with a decreased ability to tolerate therapies, reduction in ambulation, reduced quality of life, and increased mortality. Cachexia appears intricately linked to the activation of the acute phase response and is a drain on metabolic resources. Work has begun to focus on the important inflammatory factors associated with the acute phase response and their role in the immune activation of cachexia. Furthermore, data supporting the liver, lung, skeletal muscle, and tumor as all playing a role in activation of the acute phase are emerging. Although the acute phase is increasingly being recognized as being involved in cachexia, work in understanding underlying mechanisms of cachexia associated with the acute phase response remains an active area of investigation and still lack a holistic understanding and a clear causal link. Studies to date are largely correlative in nature, nonetheless suggesting the possibility for a role for various acute phase reactants. Herein, we examine the current literature regarding the acute phase response proteins, the evidence these proteins play in the promotion and exacerbation of cachexia, and current evidence of a therapeutic potential for patients.
Collapse
Affiliation(s)
- Tyler P. Robinson
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Tewfik Hamidi
- Department of Surgery, Oregon Health Sciences University, Portland, OR, United States
| | - Brittany Counts
- Department of Surgery, Oregon Health Sciences University, Portland, OR, United States
| | - Denis C. Guttridge
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Michael C. Ostrowski
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Teresa A. Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Surgery, Oregon Health Sciences University, Portland, OR, United States
| | - Leonidas G. Koniaris
- Department of Surgery, Oregon Health Sciences University, Portland, OR, United States
| |
Collapse
|
4
|
Grazzini S, Rizzo C, Conticini E, D'Alessandro R, La Barbera L, D'Alessandro M, Falsetti P, Bargagli E, Guggino G, Cantarini L, Frediani B. The role of bDMARDs in idiopathic inflammatory myopathies: A systematic literature review. Clin Exp Rheumatol 2023; 22:103264. [PMID: 36549353 DOI: 10.1016/j.autrev.2022.103264] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022]
Abstract
Idiopathic inflammatory myopathies (IIM) are a group of different conditions typically affecting striate muscle, lung, joints, skin and gastrointestinal tract. Treatment typically relies on glucocorticoids and synthetic immunosuppressants, but the occurrence of refractory, difficult to treat, manifestations, may require more aggressive treatment, borrowed from other autoimmune diseases, including biologic disease modifying drugs (bDMARDs). In this regard, we conducted a systemic literature review in order to depict the current evidence about the use of bDMARDs in IIM. A total of 78 papers, published during the last 21 years, were retrieved. The majority of patients was treated with TNF-α inhibitors, whose effectiveness was assessed particularly in recalcitrant striate muscle, skin and joints involvement. Rituximab, whose evidence is supported by a large number of real-life studies and trials, seems to be an excellent option in case of ILD and anti-synthetase syndrome, while Tocilizumab, despite not meeting primary and secondary endpoints in a recently published clinical trial, proved its effectiveness in rapidly progressing ILD. Similarly, Abatacept, studied in a phase IIb clinical trial with conflicting evidence, was reported to be effective in some case reports of refractory dermatomyositis. Less data exist for anti-IL1 and anti-IL23 agents, which were employed particularly for inclusion body myositis and severe skin disease, respectively. This study provides an organ-focused assessment of bDMARDs in IIM, which display encouraging results in the treatment of refractory subsets of disease.
Collapse
Affiliation(s)
- Silvia Grazzini
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Chiara Rizzo
- Rheumatology Section, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Edoardo Conticini
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy.
| | - Roberto D'Alessandro
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Lidia La Barbera
- Rheumatology Section, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Miriana D'Alessandro
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences and Neurosciences, University of Siena, Siena, Italy
| | - Paolo Falsetti
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Elena Bargagli
- Respiratory Diseases Unit, Department of Medical and Surgical Sciences and Neurosciences, University of Siena, Siena, Italy
| | - Giuliana Guggino
- Rheumatology Section, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Luca Cantarini
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Bruno Frediani
- Rheumatology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| |
Collapse
|
5
|
Wang Y, Zheng Y, Zhao Y, Liu Y, Zhang W, Yu M, Xie Z, Hao H, Gao F, Zhang W, Wang Z, Yuan Y. Comparison of cytokine/chemokine profiles between dermatomyositis and anti-synthetase syndrome. Front Neurol 2022; 13:1042580. [PMID: 36570445 PMCID: PMC9772994 DOI: 10.3389/fneur.2022.1042580] [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: 09/12/2022] [Accepted: 11/14/2022] [Indexed: 12/13/2022] Open
Abstract
Objectives Dermatomyositis (DM) and anti-synthetase syndrome (ASS) are autoimmune diseases with multisystem involvement. Despite sharing some clinical and myopathological features, these are two diseases with different pathogeneses and prognoses. We aimed to clarify and compare cytokine/chemokine profiles in both disorders, which may help in the differential diagnosis. Materials and methods We collected clinical data and serum samples of consecutive patients with DM and ASS. Quantibody® Human Inflammation Array 3 for cytokines/chemokines was performed in the serum of all participants. Receiver operating characteristic analysis with the area under the curve and Youden's index were performed. Results Eight newly diagnosed and treatment-naïve patients with DM, nine newly diagnosed and treatment-naïve patients with ASS, and 14 healthy controls were enrolled. Serum C-C motif chemokine ligand (CCL) 2, CCL4, C-X-C motif chemokine ligand (CXCL) 13, and tumor necrosis factor receptor 2 (TNFR2) were increased in patients with both DM and ASS. Serum interleukin (IL)-1 receptor type 1 (IL-1ra), IL-1b, CCL1, CXCL11, and CCL3 were modulated in patients with DM only, and IL-8, CXCL9, and tissue inhibitors of metalloproteinases-1 (TIMP-1) in patients with ASS only. Serum CCL2, CXCL13, and TNFR2 accurately distinguished patients with DM and ASS from healthy controls, as shown by the area under the curve >0.80. Moreover, receiver operating characteristic analysis showed that, as biomarkers for discrimination between DM and ASS, the combination of IL-1ra and TIMP-1, had an area under the curve of 0.944, a sensitivity of 87.5%, and a specificity of 88.9%. Conclusion Our study demonstrated that serum levels of cytokines/chemokines showed a different pattern in newly diagnosed patients with DM and ASS, in which serum IL-1ra and TIMP-1 could be used to distinguish between the two diseases.
Collapse
Affiliation(s)
- Yikang Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yiming Zheng
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yawen Zhao
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Yilin Liu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Wenhao Zhang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Meng Yu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Zhiying Xie
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Hongjun Hao
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Feng Gao
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Wei Zhang
- Department of Neurology, Peking University First Hospital, Beijing, China,Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, China,Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China,Beijing Key Laboratory of Neurovascular Disease Discovery, Beijing, China,*Correspondence: Yun Yuan
| |
Collapse
|
6
|
Jia Q, Hao RJL, Lu XJ, Sun SQ, Shao JJ, Su X, Huang QF. Identification of hub biomarkers and immune cell infiltration characteristics of polymyositis by bioinformatics analysis. Front Immunol 2022; 13:1002500. [PMID: 36225941 PMCID: PMC9548705 DOI: 10.3389/fimmu.2022.1002500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/09/2022] [Indexed: 11/13/2022] Open
Abstract
Background Polymyositis (PM) is an acquirable muscle disease with proximal muscle involvement of the extremities as the main manifestation; it is a category of idiopathic inflammatory myopathy. This study aimed to identify the key biomarkers of PM, while elucidating PM-associated immune cell infiltration and immune-related pathways. Methods The gene microarray data related to PM were downloaded from the Gene Expression Omnibus database. The analyses using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes, gene set enrichment analysis (GSEA), and protein-protein interaction (PPI) networks were performed on differentially expressed genes (DEGs). The hub genes of PM were identified using weighted gene co-expression network analysis (WGCNA) and least absolute shrinkage and selection operator (LASSO) algorithm, and the diagnostic accuracy of hub markers for PM was assessed using the receiver operating characteristic curve. In addition, the level of infiltration of 28 immune cells in PM and their interrelationship with hub genes were analyzed using single-sample GSEA. Results A total of 420 DEGs were identified. The biological functions and signaling pathways closely associated with PM were inflammatory and immune processes. A series of four expression modules were obtained by WGCNA analysis, with the turquoise module having the highest correlation with PM; 196 crossover genes were obtained by combining DEGs. Subsequently, six hub genes were finally identified as the potential biomarkers of PM using LASSO algorithm and validation set verification analysis. In the immune cell infiltration analysis, the infiltration of T lymphocytes and subpopulations, dendritic cells, macrophages, and natural killer cells was more significant in the PM. Conclusion We identified the hub genes closely related to PM using WGCNA combined with LASSO algorithm, which helped clarify the molecular mechanism of PM development and might have great significance for finding new immunotherapeutic targets, and disease prevention and treatment.
Collapse
Affiliation(s)
- Qi Jia
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Rui-Jin-Lin Hao
- Medical School of Nantong University, Nantong, China
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiao-Jian Lu
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Shu-Qing Sun
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Jun-Jie Shao
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Xing Su
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
- *Correspondence: Qing-Feng Huang, ; Xing Su,
| | - Qing-Feng Huang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
- *Correspondence: Qing-Feng Huang, ; Xing Su,
| |
Collapse
|
7
|
Pathophysiological Mechanisms and Treatment of Dermatomyositis and Immune Mediated Necrotizing Myopathies: A Focused Review. Int J Mol Sci 2022; 23:ijms23084301. [PMID: 35457124 PMCID: PMC9030619 DOI: 10.3390/ijms23084301] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/31/2022] [Accepted: 04/05/2022] [Indexed: 12/15/2022] Open
Abstract
Idiopathic inflammatory myopathies (IIM), collectively known as myositis, are a composite group of rare autoimmune diseases affecting mostly skeletal muscle, although other organs or tissues may also be involved. The main clinical feature of myositis is subacute, progressive, symmetrical muscle weakness in the proximal arms and legs, whereas subtypes of myositis may also present with extramuscular features, such as skin involvement, arthritis or interstitial lung disease (ILD). Established subgroups of IIM include dermatomyositis (DM), immune-mediated necrotizing myopathy (IMNM), anti-synthetase syndrome (ASyS), overlap myositis (OM) and inclusion body myositis (IBM). Although these subgroups have overlapping clinical features, the widespread variation in the clinical manifestations of IIM suggests different pathophysiological mechanisms. Various components of the immune system are known to be important immunopathogenic pathways in IIM, although the exact pathophysiological mechanisms causing the muscle damage remain unknown. Current treatment, which consists of glucocorticoids and other immunosuppressive or immunomodulating agents, often fails to achieve a sustained beneficial response and is associated with various adverse effects. New therapeutic targets have been identified that may improve outcomes in patients with IIM. A better understanding of the overlapping and diverging pathophysiological mechanisms of the major subgroups of myositis is needed to optimize treatment. The aim of this review is to report on recent advancements regarding DM and IMNM.
Collapse
|
8
|
Patwardhan A, Spencer CH. Biologics in refractory idiopathic inflammatory myositis (IIM): What experience in juvenile vs adult myositis tells us about the use of biologics in pediatric IIM. Mod Rheumatol 2021; 31:933-948. [PMID: 33499694 DOI: 10.1080/14397595.2021.1881027] [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] [Indexed: 10/22/2022]
Abstract
Juvenile dermatomyositis (JDM) is an extremely heterogeneous orphan disease with limited amount of dedicated research on the subject matter. Recent research suggests that JDM may not just be the classic antibody driven complements mediated microangiopathy as was thought to be in the past. The etiopathogenesis of JDM also involves inappropriate stimulation of innate immune system followed by dysregulation of the adaptive immune response through dendritic cells. Many variable immune factors such as genetics, major histocompatibility complex expressions, immunohistochemical variabilities, and diversity in specific and associated autoantibodies may make individual IIM and JDM cases unique. The diversity in IIM and JDM also explains individual variability in response to specific therapies. Classifying and matching the right patients to the right treatment is crucial to the successful treatment of these patients with better outcomes. Sub-type specific biologic therapy may be the best current treatment that can match the patient to the best treatment options. A PubMed search was performed to find all the available cases of refractory myositis patients treated with biologics up to July 2020. Using this search this article reviews all the current biologic treatment options and experiences for both adults and children in the context of recent basic science to assist pediatric rheumatologists in choosing the optimal biologic therapy for a child with recalcitrant JDM.
Collapse
Affiliation(s)
| | - Charles H Spencer
- University of Mississippi Medical Center, Batson Children's Hospital, Jackson, MS, USA
| |
Collapse
|
9
|
Cavalli G, Colafrancesco S, Emmi G, Imazio M, Lopalco G, Maggio MC, Sota J, Dinarello CA. Interleukin 1α: a comprehensive review on the role of IL-1α in the pathogenesis and treatment of autoimmune and inflammatory diseases. Autoimmun Rev 2021; 20:102763. [PMID: 33482337 DOI: 10.1016/j.autrev.2021.102763] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 12/08/2020] [Indexed: 12/17/2022]
Abstract
The interleukin (IL)-1 family member IL-1α is a ubiquitous and pivotal pro-inflammatory cytokine. The IL-1α precursor is constitutively present in nearly all cell types in health, but is released upon necrotic cell death as a bioactive mediator. IL-1α is also expressed by infiltrating myeloid cells within injured tissues. The cytokine binds the IL-1 receptor 1 (IL-1R1), as does IL-1β, and induces the same pro-inflammatory effects. Being a bioactive precursor released upon tissue damage and necrotic cell death, IL-1α is central to the pathogenesis of numerous conditions characterized by organ or tissue inflammation. These include conditions affecting the lung and respiratory tract, dermatoses and inflammatory skin disorders, systemic sclerosis, myocarditis, pericarditis, myocardial infarction, coronary artery disease, inflammatory thrombosis, as well as complex multifactorial conditions such as COVID-19, vasculitis and Kawasaki disease, Behcet's syndrome, Sjogren Syndrome, and cancer. This review illustrates the clinical relevance of IL-1α to the pathogenesis of inflammatory diseases, as well as the rationale for the targeted inhibition of this cytokine for treatment of these conditions. Three biologics are available to reduce the activities of IL-1α; the monoclonal antibody bermekimab, the IL-1 soluble receptor rilonacept, and the IL-1 receptor antagonist anakinra. These advances in mechanistic understanding and therapeutic management make it incumbent on physicians to be aware of IL-1α and of the opportunity for therapeutic inhibition of this cytokine in a broad spectrum of diseases.
Collapse
Affiliation(s)
- Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy, and Rare Diseases, IRCCS San Raffaele Scientific Institute; Vita-Salute San Raffaele University, Milan, Italy.
| | - Serena Colafrancesco
- Dipartimento of Clinical Sciences (Internal Medicine, Anesthesia and Resuscitation, and Cardiology), Rheumatology Unit, Sapienza University of Rome, Rome, Italy
| | - Giacomo Emmi
- Department of Experimental and Clinical Medicine, Careggi University Hospital, Firenze, Italy
| | - Massimo Imazio
- University Division of Cardiology, Cardiovascular and Throracic Department, AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - Giuseppe Lopalco
- Department of Emergency and Organ Transplantation, Rheumatology Unit, University of Bari, Bari, Italy
| | - Maria Cristina Maggio
- Department of Health Promotion, Maternal and Infantile Care, Department of Internal Medicine and Medical Specialties "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Jurgen Sota
- Research Center of Systemic Autoinflammatory Diseases and Behçet's Disease Clinic, Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA.
| |
Collapse
|
10
|
Yang CA, Chiang BL. Inflammasomes and Childhood Autoimmune Diseases: A Review of Current Knowledge. Clin Rev Allergy Immunol 2020; 61:156-170. [PMID: 33236284 PMCID: PMC7685913 DOI: 10.1007/s12016-020-08825-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2020] [Indexed: 02/08/2023]
Abstract
Inflammasomes are multiprotein complexes capable of sensing pathogen-associated molecular patterns (PAMPs), danger-associated molecular patterns (DAMPs), and cellular perturbations. Upon stimulation, the inflammasomes activate the production of the pro-inflammatory cytokines IL-1β and IL-18 and induce gasdermin D-mediated pyroptosis. Dysregulated inflammasome signaling could lead to hyperinflammation in response to environmental triggers, thus contributing to the pathogenesis of childhood autoimmune/autoinflammatory diseases. In this review, we group childhood rheumatic diseases into the autoinflammation to autoimmunity spectrum and discuss about the involvement of inflammasomes in disease mechanisms. Genetic mutations in inflammasome components cause monogenic autoinflammatory diseases, while inflammasome-related genetic variants have been implicated in polygenic childhood rheumatic diseases. We highlight the reported associations of inflammasome signaling-related genetic polymorphisms/protein levels with pediatric autoimmune disease susceptibility and disease course. Furthermore, we discuss about the use of IL-1 receptor antagonist as an adjunctive therapy in several childhood autoimmune diseases, including macrophage activation syndrome (MAS) and multisystem inflammatory syndrome in children (MIS-C) related to COVID-19. A comprehensive multi-cohort comparison on inflammasome gene expression profile in different pediatric rheumatic diseases is needed to identify patient subsets that might benefit from the adjunctive therapy of IL-1β inhibitors.
Collapse
Affiliation(s)
- Chin-An Yang
- Division of Laboratory Medicine and Division of Pediatrics, China Medical University Hsinchu Hospital, Jubei, Hsinchu, Taiwan
- College of Medicine, China Medical University, Taichung, Taiwan
| | - Bor-Luen Chiang
- Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
11
|
Antisynthetase syndrome and cardiac involvement: a rare association. Joint Bone Spine 2019; 86:517-518. [DOI: 10.1016/j.jbspin.2018.09.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 09/26/2018] [Indexed: 11/22/2022]
|
12
|
Meudec L, Ottaviani S. Reply to the comment of Sartorelli et al. “Diagnostic approach and novel therapeutic option for cardiac inflammatory disorders”. Joint Bone Spine 2019; 86:409. [DOI: 10.1016/j.jbspin.2019.01.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 01/30/2019] [Indexed: 10/27/2022]
|
13
|
Abstract
Pain is a complex subjective experience, associated with neurovegetative, affective and cognitive rapid changes. Biological, psychosocial and contextual factors may contribute. Chronic inflammation, of whatever cause, is the leading contributor to chronic pain. The mast cell directs both the inflammatory process and the shift to chronic pain, mediating through the production of Nerve Growth Factor (NGF) and other neurotrophic molecules. Women, in the fertile age, are biologically more vulnerable to chronic inflammation, as fluctuations of estrogens are agonist factors of mast cells degranulation, mostly in the premenstrual phase. Pain is defined as “nociceptive” when it indicates an ongoing damage; “neuropathic” when it becomes a disease per se. Chronic Pelvic Pain (CPP) indicates an invalidating, persistent or recurrent pelvic pain, persisting for more than 6 months. CPP is the main complaint of 10–15% of gynaecologic consultations, leading to 40% of diagnostic laparoscopies and 15% of hysterectomies. Comorbidity, i.e. the coexistence of pathologies and painful syndromes in different pelvic organs, is another common feature. Cystitis, vulvar vestibulitis, endometriosis, irritable bowel syndrome all play an important role and contribute to identifying the hyperactive mast cell and related chronic inflammation as the common pathophysiologic factor. The paper reviews nociception characteristics, the emerging role of mast cells, the pathophysiology of comorbidity, biological, psychosexual and contextual predictors, and stresses the need to move from a “hyperspecialistic” perspective to a multisystemic reading of CPP, with special attention to the urologic perspective.
Collapse
Affiliation(s)
- A. Graziottin
- Centro di Ginecologia e Sessuologia Medica, “H. San Raffaele Resnati”, Milano
| |
Collapse
|
14
|
Song A, Zhu L, Gorantla G, Berdysz O, Amici SA, Guerau-de-Arellano M, Madalena KM, Lerch JK, Liu X, Quan N. Salient type 1 interleukin 1 receptor expression in peripheral non-immune cells. Sci Rep 2018; 8:723. [PMID: 29335509 PMCID: PMC5768710 DOI: 10.1038/s41598-018-19248-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 12/21/2017] [Indexed: 12/26/2022] Open
Abstract
Interleukin 1 is a pleiotropic cytokine that mediates diverse functions through its receptor, type I interleukin 1 receptor (IL-1R1). Most previous studies have focused on the expression and function of IL-1R1 in immune cells. Here we performed a comprehensive mapping of IL-1R1 distribution in multiple peripheral tissues using our IL-1R1 reporter (IL-1R1GR/GR) mice. This method yielded the highest sensitivity of in situ detection of IL-1R1 mRNA and protein. Besides validating previously reported IL-1R1 expression in the endocrine tissues including pituitary and pancreas, our results refuted previously reported exclusive IL-1R1 expression in neurons of the spinal cord dorsal horn and dorsal root ganglia (DRG). Instead, IL-1R1 expression was detected in endothelial cells within DRG, spinal cord, pancreas, colon, muscles and many immune organs. In addition, gp38+ fibroblastic reticular cells (FRCs), rather than tissue macrophages or other immune cells, were found to express high levels of IL-1R1 in colon and many immune organs. A functional test of spleen FRCs showed that they responded rapidly to systemic IL-1β stimulation in vivo. Taken together, this study provides a rigorous re-examination of IL-1R1 expression in peripheral tissues and reveals tissue FRCs as a previously unappreciated novel high IL-1R1-expressing cell type in peripheral IL-1 signaling.
Collapse
Affiliation(s)
- Anping Song
- Department of Oncolgy, Tongji Hospital, Huazhong University of Science and Technology Tongji Medical College, Wuhan, Hubei, 430030, P. R. China
| | - Ling Zhu
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.,West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
| | - Gowthami Gorantla
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Olimpia Berdysz
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Stephanie A Amici
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.,School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Mireia Guerau-de-Arellano
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA.,School of Health and Rehabilitation Sciences, Division of Medical Laboratory Science, College of Medicine, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA.,Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA.,Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA
| | - Kathryn M Madalena
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA.,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, 43210, USA
| | - Jessica K Lerch
- Department of Neuroscience, The Ohio State University, Columbus, OH, 43210, USA.,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, 43210, USA
| | - Xiaoyu Liu
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA. .,Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, 43210, USA. .,Institute for Behavioral Medicine Research, 460 Medical Center Drive, Columbus, OH, 43210, USA.
| | - Ning Quan
- Institute for Behavioral Medicine Research, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA. .,Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, 43210, USA. .,Institute for Behavioral Medicine Research, 460 Medical Center Drive, Columbus, OH, 43210, USA.
| |
Collapse
|
15
|
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
|
16
|
López De Padilla CM, Crowson CS, Hein MS, Pendegraft RS, Strausbauch MA, Niewold TB, Ernste FC, Peterson E, Baechler EC, Reed AM. Gene Expression Profiling in Blood and Affected Muscle Tissues Reveals Differential Activation Pathways in Patients with New-onset Juvenile and Adult Dermatomyositis. J Rheumatol 2016; 44:117-124. [PMID: 27803134 DOI: 10.3899/jrheum.160293] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2016] [Indexed: 01/10/2023]
Abstract
OBJECTIVE To identify shared and differential molecular pathways in blood and affected muscle between adult dermatomyositis (DM) and juvenile DM, and their association with clinical disease activity measures. METHODS Gene expression of transcription factors and cytokines involved in differentiation and effector function of T cell subsets, regulatory T cells and follicular Th cells, were analyzed in the blood from 21 newly diagnosed adult and 26 juvenile DM subjects and in 15 muscle specimens (7 adult and 8 juvenile DM) using a custom RT2 Profiler PCR Array. Disease activity was determined and measured by established disease activity tools. RESULTS The most prominent finding was the higher blood expression of Th17-related cytokines [retinoic acid-related orphan receptor-γ, interferon regulatory factor 4, interleukin (IL)-23A, IL-6, IL-17F, and IL-21] in juvenile DM at baseline. In contrast, adult patients with DM showed increased blood levels of STAT3 and BCL6 compared with juvenile DM. In muscle, GATA3, IL-13, and STAT5B were found at higher levels in juvenile patients with DM compared with adult DM. Among 25 patients (11 adult and 14 juvenile DM) who had blood samples at baseline and at 6 months, increased expression of IL-1β, STAT3, STAT6, STAT5B, and BCL6 was associated with an improvement in global extramuscular disease activity. CONCLUSION We observed differences in gene expression profiling in blood and muscle between new-onset adult and juvenile DM. Cytokine expression in the blood of juvenile patients with new-onset DM was dominated by Th17-related cytokines compared with adult patients with DM. This may reflect the activation of different Th pathways between muscle and blood.
Collapse
Affiliation(s)
- Consuelo M López De Padilla
- From the Division of Rheumatology, Department of Pediatrics, Mayo Clinic; Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; Department of Surgical Research, Mayo Clinic; Division of Rheumatology and Department of Immunology, Mayo Clinic; Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine, Rochester; Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center, Durham, North Carolina, USA.,C.M. López De Padilla, MD, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; C.S. Crowson, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.S. Hein, BS, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; R.S. Pendegraft, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.A. Strausbauch, MS, Department of Surgical Research, Mayo Clinic; T.B. Niewold, MD, Division of Rheumatology and Department of Immunology, Mayo Clinic; F.C. Ernste, MD, Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine; E. Peterson, MD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; E.C. Baechler, PhD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; A.M. Reed, MD, Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center
| | - Cynthia S Crowson
- From the Division of Rheumatology, Department of Pediatrics, Mayo Clinic; Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; Department of Surgical Research, Mayo Clinic; Division of Rheumatology and Department of Immunology, Mayo Clinic; Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine, Rochester; Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center, Durham, North Carolina, USA.,C.M. López De Padilla, MD, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; C.S. Crowson, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.S. Hein, BS, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; R.S. Pendegraft, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.A. Strausbauch, MS, Department of Surgical Research, Mayo Clinic; T.B. Niewold, MD, Division of Rheumatology and Department of Immunology, Mayo Clinic; F.C. Ernste, MD, Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine; E. Peterson, MD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; E.C. Baechler, PhD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; A.M. Reed, MD, Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center
| | - Molly S Hein
- From the Division of Rheumatology, Department of Pediatrics, Mayo Clinic; Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; Department of Surgical Research, Mayo Clinic; Division of Rheumatology and Department of Immunology, Mayo Clinic; Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine, Rochester; Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center, Durham, North Carolina, USA.,C.M. López De Padilla, MD, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; C.S. Crowson, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.S. Hein, BS, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; R.S. Pendegraft, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.A. Strausbauch, MS, Department of Surgical Research, Mayo Clinic; T.B. Niewold, MD, Division of Rheumatology and Department of Immunology, Mayo Clinic; F.C. Ernste, MD, Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine; E. Peterson, MD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; E.C. Baechler, PhD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; A.M. Reed, MD, Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center
| | - Richard S Pendegraft
- From the Division of Rheumatology, Department of Pediatrics, Mayo Clinic; Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; Department of Surgical Research, Mayo Clinic; Division of Rheumatology and Department of Immunology, Mayo Clinic; Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine, Rochester; Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center, Durham, North Carolina, USA.,C.M. López De Padilla, MD, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; C.S. Crowson, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.S. Hein, BS, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; R.S. Pendegraft, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.A. Strausbauch, MS, Department of Surgical Research, Mayo Clinic; T.B. Niewold, MD, Division of Rheumatology and Department of Immunology, Mayo Clinic; F.C. Ernste, MD, Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine; E. Peterson, MD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; E.C. Baechler, PhD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; A.M. Reed, MD, Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center
| | - Michael A Strausbauch
- From the Division of Rheumatology, Department of Pediatrics, Mayo Clinic; Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; Department of Surgical Research, Mayo Clinic; Division of Rheumatology and Department of Immunology, Mayo Clinic; Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine, Rochester; Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center, Durham, North Carolina, USA.,C.M. López De Padilla, MD, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; C.S. Crowson, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.S. Hein, BS, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; R.S. Pendegraft, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.A. Strausbauch, MS, Department of Surgical Research, Mayo Clinic; T.B. Niewold, MD, Division of Rheumatology and Department of Immunology, Mayo Clinic; F.C. Ernste, MD, Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine; E. Peterson, MD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; E.C. Baechler, PhD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; A.M. Reed, MD, Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center
| | - Timothy B Niewold
- From the Division of Rheumatology, Department of Pediatrics, Mayo Clinic; Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; Department of Surgical Research, Mayo Clinic; Division of Rheumatology and Department of Immunology, Mayo Clinic; Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine, Rochester; Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center, Durham, North Carolina, USA.,C.M. López De Padilla, MD, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; C.S. Crowson, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.S. Hein, BS, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; R.S. Pendegraft, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.A. Strausbauch, MS, Department of Surgical Research, Mayo Clinic; T.B. Niewold, MD, Division of Rheumatology and Department of Immunology, Mayo Clinic; F.C. Ernste, MD, Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine; E. Peterson, MD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; E.C. Baechler, PhD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; A.M. Reed, MD, Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center
| | - Floranne C Ernste
- From the Division of Rheumatology, Department of Pediatrics, Mayo Clinic; Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; Department of Surgical Research, Mayo Clinic; Division of Rheumatology and Department of Immunology, Mayo Clinic; Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine, Rochester; Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center, Durham, North Carolina, USA.,C.M. López De Padilla, MD, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; C.S. Crowson, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.S. Hein, BS, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; R.S. Pendegraft, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.A. Strausbauch, MS, Department of Surgical Research, Mayo Clinic; T.B. Niewold, MD, Division of Rheumatology and Department of Immunology, Mayo Clinic; F.C. Ernste, MD, Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine; E. Peterson, MD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; E.C. Baechler, PhD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; A.M. Reed, MD, Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center
| | - Erik Peterson
- From the Division of Rheumatology, Department of Pediatrics, Mayo Clinic; Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; Department of Surgical Research, Mayo Clinic; Division of Rheumatology and Department of Immunology, Mayo Clinic; Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine, Rochester; Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center, Durham, North Carolina, USA.,C.M. López De Padilla, MD, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; C.S. Crowson, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.S. Hein, BS, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; R.S. Pendegraft, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.A. Strausbauch, MS, Department of Surgical Research, Mayo Clinic; T.B. Niewold, MD, Division of Rheumatology and Department of Immunology, Mayo Clinic; F.C. Ernste, MD, Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine; E. Peterson, MD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; E.C. Baechler, PhD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; A.M. Reed, MD, Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center
| | - Emily C Baechler
- From the Division of Rheumatology, Department of Pediatrics, Mayo Clinic; Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; Department of Surgical Research, Mayo Clinic; Division of Rheumatology and Department of Immunology, Mayo Clinic; Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine, Rochester; Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center, Durham, North Carolina, USA.,C.M. López De Padilla, MD, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; C.S. Crowson, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.S. Hein, BS, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; R.S. Pendegraft, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.A. Strausbauch, MS, Department of Surgical Research, Mayo Clinic; T.B. Niewold, MD, Division of Rheumatology and Department of Immunology, Mayo Clinic; F.C. Ernste, MD, Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine; E. Peterson, MD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; E.C. Baechler, PhD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; A.M. Reed, MD, Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center
| | - Ann M Reed
- From the Division of Rheumatology, Department of Pediatrics, Mayo Clinic; Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; Department of Surgical Research, Mayo Clinic; Division of Rheumatology and Department of Immunology, Mayo Clinic; Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine, Rochester; Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center, Durham, North Carolina, USA. .,C.M. López De Padilla, MD, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; C.S. Crowson, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.S. Hein, BS, Division of Rheumatology, Department of Pediatrics, Mayo Clinic; R.S. Pendegraft, MS, Division of Rheumatology and Division of Biomedical Statistics and Informatics, Department of Health Science Research, Mayo Clinic; M.A. Strausbauch, MS, Department of Surgical Research, Mayo Clinic; T.B. Niewold, MD, Division of Rheumatology and Department of Immunology, Mayo Clinic; F.C. Ernste, MD, Division of Rheumatology, Department of Internal Medicine and Department of Pediatrics, Mayo Clinic College of Medicine; E. Peterson, MD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; E.C. Baechler, PhD, Division of Rheumatic and Autoimmune diseases, Department of Medicine, University of Minnesota; A.M. Reed, MD, Department of Pediatrics, Duke Children's Hospital, Duke University Medical Center.
| |
Collapse
|
17
|
Yan W, Fan W, Chen C, Wu Y, Fan Z, Chen J, Chen Z, Chen H. IL-15 up-regulates the MMP-9 expression levels and induces inflammatory infiltration of macrophages in polymyositis through regulating the NF-kB pathway. Gene 2016; 591:137-147. [PMID: 27374114 DOI: 10.1016/j.gene.2016.06.055] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 06/15/2016] [Accepted: 06/28/2016] [Indexed: 02/04/2023]
Abstract
This study was aimed to research the effects of IL-15 on inducing inflammatory infiltration of macrophages in polymyositis (PM) through the NF-kB pathway, and whether IL-15 was able to further regulate MMP-9 expression levels. Prepared PM cells, collected from the patients suffering from PM, were administered to SD rats. Also, a group of healthy SD rats was undergoing the same treatment as the control group. The test animals were treated with either anti-IL-15, IL-15, MMP-9 siRNA or ERK1/2 inhibitor. The blood toxicological parameters creatine kinase (CK) and CD163 were tested by using ELISA and immunohistochemistry assay. In addition, NF-kB expression in macrophages was measured by immunocytochemical assay. To measure the degree of cell infiltration the Transwell assay was performed. Lastly, western blot and zymography were carried out to compare MMP-9 and ERK expression levels between the two groups, both in vivo and in vitro. The results showed that S-CK, IL-15 and IL-15Rα levels increased rapidly after the conventional treatment was introduced to the PM infected SD rats. The PM model establishment and IL-15 treatment significantly increased the expressions of IL-15Rα, MMP-9, p-ERK and p-IKBα. However, the same effect can be suppressed by using anti-IL-15, MMP-9 siRNA or ERK1/2 inhibitor (P < 0.05). In addition, IL-15 is proved to increase cell migration and nucleus expression of NF-kB in the macrophages. IL-15 is able to significantly regulate the inflammatory infiltration of macrophages in PM patients through affecting the NF-kB pathway and MMP-9 expression levels.
Collapse
Affiliation(s)
- Wang Yan
- Department of Neurological Internal Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang, 315000, China
| | - Weinv Fan
- Department of Neurological Internal Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang, 315000, China
| | - Caijing Chen
- Department of Neurological Internal Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang, 315000, China
| | - Yunqin Wu
- Department of Neurological Internal Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang, 315000, China
| | - Zhenyi Fan
- Department of Neurological Internal Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang, 315000, China
| | - Jiaqi Chen
- Department of Neurological Internal Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang, 315000, China
| | - Zhaoying Chen
- Department of Neurological Internal Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang, 315000, China
| | - Huimin Chen
- Department of Neurological Internal Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang, 315000, China.
| |
Collapse
|
18
|
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
|
19
|
Selva-O'Callaghan A, Ramos Casals M, Grau Junyent JM. [Biologic therapy in idiopathic inflammatory myopathy]. Med Clin (Barc) 2014; 143:275-80. [PMID: 24393419 DOI: 10.1016/j.medcli.2013.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 11/06/2013] [Accepted: 11/07/2013] [Indexed: 10/25/2022]
Abstract
The aim of this article is to study the evidence-based knowledge related to the use of biological therapies in patients diagnosed with idiopathic inflammatory myopathy (dermatomyositis, polymyositis and inclusion body myositis). In this review the leading published studies related to the use of biological therapy in patients with myositis are analysed; mainly those with high methodological standards, that means randomized and controlled studies. Methodological drawbacks due to the rarity and heterogeneity of these complex diseases are also addressed. Up to now is not possible to ascertain the biologics as a recommended therapy in patients with myositis, at least based in the current evidence-based knowledge, although it can not be neglected as a therapeutic option in some clinical situations, taking into account the scarce of effective treatments in those patients, especially in refractory myositis. Future studies probably will help to better define the role of biological therapies in patients with idiopathic inflammatory myopathy.
Collapse
Affiliation(s)
- Albert Selva-O'Callaghan
- Servicio de Medicina Interna, Hospital General Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, España.
| | - Manel Ramos Casals
- Laboratorio de Enfermedades Autoinmunes Josep Font, CELLEX-IDIBAPS, Servicio de Enfermedades Autoinmunes, Institut Clínic de Medicina i Dermatologia (ICMiD), Hospital Clínic, Barcelona, España
| | - Josep M Grau Junyent
- Grup de Recerca Muscular, Hospital Clínic, Institut D'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS) y Fundació CELLEX, Universitat de Barcelona, Barcelona, España
| |
Collapse
|
20
|
Friedrich O, Yi B, Edwards JN, Reischl B, Wirth-Hücking A, Buttgereit A, Lang R, Weber C, Polyak F, Liu I, von Wegner F, Cully TR, Lee A, Most P, Völkers M. IL-1α reversibly inhibits skeletal muscle ryanodine receptor. a novel mechanism for critical illness myopathy? Am J Respir Cell Mol Biol 2014; 50:1096-106. [PMID: 24400695 DOI: 10.1165/rcmb.2013-0059oc] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Critical illness myopathies in patients with sepsis or sustained mechanical ventilation prolong intensive care treatment and threaten both patients and health budgets; no specific therapy is available. Underlying pathophysiological mechanisms are still patchy. We characterized IL-1α action on muscle performance in "skinned" muscle fibers using force transducers and confocal Ca(2+) fluorescence microscopy for force/Ca(2+) transients and Ca(2+) sparks. Association of IL-1α with sarcoplasmic reticulum (SR) release channel, ryanodine receptor (RyR) 1, was investigated with coimmunoprecipitation and confocal immunofluorescence colocalization. Membrane integrity was studied in single, intact fibers challenged with IL-1α. IL-1α reversibly stabilized Mg(2+) inhibition of Ca(2+) release. Low Mg(2+)-induced force and Ca(2+) transients were reversibly abolished by IL-1α. At normal Mg(2+), IL-1α reversibly increased caffeine-induced force and Ca(2+) transients. IL-1α reduced SR Ca(2+) leak via RyR1, as judged by (1) increased SR Ca(2+) retention, (2) increased IL-1α force transients being reproduced by 25 μM tetracaine, and (3) reduced Ca(2+) spark frequencies by IL-1α or tetracaine. Coimmunoprecipitation confirmed RyR1/IL-1 association. RyR1/IL-1 immunofluorescence patterns perfectly colocalized. Long-term, 8-hour IL-1α challenge of intact muscle fibers compromised membrane integrity in approximately 50% of fibers, and confirmed intracellular IL-1α deposition. IL-1α exerts a novel, specific, and reversible interaction mechanism with the skeletal muscle RyR1 macromolecular release complex without the need to act via its membrane IL-1 receptor, as IL-1R membrane expression levels were not detectable in Western blots or immunostaining of single fibers. We present a potential explanation of how the inflammatory mediator, IL-1α, may contribute to muscle weakness in critical illness.
Collapse
Affiliation(s)
- Oliver Friedrich
- 1 Institute of Medical Biotechnology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Shinjo SK, de Souza FHC, de Moraes JCB. Dermatomyositis and polymyositis: from immunopathology to immunotherapy (immunobiologics). REVISTA BRASILEIRA DE REUMATOLOGIA 2014; 53:101-10. [PMID: 23588520 DOI: 10.1016/s2255-5021(13)70010-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 11/26/2012] [Indexed: 11/19/2022] Open
Abstract
Idiopathic inflammatory myopathies (IIM), which include dermatomyositis (DM) and polymyositis (PM), are chronic systemic diseases associated with high morbidity and functional disability. Current treatment is based on the use of glucocorticoids and immunosuppressive drugs, but a considerable number of patients is refractory to traditional therapy. That has led to the attempted use of biologics based on the physiopathogenesis of IIM. From the immunopathological viewpoint, PM and DM differ: the former is more related to cellular immunity, while the latter, to humoral immunity. In both, however, elevated concentrations of proinflammatory interleukins (TNF, IL-1, IL-6) and increased expression of molecules related to costimulation of T lymphocytes have been described; thus, the use of biologics in those conditions seems reasonable. Considering the biologics available, open-label studies are scarce, comprising mainly case reports and series. TNF blockers have yielded conflicting results, with no evidence of good response to treatment. The anti-CD20 therapy has the most promising results. Data on T lymphocyte costimulation blockade and anti-IL-6 therapy are extremely scarce, preventing any consideration. Thus, the use of biologics in IIM still remains an unconquered frontier. Biologics may have an important role in the management of IIM refractory to conventional therapy, but further prospective studies based on objective parameters of response to treatment are needed. So far, anti-CD20 therapy seems to be the most promising treatment for refractory IIM.
Collapse
Affiliation(s)
- Samuel Katsuyuki Shinjo
- Service of Rheumatology,, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, Brazil.
| | | | | |
Collapse
|
22
|
Bodoki L, Nagy-Vincze M, Griger Z, Péter A, András C, Dankó K. [Biological therapy in idiopathic inflammatory myopathies]. Orv Hetil 2014; 155:3-10. [PMID: 24379090 DOI: 10.1556/oh.2014.29787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Idiopathic inflammatory myopathies are systemic, immune-mediated diseases characterized by proximal, symmetrical, progressive muscle weakness. The aim of this work is to give an overview of the biological therapy used in the treatment of idiopathic inflammatory myopathies. The authors also focus on novel results in the therapy directed against the B- and T-cells. They emphasize the importance of new trials in these diseases which may lead to the introduction of novel therapeutic options in these disorders.
Collapse
Affiliation(s)
- Levente Bodoki
- Debreceni Egyetem, Orvos- és Egészségtudományi Centrum Belgyógyászati Intézet, Klinikai Immunológia Tanszék Debrecen Móricz Zsigmond körút 22. 4032
| | - Melinda Nagy-Vincze
- Debreceni Egyetem, Orvos- és Egészségtudományi Centrum Belgyógyászati Intézet, Klinikai Immunológia Tanszék Debrecen Móricz Zsigmond körút 22. 4032
| | - Zoltán Griger
- Debreceni Egyetem, Orvos- és Egészségtudományi Centrum Belgyógyászati Intézet, Klinikai Immunológia Tanszék Debrecen Móricz Zsigmond körút 22. 4032
| | - Andrea Péter
- Debreceni Egyetem, Orvos- és Egészségtudományi Centrum Kardiológiai Klinika Debrecen
| | - Csilla András
- Debreceni Egyetem, Orvos- és Egészségtudományi Centrum Onkológiai Tanszék Debrecen
| | - Katalin Dankó
- Debreceni Egyetem, Orvos- és Egészségtudományi Centrum Belgyógyászati Intézet, Klinikai Immunológia Tanszék Debrecen Móricz Zsigmond körút 22. 4032
| |
Collapse
|
23
|
Abstract
The IL-1 family of ligands and receptors has a central role in both innate and adaptive immune responses and is tightly controlled by antagonists, decoy receptors, scavengers, dominant negative molecules, miRNAs and other mechanisms, acting extracellularly or intracellularly. During evolution, the development of multiple mechanisms of negative regulation reveals the need for tight control of the biological consequences of IL-1 family ligands in order to balance local and systemic inflammation and limit immunopathology. Indeed, studies with gene targeted mice for negative regulators and genetic studies in humans provide evidence for their non-redundant role in controlling inflammation, tissue damage and adaptive responses. In addition, studies have revealed the need of negative regulation of the IL-1 family not only in disease, but also in homeostatic conditions. In this review, the negative regulation mediated by decoy receptors are presented and include IL-1R2 and IL-IL-18BP as well as atypical receptors, which include TIR8/SIGIRR, IL-1RAcPb, TIGIRR-1 and IL-1RAPL. Particular emphasis is given to IL-1R2, since its discovery is the basis for the formulation of the decoy paradigm, now considered a general strategy to counter the primary inflammatory activities of cytokines and chemokines. Emphasis is also given to TIR8, a prototypical negative regulatory receptor having non-redundant roles in limiting inflammation and adaptive responses.
Collapse
Affiliation(s)
- Cecilia Garlanda
- Humanitas Clinical and Research Center, Department of Inflammation and Immunology, Rozzano, Italy.
| | - Federica Riva
- Department of Veterinary Science and Public Health, University of Milan, Italy
| | - Eduardo Bonavita
- Humanitas Clinical and Research Center, Department of Inflammation and Immunology, Rozzano, Italy
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Department of Inflammation and Immunology, Rozzano, Italy; Department of Biotechnology and Translational Medicine, University of Milan, Rozzano (Milano), Italy
| |
Collapse
|
24
|
Garlanda C, Riva F, Bonavita E, Gentile S, Mantovani A. Decoys and Regulatory "Receptors" of the IL-1/Toll-Like Receptor Superfamily. Front Immunol 2013; 4:180. [PMID: 23847621 PMCID: PMC3705552 DOI: 10.3389/fimmu.2013.00180] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 06/22/2013] [Indexed: 11/24/2022] Open
Abstract
Members of the IL-1 family play a key role in innate and adaptive immunity and in the pathogenesis of diverse diseases. Members of IL-1R like receptor (ILR) family include signaling molecules and negative regulators. The latter include decoy receptors (IL-1RII; IL-18BP) and “receptors” with regulatory function (TIR8/SIGIRR; IL-1RAcPb; DIGIRR). Structural considerations suggest that also TIGIRR-1 and IL-1RAPL may have regulatory function. The presence of multiple pathways of negative regulation of members of the IL-1/IL-1R family emphasizes the need for a tight control of members of this fundamental system.
Collapse
Affiliation(s)
- Cecilia Garlanda
- Department of Inflammation and Immunology, Humanitas Clinical and Research Center , Rozzano , Italy
| | | | | | | | | |
Collapse
|
25
|
Zong M, Dorph C, Dastmalchi M, Alexanderson H, Pieper J, Amoudruz P, Barbasso Helmers S, Nennesmo I, Malmström V, Lundberg IE. Anakinra treatment in patients with refractory inflammatory myopathies and possible predictive response biomarkers: a mechanistic study with 12 months follow-up. Ann Rheum Dis 2013; 73:913-20. [DOI: 10.1136/annrheumdis-2012-202857] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
26
|
Katsuyuki Shinjo S, Carlos de Souza FH, Bertacini de Moraes JC. Dermatomiosite e polimiosite: da imunopatologia à imunoterapia (imunobiológicos). REVISTA BRASILEIRA DE REUMATOLOGIA 2013. [DOI: 10.1590/s0482-50042013000100010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
27
|
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]
|
28
|
Curra M, Martins MAT, Lauxen IS, Pellicioli ACA, Sant'Ana Filho M, Pavesi VCS, Carrard VC, Martins MD. Effect of topical chamomile on immunohistochemical levels of IL-1β and TNF-α in 5-fluorouracil-induced oral mucositis in hamsters. Cancer Chemother Pharmacol 2012; 71:293-9. [PMID: 23096219 DOI: 10.1007/s00280-012-2013-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 10/14/2012] [Indexed: 10/27/2022]
Abstract
PURPOSE The aim of the present study was to evaluate the effect of topical chamomile and corticosteroid treatment on the profile of tissue cytokines (IL-1β and TNF-α) in 5-fluorouracil-induced oral mucositis in hamsters. METHODS Thirty-six hamsters were randomly separated into three groups (12 animals each): Group I--without treatment (control); Group II-treatment with chamomile (Ad-Muc(®)); and Group III--treatment with corticosteroid (betamethasone elixir- Celestone(®)). The animals received an intraperitoneal injection of 5--fluorouracil on Days 0 and 2. On Days 3 and 4, the buccal mucosa was scratched and therapy was initiated on Day 5. Three animals from each group were killed on Days 0, 5, 10, and 14 and the buccal mucosa was removed. The streptavidin-biotin complex method was used to delineate the in situ distribution, localization, and semiquantitative analysis of IL-1β and TNF-α. Data from the semiquantitative analysis of immunohistochemical staining were comparatively analyzed using the Kruskal-Wallis test, followed by Dunn's multiple comparisons test. RESULTS The distribution and localization of IL-1β and TNF-α immunolabeling were similar. These proteins exhibited a diffuse pattern distributed throughout the connective tissue. The epithelium and adipose tissue were negative for both proteins. The semiquantitative analysis revealed that immunolabeling of IL-1β and TNF-α increased in all groups with the development of mucositis. On Day 10 (period of peak mucositis), the group treated with chamomile had lower scores for both pro-inflammatory cytokines. CONCLUSIONS Treatment with topical chamomile reduced the tissue levels of IL-1β and TNF-α, thereby demonstrating anti-inflammatory action in oral mucositis in hamsters.
Collapse
Affiliation(s)
- Marina Curra
- Oral Pathology Department, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Colombo E, Romaggi S, Blasevich F, Mora M, Falcone C, Lochmüller H, Morandi L, Farina C. The neurotrophin receptor p75NTR is induced on mature myofibres in inflammatory myopathies and promotes myotube survival to inflammatory stress. Neuropathol Appl Neurobiol 2012; 38:367-78. [PMID: 21851375 DOI: 10.1111/j.1365-2990.2011.01212.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
AIMS Recent studies propose the neurotrophin receptor p75NTR as a marker for muscle satellite cells and a key regulator of regenerative processes after injury. Here, we investigated the contribution of cellular compartments other than satellite cells and regenerating myofibres to p75NTR signal in diseased skeletal muscle. METHODS We checked regulation of p75NTR expression in muscle biopsies from patients with inflammatory myopathies (polymyositis, dermatomyositis and inclusion body myositis), or Becker muscular dystrophy, and in nonmyopathic tissues. Quantitative PCR, immunohistochemistry, immunofluorescence or electron microscopy were used. RNA interference approaches were applied to myotubes to explore p75NTR function. RESULTS We found p75NTR transcript and protein upregulation in all inflammatory myopathies but not in dystrophic muscle, suggesting a role for inflammatory mediators in induction of p75NTR expression. In inflamed muscle p75NTR was localized on distinct cell types, including immune cells and mature myofibres. In vitro assays on human myotubes confirmed that inflammatory factors such as IL-1 could induce p75NTR. Finally, RNA interference experiments in differentiated cells showed that, in the absence of p75NTR, myotubes were more susceptible to apoptosis when exposed to inflammatory stimuli. CONCLUSIONS Our observations that p75NTR is upregulated on skeletal myofibres in inflammatory myopathies in vivo and promotes resistance to inflammatory mediators in vitro suggest that neurotrophin signalling through p75NTR may mediate a tissue-protective response to inflammation in skeletal myofibres.
Collapse
Affiliation(s)
- E Colombo
- Neuroimmunology and Neuromuscular Disorders, Foundation IRCCS Neurological Institute Carlo Besta, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Sugihara T, Okiyama N, Watanabe N, Miyasaka N, Kohsaka H. Interleukin-1 and tumor necrosis factor α blockade treatment of experimental polymyositis in mice. ACTA ACUST UNITED AC 2012; 64:2655-62. [DOI: 10.1002/art.34465] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
31
|
Stübgen JP. Targeted immunotherapy trials for idiopathic inflammatory myopathies. J Neurol 2012; 260:368-85. [DOI: 10.1007/s00415-012-6590-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 06/11/2012] [Accepted: 06/12/2012] [Indexed: 12/13/2022]
|
32
|
Robinson AB, Reed AM. Clinical features, pathogenesis and treatment of juvenile and adult dermatomyositis. Nat Rev Rheumatol 2011; 7:664-75. [PMID: 21947177 DOI: 10.1038/nrrheum.2011.139] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Juvenile and adult dermatomyositis (DM) have multiple commonalities, yet display differing prevalence of features, outcomes and comorbidities. In general, compared with the disease in adults, children with DM have more vasculopathy and a greater likelihood of calcinosis, periungual and gingival telangiectasias, and ulceration, but have a better long-term prognosis with improved survival. Adults with DM are more likely to have myositis-specific antibodies, develop interstitial lung disease, have amyopathic disease, and have a marked association with malignancy and other comorbidities. Both diseases have similar features on muscle biopsy and interferon gene signature, although subtle differences can exist in pathogenesis and pathology, such as more capillary loss and a greater degree of C5b-9 complement deposition in affected muscle of juvenile patients. Initiatives are underway to improve classification, markers of disease activity and ability to predict outcome of juvenile and adult DM. The purpose of this Review is to compare and contrast the unique features between juvenile and adult disease and to outline new initiatives in the field.
Collapse
Affiliation(s)
- Angela B Robinson
- Department of Pediatrics, Case Western Reserve University Hospital, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | | |
Collapse
|
33
|
Nagaraju K, Lundberg IE. Polymyositis and dermatomyositis: pathophysiology. Rheum Dis Clin North Am 2011; 37:159-71, v. [PMID: 21444017 DOI: 10.1016/j.rdc.2011.01.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Recent advances have increased the understanding of the pathogenesis of polymyositis and dermatomyositis. Clearly, the pathogenesis is complex, and adaptive (eg, autoimmune) and innate and nonimmune pathways play a role in the disease mechanisms, but the relative contribution may vary between patients and in different phases of the disease. Phenotyping patients using autoantibody profiling has resulted in information on molecular pathways that may be relevant in certain subsets of patients with polymyositis or dermatomyositis, but combining the autoantibody profiles with molecular signatures of innate and nonimmune mechanisms would enhance our ability to classify, diagnose, and treat these disorders more effectively.
Collapse
Affiliation(s)
- Kanneboyina Nagaraju
- Research Center for Genetic Medicine, Children's National Medical Center and Department of Integrative Systems Biology, The George Washington University Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | | |
Collapse
|
34
|
Andres Cerezo L, Kuncova K, Mann H, Tomcik M, Zamecnik J, Lukanidin E, Neidhart M, Gay S, Grigorian M, Vencovsky J, Senolt L. The metastasis promoting protein S100A4 is increased in idiopathic inflammatory myopathies. Rheumatology (Oxford) 2011; 50:1766-72. [DOI: 10.1093/rheumatology/ker218] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
35
|
Zong M, Lundberg IE. Pathogenesis, classification and treatment of inflammatory myopathies. Nat Rev Rheumatol 2011; 7:297-306. [DOI: 10.1038/nrrheum.2011.39] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
36
|
Kreiner F, Langberg H, Galbo H. Increased muscle interstitial levels of inflammatory cytokines in polymyalgia rheumatica. ACTA ACUST UNITED AC 2011; 62:3768-75. [PMID: 20812339 DOI: 10.1002/art.27728] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
OBJECTIVE Polymyalgia rheumatica (PMR) is characterized by aching of the proximal muscles and increased blood levels of markers of inflammation. Despite the muscle complaints, the current view is that symptoms are caused by inflammation in synovial structures. The purpose of this study was to elucidate the disease mechanisms in symptomatic muscles by measuring interstitial levels of cytokines before and after prednisolone treatment. METHODS Twenty glucocorticoid-naive patients newly diagnosed as having PMR and 20 control subjects were studied before and after 14 days of prednisolone therapy (20 mg/day). Interstitial concentrations of interleukin-1α/β (IL-1α/β), IL-1 receptor antagonist, IL-6, IL-8, tumor necrosis factor α (TNFα), and monocyte chemoattractant protein 1 were measured in symptomatic vastus lateralis and trapezius muscles using the microdialysis technique. Plasma levels were measured simultaneously. RESULTS Prednisolone abolished symptoms in all of the PMR patients within 1-2 days; the erythrocyte sedimentation rate and C-reactive protein levels were normalized on day 14. In both muscles, interstitial concentrations of all cytokines were markedly higher (P < 0.05) in the PMR patients than in the controls before treatment. In both patients and controls, interstitial levels of most cytokines were higher than plasma levels, with the exception of IL-1α and TNFα, which were lower in both groups. In the PMR patients, interstitial concentrations were normalized after prednisolone treatment. CONCLUSION This study introduces a novel view of PMR, indicating that increased interstitial levels of inflammatory cytokines in symptomatic muscles play a role in the pathophysiology of the disease and that cytokines may be released locally. To explore the disease specificity, similar studies in other primary inflammatory conditions are warranted.
Collapse
Affiliation(s)
- Frederik Kreiner
- Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.
| | | | | |
Collapse
|
37
|
Loell I, Lundberg IE. Can muscle regeneration fail in chronic inflammation: a weakness in inflammatory myopathies? J Intern Med 2011; 269:243-57. [PMID: 21205023 DOI: 10.1111/j.1365-2796.2010.02334.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Idiopathic inflammatory myopathies (IIMs), collectively termed myositis, include three major subgroups: polymyositis, dermatomyositis and inclusion body myositis. IIMs are characterized clinically by muscle weakness and reduced muscle endurance preferentially affecting the proximal skeletal muscle. In typical cases, inflammatory cell infiltrates and proinflammatory cytokines, alarmins and eicosanoids are present in muscle tissue. Treatment with glucocorticoids and other immunosuppressants results in improved performance, but complete recovery is rarely seen. The mechanisms that cause muscle weakness and reduced muscle endurance are multi-factorial, and different mechanisms predominate in different phases of disease. It is likely that a combination of immune-mediated and nonimmune-mediated mechanisms contributes to clinical muscle symptoms. Immune-mediated mechanisms include immune cell-mediated muscle fibre necrosis as well as direct effects of various cytokines on muscle fibre contractility. Among the nonimmune-mediated mechanisms, an acquired metabolic myopathy and so-called endoplasmic reticulum stress may be important. There is also a possibility of defective repair mechanisms, with an influence of both disease-related factors and glucocorticoid treatment. Several proinflammatory molecules observed in muscle tissue of myositis patients, including interleukin (IL)-1, IL-15, tumour necrosis factor, high-mobility group box-1 and eicosanoids, have a role in muscle fibre regeneration, and blocking these molecule may impair muscle repair and recovery. The delicate balance between immunosuppressive treatment to downregulate proinflammatory molecules and an inhibitory effect on muscle fibre regeneration needs to be further understood. This would also be relevant for other chronic inflammatory diseases.
Collapse
Affiliation(s)
- I Loell
- Department of Medicine, Karolinska University Hospital, Solna, Stockholm, Sweden
| | | |
Collapse
|
38
|
Nader GA, Dastmalchi M, Alexanderson H, Grundtman C, Gernapudi R, Esbjörnsson M, Wang Z, Rönnelid J, Hoffman EP, Nagaraju K, Lundberg IE. A longitudinal, integrated, clinical, histological and mRNA profiling study of resistance exercise in myositis. Mol Med 2010; 16:455-64. [PMID: 20809047 DOI: 10.2119/molmed.2010.00016] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2010] [Accepted: 08/11/2010] [Indexed: 12/20/2022] Open
Abstract
Polymyositis and dermatomyositis are orphan, chronic skeletal muscle disorders characterized by weakness, infiltrations by mononuclear inflammatory cells, and fibrosis. Until recently, patients were advised to refrain from physical activity because of fears of exacerbation of muscle inflammation. However, recent studies have shown that moderate exercise training in combination with immunosuppressive drugs can improve muscle performance. Despite the positive effects of exercise training, the molecular mechanisms underlying the exercise-associated clinical improvements remain poorly understood. The present study was designed to define, at the molecular level, the effects of resistance exercise training on muscle performance and disease progression in myositis patients. We evaluated changes in muscle strength, histology and genome-wide mRNA profiles to determine the beneficial effects of exercise and determine the possible molecular changes associated with improved muscle performance. A total of 8 myositis patients underwent a 7-wk resistance exercise training program that resulted in improved muscle strength and increased maximal oxygen uptake (VO(2max)). Training also resulted in marked reductions in gene expression, reflecting reductions in proinflammatory and profibrotic gene networks, changes that were also accompanied by a reduction in tissue fibrosis. Consistent with the exercise-associated increase in VO(2max), a subset of transcripts was associated with a shift toward oxidative metabolism. The changes in gene expression reported in the present study are in agreement with the performance improvements induced by exercise and suggest that resistance exercise training can induce a reduction in inflammation and fibrosis in skeletal muscle.
Collapse
Affiliation(s)
- Gustavo A Nader
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, DC, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Chahbouni M, Escames G, Venegas C, Sevilla B, García JA, López LC, Muñoz-Hoyos A, Molina-Carballo A, Acuña-Castroviejo D. Melatonin treatment normalizes plasma pro-inflammatory cytokines and nitrosative/oxidative stress in patients suffering from Duchenne muscular dystrophy. J Pineal Res 2010; 48:282-289. [PMID: 20210854 DOI: 10.1111/j.1600-079x.2010.00752.x] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Duchenne muscular dystrophy (DMD), a lethal disorder characterized by dystrophin absence, courses with chronic inflammation, sarcolemmal damage, and skeletal muscle degeneration. Among the multiple pathogenic mechanisms proposed for DMD, oxidative stress and inflammation are directly involved in the dystrophic process. Unfortunately, there is no current treatment for DMD, and the inflammatory process is an important target for therapies. Based on the antioxidant and anti-inflammatory properties of melatonin, we investigated whether melatonin treatment may reduce the dystrophic process. Ten DMD patients aged 12.8 +/- 0.98 yr, were treated with melatonin (60 mg at 21:00 hr plus 10 mg at 09:00 hr), and plasma levels of lipid peroxidation (LPO), nitrites (NO(x)), interleukin (IL)-1beta, IL-2, IL-6, tumor necrosis factor-alpha, interferon-gamma, and plasma markers of muscle injury, were determined at 3, 6 and 9 months of treatment. Healthy age- and sex-matched subjects were used as controls. The results show a significant increase in LPO, NO(x), and cytokine levels in plasma of DMD patients compared with controls. Melatonin administration reduced these values to control levels at 3 months of treatment, decreasing further 9 months later. In parallel, melatonin also reduced plasma levels of creatine kinase (CK; 50%), lactate dehydrogenase (28%), aspartate aminotransferase (28%), alanine aminotransferase (20%), and myoglobin (13%). These findings strongly support the conclusion that melatonin administration significantly reduced the hyperoxidative and inflammatory process in DMD patients, reducing the muscle degenerative process.
Collapse
Affiliation(s)
- Mariam Chahbouni
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Armilla, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Germaine Escames
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Armilla, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Carmen Venegas
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Armilla, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Belén Sevilla
- Unidad de Gestión Clínica de Pediatría, Hospital Universitario San Cecilio, Granada, Spain
| | - José Antonio García
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Armilla, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Luis C López
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Armilla, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - Antonio Muñoz-Hoyos
- Unidad de Gestión Clínica de Pediatría, Hospital Universitario San Cecilio, Granada, Spain
| | | | - Darío Acuña-Castroviejo
- Instituto de Biotecnología, Centro de Investigación Biomédica, Parque Tecnológico de Ciencias de la Salud, Universidad de Granada, Armilla, Granada, Spain
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Servicio de Análisis Clínicos, Hospital Universitario San Cecilio, Granada, Spain
| |
Collapse
|
40
|
Evans NP, Misyak SA, Robertson JL, Bassaganya-Riera J, Grange RW. Immune-mediated mechanisms potentially regulate the disease time-course of duchenne muscular dystrophy and provide targets for therapeutic intervention. PM R 2009; 1:755-68. [PMID: 19695529 DOI: 10.1016/j.pmrj.2009.04.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Revised: 04/23/2009] [Accepted: 04/28/2009] [Indexed: 11/19/2022]
Abstract
Duchenne muscular dystrophy is a lethal muscle-wasting disease that affects boys. Mutations in the dystrophin gene result in the absence of the dystrophin glycoprotein complex (DGC) from muscle plasma membranes. In healthy muscle fibers, the DGC forms a link between the extracellular matrix and the cytoskeleton to protect against contraction-induced membrane lesions and to regulate cell signaling. The absence of the DGC results in aberrant regulation of inflammatory signaling cascades. Inflammation is a key pathological characteristic of dystrophic muscle lesion formation. However, the role and regulation of this process in the disease time-course has not been sufficiently examined. The transcription factor nuclear factor-kappaB has been shown to contribute to the disease process and is likely involved with increased inflammatory gene expression, including cytokines and chemokines, found in dystrophic muscle. These aberrant signaling processes may regulate the early time-course of inflammatory events that contribute to the onset of disease. This review critically evaluates the possibility that dystrophic muscle lesions in both patients with Duchenne muscular dystrophy and mdx mice are the result of immune-mediated mechanisms that are regulated by inflammatory signaling and also highlights new therapeutic directions.
Collapse
Affiliation(s)
- Nicholas P Evans
- Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061-0002, USA.
| | | | | | | | | |
Collapse
|
41
|
Abstract
The idiopathic inflammatory myopathies (IIM) are systemic autoimmune diseases that have predominant mononuclear inflammatory cell infiltrates in the skeletal muscle. The cells that are typically involved in the pathogenesis of disease are B-lymphocytes, T-lymphocytes, macrophages, dendritic cells, and natural killer cells. However, in addition to these immune cells, cells of nonimmunologic origin, such as myocytes, may be directly involved in the immune response. The local milieu also consists of distinct cytokine and chemokine profiles considered related to type 1 interferon stimulation. Tumor necrosis factor and interleukin 1 are also prominent, proinflammatory cytokines involved in the evolution of IIM. Although the pathologic processes involved in IIM have yet to be fully elucidated, we understand the inflammatory milieu is a model of dynamic flux made of diverse cytokine and chemokine expressions leading to alterations in muscle fiber structure and function.
Collapse
Affiliation(s)
- Ann M Reed
- Division of Rheumatology, Departments of Pediatrics and Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| | | |
Collapse
|
42
|
Grundtman C, Bruton J, Yamada T, Ostberg T, Pisetsky DS, Harris HE, Andersson U, Lundberg IE, Westerblad H. Effects of HMGB1 on in vitro responses of isolated muscle fibers and functional aspects in skeletal muscles of idiopathic inflammatory myopathies. FASEB J 2009; 24:570-8. [PMID: 19837864 DOI: 10.1096/fj.09-144782] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Idiopathic inflammatory myopathies (IIMs) are heterogeneous rheumatic disorders of unknown cause characterized by muscle weakness, inflammatory cell infiltrates, and major histocompatibility complex (MHC) class I expression on muscle fibers. The nonhistone nuclear protein alarmin high-mobility group box 1 protein (HMGB1) has been detected extranuclearly in muscle biopsies from patients with IIMs. We hypothesize that HMGB1 has a central role in the cause of muscle weakness, particularly in the early phases of IIMs. Experiments were performed on skeletal muscle fibers isolated from adult mice, which were exposed to recombinant interferon (IFN)-gamma or HMGB1. The myoplasmic free [Ca(2+)] was measured. Stimulation with IFN-gamma resulted in increased HMGB1 expression in muscle nuclei and the myoplasm. Exposure to HMGB1 induced a reversible up-regulation of MHC class I in the muscle fibers. However, HMGB1 exposure caused an irreversible decrease in Ca(2+) release from the sarcoplasmic reticulum during fatigue, induced by repeated tetanic contractions. HMGB1 and MHC class I were frequently colocalized in the myoplasm of muscle fibers in muscle biopsies from patients with early IIMs. However, HMGB1-expressing fibers outnumbered fibers expressing MHC class I. Our data indicate that HMGB1 could be an early inducer of skeletal muscle dysfunction in IIMs.
Collapse
Affiliation(s)
- Cecilia Grundtman
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital-Solna, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Li W, Moylan JS, Chambers MA, Smith J, Reid MB. Interleukin-1 stimulates catabolism in C2C12 myotubes. Am J Physiol Cell Physiol 2009; 297:C706-14. [PMID: 19625606 DOI: 10.1152/ajpcell.00626.2008] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Interleukin-1 (IL-1) is an inflammatory cytokine that has been linked to muscle catabolism, a process regulated by muscle-specific E3 proteins of the ubiquitin-proteasome pathway. To address cellular mechanism, we tested the hypothesis that IL-1 induces myofibrillar protein loss by acting directly on muscle to increase expression of two critical E3 proteins, atrogin1/muscle atrophy F-box (MAFbx) and muscle RING-finger 1 (MuRF1). Experiments were conducted using mature C2C12 myotubes to eliminate systemic cytokine effects and avoid paracrine signaling by nonmuscle cell types. Time-course protocols were used to define the sequence of cellular responses. We found that atrogin1/MAFbx mRNA and MuRF1 mRNA are elevated 60-120 min after myotube exposure to either IL-1alpha or IL-1beta. These responses are preceded by signaling events that promote E3 expression. Both IL-1 isoforms stimulate phosphorylation of p38 mitogen-activated protein kinase and stimulate nuclear factor-kappaB (NF-kappaB) signaling; I-kappaB levels fall and NF-kappaB DNA binding activity increases. Other regulators of E3 expression are unaffected by IL-1 [cytosolic oxidant activity, Forkhead-O (Foxo) activity] or respond paradoxically (AKT). Chronic exposure of C2C12 myotubes over 48 h resulted in reduced myotube width and loss of sarcomeric actin. We conclude that IL-1alpha and IL-1beta act via an oxidant- and AKT/Foxo-independent mechanism to activate p38 MAPK, stimulate NF-kappaB signaling, increase expression of atrogin1/MAFbx and MuRF1, and reduce myofibrillar protein in differentiated myotubes.
Collapse
Affiliation(s)
- Wei Li
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536-0298, USA
| | | | | | | | | |
Collapse
|
44
|
Lundberg IE, Grundtman C. Developments in the scientific and clinical understanding of inflammatory myopathies. Arthritis Res Ther 2008; 10:220. [PMID: 18947371 PMCID: PMC2592786 DOI: 10.1186/ar2501] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The idiopathic inflammatory myopathies are chronic autoimmune disorders sharing the clinical symptom of muscle weakness and, in typical cases, inflammatory cell infiltrates in muscle tissue. During the last decade, novel information has accumulated supporting a role of both the innate and adaptive immune systems in myositis and suggesting that different molecular pathways predominate in different subsets of myositis. The type I interferon activity is one such novel pathway identified in some subsets of myositis. Furthermore, nonimmunological pathways have been identified, suggesting that factors other than direct T cell-mediated muscle fibre necrosis could have a role in the development of muscle weakness.
Collapse
Affiliation(s)
- Ingrid E Lundberg
- Rheumatology Unit, Department of Medicine, Karolinska University Hospital - Solna, Karolinska Institutet, SE-171 76 Stockholm, Sweden.
| | | |
Collapse
|
45
|
Okiyama N, Kohsaka H. [Recent research developments in polymyositis/dermatomyositis]. ACTA ACUST UNITED AC 2008; 31:85-92. [PMID: 18446010 DOI: 10.2177/jsci.31.85] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The idiopathic inflammatory myopathies, polymyositis (PM) and dermatomyositis (DM), are evaluated as systemic autoimmune diseases without the pathology determined. Past immunohistochemical findings suggested that the effector response is driven predominantly by CD4 T cells and by humoral immunity in DM, and by cytotoxic T cells in PM. However, histological observations of muscle tissue do not necessarily distinguish DM and PM. Thus, the two diseases including amyopathic DM might represent a spectrum of illness in which some patients suffer only from a muscle disease or from a skin disease. In comparison with research studies on other rheumatic diseases, there are much fewer research studies conducted on PM/DM. The relationship between PM and DM is not clear yet. We reviewed past clinical and basic research on the pathology of PM/DM, including research on relevant T cells, B cells and cytokines.
Collapse
Affiliation(s)
- Naoko Okiyama
- Department of Dermatology, Tokyo Medical and Dental University
| | | |
Collapse
|
46
|
Baird GS, Montine TJ. Multiplex immunoassay analysis of cytokines in idiopathic inflammatory myopathy. Arch Pathol Lab Med 2008; 132:232-8. [PMID: 18251582 DOI: 10.5858/2008-132-232-miaoci] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2007] [Indexed: 11/06/2022]
Abstract
CONTEXT Idiopathic inflammatory myopathies (IIMs), including dermatomyositis, polymyositis, and inclusion-body myositis, can be difficult to diagnose. OBJECTIVE To determine if a multiplex immunoassay for markers of inflammation in muscle homogenates correlates with a diagnosis of IIM. DESIGN Frozen archived muscle biopsy specimens from 30 patients with IIM and 34 patients without IIM were homogenized and analyzed for cytokine content with a multiplex microbead-based immunoassay system. Analyte concentrations were normalized to total lysate protein concentration prior to comparison. RESULTS Two cytokines, interleukin 1ra and monocyte chemoattractant protein 1, and 1 soluble adhesion molecule, intracellular adhesion molecule 1, were found at significantly greater concentrations in muscle samples from patients with IIM. Intracellular adhesion molecule 1 levels alone were 83% sensitive and 91% specific for IIM at a cutoff of 1240 pg/mg muscle protein. CONCLUSIONS Immunoassays for selected inflammatory markers can serve in conjunction with histopathologic analysis as sensitive and specific tools for the diagnosis of IIM.
Collapse
Affiliation(s)
- Geoffrey S Baird
- Department of Pathology, University of Washington, Harborview Medical Center, Box 359645, Seattle, WA 98104-2499, USA.
| | | |
Collapse
|
47
|
Abstract
The clinical spectrum and immunopathogenesis of inflammatory myopathies are summarized with an update on possible triggering factors, cell degeneration, and emerging new therapies.
Collapse
Affiliation(s)
- Marinos C Dalakas
- Neuromuscular Diseases Section, US National Institute of Neurological Disorders and Stroke, US National Institutes of Health, Building 10, Room 4N248, Bethesda, MD 20892, USA.
| |
Collapse
|
48
|
Abstract
After background information about pathologic findings, this review focuses on the cytokine response in the pathogenesis of polymyositis and dermatomyositis. Cytokines are important mediators of the immune response and play a key role in these diseases by acting on inflammatory immune cells, muscle cells, and vessel cells. Various cytokines are found in myositis samples, in particular interleukin-1 and tumor necrosis factor-alpha, which are associated with the migration, differentiation, and maturation of inflammatory cells. Recent advances indicate that the muscle cell itself could participate in the inflammatory process. Cytokines promote changes in muscle metabolism resulting in a self-sustaining inflammatory response. Accordingly, cytokines may represent new targets for therapies.
Collapse
Affiliation(s)
- Anne Tournadre
- Clinical Immunology Unit, Department of Immunology and Rheumatology, Hospital Edouard Herriot, Lyon Cedex 03, France
| | | |
Collapse
|
49
|
Cooper DM, Radom-Aizik S, Schwindt C, Zaldivar F. Dangerous exercise: lessons learned from dysregulated inflammatory responses to physical activity. J Appl Physiol (1985) 2007; 103:700-9. [PMID: 17495117 DOI: 10.1152/japplphysiol.00225.2007] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Exercise elicits an immunological “danger” type of stress and inflammatory response that, on occasion, becomes dysregulated and detrimental to health. Examples include anaphylaxis, exercise-induced asthma, overuse syndromes, and exacerbation of intercurrent illnesses. In dangerous exercise, the normal balance between pro- and anti-inflammatory responses is upset. A possible pathophysiological mechanism is characterized by the concept of exercise modulation of previously activated leukocytes. In this model, circulating leukocytes are rendered more responsive than normal to the immune stimulus of exercise. For example, in the case of exercise anaphylaxis, food-sensitized immune cells may be relatively innocuous until they are redistributed during exercise from gut-associated circulatory depots, like the spleen, into the central circulation. In the case of asthma, the prior activation of leukocytes may be the result of genetic or environmental factors. In the case of overuse syndromes, the normally short-lived neutrophil may, because of acidosis and hypoxia, inhibit apoptosis and play a role in prolongation of inflammation rather than healing. Dangerous exercise demonstrates that the stress/inflammatory response caused by physical activity is robust and sufficiently powerful, perhaps, to alter subsequent responses. These longer term effects may occur through as yet unexplored mechanisms of immune “tolerance” and/or by a training-associated reduction in the innate immune response to brief exercise. A better understanding of sometimes failed homeostatic physiological systems can lead to new insights with significant implication for clinical translation.
Collapse
Affiliation(s)
- Dan Michael Cooper
- Pediatric Exercise Research Center, Department of Pediatrics, University of California, Irvine, California, USA.
| | | | | | | |
Collapse
|