1
|
Carstens PO, Müllar LM, Wrede A, Zechel S, Wachowski MM, Brandis A, Krause S, Zierz S, Schmidt J. Skeletal muscle fibers produce B-cell stimulatory factors in chronic myositis. Front Immunol 2023; 14:1177721. [PMID: 37731487 PMCID: PMC10508232 DOI: 10.3389/fimmu.2023.1177721] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/16/2023] [Indexed: 09/22/2023] Open
Abstract
Introduction We aimed to identify B-cell-mediated immunomechanisms in inclusion body myositis (IBM) and polymyositis (PM) as part of the complex pathophysiology. Materials and methods Human primary myotube cultures were derived from orthopedic surgery. Diagnostic biopsy specimens from patients with IBM (n=9) and PM (n=9) were analyzed for markers of B cell activation (BAFF and APRIL) and for chemokines that control the recruitment of B cells (CXCL-12 and CXCL-13). Results were compared to biopsy specimens without myopathic changes (n=9) and hereditary muscular dystrophy (n=9). Results The mRNA expression of BAFF, APRIL, and CXCL-13 was significantly higher in IBM and PM compared to controls. Patients with IBM displayed the highest number of double positive muscle fibers for BAFF and CXCL-12 (48%) compared to PM (25%), muscular dystrophy (3%), and non-myopathic controls (0%). In vitro, exposure of human myotubes to pro-inflammatory cytokines led to a significant upregulation of BAFF and CXCL-12, but APRIL and CXCL-13 remained unchanged. Conclusion The results substantiate the hypothesis of an involvement of B cell-associated mechanisms in the pathophysiology of IBM and PM. Muscle fibers themselves seem to contribute to the recruitment of B cells and sustain inflammation.
Collapse
Affiliation(s)
- Per-Ole Carstens
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Luisa M. Müllar
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Arne Wrede
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
- Institute of Neuropathology, Saarland University Medical Center and Medical Faculty of Saarland University, Homburg, Germany
| | - Sabrina Zechel
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Martin M. Wachowski
- Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Almuth Brandis
- Department of Pathology, Klinikum Region Hannover, Hannover, Germany
- Institute of Pathology and Neuropathology, Medical University Hannover, Hannover, Germany
| | - Sabine Krause
- Friedrich-Baur-Institute, Department of Neurology, Ludwig-Maximilians-University of München, München, Germany
| | - Stephan Zierz
- Department of Neurology, University Hospital Halle/Saale, Halle, Germany
| | - Jens Schmidt
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Department of Neurology and Pain Treatment, Neuromuscular Center, Center for Translational Medicine, Immanuel Klinik Rüdersdorf, University Hospital of the Brandenburg Medical School Theodor Fontane, Rüdersdorf bei Berlin, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, Rüdersdorf bei, Berlin, Germany
| |
Collapse
|
2
|
Lia A, Annese T, Fornaro M, Giannini M, D'Abbicco D, Errede M, Lorusso L, Amati A, Tampoia M, Trojano M, Virgintino D, Ribatti D, Serlenga L, Iannone F, Girolamo F. Perivascular and endomysial macrophages expressing VEGF and CXCL12 promote angiogenesis in anti-HMGCR immune-mediated necrotizing myopathy. Rheumatology (Oxford) 2021; 61:3448-3460. [PMID: 34864921 DOI: 10.1093/rheumatology/keab900] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/29/2021] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES To study the phenotype of macrophage infiltrates and their role in angiogenesis in different Idiopathic Inflammatory Myopathies (IIMs). METHODS The density and distribution of the subpopulations of macrophages subsets (M1, inducible nitric oxide+, CD11c+; M2, arginase-1+), endomysial capillaries (CD31+, FLK1+), degenerating (C5b-9+), and regenerating (NCAM+) myofibers, were investigated by immunohistochemistry in human muscle samples of diagnostic biopsies from a large cohort of untreated patients (n: 81) suffering from anti-3-hydroxy-3-methylglutaryl coenzyme A reductase (anti-HMGCR)+ Immune Mediated Necrotizing Myopathy (IMNM), anti-signal recognition particle (anti-SRP)+ IMNM, seronegative IMNM, Dermatomyositis, Polymyositis, Polymyositis with mitochondrial pathology, sporadic Inclusion Body Myositis, Scleromyositis, and anti-Synthetase Syndrome. The samples were compared with mitochondrial myopathy and control muscle samples. RESULTS Compared with the other IIMs and controls, endomysial capillary density (CD) was higher in anti-HMGCR+ IMNM, where M1 and M2 macrophages, detected by confocal microscopy, infiltrated perivascular endomysium and expressed angiogenic molecules such as VEGF-A and CXCL12. These angiogenic macrophages were preferentially associated with CD31+ FLK1+ microvessels in anti-HMGCR+ IMNM. The VEGF-A+ M2 macrophage density was significantly correlated with CD (rS: 0.98; p: 0.0004). Western blot analyses revealed increased expression levels of VEGF-A, FLK1, HIF-1α, and CXCL12 in anti-HMGCR+ IMNM. CD and expression levels of these angiogenic molecules were not increased in anti-SRP+ and seronegative IMNM, offering additional, useful information for differential diagnosis among these IIM subtypes. CONCLUSION Our findings suggest that in IIMs, infiltrating macrophages and microvascular cells interactions play a pivotal role in coordinating myogenesis and angiogenesis. This reciprocal crosstalk seems to distinguish anti-HMGCR associated IMNM.
Collapse
Affiliation(s)
- Anna Lia
- Unit of Neurophysiopathology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Tiziana Annese
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Marco Fornaro
- Unit of Rheumatology, Department of Emergency and Organ Transplantation, University of Bari, Italy
| | - Margherita Giannini
- Unit of Rheumatology, Department of Emergency and Organ Transplantation, University of Bari, Italy.,Service de Physiologie, Unité d'Explorations Fonctionnelles Musculaires, Hôpitaux Universitaires de Strasbourg, France
| | - Dario D'Abbicco
- Institute of General Surgery "G. Marinaccio", Department of Emergency and Organ Transplantation, University of Bari
| | - Mariella Errede
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Loredana Lorusso
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Angela Amati
- Unit of Neurophysiopathology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Marilina Tampoia
- Unit of Clinical Pathology, Ospedale SS., Annunziata, Taranto, Italy
| | - Maria Trojano
- Unit of Neurophysiopathology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Daniela Virgintino
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Domenico Ribatti
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Luigi Serlenga
- Unit of Neurophysiopathology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| | - Florenzo Iannone
- Unit of Rheumatology, Department of Emergency and Organ Transplantation, University of Bari, Italy
| | - Francesco Girolamo
- Unit of Human Anatomy and Histology, Department of Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari, Italy
| |
Collapse
|
3
|
Song T, Manoharan P, Millay DP, Koch SE, Rubinstein J, Heiny JA, Sadayappan S. Dilated cardiomyopathy-mediated heart failure induces a unique skeletal muscle myopathy with inflammation. Skelet Muscle 2019; 9:4. [PMID: 30678732 PMCID: PMC6345027 DOI: 10.1186/s13395-019-0189-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 01/10/2019] [Indexed: 02/02/2023] Open
Abstract
Background Skeletal muscle myopathy and exercise intolerance are diagnostic hallmarks of heart failure (HF). However, the molecular adaptations of skeletal muscles during dilated cardiomyopathy (DCM)-mediated HF are not completely understood. Methods Skeletal muscle structure and function were compared in wild-type (WT) and cardiac myosin binding protein-C null mice (t/t), which develop DCM-induced HF. Cardiac function was examined by echocardiography. Exercise tolerance was measured using a graded maximum treadmill running test. Hindlimb muscle function was assessed in vivo from measurements of plantar flexor strength. Inflammatory status was evaluated from the expression of inflammatory markers and the presence of specific immune cell types in gastrocnemius muscles. Muscle regenerative capacityat days 3, 7, and 14 after eccentric contraction-induced injury was determined from the number of phenotypically new and adult fibers in the gastrocnemius, and functional recovery of plantar flexion torque. Results t/t mice developed DCM-induced HF in association with profound exercise intolerance, consistent with previous reports. Compared to WT, t/t mouse hearts show significant hypertrophy of the atria and ventricles and reduced fractional shortening, both systolic and diastolic. In parallel, the skeletal muscles of t/t mice exhibit weakness and myopathy. Compared to WT, plantar flexor muscles of t/t null mice produce less peak isometric plantar torque (Po), develop torque more slowly (+ dF/dt), and relax more slowly (− dF/dt, longer half-relaxation times,1/2RT). Gastrocnemius muscles of t/t mice have a greater number of fibers with smaller diameters and central nuclei. Oxidative fibers, both type I and type IIa, show significantly smaller cross-sectional areas and more central nuclei. These fiber phenotypes suggest ongoing repair and regeneration under homeostatic conditions. In addition, the ability of muscles to recover and regenerate after acute injury is impaired in t/t mice. Conclusions Our studies concluded that DCM-induced HF induces a unique skeletal myopathy characterized by decreased muscle strength, atrophy of oxidative fiber types, ongoing inflammation and damage under homeostasis, and impaired regeneration after acute muscle injury. Furthermore, this unique myopathy in DCM-induced HF likely contributes to and exacerbates exercise intolerance. Therefore, efforts to develop therapeutic interventions to treat skeletal myopathy during DCM-induced HF should be considered. Electronic supplementary material The online version of this article (10.1186/s13395-019-0189-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Taejeong Song
- Heart Lung Vascular Institute, Division of Cardiology, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Palanikumar Manoharan
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Douglas P Millay
- Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, Cincinnati, OH, 45229, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Sheryl E Koch
- Heart Lung Vascular Institute, Division of Cardiology, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Jack Rubinstein
- Heart Lung Vascular Institute, Division of Cardiology, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Judith A Heiny
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Sakthivel Sadayappan
- Heart Lung Vascular Institute, Division of Cardiology, University of Cincinnati, Cincinnati, OH, 45267, USA. .,Department of Internal Medicine, Heart, Lung and Vascular Institute, Division of Cardiovascular Health and Sciences, College of Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA.
| |
Collapse
|
4
|
Lopez MJ, Seyed-Razavi Y, Jamali A, Harris DL, Hamrah P. The Chemokine Receptor CXCR4 Mediates Recruitment of CD11c+ Conventional Dendritic Cells Into the Inflamed Murine Cornea. Invest Ophthalmol Vis Sci 2018; 59:5671-5681. [PMID: 30489627 PMCID: PMC6266730 DOI: 10.1167/iovs.18-25084] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/19/2018] [Indexed: 12/13/2022] Open
Abstract
Purpose The cornea contains distinct populations of antigen-presenting cells (APCs), including conventional dendritic cells (cDCs). Little is known about the molecular mechanisms involved in cDCs homing and recruitment into the naïve and inflamed cornea. The purpose of this study was to investigate the presence of CXCR4 and its ligand CXCL12 in the murine cornea and its role in cDC migration during corneal inflammation. Methods The expression of CXCR4 and CXCL12 in naïve and suture-inflamed murine corneas was assessed by whole-mount staining, flow cytometry, and quantitative PCR. The role of CXCR4 in recruitment into inflamed corneas was investigated using adoptive transfer of cDCs blocked with neutralizing antibody against CXCR4. Results We show the chemokine receptor CXCR4 to be expressed on 51.7% and 64.8% of total corneal CD11c+ cDCs, equating to 98.6 ± 12.5 cells/mm2 in the peripheral and 64.7 ± 10.6 cells/mm2 in the central naïve cornea, respectively. Along with a 4.5-fold increase in CXCL12 expression during inflammation (P < 0.05), infiltrating cDCs also expressed CXCR4 in both the peripheral (222.6 ± 33.3 cells/mm2; P < 0.001) and central cornea (161.9 ± 23.8 cells/mm2; P = 0.001), representing a decrease to 31.0% and 37.3% in the cornea, respectively. Further, ex vivo blockade (390.1 ± 40.1 vs. 612.1 ± 78.3; P = 0.008) and local blockade (263.5 ± 27.1 vs. 807.5 ± 179.5, P < 0.001) with anti-CXCR4 neutralizing antibody resulted in a decrease in cDCs homing into the cornea compared with cells pretreated with isotype controls. Conclusions Our results demonstrate that corneal CXCL12 plays a direct role in CXCR4+ cDC recruitment into the cornea. The CXCR4/CXCL12 axis is therefore a potential target to modulate corneal inflammatory responses.
Collapse
Affiliation(s)
- Maria J. Lopez
- Center for Translational Ocular Immunology, Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States
- Schepens Eye Research Institute/Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Yashar Seyed-Razavi
- Center for Translational Ocular Immunology, Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States
- Schepens Eye Research Institute/Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Arsia Jamali
- Center for Translational Ocular Immunology, Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States
- Schepens Eye Research Institute/Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Deshea L. Harris
- Center for Translational Ocular Immunology, Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States
- Schepens Eye Research Institute/Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Pedram Hamrah
- Center for Translational Ocular Immunology, Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States
- Schepens Eye Research Institute/Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
- Program in Immunology, Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, Massachusetts, United States
- Cornea Service, New England Eye Center, Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts, United States
- Cornea Service, Massachusetts Eye & Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
5
|
Role of the chemokine receptors CXCR3, CXCR4 and CCR7 in the intramuscular recruitment of plasmacytoid dendritic cells in dermatomyositis. J Neuroimmunol 2018; 319:142-148. [PMID: 29366593 DOI: 10.1016/j.jneuroim.2018.01.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 10/13/2017] [Accepted: 01/08/2018] [Indexed: 12/18/2022]
Abstract
To explore the possible mechanism implicated in the recruitment of plasmacytoid dendritic cells (pDCs), we investigated the expression of the chemokine receptors CXCR3, CXCR4, and CCR7 on intramuscular and circulating pDCs from patients with dermatomyositis (DM). Using immunohistochemistry, preferential expression of CXCR3, CXCR4 and CCR7 was identified in the perivascular inflammatory infiltrates within the perimysium in DM muscle. Western-blot analysis showed marked up-regulation of expression of CXCR3, CXCR4 and CCR7 in muscle homogenate from patients with DM compared with that in non-diseased controls. Co-localization of CD303+ pDCs with these chemokine receptors was further examined by double immunofluorescence staining, which showed extensive co-localization of CD303 with CXCR3/CXCR4/CCR7 in DM biopsies. Flow cytometry was then used to investigate the proportion of pDCs among the total PBMCs and the expression of CXCR3, CXCR4 and CCR7 on circulating pDCs. The proportion of CD123+CD303+ pDCs in peripheral blood from DM patients was markedly decreased compared to that from polymyositis (PM) patients and normal controls. Significantly increased expression of CXCR3, but not CXCR4 or CCR7, was further identified on circulating pDCs in DM. Correlation analysis showed that the expression of CXCR3 correlated inversely with the frequency of pDCs in peripheral blood. Our findings indicate that the chemokine receptors, CXCR3, CXCR4 and CCR7 may be involved in the recruitment of pDCs from peripheral blood to muscle tissues in DM via different mechanisms, and in which CXCR3 may play an important role under DM conditions.
Collapse
|
6
|
Puchert M, Adams V, Linke A, Engele J. Evidence for the involvement of the CXCL12 system in the adaptation of skeletal muscles to physical exercise. Cell Signal 2016; 28:1205-1215. [DOI: 10.1016/j.cellsig.2016.05.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 05/19/2016] [Accepted: 05/24/2016] [Indexed: 12/23/2022]
|
7
|
De Paepe B, Zschüntzsch J. Scanning for Therapeutic Targets within the Cytokine Network of Idiopathic Inflammatory Myopathies. Int J Mol Sci 2015; 16:18683-713. [PMID: 26270565 PMCID: PMC4581266 DOI: 10.3390/ijms160818683] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/13/2015] [Accepted: 07/15/2015] [Indexed: 12/17/2022] Open
Abstract
The idiopathic inflammatory myopathies (IIM) constitute a heterogeneous group of chronic disorders that include dermatomyositis (DM), polymyositis (PM), sporadic inclusion body myositis (IBM) and necrotizing autoimmune myopathy (NAM). They represent distinct pathological entities that, most often, share predominant inflammation in muscle tissue. Many of the immunopathogenic processes behind the IIM remain poorly understood, but the crucial role of cytokines as essential regulators of the intramuscular build-up of inflammation is undisputed. This review describes the extensive cytokine network within IIM muscle, characterized by strong expression of Tumor Necrosis Factors (TNFα, LTβ, BAFF), Interferons (IFNα/β/γ), Interleukins (IL-1/6/12/15/18/23) and Chemokines (CXCL9/10/11/13, CCL2/3/4/8/19/21). Current therapeutic strategies and the exploration of potential disease modifying agents based on manipulation of the cytokine network are provided. Reported responses to anti-TNFα treatment in IIM are conflicting and new onset DM/PM has been described after administration of anti-TNFα agents to treat other diseases, pointing to the complex effects of TNFα neutralization. Treatment with anti-IFNα has been shown to suppress the IFN type 1 gene signature in DM/PM patients and improve muscle strength. Beneficial effects of anti-IL-1 and anti-IL-6 therapy have also been reported. Cytokine profiling in IIM aids the development of therapeutic strategies and provides approaches to subtype patients for treatment outcome prediction.
Collapse
Affiliation(s)
- Boel De Paepe
- Neuromuscular Reference Center, Laboratory for Neuropathology, 10K12E, Ghent University Hospital, 9000 Ghent, Belgium.
| | - Jana Zschüntzsch
- Department of Neurology, University Medical Centre, Göttingen University, 37075 Göttingen, Germany.
| |
Collapse
|
8
|
Fujiyama T, Ito T, Ogawa N, Suda T, Tokura Y, Hashizume H. Preferential infiltration of interleukin-4-producing CXCR4+ T cells in the lesional muscle but not skin of patients with dermatomyositis. Clin Exp Immunol 2014; 177:110-20. [PMID: 24580543 DOI: 10.1111/cei.12311] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2014] [Indexed: 11/30/2022] Open
Abstract
Dermatomyositis (DM) and polymyositis (PM) are collectively termed autoimmune myopathy. To investigate the difference between muscle- and skin-infiltrating T cells and to address their role for myopathy, we characterized T cells that were directly expanded from the tissues. Enrolled into this study were 25 patients with DM and three patients with PM. Muscle and skin biopsied specimens were immersed in cRPMI medium supplemented with interleukin (IL)-2 and anti-CD3/CD28 antibody-conjugated microbeads. The expanded cells were subjected to flow cytometry to examine their phenotypes. We analysed the cytokine concentration in the culture supernatants from the expanded T cells and the frequencies of cytokine-bearing cells by intracellular staining. There was non-biased in-vitro expansion of tissue-infiltrating CD4(+) and CD8(+) T cells from the muscle and skin specimens. The majority of expanded T cells were chemokine receptor (CCR) type 7(-) CD45RO(+) effecter memory cells with various T cell receptor (TCR) Vβs. The skin-derived but not muscle-derived T cells expressed cutaneous lymphocyte antigen (CLA) and CCR10 and secreted large amounts of IL-17A, suggesting that T helper type 17 (Th17) cells may have a crucial role in the development of skin lesions. Notably, the frequency of IL-4-producing chemokine (C-X-C motif) receptor (CXCR)4(+) Th2 cells was significantly higher in the muscle-derived cells and correlated inversely with the serum creatine phosphokinase (CPK) and lactate dehydrogenase (LDH) levels. stromal-derived factor (SDF)-1/CXCL12, a ligand for CXCR4, was expressed at a high level in the vascular endothelial cells between muscular fasciculi. Our study suggests that T cell populations in the muscle and skin are different, and the Th2 cell infiltrate in the muscle is associated with the low severity of myositis in DM.
Collapse
Affiliation(s)
- T Fujiyama
- Department of Dermatology, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | | | | | | | | |
Collapse
|
9
|
Saclier M, Cuvellier S, Magnan M, Mounier R, Chazaud B. Monocyte/macrophage interactions with myogenic precursor cells during skeletal muscle regeneration. FEBS J 2013; 280:4118-30. [PMID: 23384231 DOI: 10.1111/febs.12166] [Citation(s) in RCA: 184] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 01/27/2013] [Accepted: 01/31/2013] [Indexed: 12/14/2022]
Abstract
Adult skeletal muscle has the remarkable property of regenerating after damage, owing to satellite cells and myogenic precursor cells becoming committed to adult myogenesis to rebuild the muscle. This process is accompanied by the continuing presence of macrophages, from the phagocytosis of damaged myofibres to the full re-formation of new myofibres. In recent years, there has been huge progress in our understanding of the roles of macrophages during skeletal muscle regeneration, notably concerning their effects on myogenic precursor cells. Here, we review the most recent knowledge acquired on monocyte entry into damaged muscle, the various macrophage subpopulations, and their respective roles during the sequential phases of muscle repair. We also discuss the role of macrophages after exercise-induced muscle damage, notably in humans.
Collapse
|
10
|
Creus KK, De Paepe B, Weis J, De Bleecker JL. The multifaceted character of lymphotoxin β in inflammatory myopathies and muscular dystrophies. Neuromuscul Disord 2012; 22:712-9. [PMID: 22652080 DOI: 10.1016/j.nmd.2012.04.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 04/22/2012] [Accepted: 04/30/2012] [Indexed: 11/18/2022]
Abstract
Lymphotoxin beta (LTβ) regulates some inflammatory mechanisms that could be operative in idiopathic inflammatory myopathies (IM). We studied LTβ and LTβR in inflammatory myopathies, normal and disease controls with immunohistochemistry, Western blotting and in situ hybridisation. LTβ occurs in myonuclei of normal controls, implying its role in normal muscle physiology. LTβ is strongly upregulated in regenerating muscle fibres in all myopathies, but not in denervated myofibres. Normal-appearing myofibres in inflammatory myopathies and muscular dystrophies express LTβ possibly reflecting early myofibre damage, representing a hitherto undescribed pathologic hallmark. Furthermore, we visualised LTβ in several inflammatory cell types in inflammatory myopathies, suggesting its involvement in the different inflammatory mechanisms underlying inflammatory myopathy subgroups.
Collapse
Affiliation(s)
- Kim K Creus
- Laboratory for Neuropathology, Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | | | | | | |
Collapse
|
11
|
Oliver JA, Maarouf O, Cheema FH, Liu C, Zhang QY, Kraus C, Zeeshan Afzal M, Firdous M, Klinakis A, Efstratiadis A, Al-Awqati Q. SDF-1 activates papillary label-retaining cells during kidney repair from injury. Am J Physiol Renal Physiol 2012; 302:F1362-73. [PMID: 22461304 DOI: 10.1152/ajprenal.00202.2011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The adult kidney contains a population of low-cycling cells that resides in the papilla. These cells retain for long periods S-phase markers given as a short pulse early in life; i.e., they are label-retaining cells (LRC). In previous studies in adult rat and mice, we found that shortly after acute kidney injury many of the quiescent papillary LRC started proliferating (Oliver JA, Klinakis A, Cheema FH, Friedlander J, Sampogna RV, Martens TP, Liu C, Efstratiadis A, Al-Awqati Q. J Am Soc Nephrol 20: 2315-2327, 2009; Oliver JA, Maarouf O, Cheema FH, Martens TP, Al-Awqati Q. J Clin Invest 114: 795-804, 2004) and, with cell-tracking experiments, we found upward migration of some papillary cells including LRC (Oliver JA, Klinakis A, Cheema FH, Friedlander J, Sampogna RV, Martens TP, Liu C, Efstratiadis A, Al-Awqati Q. J Am Soc Nephrol 20: 2315-2327, 2009). To identify molecular cues involved in the activation (i.e., proliferation and/or migration) of the papillary LRC that follows injury, we isolated these cells from the H2B-GFP mice and found that they migrated and proliferated in response to the cytokine stromal cell-derived factor-1 (SDF-1). Moreover, in a papillary organ culture assay, the cell growth out of the upper papilla was dependent on the interaction of SDF-1 with its receptor Cxcr4. Interestingly, location of these two proteins in the kidney revealed a complementary location, with SDF-1 being preferentially expressed in the medulla and Cxcr4 more abundant in the papilla. Blockade of Cxcr4 in vivo prevented mobilization of papillary LRC after transient kidney ischemic injury and worsened its functional consequences. The data indicate that the SDF-1/Cxcr4 axis is a critical regulator of papillary LRC activation following transient kidney injury and during organ repair.
Collapse
Affiliation(s)
- Juan A Oliver
- 1Department of Medicine, Columbia University, 630 West 168th St., New York, NY 10032, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Shadrach JL, Wagers AJ. Stem cells for skeletal muscle repair. Philos Trans R Soc Lond B Biol Sci 2011; 366:2297-306. [PMID: 21727135 DOI: 10.1098/rstb.2011.0027] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle is a highly specialized tissue composed of non-dividing, multi-nucleated muscle fibres that contract to generate force in a controlled and directed manner. Skeletal muscle is formed during embryogenesis from a subset of muscle precursor cells, which generate both differentiated muscle fibres and specialized muscle-forming stem cells known as satellite cells. Satellite cells remain associated with muscle fibres after birth and are responsible for muscle growth and repair throughout life. Failure in satellite cell function can lead to delayed, impaired or failed recovery after muscle injury, and such failures become increasingly prominent in cases of progressive muscle disease and in old age. Recent progress in the isolation of muscle satellite cells and elucidation of the cellular and molecular mediators controlling their activity indicate that these cells represent promising therapeutic targets. Such satellite cell-based therapies may involve either direct cell replacement or development of drugs that enhance endogenous muscle repair mechanisms. Here, we discuss recent breakthroughs in understanding both the cell intrinsic and extrinsic regulators that determine the formation and function of muscle satellite cells, as well as promising paths forward to realizing their full therapeutic potential.
Collapse
Affiliation(s)
- Jennifer L Shadrach
- Department of Stem Cell and Regenerative Biology, Howard Hughes Medical Institute, Harvard University and Joslin Diabetes Center, Bauer Center, 7 Divinity Avenue, Cambridge, MA 02138, USA
| | | |
Collapse
|
13
|
Karouzakis E, Rengel Y, Jüngel A, Kolling C, Gay RE, Michel BA, Tak PP, Gay S, Neidhart M, Ospelt C. DNA methylation regulates the expression of CXCL12 in rheumatoid arthritis synovial fibroblasts. Genes Immun 2011; 12:643-52. [PMID: 21753787 DOI: 10.1038/gene.2011.45] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In the search for specific genes regulated by DNA methylation in rheumatoid arthritis (RA), we investigated the expression of CXCL12 in synovial fibroblasts (SFs) and the methylation status of its promoter and determined its contribution to the expression of matrix metalloproteinases (MMPs). DNA was isolated from SFs and methylation was analyzed by bisulfite sequencing and McrBC assay. CXCL12 protein was quantified by enzyme-linked immunosorbent assay before and after treatment with 5-azacytidine. RASFs were transfected with CXCR7-siRNA and stimulated with CXCL12. Expression of MMPs was analyzed by real-time PCR. Basal expression of CXCL12 was higher in RASFs than osteoarthritis (OA) SFs. 5-azacytidine demethylation increased the expression of CXCL12 and reduced the methylation of CpG nucleotides. A lower percentage of CpG methylation was found in the CXCL12 promoter of RASFs compared with OASFs. Overall, we observed a significant correlation in the mRNA expression and the CXCL12 promoter DNA methylation. Stimulation of RASFs with CXCL12 increased the expression of MMPs. CXCR7 but not CXCR4 was expressed and functional in SFs. We show here that RASFs produce more CXCL12 than OASFs due to promoter methylation changes and that stimulation with CXCL12 activates MMPs via CXCR7 in SFs. Thereby we describe an endogenously activated pathway in RASFs, which promotes joint destruction.
Collapse
Affiliation(s)
- E Karouzakis
- Center of Experimental Rheumatology and Zurich Center of Integrative Human Physiology (ZIHP), University Hospital Zurich, Zurich, Switzerland.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Heinisch S, Kirby LG. SDF-1alpha/CXCL12 enhances GABA and glutamate synaptic activity at serotonin neurons in the rat dorsal raphe nucleus. Neuropharmacology 2010; 58:501-14. [PMID: 19755127 PMCID: PMC2813394 DOI: 10.1016/j.neuropharm.2009.08.022] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Revised: 08/21/2009] [Accepted: 08/31/2009] [Indexed: 12/11/2022]
Abstract
The serotonin (5-hydroxytryptamine; 5-HT) system has a well-characterized role in depression. Recent reports describe comorbidities of mood-immune disorders, suggesting an immunological component may contribute to the pathogenesis of depression as well. Chemokines, immune proteins which mediate leukocyte trafficking, and their receptors are widely distributed in the brain, mediate neuronal patterning, and modulate various neuropathologies. The purpose of this study was to investigate the neuroanatomical relationship and functional impact of the chemokine stromal cell-derived factor-1alpha/CXCL12 and its receptor, CXCR4, on the serotonin dorsal raphe nucleus (DRN) system in the rat using anatomical and electrophysiological techniques. Immunohistochemical analysis indicates that over 70% of 5-HT neurons colocalize with CXCL12 and CXCR4. At a subcellular level, CXCL12 localizes throughout the cytoplasm whereas CXCR4 concentrates to the outer membrane and processes of 5-HT neurons. CXCL12 and CXCR4 also colocalize on individual DRN cells. Furthermore, electrophysiological studies demonstrate CXCL12 depolarization of 5-HT neurons indirectly via glutamate synaptic inputs. CXCL12 also enhances the frequency of spontaneous inhibitory and excitatory postsynaptic currents (sIPSC and sEPSC). CXCL12 concentration-dependently increases evoked IPSC amplitude and decreases evoked IPSC paired-pulse ratio selectively in 5-HT neurons, effects blocked by the CXCR4 antagonist AMD3100. These data indicate presynaptic enhancement of GABA and glutamate release at 5-HT DRN neurons by CXCL12. Immunohistochemical analysis further shows CXCR4 localization to DRN GABA neurons, providing an anatomical basis for CXCL12 effects on GABA release. Thus, CXCL12 indirectly modulates 5-HT neurotransmission via GABA and glutamate synaptic afferents. Future therapies targeting CXCL12 and other chemokines may treat serotonin related mood disorders, particularly depression experienced by immune-compromised individuals.
Collapse
Affiliation(s)
- Silke Heinisch
- Department of Anatomy and Cell Biology & Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | |
Collapse
|
15
|
|
16
|
Creus KK, De Paepe B, Werbrouck BF, Vervaet V, Weis J, De Bleecker JL. Distribution of the NF-κB Complex in the Inflammatory Exudates Characterizing the Idiopathic Inflammatory Myopathies. Ann N Y Acad Sci 2009; 1173:370-7. [DOI: 10.1111/j.1749-6632.2009.04874.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
17
|
Laird DJ, von Andrian UH, Wagers AJ. Stem cell trafficking in tissue development, growth, and disease. Cell 2008; 132:612-30. [PMID: 18295579 DOI: 10.1016/j.cell.2008.01.041] [Citation(s) in RCA: 233] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Regulated movement of stem cells is critical for organogenesis during development and for homeostasis and repair in adulthood. Here we analyze the biological significance and molecular mechanisms underlying stem cell trafficking in the generation of the germline, and the generation and regeneration of blood and muscle. Comparison across organisms and lineages reveals remarkable conservation as well as specialization in homing and migration mechanisms used by mature leukocytes, adult and fetal stem cells, and cancer stem cells. In vivo trafficking underpins the successful therapeutic application of hematopoietic stem cells for bone-marrow transplant, and further elucidation of homing and migration pathways in other systems will enable broader application of stem cells for targeted cell therapy and drug delivery.
Collapse
Affiliation(s)
- Diana J Laird
- Developmental Biology Program, Sloan Kettering Institute, 1275 York Avenue, New York, NY 10058, USA
| | | | | |
Collapse
|
18
|
Morosetti R, Mirabella M, Gliubizzi C, Broccolini A, De Angelis L, Tagliafico E, Sampaolesi M, Gidaro T, Papacci M, Roncaglia E, Rutella S, Ferrari S, Tonali PA, Ricci E, Cossu G. MyoD expression restores defective myogenic differentiation of human mesoangioblasts from inclusion-body myositis muscle. Proc Natl Acad Sci U S A 2006; 103:16995-7000. [PMID: 17077152 PMCID: PMC1636567 DOI: 10.1073/pnas.0603386103] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2006] [Indexed: 11/18/2022] Open
Abstract
Inflammatory myopathies (IM) are acquired diseases of skeletal muscle comprising dermatomyositis (DM), polymyositis (PM), and inclusion-body myositis (IBM). Immunosuppressive therapies, usually beneficial for DM and PM, are poorly effective in IBM. We report the isolation and characterization of mesoangioblasts, vessel-associated stem cells, from diagnostic muscle biopsies of IM. The number of cells isolated, proliferation rate and lifespan, markers expression, and ability to differentiate into smooth muscle do not differ among normal and IM mesoangioblasts. At variance with normal, DM and PM mesoangioblasts, cells isolated from IBM, fail to differentiate into skeletal myotubes. These data correlate with lack in connective tissue of IBM muscle of alkaline phosphatase (ALP)-positive cells, conversely dramatically increased in PM and DM. A myogenic inhibitory basic helix-loop-helix factor B3 is highly expressed in IBM mesoangioblasts. Indeed, silencing this gene or overexpressing MyoD rescues the myogenic defect of IBM mesoangioblasts, opening novel cell-based therapeutic strategies for this crippling disorder.
Collapse
MESH Headings
- Alkaline Phosphatase/metabolism
- Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Cell Differentiation
- Cells, Cultured
- Gene Expression
- Gene Silencing
- Humans
- Muscle Development
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- MyoD Protein/genetics
- MyoD Protein/metabolism
- Myoblasts, Skeletal/metabolism
- Myoblasts, Skeletal/pathology
- Myositis, Inclusion Body/metabolism
- Myositis, Inclusion Body/pathology
- Myositis, Inclusion Body/therapy
- RNA, Small Interfering/genetics
Collapse
Affiliation(s)
- Roberta Morosetti
- *Department of Neurosciences and
- Interdisciplinary Laboratory for Stem Cell Research and Cellular Therapy, Catholic University, Largo A. Gemelli 8, 00168 Rome, Italy
| | | | - Carla Gliubizzi
- *Department of Neurosciences and
- Fondazione Don Carlo Gnocchi, 00194 Rome, Italy
| | | | - Luciana De Angelis
- Department of Histology and Embriology, University “La Sapienza,” 00161 Rome, Italy
| | - Enrico Tagliafico
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | | | | | | | - Enrica Roncaglia
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | - Sergio Rutella
- Institute of Hematology, Catholic University, 00168 Rome, Italy; and
| | - Stefano Ferrari
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, 41100 Modena, Italy
| | | | - Enzo Ricci
- *Department of Neurosciences and
- Fondazione Don Carlo Gnocchi, 00194 Rome, Italy
| | - Giulio Cossu
- Institute of Cell Biology and Tissue Engineering, San Raffaele Biomedical Science Park, 00128 Rome, Italy
- **Stem Cell Research Institute, San Raffaele Hospital, 20132 Milan, Italy
- Department of Biology, University of Milan, 20133 Milan, Italy
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW We provide an update of progress gained from research into sporadic inclusion body myositis (s-IBM). RECENT FINDINGS Most research on s-IBM has focused on the inflammatory reaction or the accumulation of pathological proteins in vacuolated muscle fibres. The inflammatory reaction is characterized by clonal expansions of lymphocytes, predominantly CD8 cytotoxic T cells, which invade and destroy muscle fibres. That costimulatory molecules have been identified demonstrates that muscle fibres can act as antigen presenting cells, and the expression of various chemokines in muscle indicates their importance in the immunopathogenesis of s-IBM. The region of interest for a susceptibility gene in the major histocompatibility complex has been narrowed, and for the first time it has been demonstrated that a chronic viral infection can trigger the inflammatory process leading to s-IBM. The nature of the accumulated material associated with the vacuoles has been extensively investigated over the past few years. Amyloid-beta and phosphorylated tau protein in intracellular inclusions are a characteristic finding in s-IBM, which may lead to calcium dyshomeostasis and endoplasmic reticulum stress. The proteasomal system is upregulated, including immunoproteasomes. 'Molecular misreading' leading to ubiquitin mRNA mutations and accumulation of pathological ubiquitin in muscle fibres may be associated with proteasomal dysfunction. There is still no efficient treatment for s-IBM, but the effects of new, more specific immunotherapies have begun to be explored. SUMMARY Recent findings indicate that both inflammatory reaction and abnormal protein accumulation are important for the pathogenesis in s-IBM. The link between them continues to await elucidation.
Collapse
Affiliation(s)
- Anders Oldfors
- Göteborg Neuromuscular Center, Department of Pathology, Sahlgrenska University Hospital, Göteborg, Sweden.
| | | |
Collapse
|
20
|
Schreiner B, Voss J, Wischhusen J, Dombrowski Y, Steinle A, Lochmüller H, Dalakas M, Melms A, Wiendl H. Expression of toll-like receptors by human muscle cells in vitro and in vivo: TLR3 is highly expressed in inflammatory and HIV myopathies, mediates IL-8 release and up-regulation of NKG2D-ligands. FASEB J 2005; 20:118-20. [PMID: 16293707 DOI: 10.1096/fj.05-4342fje] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The particular microenvironment of the skeletal muscle can be the site of complex immune reactions. Toll-like receptors (TLRs) mediate inflammatory stimuli from pathogens and endogenous danger signals and link the innate and adaptive immune system. We investigated innate immune responses in human muscle. Analyzing TLR1-9 mRNA in cultured myoblasts and rhabdomyosarcoma cells, we found constitutive expression of TLR3. The TLR3 ligand Poly (I:C), a synthetic analog of dsRNA, and IFN-gamma increased TLR3 levels. TLR3 was mainly localized intracellularly and regulated at the protein level. Poly (I:C) challenge 1) activated nuclear factor-kappaB (NF-kappaB), 2) increased IL-8 release, and 3) up-regulated NKG2D ligands and NK-cell-mediated lysis of muscle cells. We examined muscle biopsy specimens of 6 HIV patients with inclusion body myositis/polymyositis (IBM/PM), 7 cases of sporadic IBM and 9 nonmyopathic controls for TLR3 expression. TLR3 mRNA levels were elevated in biopsy specimens from patients with IBM and HIV-myopathies. Muscle fibers in inflammatory myopathies expressed TLR3 in close proximity of infiltrating mononuclear cells. Taken together, our study suggests an important role of TLR3 in the immunobiology of muscle, and has substantial implications for the understanding of the pathogenesis of inflammatory myopathies or therapeutic interventions like vaccinations or gene transfer.
Collapse
Affiliation(s)
- Bettina Schreiner
- Department of General Neurology, Hertie-Institute for Clinical Brain Research, Eberhard-Karls-University Tuebingen, Tuebingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
De Paepe B, De Keyzer K, Martin JJ, De Bleecker JL. Alpha-chemokine receptors CXCR1-3 and their ligands in idiopathic inflammatory myopathies. Acta Neuropathol 2005; 109:576-82. [PMID: 15937690 DOI: 10.1007/s00401-005-0989-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2004] [Revised: 01/14/2005] [Accepted: 01/14/2005] [Indexed: 12/18/2022]
Abstract
The idiopathic inflammatory myopathies (IIM) are a heterogeneous group of neuromuscular disorders subdivided into polymyositis (PM), sporadic inclusion body myositis (sIBM) and dermatomyositis (DM). Chemokines play an essential role in sustained inflammation associated with IIM. We studied the distribution of the alpha-chemokine receptors CXCR1, 2, 3 and their ligands interferon-gamma (IFN-gamma)-inducible T cell alpha chemoattractant (I-TAC), IFN-gamma-inducible protein of 10 kDa (IP-10), monokine induced by IFN-gamma (MIG) and growth-related oncogene (GRO) in IIM using immunohistochemistry, immunofluorescence and Western blotting. Abundant expression of IP-10 was observed on macrophages and T cells surrounding and invading non-necrotic muscle fibers in PM and sIBM and in T cells in perimysial infiltrates of DM. IP-10 was also localized to blood vessel endothelial cells in all inflammatory and normal muscle tissues. The distribution of other alpha-chemokines was variable: Only low levels of MIG and I-TAC were detected; GRO was localized to the endomysial infiltrates of some PM and sIBM samples, but not in DM. Muscle tissues were invariably CXCR1 negative, while a subset of inflammatory cells in all IIM were CXCR2 positive. Strong CXCR3 expression was observed on the majority of T cells in each IIM. We describe the differential repertoire of alpha-chemokines in IIM, and offer additional proof of the predominance of Th1-driven reactions in the immunopathogenesis of all three diagnostic subgroups. We suggest the Th1-mediated immunity in general, and the CXCR3/IP-10 interaction in particular, as potential targets for novel therapeutic intervention in IIM.
Collapse
Affiliation(s)
- Boel De Paepe
- Department of Neurology, Ghent University Hospital, De Pintelaan 185, 9000, Ghent, Belgium
| | | | | | | |
Collapse
|
22
|
Abstract
PURPOSE OF REVIEW The etiology and much about the pathogenesis of the inflammatory myopathies remain a mystery. In this review, we investigate recent research efforts to understand the pathogenesis of the diverse entities of polymyositis (PM), dermatomyositis (DM), and inclusion body myositis (IBM), diseases that result from interactions between environmental risk factors and genetic susceptibility. RECENT FINDINGS Over the past year, there has been considerable progress toward better understanding of IBM, with relatively few developments toward understanding PM and DM. Although these diseases may share some common clinical phenotypic and serologic components, they differ on a molecular and cellular level. SUMMARY The need for definitive, safer therapies in these diseases makes vital the search for defining detailed pathogenesis of inflammation and muscle fiber damage at the molecular level.
Collapse
Affiliation(s)
- Lisa Christopher-Stine
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | |
Collapse
|
23
|
De Paepe B, De Bleecker JL. ?-chemokine receptor expression in idiopathic inflammatory myopathies. Muscle Nerve 2005; 31:621-7. [PMID: 15772970 DOI: 10.1002/mus.20294] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Beta-chemokines attract and activate T cells and monocytes and have a key role in chronic inflammation. Certain beta-chemokines, such as monocyte chemoattractant protein-1 (MCP-1), have been reported to be upregulated in the idiopathic inflammatory myopathies (IIM). We studied the distribution of beta-chemokine receptors in polymyositis (PM), sporadic inclusion-body myositis (sIBM), dermatomyositis (DM), and control samples. CCR1-5 were localized to blood vessels in all samples. In addition, increased endothelial expression of CCR2A was observed in IIM. Subsets of inflammatory cells, identified as macrophages and T cells, in all three types of IIM expressed CCR2A, CCR2B, CCR3, CCR4, and CCR5. In contrast to an earlier report, we found CCR2B to be the most prominent MCP-1 receptor on inflammatory cells in IIM, especially in PM and sIBM. Strong CCR4 expression was present on myonuclei of regenerating muscle fibers. The prominence of the CCR2 receptors further underlines the importance of the interaction with their ligand MCP-1 in the immunopathogenesis of IIM and puts CCR2B forward as a potential target for future therapeutic intervention.
Collapse
MESH Headings
- Blood Vessels/immunology
- Blood Vessels/pathology
- Blotting, Western
- Chemokine CCL2/metabolism
- Chemotaxis, Leukocyte/immunology
- Endothelial Cells/immunology
- Endothelial Cells/metabolism
- Humans
- Immunohistochemistry
- Macrophages/immunology
- Muscle, Skeletal/blood supply
- Muscle, Skeletal/immunology
- Muscle, Skeletal/pathology
- Myositis/immunology
- Myositis/pathology
- Receptors, CCR2
- Receptors, CCR4
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/immunology
- Receptors, Chemokine/metabolism
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Boel De Paepe
- Department of Neurology, Neuromuscular Laboratory, Ghent University Hospital, De Pintelaan 185, B-9000 Ghent, Belgium
| | | |
Collapse
|