1
|
Malaviya R, Laskin JD, Businaro R, Laskin DL. Targeting Tumor Necrosis Factor Alpha to Mitigate Lung Injury Induced by Mustard Vesicants and Radiation. Disaster Med Public Health Prep 2023; 17:e553. [PMID: 37848400 PMCID: PMC10841250 DOI: 10.1017/dmp.2023.178] [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] [Indexed: 10/19/2023]
Abstract
Pulmonary injury induced by mustard vesicants and radiation is characterized by DNA damage, oxidative stress, and inflammation. This is associated with increases in levels of inflammatory mediators, including tumor necrosis factor (TNF)α in the lung and upregulation of its receptor TNFR1. Dysregulated production of TNFα and TNFα signaling has been implicated in lung injury, oxidative and nitrosative stress, apoptosis, and necrosis, which contribute to tissue damage, chronic inflammation, airway hyperresponsiveness, and tissue remodeling. These findings suggest that targeting production of TNFα or TNFα activity may represent an efficacious approach to mitigating lung toxicity induced by both mustards and radiation. This review summarizes current knowledge on the role of TNFα in pathologies associated with exposure to mustard vesicants and radiation, with a focus on the therapeutic potential of TNFα-targeting agents in reducing acute injury and chronic disease pathogenesis.
Collapse
Affiliation(s)
- Rama Malaviya
- Departments of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Jeffrey D. Laskin
- Departments of Environmental and Occupational Health and Justice, School of Public Health, Rutgers University, Piscataway, NJ, USA
| | - Rita Businaro
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Debra L. Laskin
- Departments of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
2
|
Jung S, Gavriiloglou M, Séverac F, Haumesser L, Sayeh A, Chatelus E, Martin T, Huck O. Influence of systemic sclerosis on periodontal health: A case-control study. J Clin Periodontol 2023; 50:1348-1359. [PMID: 37431838 DOI: 10.1111/jcpe.13846] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/12/2023]
Abstract
AIM Patients with systemic sclerosis (SSc) present various clinical and radiological oral manifestations. However, precise evaluation of the oral features associated with diffuse cutaneous SSc (dcSSc) and limited cutaneous SSc (lcSSc) is limited. The objective of this study was to evaluate the periodontal ligament (PDL) surface in SSc patients in comparison with controls. Assessment of oral-health-related quality of life (OHRQoL) and the levels of different biomarkers in the gingival crevicular fluid (GCF) was performed. MATERIALS AND METHODS SSc patients and matched controls underwent standardized oral examination and cone-beam computed tomography (CBCT). Levels of interleukin-6 (IL-6), chemokine (C-X-C motif) ligand 4 (CXCL-4) and matrix metalloproteinase-9 (MMP-9) in the GCF were determined by enzyme-linked immunosorbent assay. PDL surface was measured on CBCT axial views. OHRQoL was quantified using the Mouth Handicap in SSc Scale (MHISS). RESULTS Thirty-nine SSc patients and 39 controls were included. SSc patients exhibited increased PDL surface, higher number of missing teeth as well as elevated IL-6, MMP-9 and CXCL-4 levels. Reduced mouth opening was observed in dcSSc but not in lcSSc patients. MHISS score was higher in dcSSc than in lcSSc patients. Although worse periodontal parameters were found in both subgroups compared with controls, dcSSc patients presented lower gingival inflammation. CONCLUSIONS SSc is associated with PDL space widening, impaired oral health and OHRQoL.
Collapse
Affiliation(s)
- Sophie Jung
- Faculté de Chirurgie Dentaire Robert Frank, Université de Strasbourg, Strasbourg, France
- Pôle de Médecine et de Chirurgie bucco-dentaires, Centre de Référence des maladies rares orales et dentaires (O-Rares), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- INSERM UMR_S 1109 "Molecular ImmunoRheumatology", Fédération de Médecine Translationnelle de Strasbourg (FMTS), LabEx TRANSPLANTEX, Strasbourg, France
| | - Marianna Gavriiloglou
- Faculté de Chirurgie Dentaire Robert Frank, Université de Strasbourg, Strasbourg, France
- Unité Fonctionnelle de Parodontologie, Pôle de Médecine et de Chirurgie bucco-dentaires, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - François Séverac
- Groupe Méthodes en Recherche Clinique (GMRC), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Lucille Haumesser
- Groupe Méthodes en Recherche Clinique (GMRC), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Amira Sayeh
- Unité Fonctionnelle de Radiologie, Pôle de Médecine et de Chirurgie bucco-dentaires, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Emmanuel Chatelus
- Service de Rhumatologie, Centre de Référence des maladies autoimmunes systémiques rares (RESO), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Thierry Martin
- INSERM UMR_S 1109 "Molecular ImmunoRheumatology", Fédération de Médecine Translationnelle de Strasbourg (FMTS), LabEx TRANSPLANTEX, Strasbourg, France
- Faculté de Médecine, Université de Strasbourg, Strasbourg, France
- Service d'Immunologie Clinique, Centre de Référence des Maladies Auto-immunes Systémiques Rares (RESO), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Olivier Huck
- Faculté de Chirurgie Dentaire Robert Frank, Université de Strasbourg, Strasbourg, France
- Unité Fonctionnelle de Parodontologie, Pôle de Médecine et de Chirurgie bucco-dentaires, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS), INSERM UMR 1260 "Regenerative Nanomedicine", Strasbourg, France
| |
Collapse
|
3
|
Papazoglou A, Huang M, Bulik M, Lafyatis A, Tabib T, Morse C, Sembrat J, Rojas M, Valenzi E, Lafyatis R. Epigenetic Regulation of Profibrotic Macrophages in Systemic Sclerosis-Associated Interstitial Lung Disease. Arthritis Rheumatol 2022; 74:2003-2014. [PMID: 35849803 PMCID: PMC9771864 DOI: 10.1002/art.42286] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/26/2022] [Accepted: 06/23/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Systemic sclerosis-associated interstitial lung disease (SSc-ILD) is the leading cause of death in patients with SSc with unclear pathogenesis and limited treatment options. Evidence strongly supports an important role for profibrotic secreted phosphoprotein 1 (SPP1)-expressing macrophages in SSc-ILD. This study was undertaken to define the transcriptome and chromatin structural changes of SPP1 SSc-ILD macrophages in order to better understand their role in promoting fibrosis and to identify transcription factors associated with open chromatin driving their altered phenotype. METHODS We performed single-cell RNA sequencing (scRNA-Seq) on 11 explanted SSc-ILD and healthy control lung samples, as well as single-cell assay for transposase-accessible chromatin sequencing on 5 lung samples to define altered chromatin accessibility of SPP1 macrophages. We predicted transcription factors regulating SPP1 macrophages using single-cell regulatory network inference and clustering (SCENIC) and determined transcription factor binding sites associated with global alterations in SPP1 chromatin accessibility using Signac/Seurat. RESULTS We identified distinct macrophage subpopulations using scRNA-Seq analysis in healthy and SSc-ILD lungs and assessed gene expression changes during the change of healthy control macrophages into SPP1 macrophages. Analysis of open chromatin validated SCENIC predictions, indicating that microphthalmia-associated transcription factor, transcription factor EB, activating transcription factor 6, sterol regulatory element binding transcription factor 1, basic helix-loop-helix family member E40, Kruppel-like factor 6, ETS variant transcription factor 5, and/or members of the activator protein 1 family of transcription factors regulate SPP1 macrophage differentiation. CONCLUSION Our findings shed light on the underlying changes in chromatin structure and transcription factor regulation of profibrotic SPP1 macrophages in SSc-ILD. Similar alterations in SPP1 macrophages may underpin fibrosis in other organs involved in SSc and point to novel targets for the treatment of SSc-ILD, specifically targeting profibrotic macrophages.
Collapse
Affiliation(s)
- Anna Papazoglou
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mengqi Huang
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Melissa Bulik
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Annika Lafyatis
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Tracy Tabib
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Christina Morse
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - John Sembrat
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mauricio Rojas
- Division of Pulmonary, Critical Care and Sleep Medicine, Ohio State University, Columbus, OH, USA
| | - Eleanor Valenzi
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Alpoim-Moreira J, Fernandes C, Pimenta J, Bliebernicht M, Rebordão MR, Castelo-Branco P, Szóstek-Mioduchowska A, Skarzynski DJ, Ferreira-Dias G. Metallopeptidades 2 and 9 genes epigenetically modulate equine endometrial fibrosis. Front Vet Sci 2022; 9:970003. [PMID: 36032279 PMCID: PMC9412240 DOI: 10.3389/fvets.2022.970003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/22/2022] [Indexed: 12/13/2022] Open
Abstract
Endometrium type I (COL1) and III (COL3) collagen accumulation, periglandular fibrosis and mare infertility characterize endometrosis. Metalloproteinase-2 (MMP-2), MMP-9 and tissue inhibitors of metalloproteinases (TIMP-1 and TIMP-2) are involved in collagen turnover. Since epigenetic changes may control fibroproliferative diseases, we hypothesized that epigenetic mechanisms could modulate equine endometrosis. Epigenetic changes can be reversed and therefore extremely promising for therapeutic use. Methylation pattern analysis of a particular gene zone is used to detect epigenetic changes. DNA methylation commonly mediates gene repression. Thus, this study aimed to evaluate if the transcription of some genes involved in equine endometrosis was altered with endometrial fibrosis, and if the observed changes were epigenetically modulated, through DNA methylation analysis. Endometrial biopsies collected from cyclic mares were histologically classified (Kenney and Doig category I, n = 6; category IIA, n = 6; category IIB, n = 6 and category III, n = 6). Transcription of COL1A1, COL1A2, COL3A1, MMP2, MMP9, TIMP1, and TIMP2 genes and DNA methylation pattern by pyrosequencing of COL1A1, MMP2, MMP9, TIMP1 genes were evaluated. Both MMP2 and MMP9 transcripts decreased with fibrosis, when compared with healthy endometrium (category I) (P < 0.05). TIMP1 transcripts were higher in category III, when compared to category I endometrium (P < 0.05). No differences were found for COL1A1, COL1A2, COL3A1 and TIMP2 transcripts between endometrial categories. There were higher methylation levels of (i) COL1A1 in category IIB (P < 0.05) and III (P < 0.01), when compared to category I; (ii) MMP2 in category III, when compared to category I (P < 0.001) and IIA (P < 0.05); and (iii) MMP9 in category III, when compared to category I and IIA (P < 0.05). No differences in TIMP1 methylation levels were observed between endometrial categories. The hypermethylation of MMP2 and MMP9, but not of COL1A1 genes, occurred simultaneously with a decrease in their mRNA levels, with endometrial fibrosis, suggesting that this hypermethylation is responsible for repressing their transcription. Our results show that endometrosis is epigenetically modulated by anti-fibrotic genes (MMP2 and MMP9) inhibition, rather than fibrotic genes activation and therefore, might be promising targets for therapeutic use.
Collapse
Affiliation(s)
- Joana Alpoim-Moreira
- CIISA - Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Carina Fernandes
- CIISA - Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Jorge Pimenta
- CIISA - Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Unidade Estratégica de Investigação e Serviços de Biotecnologia e Recursos Genéticos (UEISBR), Instituto Nacional de Investigação Agrária e Veterinária, I. P. (INIAV), Vairão, Portugal
| | | | - Maria Rosa Rebordão
- CIISA - Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
- Polytechnic of Coimbra, Coimbra Agriculture School, Coimbra, Portugal
| | - Pedro Castelo-Branco
- Faculty of Medicine and Biomedical Sciences (FMCB), University of Algarve, Faro, Portugal
- Algarve Biomedical Center Research Institute (ABC-RI), Faro, Portugal
| | | | | | - Graça Ferreira-Dias
- CIISA - Center for Interdisciplinary Research in Animal Health, Faculty of Veterinary Medicine, University of Lisbon, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
- *Correspondence: Graça Ferreira-Dias
| |
Collapse
|
5
|
Li JY, Feng YH, He YT, Hu LF, Liang L, Zhao ZQ, Chen BZ, Guo XD. Thermosensitive hydrogel microneedles for controlled transdermal drug delivery. Acta Biomater 2022; 153:308-319. [DOI: 10.1016/j.actbio.2022.08.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/05/2022] [Accepted: 08/25/2022] [Indexed: 11/01/2022]
|
6
|
Assessment of disease outcome measures in systemic sclerosis. Nat Rev Rheumatol 2022; 18:527-541. [PMID: 35859133 DOI: 10.1038/s41584-022-00803-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2022] [Indexed: 01/08/2023]
Abstract
The assessment of disease activity in systemic sclerosis (SSc) is challenging owing to its heterogeneous manifestations across multiple organ systems, the variable rate of disease progression and regression, and the relative paucity of patients in early-phase therapeutic trials. Despite some recent successes, most clinical trials have failed to show efficacy, underscoring the need for improved outcome measures linked directly to disease pathogenesis, particularly applicable for biomarker studies focused on skin disease. Current outcome measures in SSc-associated interstitial lung disease and SSc skin disease are largely adequate, although advancing imaging technology and the incorporation of skin mRNA biomarkers might provide opportunities for earlier detection of the therapeutic effect. Biomarkers can further inform pathogenesis, enabling early phase trials to act as reverse translational studies through the incorporation of routine high-throughput sequencing.
Collapse
|
7
|
Tomassetti S, Colby TV, Wells AU, Poletti V, Costabel U, Matucci-Cerinic M. Bronchoalveolar lavage and lung biopsy in connective tissue diseases, to do or not to do? Ther Adv Musculoskelet Dis 2021; 13:1759720X211059605. [PMID: 34900002 PMCID: PMC8664307 DOI: 10.1177/1759720x211059605] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/26/2021] [Indexed: 12/25/2022] Open
Abstract
Bronchoalveolar lavage and lung biopsy (LBx) are helpful in patients with connective tissue diseases (CTD) and interstitial lung diseases (ILD) regardless of cause, including infectious, noninfectious, immunologic, or malignant. The decision whether to perform only bronchoalveolar lavage (BAL), and eventually a subsequent LBx in case of a nondiagnostic lavage, or one single bronchoscopy combining both sampling methods depends on the clinical suspicion, on patient’s characteristics (e.g. increased biopsy risk) and preferences, and on the resources and biopsy techniques available locally (e.g. regular forceps versus cryobiopsy). In CTD-ILD, BAL has major clinical utility in excluding infections and in the diagnosis of specific patterns of acute lung damage (e.g. alveolar hemorrhage, diffuse alveolar damage, and organizing pneumonia). LBx is indicated to exclude neoplasm or diagnose lymphoproliferative lung disorders that in CTD patients are more common than in the general population. Defining BAL cellularity and characterizing the CTD-ILD histopathologic pattern by LBx can be helpful in the differential diagnosis of cases without established CTD [e.g. ILD preceding full-blown CTD, interstitial pneumonia with autoimmune features (IPAF)], but the prognostic and theragnostic role of those findings remains unclear. Few studies in the pretranscriptomics era have investigated the diagnostic and prognostic role of BAL and LBx in CTD-ILD, and it is reasonable to hypothesize that future studies conducted applying innovative techniques on BAL and LBx might open new and unexpected avenues in pathogenesis, diagnosis, and treatment approach to CTD-ILD. This is particularly desirable now that a new drug treatment era is emerging, in which we have more than one therapeutic choice (immunosuppressive agents, antifibrotic drugs, and biological agents). We hope that future research will pave the path toward precision medicine providing data for a more accurate ILD-CTD endotyping that will guide the physicians through targeted therapeutic choices, rather than to the approximative approach ‘one drug fits them all’.
Collapse
Affiliation(s)
- Sara Tomassetti
- Department of Experimental and Clinical Medicine, Careggi University Hospital and University of Florence, 50121 Florence, Italy
| | - Thomas V Colby
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Athol U Wells
- ILD Unit, Pulmonary Medicine, Royal Brompton Hospital, London, UK
| | - Venerino Poletti
- Department of Diseases of the Thorax, GB Morgagni Hospital, Forlì, Italy
| | - Ulrich Costabel
- Center for Interstitial and Rare Lung Diseases, Pneumology Department, Ruhrlandklinik, University Medicine Essen, Essen, Germany
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Careggi University Hospital, Florence, ItalyUnit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
8
|
Leong E, Bezuhly M, Marshall JS. Distinct Metalloproteinase Expression and Functions in Systemic Sclerosis and Fibrosis: What We Know and the Potential for Intervention. Front Physiol 2021; 12:727451. [PMID: 34512395 PMCID: PMC8432940 DOI: 10.3389/fphys.2021.727451] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Systemic sclerosis (SSc) is a chronic debilitating idiopathic disorder, characterized by deposition of excessive extracellular matrix (ECM) proteins such as collagen which leads to fibrosis of the skin and other internal organs. During normal tissue repair and remodeling, the accumulation and turnover of ECM proteins are tightly regulated by the interaction of matrix metalloproteinases (MMPs) and endogenous tissue inhibitors of metalloproteinases (TIMPs). SSc is associated with dysregulation of the activity of these proteolytic and inhibitory proteins within the tissue microenvironment, tipping the balance toward fibrosis. The resultant ECM accumulation further perpetuates tissue stiffness and decreased function, contributing to poor clinical outcomes. Understanding the expression and function of these endogenous enzymes and inhibitors within specific tissues is therefore critical to the development of therapies for SSc. This brief review describes recent advances in our understanding of the functions and mechanisms of ECM remodeling by metalloproteinases and their inhibitors in the skin and lungs affected in SSc. It highlights recent progress on potential candidates for intervention and therapeutic approaches for treating SSc fibrosis.
Collapse
Affiliation(s)
- Edwin Leong
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Michael Bezuhly
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada.,Department of Surgery, Dalhousie University, Halifax, NS, Canada
| | - Jean S Marshall
- Department of Pathology, Dalhousie University, Halifax, NS, Canada.,Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
9
|
Gholami S, Mazidi Z, Pahlavan S, Moslem F, Hosseini M, Taei A, Hesaraki M, Barekat M, Aghdami N, Baharvand H. A Novel Insight into Endothelial and Cardiac Cells Phenotype in Systemic Sclerosis Using Patient-Derived Induced Pluripotent Stem Cell. CELL JOURNAL 2021; 23:273-287. [PMID: 34308570 PMCID: PMC8286459 DOI: 10.22074/cellj.2021.7244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 01/26/2020] [Indexed: 12/03/2022]
Abstract
Objective Systemic sclerosis (SSc) is a connective tissue disease associated with vascular damage and multi organ
fibrotic changes with unknown pathogenesis. Most SSc patients suffer from defective angiogenesis/vasculogenesis
and cardiac conditions leading to high mortality rates. We aimed to investigate the cardiovascular phenotype of SSc by
cardiogenic differentiation of SSc induced pluripotent stem cells (iPSC).
Materials and Methods In this experimental study, we generated iPSC from two diffuse SSc patients, followed by
successful differentiation into endothelial cells (ECs) and cardiomyocytes (CMs).
Results SSc-derived EC (SSc-EC) expressed KDR, a nearly EC marker, similar to healthy control-EC (C1-EC). After
sorting and culturing KDR+ cells, the resulting EC expressed CD31, a late endothelial marker, but vascular endothelial
(VE)-cadherin expression markedly dropped resulting in a functional defect as reflected in tube formation failure of
SSc-EC. Interestingly, upregulation of SNAI1 (snail family transcriptional repressor 1) was observed in SSc-EC which
might underlie VE-cadherin downregulation. Furthermore, SSc-derived CM (SSc-CM) successfully expressed cardiac-
specific markers including ion channels, resulting in normal physiological behavior and responsiveness to cardioactive
drugs.
Conclusion This study provides an insight into impaired angiogenesis observed in SSc patients by evaluating in vitro
cardiovascular differentiation of SSc iPSC.
Collapse
Affiliation(s)
- Sedigheh Gholami
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Zahra Mazidi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Fariba Moslem
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahya Hosseini
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Adeleh Taei
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahdi Hesaraki
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maryam Barekat
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nasser Aghdami
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran. .,Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| |
Collapse
|
10
|
Burgess KA, Herrick AL, Watson REB. Systemic sclerosis skin is a primed microenvironment for soft tissue calcification-a hypothesis. Rheumatology (Oxford) 2021; 60:2517-2527. [PMID: 33585894 DOI: 10.1093/rheumatology/keab156] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/26/2021] [Accepted: 02/10/2021] [Indexed: 12/28/2022] Open
Abstract
Calcinosis cutis, defined as sub-epidermal deposition of calcium salts, is a major clinical problem in patients with SSc, affecting 20-40% of patients. A number of recognized factors associated with calcinosis have been identified, including disease duration, digital ischaemia and acro-osteolysis. Yet, to date, the pathogenesis of SSc-related calcinosis remains unknown, and currently there is no effective disease-modifying pharmacotherapy. Following onset of SSc, there are marked changes in the extracellular matrix (ECM) of the skin, notably a breakdown in the microfibrillar network and accumulation of type I collagen. Our hypothesis is that these pathological changes reflect a changing cellular phenotype and result in a primed microenvironment for soft tissue calcification, with SSc fibroblasts adopting a pro-osteogenic profile, and specific driving forces promoting tissue mineralization. Considering the role of the ECM in disease progression may help elucidate the mechanism(s) behind SSc-related calcinosis and inform the development of future therapeutic interventions.
Collapse
Affiliation(s)
- Kyle A Burgess
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester & Salford Royal NHS Foundation Trust, Manchester, UK
| | - Ariane L Herrick
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester & Salford Royal NHS Foundation Trust, Manchester, UK.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Rachel E B Watson
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester & Salford Royal NHS Foundation Trust, Manchester, UK.,NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
11
|
Guiot J, Njock MS, André B, Gester F, Henket M, de Seny D, Moermans C, Malaise MG, Louis R. Serum IGFBP-2 in systemic sclerosis as a prognostic factor of lung dysfunction. Sci Rep 2021; 11:10882. [PMID: 34035374 PMCID: PMC8149825 DOI: 10.1038/s41598-021-90333-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/07/2021] [Indexed: 01/17/2023] Open
Abstract
Systemic sclerosis (SSc) is a rare connective tissue disease associated with rapid evolving interstitial lung disease (ILD), driving its mortality. Specific biomarkers associated with the progression of this lung disease are highly needed. We aimed to identify specific biomarkers of SSc-ILD to predict the evolution of the disease. For this, we compared prospectively serum levels of several biomarkers associated with lung fibrosis in SSc patients (n = 102), among which SSc-no ILD (n = 63) and SSc-ILD (n = 39), compared to healthy subjects (HS) (n = 39). We also performed a longitudinal study in a subgroup of 28 patients analyzing biomarkers variations and pulmonary function tests over a period of 2 years. Serum level of IGFBP-2 was significantly increased in SSc patients compared to HS, and negatively correlated with pulmonary function (assessed by carbon monoxide transfer coefficient (KCO)) (r = - 0.29, p < 0.01). Two-year longitudinal analysis in a subgroup of 28 SSc patients determined that IGFBP-2 variation was positively correlated with KCO at 2-year follow-up (r = 0.6, p < 0.001). SSc patients with a lower variation of IGFBP-2 (less than 22%) presented significant deterioration of pulmonary function at 2-year follow-up (p < 0.01). ROC curve analysis enabled us to identify that baseline IGFBP-2 > 105 ng/ml was associated with a poor outcome (KCO < 70% predicted) at 2-year follow-up (AUC = 0.75, p < 0.05). We showed for the first time that serum levels of IGFBP-2 might be a prognostic factor of the development of SSc-ILD.
Collapse
Affiliation(s)
- Julien Guiot
- Laboratory of Pneumology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
| | - Makon-Sébastien Njock
- Laboratory of Pneumology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium.
- Laboratory of Rheumatology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium.
| | - Béatrice André
- Laboratory of Rheumatology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
| | - Fanny Gester
- Laboratory of Pneumology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
| | - Monique Henket
- Laboratory of Pneumology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
| | - Dominique de Seny
- Laboratory of Rheumatology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
| | - Catherine Moermans
- Laboratory of Pneumology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
| | - Michel G Malaise
- Laboratory of Rheumatology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
| | - Renaud Louis
- Laboratory of Pneumology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
| |
Collapse
|
12
|
Matyushchenko AG. [Contribution of endopeptidases to changes in scleral biomechanics in axial elongation of the eye]. Vestn Oftalmol 2021; 137:102-107. [PMID: 33881270 DOI: 10.17116/oftalma2021137021102] [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: 11/17/2022]
Abstract
The article describes the main properties of the sclera in patients with axial eye elongation depending on the degree of axial myopia, and presents basic information on the structure, mechanism of action and regulatory activity of matrix metalloproteinases (MMPs). MMPs play the key role in the development of abnormal catabolism of extracellular matrix components of the fibrous capsule - the process leading to changes in scleral structure and biomechanical properties in eyes with anterior-posterior axis elongation.
Collapse
|
13
|
Zhang MY, Fang S, Gao H, Zhang X, Gu D, Liu Y, Wan J, Xie J. A critical role of AREG for bleomycin-induced skin fibrosis. Cell Biosci 2021; 11:40. [PMID: 33622407 PMCID: PMC7903615 DOI: 10.1186/s13578-021-00553-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/10/2021] [Indexed: 12/16/2022] Open
Abstract
We report our discovery of an important player in the development of skin fibrosis, a hallmark of scleroderma. Scleroderma is a fibrotic disease, affecting 70,000 to 150,000 Americans. Fibrosis is a pathological wound healing process that produces an excessive extracellular matrix to interfere with normal organ function. Fibrosis contributes to nearly half of human mortality. Scleroderma has heterogeneous phenotypes, unpredictable outcomes, no validated biomarkers, and no effective treatment. Thus, strategies to slow down scleroderma progression represent an urgent medical need. While a pathological wound healing process like fibrosis leaves scars and weakens organ function, oral mucosa wound healing is a scarless process. After re-analyses of gene expression datasets from oral mucosa wound healing and skin fibrosis, we discovered that several pathways constitutively activated in skin fibrosis are transiently induced during oral mucosa wound healing process, particularly the amphiregulin (Areg) gene. Areg expression is upregulated ~ 10 folds 24hrs after oral mucosa wound but reduced to the basal level 3 days later. During bleomycin-induced skin fibrosis, a commonly used mouse model for skin fibrosis, Areg is up-regulated throughout the fibrogenesis and is associated with elevated cell proliferation in the dermis. To demonstrate the role of Areg for skin fibrosis, we used mice with Areg knockout, and found that Areg deficiency essentially prevents bleomycin-induced skin fibrosis. We further determined that bleomycin-induced cell proliferation in the dermis was not observed in the Areg null mice. Furthermore, we found that inhibiting MEK, a downstream signaling effector of Areg, by selumetinib also effectively blocked bleomycin-based skin fibrosis model. Based on these results, we concluded that the Areg-EGFR-MEK signaling axis is critical for skin fibrosis development. Blocking this signaling axis may be effective in treating scleroderma.
Collapse
Affiliation(s)
- Mary Yinghua Zhang
- Department of Pediatrics, The Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shuyi Fang
- Department of BioHealth Informatics, School of Informatics and Computing At IUPUI, Indiana University, Indianapolis, IN, USA
| | - Hongyu Gao
- The IU Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, USA
| | - Xiaoli Zhang
- Department of Pediatrics, The Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dongsheng Gu
- Department of Pediatrics, The Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yunlong Liu
- Department of BioHealth Informatics, School of Informatics and Computing At IUPUI, Indiana University, Indianapolis, IN, USA
- The IU Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, USA
- The Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jun Wan
- Department of BioHealth Informatics, School of Informatics and Computing At IUPUI, Indiana University, Indianapolis, IN, USA
- The IU Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, USA
- The Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jingwu Xie
- Department of Pediatrics, The Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
- The IU Simon Comprehensive Cancer Center, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
14
|
Matrix Metalloproteinases MMP-2 and MMP-9, Their Inhibitors TIMP-1 and TIMP-2, Vascular Endothelial Growth Factor and sVEGFR-2 as Predictive Markers of Ischemic Retinopathy in Patients with Systemic Sclerosis-Case Series Report. Int J Mol Sci 2020; 21:ijms21228703. [PMID: 33218057 PMCID: PMC7698901 DOI: 10.3390/ijms21228703] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/27/2020] [Accepted: 11/14/2020] [Indexed: 02/06/2023] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune connective tissue disorder associated with multiple organ involvement. The aim of the study was to present two SSc patients who were diagnosed with ischemic retinopathy in both eyes. As a background to our case study, we decided to investigate the imbalance of angiogenesis factors in 25 SSc patients in relation to 25 healthy controls. Assays of matrix metalloproteinases-2 and -9 (MMP-2, MMP-9), tissue inhibitor of metalloproteinases-1 (TIMP-1) and -2 (TIMP-2), vascular endothelial growth factor (VEGF), and soluble VEGF receptor-2 (sVEGFR-2) in blood serum and tears were performed. A significantly increased levels of MMP-9 in serum and tears, (p = 0.0375 and p < 0.001, respectively) as well as VEGF/sVEGFR-2 ratio in tears (p < 0.001) were found in the whole SSc patients group compared with controls, while reduced levels of these parameters in patients with ischemic sclerodermic retinopathy were noted. We also observed decreased level MMP-2 in tears and increased levels of TIMP-2 in blood serum and tears of SSc patients with retinal ischemic changes. MMP-9, MMP-2, TIMP-2, and VEGF/sVEGFR-2 may play a crucial role in ischemic retinal degeneration or retinal reorganization in SSc.
Collapse
|
15
|
Cheikhi AM, Johnson ZI, Julian DR, Wheeler S, Feghali-Bostwick C, Conley YP, Lyons-Weiler J, Yates CC. Prediction of severity and subtype of fibrosing disease using model informed by inflammation and extracellular matrix gene index. PLoS One 2020; 15:e0240986. [PMID: 33095822 PMCID: PMC7584227 DOI: 10.1371/journal.pone.0240986] [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: 10/01/2019] [Accepted: 10/06/2020] [Indexed: 11/19/2022] Open
Abstract
Fibrosis is a chronic disease with heterogeneous clinical presentation, rate of progression, and occurrence of comorbidities. Systemic sclerosis (scleroderma, SSc) is a rare rheumatic autoimmune disease that encompasses several aspects of fibrosis, including highly variable fibrotic manifestation and rate of progression. The development of effective treatments is limited by these variabilities. The fibrotic response is characterized by both chronic inflammation and extracellular remodeling. Therefore, there is a need for improved understanding of which inflammation-related genes contribute to the ongoing turnover of extracellular matrix that accompanies disease. We have developed a multi-tiered method using Naïve Bayes modeling that is capable of predicting level of disease and clinical assessment of patients based on expression of a curated 60-gene panel that profiles inflammation and extracellular matrix production in the fibrotic disease state. Our novel modeling design, incorporating global and parametric-based methods, was highly accurate in distinguishing between severity groups, highlighting the importance of these genes in disease. We refined this gene set to a 12-gene index that can accurately identify SSc patient disease state subsets and informs knowledge of the central regulatory pathways in disease progression.
Collapse
Affiliation(s)
- Amin M. Cheikhi
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States of America
| | - Zariel I. Johnson
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States of America
| | - Dana R. Julian
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States of America
- Department of Health Promotion and Development, University of Pittsburgh School of Nursing, Pittsburgh, PA, United States of America
| | - Sarah Wheeler
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Carol Feghali-Bostwick
- Department of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, United States of America
| | - Yvette P. Conley
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States of America
| | - James Lyons-Weiler
- Genomic and Proteomic Core Laboratories, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Cecelia C. Yates
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States of America
- Department of Health Promotion and Development, University of Pittsburgh School of Nursing, Pittsburgh, PA, United States of America
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
- * E-mail:
| |
Collapse
|
16
|
Utsunomiya A, Oyama N, Hasegawa M. Potential Biomarkers in Systemic Sclerosis: A Literature Review and Update. J Clin Med 2020; 9:E3388. [PMID: 33105647 PMCID: PMC7690387 DOI: 10.3390/jcm9113388] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 12/12/2022] Open
Abstract
Systemic sclerosis (SSc) is a chronic autoimmune disease characterized by dysregulation of the immune system, vascular damage, and fibrosis of the skin and internal organs. Patients with SSc show a heterogeneous phenotype and a range of clinical courses. Therefore, biomarkers that are helpful for precise diagnosis, prediction of clinical course, and evaluation of the therapeutic responsiveness of disease are required in clinical practice. SSc-specific autoantibodies are currently used for diagnosis and prediction of clinical features, as other biomarkers have not yet been fully vetted. Krebs von den Lungen-6 (KL-6), surfactant protein-D (SP-D), and CCL18 have been considered as serum biomarkers of SSc-related interstitial lung disease. Moreover, levels of circulating brain natriuretic peptide (BNP) and N-terminal pro-brain natriuretic peptide (NT-proBNP) can provide diagnostic information and indicate the severity of pulmonary arterial hypertension. Assessment of several serum/plasma cytokines, chemokines, growth factors, adhesion molecules, and other molecules may also reflect the activity or progression of fibrosis and vascular involvement in affected organs. Recently, microRNAs have also been implicated as possible circulating indicators of SSc. In this review, we focus on several potential SSc biomarkers and discuss their clinical utility.
Collapse
Affiliation(s)
| | | | - Minoru Hasegawa
- Department of Dermatology, Divison of Medicine, Faculty of Medical Sciences, University of Fukui, 23-3, Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan; (A.U.); (N.O.)
| |
Collapse
|
17
|
Toyama S, Yamashita T, Saigusa R, Miura S, Nakamura K, Hirabayashi M, Miyagawa T, Fukui Y, Omatsu J, Yoshizaki A, Sato S, Asano Y. Decreased serum cathepsin S levels in patients with systemic sclerosis‐associated interstitial lung disease. J Dermatol 2020; 47:1027-1032. [DOI: 10.1111/1346-8138.15458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/11/2020] [Indexed: 12/26/2022]
Affiliation(s)
- Satoshi Toyama
- Department of Dermatology University of Tokyo Graduate School of Medicine Tokyo Japan
| | - Takashi Yamashita
- Department of Dermatology University of Tokyo Graduate School of Medicine Tokyo Japan
| | - Ryosuke Saigusa
- Department of Dermatology University of Tokyo Graduate School of Medicine Tokyo Japan
| | - Shunsuke Miura
- Department of Dermatology University of Tokyo Graduate School of Medicine Tokyo Japan
| | - Kouki Nakamura
- Department of Dermatology University of Tokyo Graduate School of Medicine Tokyo Japan
| | - Megumi Hirabayashi
- Department of Dermatology University of Tokyo Graduate School of Medicine Tokyo Japan
| | - Takuya Miyagawa
- Department of Dermatology University of Tokyo Graduate School of Medicine Tokyo Japan
| | - Yuki Fukui
- Department of Dermatology University of Tokyo Graduate School of Medicine Tokyo Japan
| | - Jun Omatsu
- Department of Dermatology University of Tokyo Graduate School of Medicine Tokyo Japan
| | - Ayumi Yoshizaki
- Department of Dermatology University of Tokyo Graduate School of Medicine Tokyo Japan
| | - Shinichi Sato
- Department of Dermatology University of Tokyo Graduate School of Medicine Tokyo Japan
| | - Yoshihide Asano
- Department of Dermatology University of Tokyo Graduate School of Medicine Tokyo Japan
| |
Collapse
|
18
|
Guiot J, Henket M, Andre B, Herzog M, Hardat N, Njock MS, Moermans C, Malaise M, Louis R. A new nucleosomic-based model to identify and diagnose SSc-ILD. Clin Epigenetics 2020; 12:124. [PMID: 32807242 PMCID: PMC7430109 DOI: 10.1186/s13148-020-00915-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/30/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Systemic sclerosis (SSc) is a rare connective tissue disease associated with rapid evolving interstitial lung disease (SSc-ILD), driving its mortality. Specific biomarkers associated with the evolution of the lung disease are highly needed. We aimed to identify specific biomarkers of SSc-ILD to predict the evolution of the disease. Nucleosomes are stable DNA/protein complexes that are shed into the blood stream making them ideal candidates for biomarkers. METHODS We studied circulating cell-free nucleosomes (cf-nucleosomes) in SSc patients, 31 with ILD (SSc-ILD) and 67 without ILD. We analyzed plasma levels for cf-nucleosomes and investigated whether global circulating nucleosome levels in association with or without other biomarkers of interest for systemic sclerosis or lung fibrosis (e.g., serum growth factors: IGFBP-1 and the MMP enzyme: MMP-9), could be suitable potential biomarkers for the correct identification of SSc-ILD disease. RESULTS We found that H3.1 nucleosome levels were significantly higher in patients with SSc-ILD compared SSc patients without ILD (p < 0.05) and levels of MMP-9 were significantly increased in patients with SSc-ILD compared to SSc patients without ILD (p < 0.05). Conversely, IGFBP-1 was significantly reduced in patients with SSc-ILD compared to SSc without ILD (p < 0.001). The combination of cf-nucleosomes H3.1 coupled to MMP-9 and IGFBP-1 increased the sensitivity for the differential detection of SSc-ILD. High levels of accuracy were reached with this combined model: its performances are strong with 68.4% of positive predictive value and 77.2% of negative predictive value for 90% of specificity. With our model, we identified a significant negative correlation with FVC % pred (r = -0.22) and TLC % pred (r = -0.31). The value of our model at T1 (baseline) has a predictive power over the Rodnan score at T2 (after 6-18 months), showed by a significant linear regression with R2 = 19% (p = 0.013). We identified in the sole group of SSc-ILD patients a significant linear regression with a R2 = 54.4% with the variation of DLCO between T1 and T2 (p < 0.05). CONCLUSION In our study, we identified a new blood-based model with nucleosomic biomarker in order to diagnose SSc-ILD in a SSc cohort. This model is correlated with TLC and FVC at baseline and predictive of the skin evolution and the DLCO. Further longitudinal exploration studies should be performed in order to evaluate the potential of such diagnostic and predictive model.
Collapse
Affiliation(s)
- Julien Guiot
- Pneumology Department, CHU Liège, Domaine Universitaire du Sart-Tilman, B35, B4000, Liège, Belgium.
| | - Monique Henket
- Pneumology Department, CHU Liège, Domaine Universitaire du Sart-Tilman, B35, B4000, Liège, Belgium
| | - Béatrice Andre
- Rheumatology Department, CHU Liège, Domaine Universitaire du Sart-Tilman, B35, B4000, Liège, Belgium
| | - Marielle Herzog
- Belgian Volition SPRL, Parc Scientifique Créalys, 22 rue Phocas lejeune, B5032, Isnes, Belgium
| | - Nathalie Hardat
- Belgian Volition SPRL, Parc Scientifique Créalys, 22 rue Phocas lejeune, B5032, Isnes, Belgium
| | - Makon-Sebastien Njock
- Pneumology Department, CHU Liège, Domaine Universitaire du Sart-Tilman, B35, B4000, Liège, Belgium
- Rheumatology Department, CHU Liège, Domaine Universitaire du Sart-Tilman, B35, B4000, Liège, Belgium
| | - Catherine Moermans
- Pneumology Department, CHU Liège, Domaine Universitaire du Sart-Tilman, B35, B4000, Liège, Belgium
| | - Michel Malaise
- Rheumatology Department, CHU Liège, Domaine Universitaire du Sart-Tilman, B35, B4000, Liège, Belgium
| | - Renaud Louis
- Pneumology Department, CHU Liège, Domaine Universitaire du Sart-Tilman, B35, B4000, Liège, Belgium
| |
Collapse
|
19
|
Bonhomme O, André B, Gester F, de Seny D, Moermans C, Struman I, Louis R, Malaise M, Guiot J. Biomarkers in systemic sclerosis-associated interstitial lung disease: review of the literature. Rheumatology (Oxford) 2020; 58:1534-1546. [PMID: 31292645 PMCID: PMC6736409 DOI: 10.1093/rheumatology/kez230] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 05/09/2019] [Indexed: 12/18/2022] Open
Abstract
SSc is a rare disease of unknown origin associated with multiple organ involvement. One of the major complications that drives the mortality of SSc patients is interstitial lung disease. The course of SSc-interstitial lung disease progression has a wide spectrum. Since the treatment is based on aggressive immunosuppression it should not be given to stable or non-progressing disease. The correct identification of disease with high risk of progression remains a challenge for early therapeutic intervention, and biomarkers remain urgently needed. In fact, eight categories of biomarkers have been identified and classified according to the different biological pathways involved. The purpose of this article is to describe the main biomarkers thought to be of interest with clinical value in the diagnosis and prognosis of SSc-interstitial lung disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Ingrid Struman
- Molecular Angiogenesis Laboratory, GIGA R, University of Liege, Liège, Belgium
| | | | | | | |
Collapse
|
20
|
Mayali H, Altinisik M, Sencan S, Pirildar T, Kurt E. A multimodal ophthalmic analysis in patients with systemic sclerosis using ocular response analyzer, corneal topography and specular microscopy. Int Ophthalmol 2019; 40:287-296. [PMID: 31564047 DOI: 10.1007/s10792-019-01173-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 09/19/2019] [Indexed: 11/30/2022]
Abstract
PURPOSE To conduct a multimodal ophthalmic evaluation of systemic sclerosis (SSc) in patients using ocular response analyzer (ORA), Pentacam, and specular microscopy (SM). METHODS Thirty-one SSc patients and a group of age- and sex-matched controls were enrolled in this cross-sectional study. Corneal hysteresis (CH), corneal resistance factor (CRF), corneal-compensated intraocular pressure (IOPcc), and Goldmann-correlated IOP (IOPg) were measured with ORA. Anterior chamber depth (ACD), central corneal thickness (CCT), and corneal volume (CV) measurements were obtained using Pentacam. Corneal endothelial cell density (ECD) and CCT were evaluated by SM. RESULTS SSc patients had significantly lower CH, ACD, and ECD values compared to the control group (p = 0.018; < 0.001; < 0.001, respectively). There was no significant difference regarding CRF, IOP, CV, or CCT measurements acquired by Pentacam and SM. Regarding CCT, SM and Pentacam showed relatively better agreement in SSc patients. CONCLUSIONS Multimodal imaging can provide more comprehensive and useful information regarding the ocular involvement of systemic diseases. The multimodal evaluation in our study demonstrated that the pathologic effects of SSc may manifest as reductions in ACD, corneal elasticity, and ECD before there are any detectable changes in corneal thickness or IOP.
Collapse
Affiliation(s)
- Huseyin Mayali
- Ophthalmology Department, Medical School, Manisa Celal Bayar University, Manisa, Turkey
| | - Muhammed Altinisik
- Ophthalmology Department, Medical School, Manisa Celal Bayar University, Manisa, Turkey.
| | - Secil Sencan
- Ophthalmology Department, Tinaztepe Hospital, Izmir, Turkey
| | - Timur Pirildar
- Ophthalmology Department, Medical School, Manisa Celal Bayar University, Manisa, Turkey
| | - Emin Kurt
- Ophthalmology Department, Medical School, Manisa Celal Bayar University, Manisa, Turkey
| |
Collapse
|
21
|
Martinović Kaliterna D, Petrić M. Biomarkers of skin and lung fibrosis in systemic sclerosis. Expert Rev Clin Immunol 2019; 15:1215-1223. [DOI: 10.1080/1744666x.2020.1670062] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
| | - Marin Petrić
- Department of Clinical Immunology and Rheumatology, Department of Internal Medicine, University Hospital of Split, Split, Croatia
| |
Collapse
|
22
|
Liu Y, Luo H, Wang L, Li C, Liu L, Huang L, Liu K, Liu M, Gao S, Xiao Y, Zhu H, Zuo X, Li QZ, Zhang H. Increased Serum Matrix Metalloproteinase-9 Levels are Associated with Anti-Jo1 but not Anti-MDA5 in Myositis Patients. Aging Dis 2019; 10:746-755. [PMID: 31440381 PMCID: PMC6675534 DOI: 10.14336/ad.2018.1120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/20/2018] [Indexed: 12/31/2022] Open
Abstract
Matrix metalloproteinases 9 (MMP9) is a member of the zinc-ion-dependent proteinases family and plays a pathogenic role in chronic inflammatory autoimmune diseases. However, its roles in the pathogenesis of myositis have not been elucidated. In this study, we aimed to determine the gene expression and serum level of MMP9 and their relationship with clinical features and serological parameters in myositis. Our results showed that MMP9 mRNA in peripheral blood mononuclear cells (PBMC) was upregulated in myositis patients compared to that in healthy controls. Myositis patients positive for anti-Jo1 antibodies exhibited significantly higher serum MMP9 than anti-MDA5 positive or antibody-negative patients and healthy controls. However, the presence of interstitial lung disease (ILD) did not affect MMP9 levels. We further identified that anti-Jo1-positive myositis patients showed higher numbers of white blood cells (WBC), lymphocytes and neutrophils; increased levels of creatine kinase (CK), lactate dehydrogenase (LDH), and C-reactive protein (CRP); and higher erythrocyte sedimentation rate (ESR) than anti-MDA5 positive patients. In addition, serum MMP-9 levels were positively correlated with WBCs, neutrophils, CK, CRP, ESR, and LDH in myositis patients. In vitro experiments showed that purified serum IgG from Jo-1-positive patients could stimulate PBMCs to release more MMP9 than the IgG from MDA-5-positive sera. These results indicated that increased MMP9 in anti-Jo1-positive myositis patients was associated with the extent of muscle involvement, but not pulmonary damage. The distinct pattern of serum MMP9 perhaps clarifies the differences in pathophysiology between anti-Jo1 and anti-MDA5 in patients with myositis.
Collapse
Affiliation(s)
- Yanjuan Liu
- 1Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,2Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China
| | - Hui Luo
- 1Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Li Wang
- 1Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,2Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China
| | - Caiyan Li
- 1Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,2Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China
| | - Liyun Liu
- 1Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,2Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China
| | - Li Huang
- 1Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,2Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China
| | - Ke Liu
- 2Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China
| | - Meidong Liu
- 2Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China
| | - Siming Gao
- 1Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yizhi Xiao
- 1Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Honglin Zhu
- 1Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoxia Zuo
- 1Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Quan-Zhen Li
- 1Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,3Department of Immunology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Huali Zhang
- 1Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,2Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China.,4Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Hunan, China
| |
Collapse
|
23
|
Almadori A, Griffin M, Ryan CM, Hunt DF, Hansen E, Kumar R, Abraham DJ, Denton CP, Butler PEM. Stem cell enriched lipotransfer reverses the effects of fibrosis in systemic sclerosis. PLoS One 2019; 14:e0218068. [PMID: 31314805 PMCID: PMC6636710 DOI: 10.1371/journal.pone.0218068] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 05/17/2019] [Indexed: 12/15/2022] Open
Abstract
Oro-facial fibrosis in systemic sclerosis (Scleroderma;SSc) has a major impact on mouth function, facial appearance, and patient quality of life. Lipotransfer is a method of reconstruction that can be used in the treatment of oro-facial fibrosis. The effect of this treatment not only restores oro-facial volume but has also been found to reverse the effects of oro-facial fibrosis. Adipose derived stem cells (ADSCs) within the engrafted adipose tissue have been shown to be anti-fibrotic in SSc and are proposed as the mechanism of the anti-fibrotic effect of lipotransfer. A cohort of 62 SSc patients with oro-facial fibrosis were assessed before and after stem cell enriched lipotransfer treatment. Clinical evaluation included assessment of mouth function using a validated assessment tool (Mouth Handicap in Systemic Sclerosis Scale-MHISS), validated psychological measurements and pre and post-operative volumetric assessment. In addition, to understand the mechanism by which the anti-fibrotic effect of ADSCs occur, SSc derived fibroblasts and ADSCs from this cohort of patients were co-cultured in direct and indirect culture systems and compared to monoculture controls. Cell viability, DNA content, protein secretion of known fibrotic mediators including growth factor- β1 (TGF β-1) and connective tissue growth factor (CTGF) using ELISA analysis and fibrosis gene expression using a fibrosis pathway specific qPCR array were evaluated. Mouth function (MHISS) was significantly improved (6.85±5.07) (p<0.0001) after treatment. All psychological measures were significantly improved: DAS 24 (12.1±9.5) (p<0.0001); HADS-anxiety (2.8±3.2) (p<0.0001), HADS-depression (2.0±3.1) (p<0.0001); BFNE (2.9 ± 4.3) (p<0.0001); VAS (3.56±4.1) (p<0.0001). Multiple treatments further improved mouth function (p<0.05), DAS (p<0.0001) and VAS (p = 0.01) scores. SSc fibroblast viability and proliferation was significantly reduced in co-culture compared to monoculture via a paracrine effect over 14 days (p < 0.0001). Protein secretion of transforming growth factor (TGF-β1) and connective tissue growth factor (CTGF) was significantly reduced in co-culture compared to monoculture (p < 0.0001). Multiple fibrosis associated genes were down regulated in SSc co-culture compared to monoculture after 14 days including Matrix metalloproteinase-8 (MMMP-8), Platelet derived growth factor-β (PDGF-β) and Integrin Subunit Beta 6 (ITG-β6). Autologous stem cell enriched lipotransfer significantly improved the effects of oro-facial fibrosis in SSc in this open cohort study. Lipotransfer may reduce dermal fibrosis through the suppression of fibroblast proliferation and key regulators of fibrogenesis including TG-β1 and CTGF. Our findings warrant further investigation in a randomised controlled trial.
Collapse
Affiliation(s)
- Aurora Almadori
- UCL Division of Surgery & Interventional Science, University College London, London, United Kingdom
- Department of Plastic Surgery, Royal Free London NHS Foundation Trust Hospital, London, United Kingdom
- The Charles Wolfson Center for Reconstructive Surgery, Royal Free London NHS Foundation Trust Hospital, London, United Kingdom
| | - Michelle Griffin
- UCL Division of Surgery & Interventional Science, University College London, London, United Kingdom
- Department of Plastic Surgery, Royal Free London NHS Foundation Trust Hospital, London, United Kingdom
- The Charles Wolfson Center for Reconstructive Surgery, Royal Free London NHS Foundation Trust Hospital, London, United Kingdom
- * E-mail: (MG); (PEMB)
| | - Caroline M. Ryan
- UCL Division of Surgery & Interventional Science, University College London, London, United Kingdom
- Department of Plastic Surgery, Royal Free London NHS Foundation Trust Hospital, London, United Kingdom
| | - Debbie F. Hunt
- UCL Division of Surgery & Interventional Science, University College London, London, United Kingdom
- Department of Plastic Surgery, Royal Free London NHS Foundation Trust Hospital, London, United Kingdom
| | - Esther Hansen
- Department of Plastic Surgery, Royal Free London NHS Foundation Trust Hospital, London, United Kingdom
| | - Ravi Kumar
- UCL Division of Surgery & Interventional Science, University College London, London, United Kingdom
- Department of Plastic Surgery, Royal Free London NHS Foundation Trust Hospital, London, United Kingdom
- The Charles Wolfson Center for Reconstructive Surgery, Royal Free London NHS Foundation Trust Hospital, London, United Kingdom
| | - David J. Abraham
- Centre for Rheumatology, UCL Division of Medicine and Royal Free London NHS Foundation Trust Hospital, London, United Kingdom
| | - Christopher P. Denton
- Centre for Rheumatology, UCL Division of Medicine and Royal Free London NHS Foundation Trust Hospital, London, United Kingdom
| | - Peter E. M. Butler
- UCL Division of Surgery & Interventional Science, University College London, London, United Kingdom
- Department of Plastic Surgery, Royal Free London NHS Foundation Trust Hospital, London, United Kingdom
- The Charles Wolfson Center for Reconstructive Surgery, Royal Free London NHS Foundation Trust Hospital, London, United Kingdom
- Centre for Rheumatology, UCL Division of Medicine and Royal Free London NHS Foundation Trust Hospital, London, United Kingdom
- * E-mail: (MG); (PEMB)
| |
Collapse
|
24
|
Summer R, Krishna R, Schriner D, Cuevas-Mora K, Sales D, Para R, Roman J, Nieweld C, Gochuico BR, Romero F. Matrix metalloproteinase activity in the lung is increased in Hermansky-Pudlak syndrome. Orphanet J Rare Dis 2019; 14:162. [PMID: 31272455 PMCID: PMC6610946 DOI: 10.1186/s13023-019-1143-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/25/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hermansky-Pudlak syndrome (HPS) is a rare autosomal recessive disorder characterized by oculocutaneous albinism and platelet dysfunction and can sometimes lead to a highly aggressive form of pulmonary fibrosis that mimics the fatal lung condition called idiopathic pulmonary fibrosis (IPF). Although the activities of various matrix metalloproteinases (MMPs) are known to be dysregulated in IPF, it remains to be determined whether similar changes in these enzymes can be detected in HPS. RESULTS Here, we show that transcript and protein levels as well as enzymatic activities of MMP-2 and -9 are markedly increased in the lungs of mice carrying the HPS Ap3b1 gene mutation. Moreover, immunohistochemical staining localized this increase in MMP expression to the distal pulmonary epithelium, and shRNA knockdown of the Ap3b1 gene in cultured lung epithelial cells resulted in a similar upregulation in MMP-2 and -9 expression. Mechanistically, we found that upregulation in MMP expression associated with increased activity of the serine/threonine kinase Akt, and pharmacological inhibition of this enzyme resulted in a dramatic suppression of MMP expression in Ap3b1 deficient lung epithelial cells. Similarly, levels and activity of different MMPs were also found to be increased in the lungs of mice carrying the Bloc3 HPS gene mutation and in the bronchoalveolar lavage fluid of subjects with HPS. However, an association between MMP activity and disease severity was not detected in these individuals. CONCLUSIONS In summary, our findings indicate that MMP activity is dysregulated in the HPS lung, suggesting a role for these proteases as biological markers or pathogenic players in HPS lung disease.
Collapse
Affiliation(s)
- Ross Summer
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - Rachana Krishna
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - DeLeila Schriner
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - Karina Cuevas-Mora
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - Dominic Sales
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - Rachel Para
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - Jesse Roman
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - Carl Nieweld
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
| | - Bernadette R. Gochuico
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD USA
| | - Freddy Romero
- Center for Translational Medicine and Jane and Leonard Korman Lung Center, Thomas Jefferson University, Philadelphia, USA
- Center for Translational Medicine and Jane and Leonard Korman Respiratory Institute, Thomas Jefferson University, 1020 Locust Street, JAH 354, Philadelphia, PA 19107 USA
| |
Collapse
|
25
|
Coentro JQ, Pugliese E, Hanley G, Raghunath M, Zeugolis DI. Current and upcoming therapies to modulate skin scarring and fibrosis. Adv Drug Deliv Rev 2019; 146:37-59. [PMID: 30172924 DOI: 10.1016/j.addr.2018.08.009] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/08/2018] [Accepted: 08/26/2018] [Indexed: 12/12/2022]
Abstract
Skin is the largest organ of the human body. Being the interface between the body and the outer environment, makes it susceptible to physical injury. To maintain life, nature has endowed skin with a fast healing response that invariably ends in the formation of scar at the wounded dermal area. In many cases, skin remodelling may be impaired, leading to local hypertrophic scars or keloids. One should also consider that the scarring process is part of the wound healing response, which always starts with inflammation. Thus, scarring can also be induced in the dermis, in the absence of an actual wound, during chronic inflammatory processes. Considering the significant portion of the population that is subject to abnormal scarring, this review critically discusses the state-of-the-art and upcoming therapies in skin scarring and fibrosis.
Collapse
Affiliation(s)
- João Q Coentro
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland; Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| | - Eugenia Pugliese
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland; Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| | - Geoffrey Hanley
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland; Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| | - Michael Raghunath
- Center for Cell Biology and Tissue Engineering, Institute for Chemistry and Biotechnology (ICBT), Zurich University of Applied Sciences (ZHAW), Wädenswil, Switzerland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland; Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland.
| |
Collapse
|
26
|
Farhadi E, Mahmoudi M, Rahmani F, Yousefi B, Sarafnejad A, Kavosi H, Karimizadeh E, Jamshidi A, Gharibdoost F. Attenuation of aquaporin-3 and epidermal growth factor receptor expression and activation in systemic sclerosis dermal fibroblasts. J Cell Physiol 2018; 234:12876-12883. [PMID: 30536805 DOI: 10.1002/jcp.27952] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 11/19/2018] [Indexed: 01/08/2023]
Abstract
OBJECTIVES Impaired wound healing and skin dehydration are the mainstay of systemic sclerosis (SSc) cutaneous manifestations. Aquaporin-3 (AQP3) has a pivotal role in skin hydration and wound healing. Epidermal growth factor receptor (EGFR) activation is impaired in SSc fibroblasts. It is unclear whether AQP3 downregulation or epidermal growth factor (EGF) signaling are the primary points of dysregulation in SSc patients. METHODS Skin punch biopsies were obtained from 10 SSc patients and 10 healthy subjects. The mRNA and/or protein expression levels of AQP3, EGFR/p-EGFR, matrix metalloproteinase-1/2/9 (MMP-1/2/9), and tissue inhibitors of metalloproteinase-1 (TIMP1) at baseline and after EGF and transforming growth factor-β1 (TGF-β1) treatment was evaluated in extracted fibroblasts using real-time polymerase chain reaction and western blot analysis. RESULTS SSc fibroblasts expressed lower AQP3 and EGFR, compared with normal fibroblasts. Normal fibroblasts increased AQP3 expression in response to EGF whereas AQP3 expression had no change in EGF-treated-SSc fibroblasts. Likewise, EGFR was activated in response to EGF in the normal group but not SSc group. Baseline expression of MMP-1/2/9 and TIMP1 was not different between SSc and controls. EGF treatment did not result in alteration of any MMPs expression in either of the groups. Combination treatment resulted in a significant upregulation of MMP-1 in normal fibroblasts compared with SSc fibroblasts, while in SSc fibroblasts MMP-9 expression was upregulated in response to treatment with TGF-β1 only. CONCLUSION Downregulation of AQP3 expression in SSc fibroblasts may be related to reduced EGFR expression and activation. TGF-β1 (alone or in combination with EGF) only can upregulate AQP3 expression in SSc fibroblasts so, TGF-β1 affect MMP-1 and MMP-9 just in SSc fibroblasts.
Collapse
Affiliation(s)
- Elham Farhadi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzaneh Rahmani
- Student's Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran.,NeuroImaging Network (NIN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Bahman Yousefi
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Abdolfattah Sarafnejad
- Department of Immunology, School of Public Health and Institute of Public Health Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Hoda Kavosi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Karimizadeh
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Gharibdoost
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Lin J, Wu G, Chen J, Fu C, Hong X, Li L, Liu X, Wu M. Electroacupuncture inhibits sodium nitroprusside‑mediated chondrocyte apoptosis through the mitochondrial pathway. Mol Med Rep 2018; 18:4922-4930. [PMID: 30272266 PMCID: PMC6236293 DOI: 10.3892/mmr.2018.9498] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 08/22/2018] [Indexed: 12/17/2022] Open
Abstract
In China, electroacupuncture (EA) is a therapeutic method that is extensively applied in the clinical treatment of osteoarthritis (OA); however, the underlying molecular mechanism remains unclear. Chondrocyte apoptosis may be observed in cartilage tissue in OA, and is often considered a key target for the treatment of this condition. Therefore, the present study aimed to determine the effects of EA on sodium nitroprusside (SNP)-induced chondrocyte apoptosis. Chondrocytes were obtained from the knee joints of Sprague Dawley rats by type II collagenase digestion. Following microscopic observation and authentication with type II collagen immunohistochemistry, articular cartilage cells were used in subsequent experiments. Using inverted phase contrast microscopy, DAPI staining and flow cytometry, it was revealed that chondrocytes treated with SNP became apoptotic, whereas EA inhibited SNP-induced chondrocyte apoptosis. Subsequently, JC-1 single staining, reverse transcription-quantitative polymerase chain reaction analysis, western blotting, colorimetric assays and immunofluorescence staining were performed for further investigation. The results demonstrated that, when compared with normal chondrocytes, the mitochondrial membrane potential of SNP-treated chondrocytes was markedly lowered, B-cell lymphoma 2 (Bcl-2) expression was reduced, and the expression levels of Bcl-2-associated X protein (Bax), cytochrome c, caspase-9 and caspase-3 were increased. Compared with in SNP-treated chondrocytes, the decrease in the mitochondrial membrane potential of chondrocytes treated with SNP and EA was smaller, Bcl-2 expression was increased, and the expression levels of Bax, cytochrome c, caspase-9 and caspase-3 were decreased following EA intervention. In conclusion, the present study demonstrated that EA modulated the mitochondrial pathway to suppress SNP-mediated chondrocyte apoptosis. Therefore, EA may be of value in the treatment of OA.
Collapse
Affiliation(s)
- Jie Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Guangwen Wu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jun Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Changlong Fu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Xiue Hong
- Department of Acupuncture and Moxibusion, The Second Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Li Li
- Department of Acupuncture and Moxibusion, The Second Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| | - Xianxiang Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Mingxia Wu
- Department of Acupuncture and Moxibusion, The Second Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350003, P.R. China
| |
Collapse
|
28
|
Park MJ, Moon SJ, Lee EJ, Jung KA, Kim EK, Kim DS, Lee JH, Kwok SK, Min JK, Park SH, Cho ML. IL-1-IL-17 Signaling Axis Contributes to Fibrosis and Inflammation in Two Different Murine Models of Systemic Sclerosis. Front Immunol 2018; 9:1611. [PMID: 30042768 PMCID: PMC6048384 DOI: 10.3389/fimmu.2018.01611] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 06/28/2018] [Indexed: 12/24/2022] Open
Abstract
Objective Systemic sclerosis (SSc) is a progressive fibrotic disease that affects the skin and internal organs. Despite evidence implicating increased interleukin-17 (IL-17) activity in SSc, the role of IL-17 in SSc remains uncertain. The purpose of this study was to investigate whether IL-17 plays a pathophysiological role in SSc in two different murine models of SSc. Methods Bleomycin (BLM)-induced fibrosis and chronic graft-versus-host disease (cGVHD) models were used. Histological analysis was performed using Masson's trichrome and immunohistochemical staining. Quantitative reverse transcription-polymerase chain reaction and enzyme-linked immunoassays were used to quantify the messenger RNA and protein levels of inflammatory mediators in dermal fibroblasts. Results IL-1 receptor antagonist-deficient (IL-1Ra-KO) mice were more severely affected by BLM injection, as shown by dermal and pulmonary fibrosis, compared with wild-type (WT) mice. Increased tissue fibrosis was reversed by knocking down IL-17. In vitro experiments showed that IL-1 and IL-17 exerted synergistic effects on the expression of profibrotic and inflammatory mediators. In the cGVHD model, C57BL/6 mice receiving splenocytes of IL-1Ra-KO BALB/c mice developed more severe cGVHD than did those receiving cells from WT mice. Knockdown of IL-17 in IL-1Ra-KO donor mice significantly attenuated the IL-1-induced acceleration of cGVHD severity. Conclusion Targeting IL-1 and its downstream IL-17 activity may be a novel treatment strategy for inhibiting inflammation and tissue fibrosis in SSc.
Collapse
Affiliation(s)
- Min-Jung Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Su-Jin Moon
- Divison of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Eun-Jung Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Kyung-Ah Jung
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Eun-Kyung Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Da-Som Kim
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| | - Jung-Ho Lee
- Department of Plastic and Reconstructive Surgery, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seung-Ki Kwok
- Divison of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jun-Ki Min
- Divison of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Hwan Park
- Divison of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
29
|
Fibroproliferative genes are preferentially expressed in central centrifugal cicatricial alopecia. J Am Acad Dermatol 2018; 79:904-912.e1. [PMID: 29913259 DOI: 10.1016/j.jaad.2018.05.1257] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/28/2018] [Accepted: 05/31/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Central centrifugal cicatricial alopecia (CCCA) is a primary cicatricial alopecia that most commonly affects women of African descent. Like CCCA, fibroproliferative disorders (FPDs) such as keloids, atherosclerosis, and fibroids are characterized by low-grade inflammation and irritation, resulting in end-stage fibrosis. OBJECTIVE We sought to determine whether fibroproliferative genes were up-regulated in patients with CCCA. METHODS A total of 5 patients with biopsy-proven CCCA were recruited for this study. Two scalp biopsy specimens were obtained from each patient; 1 from CCCA-affected vertex scalp and 1 from the unaffected occipital scalp. Microarray analysis was performed to determine the differential gene expression patterns. RESULTS There was an upregulation of genes implicated in FPDs in patients with CCCA. Specifically, we noted increased expression of platelet derived growth factor gene (PDGF), collagen I gene (COL I), collagen III gene (COL III), matrix metallopeptidase 1 gene (MMP1), matrix metallopeptidase 2 gene (MMP2), matrix metallopeptidase 7 gene (MMP7), and matrix metallopeptidase 9 gene (MMP9) in affected scalp compared with in unaffected scalp. Significant overlap in the canonic pathways was noted between patients with CCCA and patients with both atherosclerosis and hepatic fibrosis (P < .001). LIMITATIONS Small sample size and the use of whole skin tissue for analysis. CONCLUSION We have identified the upregulation of critical genes implicated in FPDs in the gene expression profile of patients with CCCA. These findings may help identify future therapeutic targets for this otherwise difficult-to-treat condition.
Collapse
|
30
|
Liphardt AM, Mündermann A, Andriacchi TP, Achtzehn S, Heer M, Mester J. Sensitivity of serum concentration of cartilage biomarkers to 21-days of bed rest. J Orthop Res 2018; 36:1465-1471. [PMID: 29077223 DOI: 10.1002/jor.23786] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 10/13/2017] [Indexed: 02/04/2023]
Abstract
UNLABELLED The objective of the study was to test the hypothesis that serum levels of cartilage oligomeric matrix protein (COMP) would decrease and serum levels of tumor-necrosis factor alpha (TNF-α) and selected matrix metalloproteinases (MMPs) would increase in response to bed rest (BR) and that these changes are unaffected by the intake of potassium bicarbonate or whey protein. Seven and nine healthy male subjects participated in two 21-day 6° head down tilt crossover BR-studies with nutrition interventions. Serum samples were taken before, during, and after BR and biomarker concentrations were measured using commercial enzyme-linked immunosorbent assays. MMP-3 during BR was significantly lower than at baseline (reduction greater 20%; p < 0.001). MMP-3 increased significantly from 14 to 21 days of BR (+7%; p = 0.049). COMP during BR was significantly lower than at baseline (reduction greater 20%; p < 0.001). MMP-3 and COMP returned to baseline within 1 day after BR. MMP-9 on day 3 of BR was significantly lower than at baseline (-31%; p < 0.033) and on days 3, 5, and 14 of BR significantly lower than at the end of and after BR (reduction greater 35%; p < 0.030). The nutritional countermeasures did not affect these results. The observed changes in cartilage biomarkers may be caused by altered cartilage metabolism in response to the lack of mechanical stimulus during BR and inflammatory biomarkers may play a role in changes in biomarker levels. CLINICAL RELEVANCE Immobilization independently from injury can cause altered cartilage biomarker concentration. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1465-1471, 2018.
Collapse
Affiliation(s)
- Anna-Maria Liphardt
- German Sport University Cologne (DSHS Köln), Training Science and Sport Informatics, Köln, Germany.,German Sport University Cologne (DSHS Köln), Biomechanics and Orthopaedics, Köln, Germany.,Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nuremberg (FAU) and Universitätsklinikum, Ulmenweg 18, Erlangen, 91054, Germany
| | - Annegret Mündermann
- Clinic for Orthopaedics and Traumatology, University Hospital Basel, Basel, Switzerland.,Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Thomas P Andriacchi
- Department of Mechanical Engineering, Stanford University,, Stanford, California
| | - Silvia Achtzehn
- German Sport University Cologne (DSHS Köln), Training Science and Sport Informatics, Köln, Germany
| | - Martina Heer
- Department of Nutrition and Food Science, Nutrition Physiology, University of Bonn, Bonn, Germany
| | - Joachim Mester
- German Sport University Cologne (DSHS Köln), Training Science and Sport Informatics, Köln, Germany
| |
Collapse
|
31
|
Kokosi MA, Margaritopoulos GA, Wells AU. Personalised medicine in interstitial lung diseases. Eur Respir Rev 2018; 27:27/148/170117. [DOI: 10.1183/16000617.0117-2017] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 03/05/2018] [Indexed: 12/14/2022] Open
Abstract
Interstitial lung diseases in general, and idiopathic pulmonary fibrosis in particular, are complex disorders with multiple pathogenetic pathways, various disease behaviour profiles and different responses to treatment, all facets that make personalised medicine a highly attractive concept. Personalised medicine is aimed at describing distinct disease subsets taking into account individual lifestyle, environmental exposures, genetic profiles and molecular pathways. The cornerstone of personalised medicine is the identification of biomarkers that can be used to inform diagnosis, prognosis and treatment stratification. At present, no data exist validating a personalised approach in individual diseases. However, the importance of the goal amply justifies the characterisation of genotype and pathway signatures with a view to refining prognostic evaluation and trial design, with the ultimate aim of selecting treatments according to profiles in individual patients.
Collapse
|
32
|
Taylor DK, Mittereder N, Kuta E, Delaney T, Burwell T, Dacosta K, Zhao W, Cheng LI, Brown C, Boutrin A, Guo X, White WI, Zhu J, Dong H, Bowen MA, Lin J, Gao C, Yu L, Ramaswamy M, Gaudreau MC, Woods R, Herbst R, Carlesso G. T follicular helper–like cells contribute to skin fibrosis. Sci Transl Med 2018. [DOI: 10.1126/scitranslmed.aaf5307] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
33
|
Singh SK, Prasad KN, Singh AK, Gupta KK, Singh A, Tripathi M, Gupta RK. Adhesion molecules, chemokines and matrix metallo-proteinases response after albendazole and albendazole plus steroid therapy in swine neurocysticercosis. Exp Parasitol 2017; 182:1-8. [DOI: 10.1016/j.exppara.2017.08.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 06/23/2017] [Accepted: 08/13/2017] [Indexed: 01/15/2023]
|
34
|
Toyama T, Asano Y, Miyagawa T, Nakamura K, Hirabayashi M, Yamashita T, Saigusa R, Miura S, Ichimura Y, Takahashi T, Taniguchi T, Yoshizaki A, Sato S. The impact of transcription factor Fli1 deficiency on the regulation of angiogenesis. Exp Dermatol 2017; 26:912-918. [PMID: 28370536 DOI: 10.1111/exd.13341] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2017] [Indexed: 12/18/2022]
Abstract
The insufficiency of Friend leukaemia virus integration 1 (Fli1), a member of the Ets family transcription factors, is implicated in the pathogenesis of vasculopathy associated with systemic sclerosis (SSc). Fli1 deficiency accelerates early steps of angiogenesis, including detachment of pre-existing pericytes and extracellular matrix degradation by endothelial proteinases, but the impact of Fli1 deficiency on the other steps of angiogenesis has not been investigated. Therefore, we evaluated the effect of Fli1 deficiency on migration, proliferation, cell survival and tube formation of human dermal microvascular endothelial cells (HDMECs). HDMECs transfected with FLI1 siRNA exhibited a greater migratory property in scratch assay and transwell migration assay and a higher proliferation rate in BrdU assay than HDMECs transfected with non-silencing scrambled RNA. In flow cytometry-based apoptosis assay, FLI1 siRNA-transduced HDMECs revealed the decreased number of annexin and propidium iodide-double-positive apoptotic cells compared with control cells, reflecting the promotion of cell survival. On the other hand, tubulogenic activity on Matrigel was remarkably suppressed in Fli1-deficient HDMECs relative to control cells. These results indicate that Fli1 deficiency promotes migration, proliferation and cell survival, while abating tube formation of endothelial cells, suggesting that Fli1 deficiency is potentially attributable to the development of both proliferative obliterative vasculopathy (occlusion of arterioles and small arteries) and destructive vasculopathy (loss of small vessels) characteristic of SSc vasculopathy.
Collapse
Affiliation(s)
- Tetsuo Toyama
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Yoshihide Asano
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takuya Miyagawa
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Kouki Nakamura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Megumi Hirabayashi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takashi Yamashita
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ryosuke Saigusa
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Shunsuke Miura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Yohei Ichimura
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takehiro Takahashi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takashi Taniguchi
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Ayumi Yoshizaki
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Shinichi Sato
- Department of Dermatology, University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
35
|
Multiple receptor-ligand based pharmacophore modeling and molecular docking to screen the selective inhibitors of matrix metalloproteinase-9 from natural products. J Comput Aided Mol Des 2017. [PMID: 28623487 DOI: 10.1007/s10822-017-0028-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Matrix metalloproteinase-9 (MMP-9) is an attractive target for cancer therapy. In this study, the pharmacophore model of MMP-9 inhibitors is built based on the experimental binding structures of multiple receptor-ligand complexes. It is found that the pharmacophore model consists of six chemical features, including two hydrogen bond acceptors, one hydrogen bond donor, one ring aromatic regions, and two hydrophobic (HY) features. Among them, the two HY features are especially important because they can enter the S1' pocket of MMP-9 which determines the selectivity of MMP-9 inhibitors. The reliability of pharmacophore model is validated based on the two different decoy sets and relevant experimental data. The virtual screening, combining pharmacophore model with molecular docking, is performed to identify the selective MMP-9 inhibitors from a database of natural products. The four novel MMP-9 inhibitors of natural products, NP-000686, NP-001752, NP-014331, and NP-015905, are found; one of them, NP-000686, is used to perform the experiment of in vitro bioassay inhibiting MMP-9, and the IC50 value was estimated to be only 13.4 µM, showing the strongly inhibitory activity of NP-000686 against MMP-9, which suggests that our screening results should be reliable. The binding modes of screened inhibitors with MMP-9 active sites were discussed. In addition, the ADMET properties and physicochemical properties of screened four compounds were assessed. The found MMP-9 inhibitors of natural products could serve as the lead compounds for designing the new MMP-9 inhibitors by carrying out structural modifications in the future.
Collapse
|
36
|
Rentka A, Harsfalvi J, Szucs G, Szekanecz Z, Szodoray P, Koroskenyi K, Kemeny-Beke A. Membrane array and multiplex bead analysis of tear cytokines in systemic sclerosis. Immunol Res 2016; 64:619-26. [PMID: 26687127 DOI: 10.1007/s12026-015-8763-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Although serious ocular manifestations of systemic sclerosis (SSc) have been described, tear analysis of patients with SSc has not been performed in previous studies. Our aim was to measure a wide panel of cytokines and chemokines in tears of patients with SSc and to assess the most significant molecules with a more sensitive and specific method. Unstimulated tear samples were collected from nine patients with SSc and 12 age- and gender-matched healthy controls. The relative levels of 102 different cytokines were determined by a cytokine array, and then absolute levels of four key cytokines were determined by a magnetic bead assay. Array results revealed shifted cytokine profile characterized by predominance of inflammatory mediators. Of the 102 analyzed molecules, nine were significantly increased in tears of patients with SSc. Based on the multiplex bead results, C-reactive protein, interferon-γ-inducible protein-10, and monocyte chemoattractant protein-1 levels were significantly higher in tears of patients with SSc. Our current data depict a group of inflammatory mediators, which play a significant role in ocular pathology of SSc; furthermore, they might function as excellent candidates for future therapeutic targets in SSc patients with ocular manifestations.
Collapse
Affiliation(s)
- Aniko Rentka
- Department of Ophthalmology, University of Debrecen, Debrecen, Hungary
| | - Jolan Harsfalvi
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Gabriella Szucs
- Department of Rheumatology, Institute of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Zoltan Szekanecz
- Department of Rheumatology, Institute of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Peter Szodoray
- Institute of Immunology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Krisztina Koroskenyi
- Department of Biochemistry and Molecular Biology, Signaling and Apoptosis Research Group, Hungarian Academy of Sciences, Research Center of Molecular Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Adam Kemeny-Beke
- Department of Ophthalmology, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
37
|
George JA, Bashir G, Qureshi MM, Mohamed YA, Azzi J, Al-Ramadi BK, Fernández-Cabezudo MJ. Cholinergic Stimulation Prevents the Development of Autoimmune Diabetes: Evidence for the Modulation of Th17 Effector Cells via an IFNγ-Dependent Mechanism. Front Immunol 2016; 7:419. [PMID: 27790217 PMCID: PMC5061850 DOI: 10.3389/fimmu.2016.00419] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/27/2016] [Indexed: 12/12/2022] Open
Abstract
Type I diabetes (T1D) results from T cell-mediated damage of pancreatic β-cells and loss of insulin production. The cholinergic anti-inflammatory pathway represents a physiological link connecting the central nervous and immune systems via vagus nerve, and functions to control the release of proinflammatory cytokines. Using the multiple low-dose streptozotocin (MLD-STZ) model to induce experimental autoimmune diabetes, we investigated the potential of regulating the development of hyperglycemia through administration of paraoxon, a highly specific acetylcholinesterase inhibitor (AChEI). We demonstrate that pretreatment with paraoxon prevented hyperglycemia in STZ-treated C57BL/6 mice. This correlated with a reduction in T cell infiltration into pancreatic islets and preservation of the structure and functionality of β-cells. Gene expression analysis of pancreatic tissue revealed that increased peripheral cholinergic activity prevented STZ-mediated loss of insulin production, this being associated with a reduction in IL-1β, IL-6, and IL-17 proinflammatory cytokines. Intracellular cytokine analysis in splenic T cells demonstrated that inhibition of AChE led to a shift in STZ-induced immune response from a predominantly disease-causing IL-17-expressing Th17 cells to IFNγ-positive Th1 cells. Consistent with this conclusion, inhibition of AChE failed to prevent STZ-induced hyperglycemia in IFNγ-deficient mice. Our results provide mechanistic evidence for the prevention of murine T1D by inhibition of AChE and suggest a promising strategy for modulating disease severity.
Collapse
Affiliation(s)
- Junu A George
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab University , Al-Ain , UAE
| | - Ghada Bashir
- Department of Medical Microbiology & Immunology, College of Medicine and Health Sciences, United Arab University , Al-Ain , UAE
| | - Mohammed M Qureshi
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab University , Al-Ain , UAE
| | - Yassir A Mohamed
- Department of Medical Microbiology & Immunology, College of Medicine and Health Sciences, United Arab University , Al-Ain , UAE
| | - Jamil Azzi
- Renal Division, Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School , Boston, MA , USA
| | - Basel K Al-Ramadi
- Department of Medical Microbiology & Immunology, College of Medicine and Health Sciences, United Arab University , Al-Ain , UAE
| | - Maria J Fernández-Cabezudo
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab University , Al-Ain , UAE
| |
Collapse
|
38
|
Ligon C, Hummers LK. Biomarkers in Scleroderma: Progressing from Association to Clinical Utility. Curr Rheumatol Rep 2016; 18:17. [DOI: 10.1007/s11926-016-0565-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
39
|
Şahin M, Yüksel H, Şahin A, Cingü AK, Türkcü FM, Kaya S, Yazmalar L, Batmaz İ. Evaluation of the Anterior Segment Parameters of the Patients with Scleroderma. Ocul Immunol Inflamm 2016; 25:233-238. [DOI: 10.3109/09273948.2015.1115079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Muhammed Şahin
- School of Medicine, Department of Ophthalmology, Dicle University, Diyarbakir, Turkey
| | - Harun Yüksel
- School of Medicine, Department of Ophthalmology, Dicle University, Diyarbakir, Turkey
| | - Alparslan Şahin
- School of Medicine, Department of Ophthalmology, Dicle University, Diyarbakir, Turkey
| | - Abdullah Kürşat Cingü
- School of Medicine, Department of Ophthalmology, Dicle University, Diyarbakir, Turkey
| | - Fatih Mehmet Türkcü
- School of Medicine, Department of Ophthalmology, Dicle University, Diyarbakir, Turkey
| | - Savaş Kaya
- Department of Immunology, Dicle University, Diyarbakir, Turkey
| | - Levent Yazmalar
- Department of Physical Medicine and Rehabilitation, Dicle University, Diyarbakir, Turkey
| | - İbrahim Batmaz
- Department of Physical Medicine and Rehabilitation, Dicle University, Diyarbakir, Turkey
| |
Collapse
|
40
|
Expression of adhesion molecules, chemokines and matrix metallo- proteinases (MMPs) in viable and degenerating stage of Taenia solium metacestode in swine neurocysticercosis. Vet Parasitol 2015; 214:59-66. [PMID: 26412140 DOI: 10.1016/j.vetpar.2015.09.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 08/27/2015] [Accepted: 09/12/2015] [Indexed: 01/13/2023]
Abstract
Neurocysticercosis (NCC) is a parasitic infection of central nervous system (CNS). Expression of adhesion molecules, chemokines and matrix metalloproteinases (MMPs) were investigated on brain tissues surrounding viable (n=15) and degenerating cysticerci (n=15) of Taenia solium in swine by real-time RT-PCR and ELISA. Gelatin gel zymography was performed for MMPs activity. ICAM-1 (intercellular adhesion molecule-1), E-selectin, MIP-1α (macrophage inflammatory protein-1α), Eotaxin-1 and RANTES (regulated on activation, normal T cell expressed and secreted) were associated with degenerating cysticerci (cysts). However, VCAM-1 (vascular cell adhesion molecule-1), MCP-1 (monocyte chemotactic protein-1), MMP-2 and MMP-9 were associated with both viable and degenerating cysts. In conclusion, viable and degenerating cysticerci have different immune molecule profiles and role of these molecules in disease pathogenesis needs to be investigated.
Collapse
|
41
|
Numan MS, Amiable N, Brown JP, Michou L. Paget's disease of bone: an osteoimmunological disorder? DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:4695-707. [PMID: 26316708 PMCID: PMC4544727 DOI: 10.2147/dddt.s88845] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Osteoimmunology represents a large area of research resulting from the cross talk between bone and immune systems. Many cytokines and signaling cascades are involved in the field of osteoimmunology, originating from various cell types. The RANK/receptor activator of nuclear factor Kappa-B ligand (RANKL)/osteoprotegerin (OPG) signaling has a pivotal role in osteoimmunology, in addition to proinflammatory cytokines such as tumor necrosis factor-α, interleukin (IL)-1, IL-6, and IL-17. Clinically, osteoimmunological disorders, such as rheumatoid arthritis, osteoporosis, and periodontitis, should be classified according to their pattern of osteoimmunological serum biomarkers. Paget’s disease of bone is a common metabolic bone disorder, resulting from an excessively increased bone resorption coupled with aberrant bone formation. With the exception of the cellular responses to measles virus nucleocapsid protein and the interferon-gamma signature, the exact role of the immune system in Paget’s disease of bone is not well understood. The cytokine profiles, such as the increased levels of IL-6 and the interferon-gamma signature observed in this disease, are also very similar to those observed in other osteoimmunological disorders. As a potential osteoimmunological disorder, the treatment of Paget’s disease of bone may also benefit from progress made in targeted therapies, in particular for receptor activator of nuclear factor Kappa-B ligand and IL-6 signaling inhibition.
Collapse
Affiliation(s)
- Mohamed S Numan
- CHU de Québec Research Centre, CHU de Québec-Université Laval, Quebec City, QC, Canada ; Division of Rheumatology, Department of Medicine, CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Nathalie Amiable
- CHU de Québec Research Centre, CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Jacques P Brown
- CHU de Québec Research Centre, CHU de Québec-Université Laval, Quebec City, QC, Canada ; Division of Rheumatology, Department of Medicine, CHU de Québec-Université Laval, Quebec City, QC, Canada ; Department of Rheumatology, CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Laëtitia Michou
- CHU de Québec Research Centre, CHU de Québec-Université Laval, Quebec City, QC, Canada ; Division of Rheumatology, Department of Medicine, CHU de Québec-Université Laval, Quebec City, QC, Canada ; Department of Rheumatology, CHU de Québec-Université Laval, Quebec City, QC, Canada
| |
Collapse
|
42
|
Update on biomarkers in systemic sclerosis: tools for diagnosis and treatment. Semin Immunopathol 2015; 37:475-87. [PMID: 26168983 PMCID: PMC4554742 DOI: 10.1007/s00281-015-0506-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 06/16/2015] [Indexed: 01/06/2023]
Abstract
Systemic sclerosis (SSc) is a complex autoimmune disease in which immune activation, vasculopathy, and extensive fibrosis of the skin and internal organs are among the principal features. SSc is a heterogeneous disease with varying manifestations and clinical outcomes. Currently, patients’ clinical evaluation often relies on subjective measures, non-quantitative methods, or requires invasive procedures as markers able to predict disease trajectory or response to therapy are lacking. Therefore, current research is focusing on the discovery of useful biomarkers reflecting ongoing inflammatory or fibrotic activity in the skin and internal organs, as well as being predictive of future disease course. Recently, remarkable progress has been made towards a better understanding of numerous mechanisms involved in the pathogenesis of SSc. This has opened new possibilities for the development of novel biomarkers and therapy. However, current proposed biomarkers that could reliably describe various aspects of SSc still require further investigation. This review will summarize studies describing the commonly used and validated biomarkers, the newly emerging and promising SSc biomarkers identified to date, and consideration of future directions in this field.
Collapse
|
43
|
Campo I, Zorzetto M, Bonella F. Facts and promises on lung biomarkers in interstitial lung diseases. Expert Rev Respir Med 2015; 9:437-57. [DOI: 10.1586/17476348.2015.1062367] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
44
|
Efficacy of cilostazol for the treatment of Raynaud's phenomenon in systemic sclerosis patients. Clin Exp Med 2015; 16:407-12. [PMID: 26088182 DOI: 10.1007/s10238-015-0370-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 06/08/2015] [Indexed: 10/23/2022]
Abstract
Cilostazol is a selective inhibitor of phosphodiesterase-III with antiplatelet, antithrombotic and vasodilating properties. The aim of our study was to evaluate the effect of the drug on vasculopathy and Raynaud's phenomenon (RP), in a series of patients with systemic sclerosis (SSc), before and after cilostazol treatment. Twenty-one consecutive SSc patients with moderate or severe RP were enrolled in an open-label study. Cilostazol was administered at the dose of 100 mg twice a day, for 12 months. Evaluations included: daily RP attack diary documenting the frequency and duration of RP episodes, Health Assessment Questionnaire-Disability Index, scleroderma visual analogue scales (VAS), flow-mediated dilation and immunological status, including endothelin 1 and interleukin 6 plasma levels. Thirteen patients completed the study. RP duration and daily number episodes recorded over a 3-week period significantly decreased after cilostazol treatment (p = 0.0049 and p = 0.0067, respectively). VAS score indicated a significant amelioration of the patients' perception of RP (p = 0.0117), and both baseline and post-ischemic brachial artery diameters were significantly increased after cilostazol treatment, as compared with basal values (p = 0.0119 and p = 0.0076, respectively). None of the patients developed digital ulcers during the study. A significant clinical improvement of RP was recorded in SSc patients undergoing cilostazol treatment. Study results indicate a potential role of cilostazol as oral maintenance therapy in SSc patients with RP.
Collapse
|
45
|
Palkina NV, Shvetsova YI, Kirichenko AK, Ruksha TG. [Inhibition of matrix metalloproteinases 9 and 13 affects the degree of lymphocytic infiltration and the expression levels of microRNA miR-21 and miR-let-7b in melanoma cells in vivo]. Arkh Patol 2015; 77:41-47. [PMID: 25868368 DOI: 10.17116/patol201577141-] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
OBJECTIVE To estimate changes in the trend of growth of primary tumor nodules, the degree of lymphocytic infiltration, and the expression levels of oncomicroRNA miR-21 and miR-let-7b when inhibiting matrix metalloproteinases 9 and 13 (MMP-9 and MMP-13) in vivo in C57B16 mice with transplantable melanoma B-16. MATERIAL AND METHODS Tumor growth was evaluated measuring the volume of primary tumor nodules; the degree of lymphocytic infiltration was microscopically estimated using hematoxylin-eosin-stained tissue specimens, by calculating intratumoral lymphocytes. The expression of oncomicroRNA was quantified by real-time PCR. RESULTS It was shown that MMP-9 and MMP-13 inhibition had no impact on the growth of primary tumor nodules; selective MMP-9 inhibition failed to affect the degree of lymphocytic infiltration of a primary tumor nodule and to change the expression of oncomicroRNA miR-21 and miR-let-7b; the concomitant inhibition of MMP-9 and MMP-13 altered the immunogenic properties of melanoma, stimulated the lymphocytic infiltration of tumor nodules, and decreased the expression of oncomicroRNA miR-21 and miR-let-7b; the degree of lymphocytic infiltration of primary tumor nodules increased in the dynamics of a tumor process and the expression levels of oncomicroRNA remained unchanged. CONCLUSION The concomitant inhibition of MMP-9 and MMP-13 affects prognosis and survival in skin melanoma.
Collapse
Affiliation(s)
- N V Palkina
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Health of Russia, Krasnoyarsk
| | - Yu I Shvetsova
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Health of Russia, Krasnoyarsk
| | - A K Kirichenko
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Health of Russia, Krasnoyarsk
| | - T G Ruksha
- Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Ministry of Health of Russia, Krasnoyarsk
| |
Collapse
|
46
|
|
47
|
Rieder F, de Bruyn JR, Pham BT, Katsanos K, Annese V, Higgins PDR, Magro F, Dotan I. Results of the 4th scientific workshop of the ECCO (Group II): markers of intestinal fibrosis in inflammatory bowel disease. J Crohns Colitis 2014; 8:1166-78. [PMID: 24726695 DOI: 10.1016/j.crohns.2014.03.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 03/12/2014] [Accepted: 03/15/2014] [Indexed: 02/08/2023]
Abstract
The fourth scientific workshop of the European Crohn's and Colitis Organization (ECCO) focused on intestinal fibrosis in inflammatory bowel disease (IBD). The objective was to better understand basic mechanisms and markers of intestinal fibrosis as well as to suggest new therapeutic targets to prevent or treat fibrosis. The results of this workshop are presented in three separate manuscripts. This section describes markers of fibrosis in IBD, identifies unanswered questions in the field and provides a framework for future studies addressing the unmet needs in the field of intestinal fibrosis.
Collapse
Affiliation(s)
- Florian Rieder
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA; Department of Gastroenterology & Hepatology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.
| | - Jessica R de Bruyn
- Academic Medical Center Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam, The Netherlands.
| | - Bao Tung Pham
- Division of Pharmaceutical Technology and Biopharmacy, Department of Pharmacy, University of Groningen, The Netherlands.
| | - Konstantinos Katsanos
- Department of Gastroenterology, University Hospital of Ioannina, Medical School of Ioannina, Greece.
| | - Vito Annese
- Division of Gastroenterology, University Hospital Careggi, Florence, Italy.
| | - Peter D R Higgins
- Division of Gastroenterology, University of Michigan, Ann Arbor, MI, USA.
| | - Fernando Magro
- Department of Pharmacology & Therapeutics, Institute for Molecular and Cell Biology, Faculty of Medicine University of Porto, Porto, Portugal; Department of Gastroenterology, Hospital de Sao Joao, Porto, Portugal.
| | - Iris Dotan
- IBD Center, Department of Gastroenterology and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
48
|
Abstract
Transforming growth factor β (TGF-β) has long been implicated in fibrotic diseases, including the multisystem fibrotic disease systemic sclerosis (SSc). Expression of TGF-β-regulated genes in fibrotic skin and lungs of patients with SSc correlates with disease activity, which points to this cytokine as the central mediator of pathogenesis. Patients with SSc often develop pulmonary arterial hypertension (PAH), a particularly lethal complication caused by vascular dysfunction. Several genetic diseases with vascular features related to SSc, such as familial PAH and hereditary haemorrhagic telangiectasia, are caused by mutations in the TGF-β-sensing ALK-1 signalling pathway. These observations suggest that increased TGF-β signalling causes both vascular and fibrotic features of SSc. The question of how latent TGF-β becomes activated in local SSc tissues is, therefore, central to the understanding of SSc. Both TGF-β1 and TGF-β3 can be activated by integrins αvβ6 and αvβ8, whose upregulation in bronchial epithelial cells can activate TGF-β in SSc lungs. Other αv integrins, thrombospondin-1 or altered TGF-β sequestration by matrix proteins might be important in other target tissues. How the immune system triggers this process remains unclear, although links between inflammation and TGF-β activation are emerging. Together, these observations provide an increasingly secure framework for understanding TGF-β in SSc pathogenesis.
Collapse
Affiliation(s)
- Robert Lafyatis
- Boston University School of Medicine, E5 Arthritis Centre, 72 E. Concord Street, Boston, MA 02118, USA
| |
Collapse
|
49
|
Castelino FV, Varga J. Current status of systemic sclerosis biomarkers: applications for diagnosis, management and drug development. Expert Rev Clin Immunol 2014; 9:1077-90. [PMID: 24168414 DOI: 10.1586/1744666x.2013.848792] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Systemic sclerosis (SSc) is a clinically heterogeneous orphan disease of unknown etiology and no effective therapy. It is characterized by protean manifestations, an unpredictable disease course and variable outcomes. Clinical manifestations reflect underlying autoimmunity, small vessel vasculopathy and progressive multi-organ fibrosis. Predicting disease progression, pattern and severity of complications and response to therapy in SSc remain major challenges both for the management of patients and for the development of effective disease-modifying therapies. This review summarizes contemporary understanding of novel and emerging biomarkers for SSc. We focus on the development of new classification criteria, the utility of SSc-specific autoantibodies as diagnostic and prognostic markers, and on biomarkers for skin and lung involvement. Finally, we review genome-wide expression analysis as a tool to predict therapeutic responses. We anticipate that the development, validation and application of these biomarkers, singly or more likely in combination, will have a transformative impact in SSc, informing early diagnosis, classification and management, as well as the design, execution and interpretation of clinical trials of novel therapeutic agents.
Collapse
Affiliation(s)
- Flavia V Castelino
- Division of Rheumatology, Massachusetts General Hospital, Harvard Medical School, Yawkey 2C-2100, 55 Fruit St, Boston, MA 02114, USA
| | | |
Collapse
|
50
|
Ciurzyński M, Bienias P, Irzyk K, Kostrubiec M, Szewczyk A, Demkow U, Siwicka M, Kurnicka K, Lichodziejewska B, Pruszczyk P. Heart diastolic dysfunction in patients with systemic sclerosis. Arch Med Sci 2014; 10:445-54. [PMID: 25097573 PMCID: PMC4107251 DOI: 10.5114/aoms.2014.43739] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 07/24/2013] [Accepted: 10/14/2013] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION There are limited data on left (LV) and right ventricular (RV) diastolic function in systemic sclerosis (SSc) patients especially in relation to biomarkers of matrix remodeling. The aim of the study was to analyze LV and RV myocardial diastolic function in SSc patients at baseline and after at least 1 year of follow-up and its relation to serum tissue inhibitors of metalloproteinase 1 (TIMP-1) level. MATERIAL AND METHODS We prospectively studied 111 SSc patients (101 female, 10 male, age 54.2 ±13.8 years) and 21 age-matched controls (18 female, 3 male, age 49.3 ±10.5 years). After at least 1 year of observation (3.0 ±1.1 years) we reevaluated 69 of the SSc patients. Transthoracic echocardiography (Philips, iE33) for assessment of LV and RV diastolic function was performed and TIMP-1 serum level was measured. RESULTS Impaired LV relaxation was observed in 38 (34%) SSc patients and in 1 (5%) of the controls (p < 0.001). The mean E/A ratio was lower in patients with SSc than in controls (p = 0.002) and significantly decreased after the follow-up period (p = 0.02). Impaired RV relaxation was detected in 25 (22.5%) SSc patients and in 1 (5%) control subject (p < 0.001) but did not deteriorate after follow-up. Mean serum level of TIMP-1 was significantly elevated in the follow-up group compared to baseline examination (p = 0.0001). Serum TIMP-1 level correlated positively with E/E', both septal and lateral (r = 0.4, p = 0.002 and r = 0.32, p = 0.01). CONCLUSIONS The LV and RV relaxation is impaired in SSc patients. Moreover, left ventricular diastolic function deteriorated after the follow-up period. The TIMP-1 serum levels correlate with echocardiographic parameters, providing a potent link for LV diastolic function and matrix remodeling in patients with SSc.
Collapse
Affiliation(s)
- Michał Ciurzyński
- Department of Internal Medicine and Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Bienias
- Department of Internal Medicine and Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Irzyk
- Department of Internal Medicine and Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Maciej Kostrubiec
- Department of Internal Medicine and Cardiology, Medical University of Warsaw, Warsaw, Poland
| | | | - Urszula Demkow
- Department of Laboratory Diagnostics and Clinical Immunology of Developmental Age, Medical University of Warsaw, Warsaw, Poland
| | - Maria Siwicka
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Kurnicka
- Department of Internal Medicine and Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Barbara Lichodziejewska
- Department of Internal Medicine and Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Piotr Pruszczyk
- Department of Internal Medicine and Cardiology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|