1
|
Garcia JPT, Tayo LL. Theoretical Studies of DNA Microarray Present Potential Molecular and Cellular Interconnectivity of Signaling Pathways in Immune System Dysregulation. Genes (Basel) 2024; 15:393. [PMID: 38674328 PMCID: PMC11049615 DOI: 10.3390/genes15040393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/03/2024] [Accepted: 03/07/2024] [Indexed: 04/28/2024] Open
Abstract
Autoimmunity is defined as the inability to regulate immunological activities in the body, especially in response to external triggers, leading to the attack of the tissues and organs of the host. Outcomes include the onset of autoimmune diseases whose effects are primarily due to dysregulated immune responses. In past years, there have been cases that show an increased susceptibility to other autoimmune disorders in patients who are already experiencing the same type of disease. Research in this field has started analyzing the potential molecular and cellular causes of this interconnectedness, bearing in mind the possibility of advancing drugs and therapies for the treatment of autoimmunity. With that, this study aimed to determine the correlation of four autoimmune diseases, which are type 1 diabetes (T1D), psoriasis (PSR), systemic sclerosis (SSc), and systemic lupus erythematosus (SLE), by identifying highly preserved co-expressed genes among datasets using WGCNA. Functional annotation was then employed to characterize these sets of genes based on their systemic relationship as a whole to elucidate the biological processes, cellular components, and molecular functions of the pathways they are involved in. Lastly, drug repurposing analysis was performed to screen candidate drugs for repositioning that could regulate the abnormal expression of genes among the diseases. A total of thirteen modules were obtained from the analysis, the majority of which were associated with transcriptional, post-transcriptional, and post-translational modification processes. Also, the evaluation based on KEGG suggested the possible role of TH17 differentiation in the simultaneous onset of the four diseases. Furthermore, clomiphene was the top drug candidate for regulating overexpressed hub genes; meanwhile, prilocaine was the top drug for regulating under-expressed hub genes. This study was geared towards utilizing transcriptomics approaches for the assessment of microarray data, which is different from the use of traditional genomic analyses. Such a research design for investigating correlations among autoimmune diseases may be the first of its kind.
Collapse
Affiliation(s)
- Jon Patrick T. Garcia
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila 1002, Philippines;
- School of Graduate Studies, Mapúa University, Manila 1002, Philippines
| | - Lemmuel L. Tayo
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila 1002, Philippines;
- Department of Biology, School of Medicine and Health Sciences, Mapúa University, Makati 1200, Philippines
| |
Collapse
|
2
|
Alesci S, Wahle M, Himsel A, Miesbach W. Factor XIII and Endothelial Dysfunction in Patients with Systemic Sclerosis. Hamostaseologie 2023; 43:411-417. [PMID: 37127039 DOI: 10.1055/a-2018-7014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023] Open
Abstract
Systemic sclerosis (SSc, scleroderma) is a severe autoimmune connective tissue disease which affects the skin and internal organs. There has been evidence that coagulation factor XIII (FXIII) has a positive impact on clinical results in patients with SSc. In a single-center cohort study, we investigated the relationship between coagulation FXIII, endothelial dysfunction, and skin infection in SSc. Fifty-six patients could be included and were divided into two groups (with and without scleroderma). Markers of inflammation, coagulation, and endothelial dysfunction like C-reactive protein, leucocytes, fibrinogen, FVIII, VWF-Ag (von Willebrand factor antigen), D-dimers, and vascular endothelial growth factor were analyzed as well as MRSS (modified Rodnan skin scores) data were evaluated. Reduced daily activities were evaluated by the Scleroderma Health Assessment Questionnaire (SHAQ). There were no significant correlations between FXIII activity, MRSS, and SHAQ score. There were correlations between FXIII activity and Raynaud's phenomenon-related symptoms and a weak but not significant positive correlation with the level of pain. A significant correlation between VWF-Ag and lung-associated complaints (n = 56; p = 0.41, p < 0.0001) was found. Moreover, the study showed a correlation between VWF-Ag and MRSS (r [N = 48] = 0.4, p = 0.01), which means that higher VWF-Ag levels come along with more severe skin involvement. A trend toward a negative correlation between FXIII activity and VWF-Ag as marker of endothelial dysfunction was found (r [N = 56] = - 0.20, p = 0.15). In our cohort, there is no FXIII deficiency in patients with SSc. FXIII might have a role in improving cutaneous manifestations indirectly by means of a moderating influence on endothelial dysfunction. Further clinical evaluation is needed.
Collapse
Affiliation(s)
- Sonja Alesci
- Frankfurt Haemophilia Centre, Goethe University Frankfurt, Frankfurt am Main, Hessen, Germany
- MVZ IMD GmbH Gerinnungszentrum Hochtaunus, Bad Homburg, Hessen, Germany
| | - Matthias Wahle
- Department of Rheumatology, Medical Clinic III, University Hospital Augsburg, Augsburg, Bayern, Germany
| | - Andrea Himsel
- Department of Rheumatology, GPR MVZ Rüsselsheim, Medical Clinic I, Rüsselsheim, Hessen, Germany
| | - Wolfgang Miesbach
- Frankfurt Haemophilia Centre, Goethe University Frankfurt, Frankfurt am Main, Hessen, Germany
| |
Collapse
|
3
|
JAK inhibitors and systemic sclerosis: A systematic review of the literature. Clin Exp Rheumatol 2022; 21:103168. [DOI: 10.1016/j.autrev.2022.103168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 01/06/2023]
|
4
|
Kidney Involvement in Systemic Sclerosis. J Pers Med 2022; 12:jpm12071123. [PMID: 35887620 PMCID: PMC9324204 DOI: 10.3390/jpm12071123] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Systemic sclerosis is a chronic multisystem autoimmune disease, characterized by diffuse fibrosis and abnormalities of microcirculation and small arterioles in the skin, joints and visceral organs. Material and Methods: We searched for the relevant articles on systemic sclerosis and kidney involvement in systemic sclerosis in the NIH library of medicine, transplant, rheumatologic and nephrological journals. Results: Half of patients with systemic sclerosis have clinical evidence of kidney involvement. Scleroderma renal crisis represents the most specific and serious renal event associated with this condition. It is characterized by an abrupt onset of moderate to marked hypertension and kidney failure. Early and aggressive treatment is mandatory to prevent irreversible organ damage and death. The advent of ACE-inhibitors revolutionized the management of scleroderma renal crisis. However, the outcomes of this serious complication are still poor, and between 20 to 50% of patients progress to end stage renal disease. Conclusions: Scleroderma renal crisis still represents a serious and life-threatening event. Thus, further studies on its prevention and on new therapeutic strategies should be encouraged.
Collapse
|
5
|
Shan M, Liu H, Hao Y, Meng T, Feng C, Song K, Wang Y. IL-4 and CCR7 play an important role in the development of keloids in patients with a family history. Am J Transl Res 2022; 14:3381-3394. [PMID: 35702126 PMCID: PMC9185030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 03/03/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND The occurrence of keloids tends to show family clusters and more severe symptoms in patients with a family history of the condition, but their pathological mechanisms remain unclear. In this study, we aimed to investigate the differences in genetic susceptibility between keloid patients with a family history of keloids and sporadic patients with sporadic keloids and explore potential therapeutic targets of keloids at the molecular level. METHODS High-throughput sequencing data were obtained from normal skin tissue of patients with a family history of keloids (FN group) and normal skin tissue from sporadic patients (N group). Bioinformatics analysis was employed to identify hub genes. Promising hub genes were identified using RT-qPCR, immunohistochemistry and immunofluorescence assays, and Western blotting. GO and KEGG pathway enrichment analysis was performed to determine the main functions between the two groups. RESULTS Patients with a family history of keloids had more severe clinical symptoms (Ρ = -0.749, P < 0.001). The expression of IL-4 and CCR7 was significantly different between patients with a family history of keloids and those with sporadic keloids. The high expression of IL-4 and the low expression of CCR7 in the FN group may be of key importance in explaining why keloids run in families (P < 0.05). CONCLUSION Having a family history of keloids is a risk factor for increased severity of keloids. IL-4 and CCR7 play an important role in the development of keloids in patients with a family history of the condition and may represent new targets for the treatment of keloids.
Collapse
Affiliation(s)
- Mengjie Shan
- Department of Plastic Surgery, Peking Union Medical College HospitalBeijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Hao Liu
- Department of Plastic Surgery, Peking Union Medical College HospitalBeijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Yan Hao
- Department of Plastic Surgery, Peking Union Medical College HospitalBeijing, China
- Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, China
| | - Tian Meng
- Department of Plastic Surgery, Peking Union Medical College HospitalBeijing, China
| | - Cheng Feng
- Department of Plastic Surgery, Peking Union Medical College HospitalBeijing, China
| | - Kexin Song
- Department of Plastic Surgery, Peking Union Medical College HospitalBeijing, China
| | - Youbin Wang
- Department of Plastic Surgery, Peking Union Medical College HospitalBeijing, China
| |
Collapse
|
6
|
Han L, Lv Q, Guo K, Li L, Zhang H, Bian H. Th17 cell-derived miR-155-5p modulates interleukin-17 and suppressor of cytokines signaling 1 expression during the progression of systemic sclerosis. J Clin Lab Anal 2022; 36:e24489. [PMID: 35545753 PMCID: PMC9169208 DOI: 10.1002/jcla.24489] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/23/2022] [Accepted: 02/16/2022] [Indexed: 11/12/2022] Open
Abstract
Background miR‐155‐5p is associated with autoimmune diseases. T helper 17 (Th17) cells, interleukin (IL)‐17, and suppressor of cytokines signaling 1 (SOCS1) have important roles in the pathogenesis of systemic sclerosis (SSc). The purpose of this study was to explore the role of miR‐155‐5p in the regulation of IL‐17 and SOCS1 expression in Th17 cells and the subsequent effect on SSc disease progression. Methods Th17 cells were isolated from peripheral blood mononuclear cells of SSc patients and healthy controls (HCs). RT‐qPCR and western blotting were used to examine the expression patterns of miR‐155‐5p, IL‐17, and SOCS1. Luciferase reporter assays were performed to confirm SOCS1 as a target of miR‐155‐5p. RNA pull‐down assays were performed to detect the interaction of IL‐17 and SOCS1 with miR‐155‐5p. In situ hybridization was performed to analyze the co‐expression pattern of miR‐155‐5p and IL17A in Th17 cells. Results The levels of Th17 cell‐derived miR‐155‐5p were significantly up‐regulated in SSc patients compared with HCs, and its levels were negatively correlated with SOCS1 levels. Meanwhile, miR‐155‐5p positively regulated IL‐17 expression levels in Th17 cells isolated from SSc patients as the disease progressed. Using pmirGLO vectors, SOCS1 was confirmed as a target of miR‐155‐5p. The binding status of IL‐17 and SOCS1 to miR‐155‐5p was related to SSc progression. An increase in the co‐localization of miR‐155‐5p and IL‐17 was associated with greater SSc progression. Conclusions IL‐17 and SOCS1 expression modulated by Th17 cell‐derived miR‐155‐5p are critical for SSc progression, which may provide novel insights into the pathogenesis of SSc.
Collapse
Affiliation(s)
- Li Han
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, China.,Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, China
| | - Qin Lv
- Department of Chinese Medicine, Nanyang Medical College, Nanyang, China
| | - Kelei Guo
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, China.,Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, China
| | - Linyun Li
- Department of Rheumatism Immunity, Nanyang Traditional Chinese Medicine Hospital, Nanyang, China
| | - Hong Zhang
- Department of Rheumatism Immunity, Nanyang Central Hospital, Nanyang, China
| | - Hua Bian
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang, China.,Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, China
| |
Collapse
|
7
|
Liu S, Chung MP, Ley B, French S, Elicker BM, Fiorentino DF, Chung LS, Boin F, Wolters PJ. Peripheral blood leucocyte telomere length is associated with progression of interstitial lung disease in systemic sclerosis. Thorax 2021; 76:1186-1192. [PMID: 34272332 DOI: 10.1136/thoraxjnl-2020-215918] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 06/10/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Peripheral blood leucocyte telomere length (PBL-TL) is associated with outcomes in patients with idiopathic pulmonary fibrosis. Whether PBL-TL is associated with progression of systemic sclerosis-associated interstitial lung disease (SSc-ILD) is unknown. METHODS A retrospective observational cohort study was performed using prospectively collected data from 213 patients with SSc followed at the University of California San Francisco (UCSF) Scleroderma Center. PBL-TL was measured by quantitative PCR of DNA isolated from peripheral blood. Associations between PBL-TL and pulmonary function test trends in patients with SSc-ILD were assessed by longitudinal analysis using Generalised Linear Mixed Models. Findings were validated in a cohort of 61 patients with SSc-ILD enrolled in the Stanford University Scleroderma Center database. RESULTS Patients with UCSF SSc with ILD were found to have shorter PBL-TL compared with those without ILD (6554±671 base pairs (bp) vs 6782±698 bp, p=0.01). Shorter PBL-TL was associated with the presence of ILD (adjusted OR 2.1 per 1000 bp TL decrease, 95% CI [1.25 to 3.70], p=0.006). PBL-TL was shorter in patients with SSc-ILD lacking SSc-specific autoantibodies compared with seropositive subjects (6237±647 bp vs 6651±653 bp, p=0.004). Shorter PBL-TL was associated with increased risk for lung function deterioration with an average of 67 mL greater loss in per year for every 1000 bp decrease in PBL-TL in the combined SSc-ILD cohorts (longitudinal analysis, adjusted model: 95% CI -104 mL to -33 mL, p<0.001). CONCLUSIONS These findings suggest that telomere dysfunction may be associated with SSc-ILD progression and that PBL-TL measurement may be useful for stratifying risk for SSc-ILD progression.
Collapse
Affiliation(s)
- Shuo Liu
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, USA.,Pulmonary and Critical Care Medicine, Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Melody P Chung
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
| | - Brett Ley
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Sarah French
- Division of Rheumatology, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Brett M Elicker
- Division of Radiology, University of California San Francisco, San Francisco, California, USA
| | - David F Fiorentino
- Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Lorinda S Chung
- Division of Immunology and Rheumatology, Department of Dermatology, Stanford University School of Medicine, Stanford, California, USA
| | - Francesco Boin
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Paul J Wolters
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
8
|
Aung WW, Wang C, Xibei J, Horii M, Mizumaki K, Kano M, Okamura A, Kobayashi T, Matsushita T. Immunomodulating role of the JAKs inhibitor tofacitinib in a mouse model of bleomycin-induced scleroderma. J Dermatol Sci 2020; 101:174-184. [PMID: 33451905 DOI: 10.1016/j.jdermsci.2020.12.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 12/03/2020] [Accepted: 12/24/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Janus kinase (JAK)-signal transducer and activator of transcription (STAT) was hyperactivated in biopsies from patients with systemic sclerosis (SSc) and in several autoimmune disease models. Tofacitinib, a pan-JAK inhibitor, blocks the downstream signaling of multiple cytokines and has exhibited therapeutic efficacy in various autoimmune diseases, although its immunomodulating property in scleroderma is unclear. OBJECTIVE To evaluate the effect of tofacitinib on the modulation of cytokine-producing T and B cells, and proinflammatory cells in a mouse model of SSc. METHODS Bleomycin (BLM)-induced SSc was generated by intradermal injection of BLM or PBS for control. Mice received intraperitoneal tofacitinib (20 mg/kg) or vehicle 3 times per week from day 0-28. Mice were sacrificed at day 28 after the last BLM/PBS injection. RESULTS Tofacitinib administration significantly alleviated fibrosis of the skin and lungs in scleroderma mouse model. Furthermore, tofacitinib suppressed adaptive and innate immune responses by reducing splenocytes, total lymphocytes, CD4+ T helper cells (especially Th2 and Th17 subtypes), IL-6-producing effector B cells, PDCA-1+ dendritic cells in the spleen, and infiltration of F4/80+, CD206+ and CD163+ macrophages in the skin and lungs. Conversely, tofacitinib increased the proportions of splenic regulatory T and B cells. The mRNA expression of extracellular matrix proteins and fibrogenic cytokines was downregulated by tofacitinib in both the skin and lungs. CONCLUSION These observations suggest JAK inhibition as a therapeutic approach for the treatment of inflammatory and fibrotic diseases, and highlight the potential of tofacitinib as a promising candidate for treating patients with scleroderma.
Collapse
Affiliation(s)
- Wah Wah Aung
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Chenyang Wang
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Jia Xibei
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Motoki Horii
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Kie Mizumaki
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Miyu Kano
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Ai Okamura
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan; Department of Plastic Surgery, Kanazawa University Hospital, Kanazawa, Japan
| | - Tadahiro Kobayashi
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Takashi Matsushita
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
9
|
Wu J, Del Duca E, Espino M, Gontzes A, Cueto I, Zhang N, Estrada YD, Pavel AB, Krueger JG, Guttman-Yassky E. RNA Sequencing Keloid Transcriptome Associates Keloids With Th2, Th1, Th17/Th22, and JAK3-Skewing. Front Immunol 2020; 11:597741. [PMID: 33329590 PMCID: PMC7719808 DOI: 10.3389/fimmu.2020.597741] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/19/2020] [Indexed: 12/23/2022] Open
Abstract
Keloids are disfiguring, fibroproliferative growths and their pathogenesis remains unclear, inhibiting therapeutic development. Available treatment options have limited efficacy and harbor safety concerns. Thus, there is a great need to clarify keloid pathomechanisms that may lead to novel treatments. In this study, we aimed to elucidate the profile of lesional and non-lesional keloid skin compared to normal skin. We performed gene (RNAseq, qRT-PCR) and protein (immunohistochemistry) expression analyses on biopsy specimens obtained from lesional and non-lesional skin of African American (AA) keloid patients compared to healthy skin from AA controls. Fold-change≥2 and false-discovery rate (FDR)<0.05 was used to define significance. We found that lesional versus normal skin showed significant up-regulation of markers of T-cell activation/migration (ICOS, CCR7), Th2- (IL-4R, CCL11, TNFSF4/OX40L), Th1- (CXCL9/CXCL10/CXCL11), Th17/Th22- (CCL20, S100As) pathways, and JAK/STAT-signaling (JAK3) (false-discovery rate [FDR]<0.05). Non-lesional skin also exhibited similar trends. We observed increased cellular infiltrates in keloid tissues, including T-cells, dendritic cells, mast cells, as well as greater IL-4rα+, CCR9+, and periostin+ immunostaining. In sum, comprehensive molecular profiling demonstrated that both lesional and non-lesional skin show significant immune alternations, and particularly Th2 and JAK3 expression. This advocates for the investigation of novel treatments targeting the Th2 axis and/or JAK/STAT-signaling in keloid patients.
Collapse
Affiliation(s)
- Jianni Wu
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- College of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, United States
| | - Ester Del Duca
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Dermatology, University of Rome Tor Vergata, Rome, Italy
| | - Michael Espino
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Alyssa Gontzes
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Inna Cueto
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, United States
| | - Ning Zhang
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Yeriel D. Estrada
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ana B. Pavel
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Biomedical Engineering, University of Mississippi, Oxford, MS, United States
| | - James G. Krueger
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, United States
| | - Emma Guttman-Yassky
- Laboratory of Inflammatory Skin Diseases, Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY, United States
| |
Collapse
|
10
|
He H, Suryawanshi H, Morozov P, Gay-Mimbrera J, Del Duca E, Kim HJ, Kameyama N, Estrada Y, Der E, Krueger JG, Ruano J, Tuschl T, Guttman-Yassky E. Single-cell transcriptome analysis of human skin identifies novel fibroblast subpopulation and enrichment of immune subsets in atopic dermatitis. J Allergy Clin Immunol 2020; 145:1615-1628. [DOI: 10.1016/j.jaci.2020.01.042] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 01/15/2020] [Accepted: 01/21/2020] [Indexed: 12/18/2022]
|
11
|
Nguyen JK, Austin E, Huang A, Mamalis A, Jagdeo J. The IL-4/IL-13 axis in skin fibrosis and scarring: mechanistic concepts and therapeutic targets. Arch Dermatol Res 2020; 312:81-92. [PMID: 31493000 PMCID: PMC7008089 DOI: 10.1007/s00403-019-01972-3] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/22/2019] [Accepted: 08/29/2019] [Indexed: 02/06/2023]
Abstract
Skin fibrosis, characterized by excessive fibroblast proliferation and extracellular matrix deposition in the dermis, is the histopathologic hallmark of dermatologic diseases such as systemic sclerosis, hypertrophic scars, and keloids. Effective anti-scarring therapeutics remain an unmet need, underscoring the complex pathophysiologic mechanisms of skin fibrosis. The Th2 cytokines interleukin (IL)-4 and IL-13 have been implicated as key mediators in the pathogenesis of fibroproliferative disorders. The goal of this article is to summarize the current understanding of the role of the IL-4/IL-13 axis in wound healing and skin fibrosis. We conducted a literature search to identify research studies investigating the roles of IL-4 and IL-13 in fibrotic skin diseases. While transforming growth factor-beta has long been regarded as the main driver of fibrotic processes, research into the cellular and molecular biology of wound healing has revealed other pathways that promote scar tissue formation. IL-4 and IL-13 are important mediators of skin fibrosis, supported by evidence from in vitro data, animal models of fibrosis, and clinical studies. Overactive signaling of the IL-4/IL-13 axis contributes to the initiation and perpetuation of fibrotic skin diseases. Further insights into the IL-4/IL-13 axis may reveal potential targets for the development of novel therapies that prevent or treat fibrotic skin diseases.
Collapse
Affiliation(s)
- Julie K Nguyen
- Department of Dermatology, SUNY Downstate Medical Center, 450 Clarkson Avenue MSC 46, Brooklyn, NY, 11203, USA
| | - Evan Austin
- Department of Dermatology, SUNY Downstate Medical Center, 450 Clarkson Avenue MSC 46, Brooklyn, NY, 11203, USA
| | - Alisen Huang
- Department of Dermatology, SUNY Downstate Medical Center, 450 Clarkson Avenue MSC 46, Brooklyn, NY, 11203, USA
| | - Andrew Mamalis
- Department of Dermatology, SUNY Downstate Medical Center, 450 Clarkson Avenue MSC 46, Brooklyn, NY, 11203, USA
| | - Jared Jagdeo
- Department of Dermatology, SUNY Downstate Medical Center, 450 Clarkson Avenue MSC 46, Brooklyn, NY, 11203, USA.
- Dermatology Service, VA New York Harbor Healthcare System, Brooklyn, NY, USA.
| |
Collapse
|
12
|
Henry TW, Mendoza FA, Jimenez SA. Role of microRNA in the pathogenesis of systemic sclerosis tissue fibrosis and vasculopathy. Autoimmun Rev 2019; 18:102396. [PMID: 31520794 DOI: 10.1016/j.autrev.2019.102396] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 05/07/2019] [Indexed: 12/11/2022]
Abstract
Systemic Sclerosis (SSc) pathogenesis involves multiple immunological, vascular and fibroproliferative abnormalities that contribute to a severe and complex clinical picture. Vasculopathy and fibroproliferative alterations are two hallmark pathological processes in SSc that are responsible for the most severe clinical manifestations of the disease and determine its clinical outcome and mortality. However, the pathogenesis of SSc vasculopathy and of the uncontrolled SSc fibrotic process remain incompletely understood. Recent investigations into the molecular pathways involved in these processes have identified an important role for epigenetic processes that contribute to overall disease progression and have emphasized microRNAs (miRNAs) as crucial epigenetic regulators. MiRNAs hold unique potential for elucidating SSc pathogenesis, improving diagnosis and developing effective targeted therapies for the disease. This review examines the important role that miRNAs play in the development and regulation of vascular and fibroproliferative alterations associated with SSc pathogenesis and their possible participation in the establishment of pathogenetic connections between these two processes. This review also emphasizes that further understanding of the involvement of miRNA in SSc fibrosis and vasculopathy will very likely provide novel future research directions and allow for the identification of groundbreaking therapeutic interventions within these processes. MiR-21, miR- 31, and miR-155 are of particular interest owing to their important involvement in both SSc vasculopathy and fibroproliferative alterations.
Collapse
Affiliation(s)
- Tyler W Henry
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, Philadelphia 19107, USA; Sidney Kimmel Medical College, Thomas Jefferson University, USA
| | - Fabian A Mendoza
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, USA; Division of Rheumatology, Department of Medicine, Thomas Jefferson University, USA
| | - Sergio A Jimenez
- Jefferson Institute of Molecular Medicine and Scleroderma Center, Thomas Jefferson University, USA.
| |
Collapse
|
13
|
Bocchino M, Bruzzese D, D'Alto M, Argiento P, Borgia A, Capaccio A, Romeo E, Russo B, Sanduzzi A, Valente T, Sverzellati N, Rea G, Vettori S. Performance of a new quantitative computed tomography index for interstitial lung disease assessment in systemic sclerosis. Sci Rep 2019; 9:9468. [PMID: 31263199 PMCID: PMC6603022 DOI: 10.1038/s41598-019-45990-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 06/21/2019] [Indexed: 01/24/2023] Open
Abstract
Quantitative high resolution computed tomography (HRCT) may objectively assess systemic sclerosis (SSc)-interstitial lung disease (ILD) extent, using three basic densitometric measures: mean lung attenuation (MLA), skewness, and kurtosis. This prospective study aimed to develop a composite index - computerized integrated index (CII) – that accounted for MLA, skewness, and kurtosis by means of Principal Component Analysis over HRCTs of 83 consecutive SSc subjects, thus eliminating redundancies. Correlations among CII, cardiopulmonary function and immune-inflammatory biomarkers (e.g. sIL-2Rα and CCL18 serum levels) were explored. ILD was detected in 47% of patients at visual HRCT assessment. These patients had worse CII values than patients without ILD. The CII correlated with lung function at both baseline and follow-up, and with sIL-2Rα and CCL18 serum levels. The best discriminating CII value for ILD was 0.1966 (AUC = 0.77; sensitivity = 0.81 [95%CI:0.68–0.92]; specificity = 0.66 [95%CI:0.52–0.80]). Thirty-four percent of patients without visual trace of ILD had a CII lower than 0.1966, and 67% of them had a diffusing lung capacity for CO <80% of predicted. We showed that this new composite CT index for SSc-ILD assessment correlates with both lung function and immune-inflammatory parameters and could be sufficiently sensitive for capturing early lung density changes in visually ILD-free patients.
Collapse
Affiliation(s)
- Marialuisa Bocchino
- Respiratory Medicine Unit, Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.
| | - Dario Bruzzese
- Department of Public Health, Federico II University, Naples, Italy
| | - Michele D'Alto
- Department of Cardiology, Monaldi Hospital - University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paola Argiento
- Department of Cardiology, Monaldi Hospital - University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Alessia Borgia
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Annalisa Capaccio
- Respiratory Medicine Unit, Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Emanuele Romeo
- Department of Cardiology, Monaldi Hospital - University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Barbara Russo
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Alessandro Sanduzzi
- Respiratory Medicine Unit, Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Tullio Valente
- Department of Radiology, Monaldi Hospital, Naples, Italy
| | - Nicola Sverzellati
- Section of Radiology, Unit of Surgical Sciences, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Gaetano Rea
- Department of Radiology, Monaldi Hospital, Naples, Italy
| | - Serena Vettori
- Rheumatology Unit, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
14
|
Mao YM, Zhao CN, Leng J, Leng RX, Ye DQ, Zheng SG, Pan HF. Interleukin-13: A promising therapeutic target for autoimmune disease. Cytokine Growth Factor Rev 2018; 45:9-23. [PMID: 30581068 DOI: 10.1016/j.cytogfr.2018.12.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 12/11/2018] [Indexed: 12/16/2022]
Abstract
Interleukin-13 (IL-13) was previously thought to be a redundant presence of IL-4, but in recent years its role in immunity, inflammation, fibrosis, and allergic diseases has become increasingly prominent. IL-13 can regulate several subtypes of T helper (Th) cells and affect their transformation, including Th1, Th2, T17, etc., thus it may play an important role in immune system. Previous studies have revealed that IL-13 is implicated in the pathogenesis of autoimmune diseases, such as systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), systemic sclerosis (SSc), ulcerative colitis (UC), type 1 diabetes (T1D), sjogren's syndrome (SS), etc. In this review, we will briefly discuss the biological features of IL-13 and summarize recent advances in the role of IL-13 in the development and pathogenesis of autoimmune diseases. This information may provide new perspectives and suggestions for the selection of therapeutic targets for autoimmune diseases.
Collapse
Affiliation(s)
- Yan-Mei Mao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, 230032, China
| | - Chan-Na Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, 230032, China
| | - Jing Leng
- Anhui Academy of Medical Sciences, 15 Yonghong Road, Hefei, Anhui, China
| | - Rui-Xue Leng
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, 230032, China
| | - Dong-Qing Ye
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, 230032, China
| | - Song Guo Zheng
- Division of Rheumatology, Penn State College of Medicine, Hershey, PA 17033, USA.
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Anhui Province Key Laboratory of Major Autoimmune Diseases, Hefei, Anhui, 230032, China.
| |
Collapse
|
15
|
Li L, Zhu H, Zuo X. Interleukin-33 in Systemic Sclerosis: Expression and Pathogenesis. Front Immunol 2018; 9:2663. [PMID: 30498500 PMCID: PMC6249369 DOI: 10.3389/fimmu.2018.02663] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 10/29/2018] [Indexed: 01/05/2023] Open
Abstract
Interleukin-33 (IL-33), a member of the IL-1 superfamily, functions as a traditional cytokine and nuclear factor. It is proposed to have an “alarmin” role. IL-33 mediates biological effects by interacting with the ST2 receptor and IL-1 receptor accessory protein, particularly in innate immune cells and T helper 2 cells. Recent articles have described IL-33 as an emerging pro-fibrotic cytokine in the immune system as well as a novel potential target for systemic sclerosis. Here, we review the available information and focus on the pleiotropic expression and pathogenesis of IL-33 in systemic sclerosis, as well as the feasibility of using IL-33 in clinical applications.
Collapse
Affiliation(s)
- Liya Li
- Department of Rheumatology and immunology, Xiangya Hospital, Central South University, Changsha, China.,The Institute of Rheumatology and Immunology, Central South University, Changsha, China
| | - Honglin Zhu
- Department of Rheumatology and immunology, Xiangya Hospital, Central South University, Changsha, China.,The Institute of Rheumatology and Immunology, Central South University, Changsha, China
| | - Xiaoxia Zuo
- Department of Rheumatology and immunology, Xiangya Hospital, Central South University, Changsha, China.,The Institute of Rheumatology and Immunology, Central South University, Changsha, China
| |
Collapse
|
16
|
van den Hombergh WMT, Kersten BE, Knaapen-Hans HKA, Thurlings RM, van der Kraan PM, van den Hoogen FHJ, Fransen J, Vonk MC. Hit hard and early: analysing the effects of high-dose methylprednisolone on nailfold capillary changes and biomarkers in very early systemic sclerosis: study protocol for a 12-week randomised controlled trial. Trials 2018; 19:449. [PMID: 30134971 PMCID: PMC6104002 DOI: 10.1186/s13063-018-2798-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 07/09/2018] [Indexed: 01/08/2023] Open
Abstract
Background Mounting evidence indicates that inflammatory mechanisms drive systemic sclerosis (SSc) vasculopathy and fibrosis, especially early in the disease. Therefore, patients with very early SSc could benefit from early treatments targeting inflammation. Glucocorticoids are among the most potent anti-inflammatory and immunosuppressive agents. Several studies have demonstrated a mixed response to treatment with glucocorticoids in SSc, probably because it is seldom initiated at very early stages of the disease. We hypothesise that by inhibiting the inflammatory process driving SSc disease progression, glucocorticoid treatments will induce remission in patients with very early SSc. Methods/design This study is a 12-week, randomised, double-blind, placebo-controlled trial analysing the effects of high-dose intravenous methylprednisolone in very early SSc. Thirty patients who fulfil the criteria for very early SSc will be randomly assigned in a 2:1 ratio to receive either intravenous methylprednisolone or a placebo on three consecutive days over three consecutive months. In this study, the primary endpoint will be the change in capillary density between the baseline and after 12 weeks of treatment. The secondary outcomes of this study are a change in selected biomarkers, other changes in the nailfold capillaries, signs of established SSc and changes in physical function, general health and utilities, as reported through questionnaires. Discussion This trial is the first aiming to treat very early SSc and is promising because it targets the very early stages of the disease process by using an inexpensive and relatively safe treatment known to be highly effective against inflammation. The use of vasculopathy and inflammatory biomarkers as well as clinical signs and symptoms as the endpoints in our study enables us to meet the patient need for markers of disease activity. If it is possible to prevent clinically significant disease in patients with very early SSc by using a safe treatment, this will cause a paradigm shift in scleroderma care and research. Trial registration ClinicalTrials.gov Identifier: NCT03059979. Registered on 20 February 2017. Electronic supplementary material The online version of this article (10.1186/s13063-018-2798-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wieneke M T van den Hombergh
- Department of Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands.
| | - Brigit E Kersten
- Department of Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands
| | - Hanneke K A Knaapen-Hans
- Department of Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands
| | - Rogier M Thurlings
- Department of Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands
| | - Peter M van der Kraan
- Experimental Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands
| | - Frank H J van den Hoogen
- Department of Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands
| | - Jaap Fransen
- Department of Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands
| | - Madelon C Vonk
- Department of Rheumatology, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525, GA, Nijmegen, The Netherlands
| |
Collapse
|
17
|
Kafaja S, Valera I, Divekar AA, Saggar R, Abtin F, Furst DE, Khanna D, Singh RR. pDCs in lung and skin fibrosis in a bleomycin-induced model and patients with systemic sclerosis. JCI Insight 2018; 3:98380. [PMID: 29720568 PMCID: PMC6012518 DOI: 10.1172/jci.insight.98380] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 04/04/2018] [Indexed: 01/01/2023] Open
Abstract
Fibrosis is the end result of most inflammatory conditions, but its pathogenesis remains unclear. We demonstrate that, in animals and humans with systemic fibrosis, plasmacytoid DCs (pDCs) are unaffected or are reduced systemically (spleen/peripheral blood), but they increase in the affected organs (lungs/skin/bronchoalveolar lavage). A pivotal role of pDCs was shown by depleting them in vivo, which ameliorated skin and/or lung fibrosis, reduced immune cell infiltration in the affected organs but not in spleen, and reduced the expression of genes and proteins implicated in chemotaxis, inflammation, and fibrosis in the affected organs of animals with bleomycin-induced fibrosis. As with animal findings, the frequency of pDCs in the lungs of patients with systemic sclerosis correlated with the severity of lung disease and with the frequency of CD4+ and IL-4+ T cells in the lung. Finally, treatment with imatinib that has been reported to reduce and/or prevent deterioration of skin and lung fibrosis profoundly reduced pDCs in lungs but not in peripheral blood of patients with systemic sclerosis. These observations suggest a role for pDCs in the pathogenesis of systemic fibrosis and identify the increased trafficking of pDCs to the affected organs as a potential therapeutic target in fibrotic diseases.
Collapse
Affiliation(s)
- Suzanne Kafaja
- Autoimmunity and Tolerance Laboratory
- Division of Rheumatology
| | - Isela Valera
- Autoimmunity and Tolerance Laboratory
- Division of Rheumatology
| | | | - Rajan Saggar
- Division of Pulmonary and Critical Care Medicine, Department of Medicine
| | | | | | | | - Ram Raj Singh
- Autoimmunity and Tolerance Laboratory
- Division of Rheumatology
- Molecular Toxicology Interdepartmental Program
- Jonsson Comprehensive Cancer Center, and
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| |
Collapse
|
18
|
McFarlane IM, Bhamra MS, Kreps A, Iqbal S, Al-Ani F, Saladini-Aponte C, Grant C, Singh S, Awwal K, Koci K, Saperstein Y, Arroyo-Mercado FM, Laskar DB, Atluri P. Gastrointestinal Manifestations of Systemic Sclerosis. ACTA ACUST UNITED AC 2018; 8. [PMID: 30057856 PMCID: PMC6059963 DOI: 10.4172/2161-1149.1000235] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Systemic sclerosis (SSc) is a rare autoimmune disease characterized by fibroproliferative alterations of the microvasculature leading to fibrosis and loss of function of the skin and internal organs. Gastrointestinal manifestations of SSc are the most commonly encountered complications of the disease affecting nearly 90% of the SSc population. Among these complications, the esophagus and the anorectum are the most commonly affected. However, this devastating disorder does not spare any part of the gastrointestinal tract (GIT), and includes the oral cavity, esophagus, stomach, small and large bowels as well as the liver and pancreas. In this review, we present the current understanding of the pathophysiologic mechanisms of SSc including vasculopathy, endothelial to mesenchymal transformation as well as the autoimmune pathogenetic pathways. We also discuss the clinical presentation and diagnosis of each part of the GIT affected by SSc. Finally, we highlight the latest developments in the management of this disease, addressing the severe malnutrition that affects this vulnerable patient population and ways to assess and improve the nutritional status of the patients.
Collapse
Affiliation(s)
- Isabel M McFarlane
- Division of Rheumatology and Gastroenterology, Department of Medicine and Pathology, Hospitals Kings County Hospital Brooklyn, State University of New York, USA
| | - Manjeet S Bhamra
- Division of Rheumatology and Gastroenterology, Department of Medicine and Pathology, Hospitals Kings County Hospital Brooklyn, State University of New York, USA
| | - Alexandra Kreps
- Division of Rheumatology and Gastroenterology, Department of Medicine and Pathology, Hospitals Kings County Hospital Brooklyn, State University of New York, USA
| | - Sadat Iqbal
- Division of Rheumatology and Gastroenterology, Department of Medicine and Pathology, Hospitals Kings County Hospital Brooklyn, State University of New York, USA
| | - Firas Al-Ani
- Division of Rheumatology and Gastroenterology, Department of Medicine and Pathology, Hospitals Kings County Hospital Brooklyn, State University of New York, USA
| | - Carla Saladini-Aponte
- Division of Rheumatology and Gastroenterology, Department of Medicine and Pathology, Hospitals Kings County Hospital Brooklyn, State University of New York, USA
| | - Christon Grant
- Division of Rheumatology and Gastroenterology, Department of Medicine and Pathology, Hospitals Kings County Hospital Brooklyn, State University of New York, USA
| | - Soberjot Singh
- Division of Rheumatology and Gastroenterology, Department of Medicine and Pathology, Hospitals Kings County Hospital Brooklyn, State University of New York, USA
| | - Khalid Awwal
- Division of Rheumatology and Gastroenterology, Department of Medicine and Pathology, Hospitals Kings County Hospital Brooklyn, State University of New York, USA
| | - Kristaq Koci
- Division of Rheumatology and Gastroenterology, Department of Medicine and Pathology, Hospitals Kings County Hospital Brooklyn, State University of New York, USA
| | - Yair Saperstein
- Division of Rheumatology and Gastroenterology, Department of Medicine and Pathology, Hospitals Kings County Hospital Brooklyn, State University of New York, USA
| | - Fray M Arroyo-Mercado
- Division of Rheumatology and Gastroenterology, Department of Medicine and Pathology, Hospitals Kings County Hospital Brooklyn, State University of New York, USA
| | - Derek B Laskar
- Division of Rheumatology and Gastroenterology, Department of Medicine and Pathology, Hospitals Kings County Hospital Brooklyn, State University of New York, USA
| | - Purna Atluri
- Division of Rheumatology and Gastroenterology, Department of Medicine and Pathology, Hospitals Kings County Hospital Brooklyn, State University of New York, USA
| |
Collapse
|
19
|
Vettori S, Tolone S, Capocotta D, Chieffo R, Giacco V, Valentini G, Docimo L. Esophageal high-resolution impedance manometry alterations in asymptomatic patients with systemic sclerosis: prevalence, associations with disease features, and prognostic value. Clin Rheumatol 2018; 37:1239-1247. [PMID: 29442260 DOI: 10.1007/s10067-018-4026-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/30/2018] [Accepted: 02/02/2018] [Indexed: 12/20/2022]
Abstract
This study aims to investigate pre-clinical esophageal involvement in systemic sclerosis (SSc) by high-resolution impedance manometry (HRiM), its associations with disease features including lung involvement, and its predictivity of esophageal symptoms overtime. Charts of 45 asymptomatic (no heartburn/regurgitation/dysphagia) SSc patients (96% females; mean age 46 years) with at least one follow-up (FU) visit and complete clinical, serological, functional, and radiological assessment, including high-resolution computed tomography (HRCT) of the chest and lung function tests, that had undergone esophageal HRiM were retrospectively evaluated. Esophagogastric junction-contractile integral (EGJ-CI) and esophageal body motility, as evaluated by mean distal contractile integral (DCI), were assessed. SSc patients had a normal esophageal motility in 7/45 cases, a defective EGJ-CI in 28, an ineffective esophageal motility (IEM) in 17, and aperistalsis in 12. Defective EGJ-CI was associated with IEM/aperistalsis in 20 cases, while 9 patients had isolated IEM. Defective EGJ-CI and/or IEM/aperistalsis were associated with a diffusing lung capacity for CO < 80% of predicted value (all p < 0.05), while defective EGJ-CI was also associated with interstitial lung disease on HRCT (p = 0.03). Prevalence of any HRiM abnormality was higher in anti-centromere antibody negative patients (all p < 0.05). IEM/aperistalsis independently increased the risk of esophageal symptoms by 2.3-fold (95% CI 1.1-5.7) and was associated with their higher cumulative incidence with respect to patients with other HRiM patterns at FU (χ2 = 4.63; p = 0.03). SSc patients asymptomatic for esophageal involvement can have HRiM abnormalities in up to 84% of cases. A baseline-impaired motility is a risk factor for symptomatic esophageal disease.
Collapse
Affiliation(s)
- Serena Vettori
- Rheumatology Section, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", c/o II Policlinico, via Pansini 5, 80131, Naples, Italy.
| | - Salvatore Tolone
- Division of General, Mini-Invasive and Bariatric Surgery, Department of Surgery, University of Campania "Luigi Vanvitelli", II Policlinico, Via Pansini 5, 80131, Naples, Italy
| | - Domenico Capocotta
- Rheumatology Section, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", c/o II Policlinico, via Pansini 5, 80131, Naples, Italy
| | - Rossella Chieffo
- Rheumatology Section, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", c/o II Policlinico, via Pansini 5, 80131, Naples, Italy
| | - Veronica Giacco
- Rheumatology Section, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", c/o II Policlinico, via Pansini 5, 80131, Naples, Italy
| | - Gabriele Valentini
- Rheumatology Section, Department of Precision Medicine, University of Campania "Luigi Vanvitelli", c/o II Policlinico, via Pansini 5, 80131, Naples, Italy
| | - Ludovico Docimo
- Division of General, Mini-Invasive and Bariatric Surgery, Department of Surgery, University of Campania "Luigi Vanvitelli", II Policlinico, Via Pansini 5, 80131, Naples, Italy
| |
Collapse
|
20
|
Virzì F, Bianca P, Giammona A, Apuzzo T, Di Franco S, Mangiapane LR, Colorito ML, Catalano D, Scavo E, Nicotra A, Benfante A, Pistone G, Caputo V, Dieli F, Pirrello R, Stassi G. Combined platelet-rich plasma and lipofilling treatment provides great improvement in facial skin-induced lesion regeneration for scleroderma patients. Stem Cell Res Ther 2017; 8:236. [PMID: 29058626 PMCID: PMC5651639 DOI: 10.1186/s13287-017-0690-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 08/05/2017] [Accepted: 10/09/2017] [Indexed: 12/26/2022] Open
Abstract
Background The use of stem cells, including mesenchymal stem cells (MSCs), for regenerative medicine is gaining interest for the clinical benefits so far obtained in patients. This study investigates the use of adipose autologous tissue in combination with platelet-rich plasma (PRP) to improve the clinical outcome of patients affected by systemic sclerosis (SSc). Methods Adipose-derived mesenchymal stem cells (AD-MSCs) and PRPs were purified from healthy donors and SSc patients. The multilineage differentiation potential of AD-MSCs and their genotypic–phenotypic features were investigated. A cytokine production profile was evaluated on AD-MSCs and PRPs from both healthy subjects and SSc patients. The adipose tissue-derived cell fraction, the so-called stromal vascular fraction (SVF), was coinjected with PRP in the perioral area of SSc patients. Results Histopathological and phenotypical analysis of adipose tissue from SSc patients revealed a disorganization of its distinct architecture coupled with an altered cell composition. Although AD-MSCs derived from SSc patients showed high multipotency, they failed to sustain a terminally differentiated progeny. Furthermore, SVFs derived from SSc patients differed from healthy donors in their MSC-like traits coupled with an aberrant cytokine production profile. Finally, the administration of PRP in combination with autologous SVF improved buccal’s rhyme, skin elasticity and vascularization for all of the SSc patients enrolled in this study. Conclusions This innovative regenerative therapy could be exploited for the treatment of chronic connective tissue diseases, including SSc. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0690-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Francesco Virzì
- Cellular and Molecular Pathophysiology Laboratory, Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90127, Italy.,Division of Plastic and Reconstructive Surgery Department of Surgical and Oncological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90127, Italy
| | - Paola Bianca
- Cellular and Molecular Pathophysiology Laboratory, Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90127, Italy
| | - Alessandro Giammona
- Cellular and Molecular Pathophysiology Laboratory, Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90127, Italy
| | - Tiziana Apuzzo
- Cellular and Molecular Pathophysiology Laboratory, Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90127, Italy
| | - Simone Di Franco
- Cellular and Molecular Pathophysiology Laboratory, Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90127, Italy
| | - Laura Rosa Mangiapane
- Cellular and Molecular Pathophysiology Laboratory, Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90127, Italy
| | - Maria Luisa Colorito
- Cellular and Molecular Pathophysiology Laboratory, Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90127, Italy
| | - Dario Catalano
- Cellular and Molecular Pathophysiology Laboratory, Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90127, Italy
| | - Emanuela Scavo
- Cellular and Molecular Pathophysiology Laboratory, Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90127, Italy
| | - Annalisa Nicotra
- Cellular and Molecular Pathophysiology Laboratory, Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90127, Italy
| | - Antonina Benfante
- Cellular and Molecular Pathophysiology Laboratory, Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90127, Italy
| | - Giuseppe Pistone
- DiBiMIS, Piazza delle Cliniche, 2, University of Palermo, Palermo, 90127, Italy
| | - Valentina Caputo
- DiBiMIS, Piazza delle Cliniche, 2, University of Palermo, Palermo, 90127, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis & Biomedical Research (CLADIBIOR), University of Palermo, Via del Vespro 131, Palermo, 90127, Italy
| | - Roberto Pirrello
- Division of Plastic and Reconstructive Surgery Department of Surgical and Oncological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90127, Italy
| | - Giorgio Stassi
- Cellular and Molecular Pathophysiology Laboratory, Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Via del Vespro 131, Palermo, 90127, Italy.
| |
Collapse
|
21
|
Yang X, Liu C, Fujino M, Yang J, Li XK, Zou H. A modified graft-versus-host-induced model for systemic sclerosis, with pulmonary fibrosis in Rag2-deficient mice. FEBS Open Bio 2017; 7:1316-1327. [PMID: 28904861 PMCID: PMC5586340 DOI: 10.1002/2211-5463.12268] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 07/03/2017] [Accepted: 07/05/2017] [Indexed: 11/10/2022] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disease that results in fibrosis in multiple organs. Various animal models for this disease have been developed, both genetic and induced. One of the induced models, sclerodermatous graft‐versus‐host disease (scl‐GvHD), exhibits the main characteristics of SSc, but involves lethal γ‐irradiation of recipients. We sought to develop a modified scl‐GvHD model. Spleen cells from B10.D2 donor mice were transplanted into immunodeficient Rag‐2 recipients on the BALB/c genetic background. Tissue fibrosis was analyzed at 3 and 9 weeks after transplantation. In addition to serum levels of anti‐Scl‐70 autoantibody and cytokines, tissue inflammation, fibrosis, expression of collagen‐I and α‐smooth muscle actin (α‐SMA), infiltration of leukocytes, mRNA expression of transforming growth factor (TGF)‐β, collagen‐I, α‐SMA, tumor necrosis factor (TNF)‐α, and interleukin (IL)‐6, the classical signal pathway of TGF‐β, Smad‐3, and p‐Smad‐3 expression in tissue were analyzed. Skin thickening and increased collagen synthesis, as well as the manifestation of tissue fibrosis, could be detected in skin, kidney, and lung of modified scl‐GvHD mouse model. Increased serum levels of anti‐Scl‐70 autoantibody, IL‐10, and TGF‐β could be detected. Increased CD4+ T cells and F4/80+ macrophage infiltration were found in skin, kidney, and lung. Gene expression of collagen‐I, TGF‐β, α‐SMA, TNF‐α, and IL‐6 was increased in tissue of the scl‐GvHD model. Moreover, TGF‐β expression and Smad‐3 phosphorylation were detected in skin, kidney, and lung of scl‐GvHD mice. Our data show that spleen cells from B10.D2 donor mice transplanted into immunodeficient Rag‐2 recipients could induce typical fibrosis not only of the skin and kidney but also of lung, which was missing from previous scl‐GvHD models. Thus, the modified scl‐GvHD model might be a promising model to explore the immunologic mechanisms of SSc and may be useful for investigation of new therapies for systemic sclerosis.
Collapse
Affiliation(s)
- Xue Yang
- Division of Rheumatology Huashan Hospital Fudan University Shanghai China.,Division of Transplantation Immunology National Research Institute for Child Health and Development Tokyo Japan.,Institute of Rheumatology, Immunology and Allergy Fudan University Shanghai China
| | - Chi Liu
- Division of Transplantation Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Masayuki Fujino
- Division of Transplantation Immunology National Research Institute for Child Health and Development Tokyo Japan.,AIDS Research Center National Institute of Infectious Diseases Tokyo Japan
| | - Ji Yang
- Department of Dermatology Zhongshan Hospital Fudan University Shanghai China
| | - Xiao-Kang Li
- Division of Transplantation Immunology National Research Institute for Child Health and Development Tokyo Japan.,Institute of Rheumatology, Immunology and Allergy Fudan University Shanghai China
| | - Hejian Zou
- Division of Rheumatology Huashan Hospital Fudan University Shanghai China.,Institute of Rheumatology, Immunology and Allergy Fudan University Shanghai China
| |
Collapse
|
22
|
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: 1.3] [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
|
23
|
Rice LM, Mantero JC, Stifano G, Ziemek J, Simms RW, Gordon J, Domsic R, Lafyatis R. A Proteome-Derived Longitudinal Pharmacodynamic Biomarker for Diffuse Systemic Sclerosis Skin. J Invest Dermatol 2016; 137:62-70. [PMID: 27640094 DOI: 10.1016/j.jid.2016.08.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 07/20/2016] [Accepted: 08/17/2016] [Indexed: 11/16/2022]
Abstract
In this study we systematically investigated alterations in the serum proteome of patients with diffuse cutaneous systemic sclerosis and identified differentially expressed proteins that correlated with disease severity. Our goal was to identify a combination of serum proteins that would provide a biological measure for the extent of skin disease and that could be combined into a longitudinal pharmacodynamic biomarker. We found that 16% of the sera proteins analyzed by SOMAscan aptamer technology, from two cohorts of patients with diffuse cutaneous systemic sclerosis, were identified as differentially regulated between diffuse cutaneous systemic sclerosis and controls and correlated with modified Rodnan skin score. This dataset showed tumor necrosis factor-α, IFN-γ, transforming growth factor-β, and IL-13 as potential upstream regulators of the serum protein patterns in the sera of patients with diffuse cutaneous systemic sclerosis. By ELISA, two analytes (ST2 and Spondin-1) best described longitudinal change in modified Rodnan skin score, using linear mixed models. This model was then validated in three independent cohorts. In this study we discovered a large array of proteins not previously associated with systemic sclerosis that provide insight into pathogenesis and potential targets for therapeutic intervention. Furthermore, we show that two of these proteins can be combined to form a robust longitudinal biomarker that might be used in clinical trials to assess changes in diffuse cutaneous systemic sclerosis skin disease over time.
Collapse
Affiliation(s)
- Lisa M Rice
- Boston University School of Medicine, Boston, Massachusetts, USA.
| | - Julio C Mantero
- Boston University School of Medicine, Boston, Massachusetts, USA
| | | | - Jessica Ziemek
- Boston University School of Medicine, Boston, Massachusetts, USA
| | - Robert W Simms
- Boston University School of Medicine, Boston, Massachusetts, USA
| | | | - Robyn Domsic
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Robert Lafyatis
- University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
24
|
Koca SS, Pehlivan Y, Kara M, Alibaz-Oner F, Oztuzcu S, Yilmaz N, Cetin GY, Kisacik B, Ozgen M, Pamuk ON, Direskeneli H, Sayarlioglu M, Onat AM. The IL-33 gene is related to increased susceptibility to systemic sclerosis. Rheumatol Int 2016; 36:579-84. [PMID: 26743213 DOI: 10.1007/s00296-015-3417-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 12/23/2015] [Indexed: 12/26/2022]
Abstract
Systemic sclerosis (SSc) is a chronic inflammatory disease characterized by widespread fibrosis of the skin and several visceral organs. The pro-fibrotic potential of interleukin (IL)-33 has been demonstrated by in both in vitro and in vivo settings; moreover, increased level of IL-33 has also been reported in patients with SSc. Therefore, the aim of the present study was to detect the potential association of IL-33 gene polymorphisms on the susceptibility of SSc. A total of 300 SSc patients and 280 healthy controls (HC) were enrolled in this multicentric preliminary candidate gene study. DNA samples were harvested using an appropriate commercial DNA isolation kit. Four single nucleotide polymorphisms (SNPs) of IL-33 gene (rs7044343, rs1157505, rs11792633 and rs1929992) were genotyped using the appropriate commercial primer/probe sets on real-time PCR. There was no significant difference in terms of the allelic distributions and minor allele frequencies of evaluated four IL-33 polymorphisms between the SSc and HC groups (P > 0.05 for all). Moreover, the genotypic distributions of rs1157505, rs11792633 and rs1929992 polymorphisms were not significantly different (P > 0.05 for all). However, CC genotype of rs7044343 SNP was significantly higher in the SSc group compared to the HC group (P = 0.013, OR 1.75, 95 % CI 1.12-2.72). This preliminary candidate gene study demonstrates that rs7044343 polymorphism of IL-33 gene is associated with the susceptibility to the SSc in Turkish population. It may be suggested that IL-33 gene may be a candidate gene to research in SSc.
Collapse
Affiliation(s)
- Suleyman Serdar Koca
- Department of Rheumatology, Faculty of Medicine, Firat University, 23119, Elazig, Turkey.
| | - Yavuz Pehlivan
- Department of Rheumatology, Faculty of Medicine, Uludag University, Bursa, Turkey
| | - Murat Kara
- Department of Medical Genetics, Faculty of Medicine, Sitki Kocman University, Mugla, Turkey
| | - Fatma Alibaz-Oner
- Department of Rheumatology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Serdar Oztuzcu
- Department of Medical Genetics, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Neslihan Yilmaz
- Department of Rheumatology, Sisli Florence Nightingale Hospital, Istanbul, Turkey
| | - Gozde Yildirim Cetin
- Department of Rheumatology, Faculty of Medicine, Sutcu Imam University, Kahramanmaras, Turkey
| | - Bunyamin Kisacik
- Department of Rheumatology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Metin Ozgen
- Department of Rheumatology, Faculty of Medicine, 19 Mayis University, Samsun, Turkey
| | - Omer Nuri Pamuk
- Department of Rheumatology, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Haner Direskeneli
- Department of Rheumatology, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Mehmet Sayarlioglu
- Department of Rheumatology, Faculty of Medicine, 19 Mayis University, Samsun, Turkey
| | - Ahmet Mesut Onat
- Department of Rheumatology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW In this review of the literature from 2014 through mid-2015, we examine new data that shed light on how macrophages and other innate immune cells and signals contribute to inflammation, vascular dysfunction, and fibrosis in scleroderma. RECENT FINDINGS Recent human studies have focused on changes early in scleroderma, and linked macrophages to inflammation in skin and progression of lung disease. Plasmacytoid dendritic cells have been implicated in vascular dysfunction. In mice, several factors have been identified that influence macrophage activation and experimental fibrosis. However, emerging data also suggest that myeloid cells can have differential effects in fibrosis. Sustained signaling through different toll-like receptors can lead to inflammation or fibrosis, and these signals can influence both immune and nonimmune cells. SUMMARY There are many types of innate immune cells that can potentially contribute to scleroderma and will be worth exploring in detail. Experimentally dissecting the roles of macrophages based on ontogeny and activation state, and the innate signaling pathways in the tissue microenvironment, may also lead to better understanding of scleroderma pathogenesis.
Collapse
Affiliation(s)
- Jennifer J Chia
- aWeill Cornell/Rockefeller/Sloan-Kettering Tri-Institutional MD-PhD Program bImmunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences cAutoimmunity and Inflammation Program dAutoimmunity and Inflammation Program and Department of Pediatric Rheumatology, Hospital for Special Surgery eDepartment of Microbiology and Immunology, Weill Cornell Medical College, New York, USA
| | | |
Collapse
|
26
|
Delantoni A, Matziari E. Osteolysis Affecting the Jaws in Systemic Sclerosis: Clinical and Osseous Changes Based on a Case Presentation. CLINICAL MEDICINE INSIGHTS-ARTHRITIS AND MUSCULOSKELETAL DISORDERS 2015; 8:65-7. [PMID: 26508895 PMCID: PMC4610725 DOI: 10.4137/cmamd.s30510] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/06/2015] [Accepted: 09/07/2015] [Indexed: 11/05/2022]
Abstract
OBJECTIVES The aim of the current paper is to present a case of systemic sclerosis of the jaws with all the characteristics and some extensive findings. METHODS Systemic sclerosis is a connective tissue condition characterized by chronic inflammatory changes, presenting with a number of symptoms. The paper aims to present a case of systemic sclerosis that had some of the characteristics of the condition unilaterally. The features were prominent and well demarcated on the panoramic radiograph. RESULTS The panoramic radiograph of the patient showed extensive condylar head resorption almost to the level of complete flattening. DISCUSSION The rarity of the condition and the awareness the oral radiologist must have upon presentation of similar images is the main reason for the presentation of the case. The features the case presents are both characteristic and well demarcated. Finally, the authors try to alert the clinician, who should be mindful of the fact that sclerodermatous involvement of organ systems is so pleotropic, that it may include the oral cavity, which is not always thoroughly observed, and is often left unattended by medical practitioners.
Collapse
Affiliation(s)
- A Delantoni
- Lecturer, Dept. of Dentoalveolar Surgery, Implant Surgery and Radiology, Faculty of Dentistry, Aristotle University of Thessaloniki, Greece
| | - E Matziari
- Lecturer, Dept. of Dentoalveolar Surgery, Implant Surgery and Radiology, Faculty of Dentistry, Aristotle University of Thessaloniki, Greece
| |
Collapse
|
27
|
Ayano M, Tsukamoto H, Kohno K, Ueda N, Tanaka A, Mitoma H, Akahoshi M, Arinobu Y, Niiro H, Horiuchi T, Akashi K. Increased CD226 Expression on CD8+ T Cells Is Associated with Upregulated Cytokine Production and Endothelial Cell Injury in Patients with Systemic Sclerosis. THE JOURNAL OF IMMUNOLOGY 2015; 195:892-900. [PMID: 26109642 DOI: 10.4049/jimmunol.1403046] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 05/26/2015] [Indexed: 02/06/2023]
Abstract
Systemic sclerosis (SSc) is an autoimmune disease characterized by vascular damage and fibrosis of the skin and internal organs. Because activated and oligoclonally expanded CD8(+) T cells can be detected in peripheral blood and lungs of SSc patients, effector memory CD8(+) T cells may play a critical role for organ involvement in SSc; however, the pathogenic functions of effector memory CD8(+) T cells remain incompletely understood. In this study, we performed DNA microarray analysis of the sort-purified effector memory CD8(+) T cells from SSc patients and healthy controls, and showed that the expression of genes related to immune response and cell adhesion, including CD226 (also known as DNAX accessory molecule-1 [DNAM-1]), was significantly altered. Moreover, detailed analysis of CD226 revealed that CD226(high)CD8(+) T cells were increased in SSc patients (mean, 50.7%) compared with healthy controls (32.9%) and were appreciably associated with the severity of skin sclerosis and interstitial lung disease. Furthermore, CD226(+)CD8(+) T cells produced higher amount of various cytokines than CD226(-) ones, and CD226(high)CD8(+) T cells from SSc patients showed upregulated IL-13 production and positive correlation with the cytotoxic capacity of CD8(+) T cells against HUVECs. Finally, the neutralization of CD226 in CD8(+) T cells impaired costimulation, cytokine productions, and cytolysis against HUVECs. These findings indicate that upregulated CD226 expression on CD8(+) T cells reflects disease severity and is involved in SSc pathogenesis via the production of various cytokines, including profibrotic IL-13 and endothelial cell injury, and that CD226 may be a useful target in the treatment of SSc.
Collapse
Affiliation(s)
- Masahiro Ayano
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiroshi Tsukamoto
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan;
| | - Kentaro Kohno
- Department of Hematology, National Kyushu Medical Center, Fukuoka 810-8563, Japan
| | - Naoyasu Ueda
- Department of Internal Medicine, Miyazaki Prefectural Miyazaki Hospital, Miyazaki 880-8510, Japan
| | - Atsushi Tanaka
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiroki Mitoma
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Mitsuteru Akahoshi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Yojiro Arinobu
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiroaki Niiro
- Clinical Education Center, Kyushu University Hospital, Fukuoka 812-8582, Japan; and
| | - Takahiko Horiuchi
- Department of Internal Medicine, Kyushu University Beppu Hospital, Beppu 874-0836, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| |
Collapse
|
28
|
Kato A, Yutani M, Terao M, Kimura A, Itoi S, Murota H, Miyoshi E, Katayama I. Oligosaccharide modification byN-acetylglucosaminyltransferase-V in macrophages are involved in pathogenesis of bleomycin-induced scleroderma. Exp Dermatol 2015; 24:585-90. [DOI: 10.1111/exd.12730] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Arisa Kato
- Department of Dermatology; Graduate School of Medicine; Osaka University; Suita Osaka Japan
- Department of Molecular Biochemistry and Clinical Investigation; Graduate School of Medicine; Osaka University; Suita Osaka Japan
| | - Mizuki Yutani
- Department of Dermatology; Graduate School of Medicine; Osaka University; Suita Osaka Japan
- Department of Molecular Biochemistry and Clinical Investigation; Graduate School of Medicine; Osaka University; Suita Osaka Japan
| | - Mika Terao
- Department of Dermatology; Graduate School of Medicine; Osaka University; Suita Osaka Japan
| | - Akihiro Kimura
- Department of Dermatology; Graduate School of Medicine; Osaka University; Suita Osaka Japan
- Department of Molecular Biochemistry and Clinical Investigation; Graduate School of Medicine; Osaka University; Suita Osaka Japan
| | - Saori Itoi
- Department of Dermatology; Graduate School of Medicine; Osaka University; Suita Osaka Japan
| | - Hiroyuki Murota
- Department of Dermatology; Graduate School of Medicine; Osaka University; Suita Osaka Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical Investigation; Graduate School of Medicine; Osaka University; Suita Osaka Japan
| | - Ichiro Katayama
- Department of Dermatology; Graduate School of Medicine; Osaka University; Suita Osaka Japan
| |
Collapse
|
29
|
Amedei A, Pimpinelli N, Grassi A, Bella CD, Niccolai E, Brancati S, Benagiano M, D'Elios S, Bosi A, D'Elios MM. Skin CD30(+) T cells and circulating levels of soluble CD30 are increased in patients with graft versus host disease. AUTOIMMUNITY HIGHLIGHTS 2015; 5:21-6. [PMID: 26000151 PMCID: PMC4389013 DOI: 10.1007/s13317-013-0054-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 07/30/2013] [Indexed: 11/17/2022]
Abstract
Objective To determine serum soluble CD30 (sCD30) levels in patients with graft versus host disease (GVHD). Methods Serum soluble CD30 levels and IgE levels were assayed by a sensitive ELISA in 57 patients with bone marrow transplantation, and in 44 healthy controls. We analyzed the type of effector T cells in patients with GVHD. Results Serum levels of sCD30 and serum IgE levels were significantly higher (p values <0.05) in patients with acute and chronic GVHD than in healthy controls. We found that CD30+ T-cells are present in the skin of patients with GVHD. Conclusion These results suggest that serum sCD30 levels may be helpful for the management of patients with bone marrow transplantation.
Collapse
Affiliation(s)
- Amedeo Amedei
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Largo Brambilla 3, 50134 Florence, Italy
| | - Nicola Pimpinelli
- Dipartimento di Chirurgia e Medicina Traslazionale, Università di Firenze, Florence, Italy
| | - Alessia Grassi
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Largo Brambilla 3, 50134 Florence, Italy
| | - Chiara Della Bella
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Largo Brambilla 3, 50134 Florence, Italy
| | - Elena Niccolai
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Largo Brambilla 3, 50134 Florence, Italy
| | - Simona Brancati
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Largo Brambilla 3, 50134 Florence, Italy
| | - Marisa Benagiano
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Largo Brambilla 3, 50134 Florence, Italy
| | - Sofia D'Elios
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Largo Brambilla 3, 50134 Florence, Italy
| | - Alberto Bosi
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Largo Brambilla 3, 50134 Florence, Italy
| | - Mario M D'Elios
- Dipartimento di Medicina Sperimentale e Clinica, Università di Firenze, Largo Brambilla 3, 50134 Florence, Italy
| |
Collapse
|
30
|
Huang XL, Wang YJ, Yan JW, Wan YN, Chen B, Li BZ, Yang GJ, Wang J. Role of anti-inflammatory cytokines IL-4 and IL-13 in systemic sclerosis. Inflamm Res 2015; 64:151-9. [PMID: 25725697 DOI: 10.1007/s00011-015-0806-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 02/16/2015] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE The aim of this paper is to review the anti-inflammatory cytokines IL-4 and IL-13 and their receptor signals; we discuss new insight into their possible roles in systemic sclerosis (SSc) and their overlapping function in SSc. INTRODUCTION SSc is a connective tissue disease characterized by fibrosis. The exact etiology of SSc is unknown, and no therapy has been proved effective in modifying its course. Recently the roles of IL-4 and IL-13 in the development of SSc have been extensively considered. The possible roles of IL-4 and IL-13, especially their overlapping function, in SSc are not well documented. METHODS A literature survey was performed using a PubMed database search to gather complete information regarding IL-4 and IL-13 and their role in inflammation. RESULTS AND CONCLUSIONS The participation of complex pathways of IL-4 and IL-13 in the process of inflammation and fibrosis action in SSc is still not very clear, and some pathogenesis of regulation found in vitro needs to be further proved. There is still more work which could be done to achieve useful developments with therapeutic benefit in SSc.
Collapse
Affiliation(s)
- Xiao-Lei Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, No. 81, Meishan Road, 230032, Hefei, Anhui, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Esclerodermia en el Caribe: características en una serie de casos dominicana. ACTA ACUST UNITED AC 2014; 10:373-9. [DOI: 10.1016/j.reuma.2014.01.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 12/17/2013] [Accepted: 01/31/2014] [Indexed: 11/23/2022]
|
32
|
Li H, Nagai T, Hasui K, Matsuyama T. Depletion of folate receptor β-expressing macrophages alleviates bleomycin-induced experimental skin fibrosis. Mod Rheumatol 2014; 24:816-22. [PMID: 24498991 DOI: 10.3109/14397595.2013.879415] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Folate receptor β (FRβ)-expressing macrophages have been identified as activated macrophages. Here, we investigated the infiltration of FRβ-expressing macrophages in a murine model of bleomycin (BLM)-induced skin fibrosis and assessed the antifibrotic effects of depletion of FRβ-expressing macrophages in this model using a recombinant immunotoxin to FRβ. METHODS A recombinant immunotoxin (anti-FRβ-PE38) was prepared by conjugating the Fv portion of the anti-mouse FRβ heavy chain with truncated Pseudomonas exotoxin A (VH-PE38) and the Fv portion of the anti-mouse FRβ light chain. BLM-induced skin fibrosis mice were intravenously treated with either anti-FRβ-PE38 or VH-PE38 as a control protein. Skin fibrosis was evaluated by the change of skin thickness and hydroxyproline content on Day 29. The TGFβ1 mRNA levels in the treated skin were assessed by quantitative real-time RT-PCR on Day 9. RESULTS Numbers of FRβ-expressing macrophages increased in BLM-injected skin. Anti-FRβ-PE38 treatment led to a dramatic reduction in the number of FRβ-expressing macrophages. Additionally, skin thickness and hydroxyproline content, were markedly reduced. TGFβ1 mRNA levels were also down-regulated after the treatment. TGFβ1 expression was enriched in FRβ-expressing macrophages compared with FRβ-negative macrophages. CONCLUSION These results indicated that anti-FRβ-PE38 treatment efficiently depleted FRβ-expressing macrophages and consequently alleviated BLM-induced skin fibrosis.
Collapse
Affiliation(s)
- Hua Li
- Department of Immunology, Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima , Japan
| | | | | | | |
Collapse
|
33
|
Early Systemic Sclerosis: Serum Profiling of Factors Involved in Endothelial, T-cell, and Fibroblast Interplay is Marked by Elevated Interleukin-33 Levels. J Clin Immunol 2014; 34:663-8. [DOI: 10.1007/s10875-014-0037-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 04/02/2014] [Indexed: 02/06/2023]
|
34
|
Dobrota R, Mihai C, Distler O. Personalized Medicine in Systemic Sclerosis: Facts and Promises. Curr Rheumatol Rep 2014; 16:425. [DOI: 10.1007/s11926-014-0425-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|