1
|
Lu J, Huang D, Liu R, Zhu H, Wang D, Zhao Y, Sun L. Extracellular Matrix-Inspired Dendrimer Nanogels Encapsulating Cyclophosphamide for Systemic Sclerosis Treatment. ACS NANO 2025; 19:4672-4683. [PMID: 39834323 DOI: 10.1021/acsnano.4c15164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Cyclophosphamide has a certain therapeutic effect on treating systemic sclerosis (SSc), while difficulties exist in controlling severe systematic side effects and enhancing targeting capacity. Here, inspired from the natural extracellular matrix composition, we propose a cyclophosphamide-encapsulated nanogel based on dendritic polymers polyamidoamine (PAMAM) for SSc treatment. We combine bovine serum albumin and generation 5 (G5) PAMAM dendrimers with polyphenol modification to obtain nanogels featured with antioxidant and anti-inflammatory effects. The nanogels can possess excellent biocompatibility and prevent fibroblasts from oxidative stress damage and TGF-β-mediated activation. Furthermore, in the bleomycin-induced SSc mouse model, dendrimer nanogels encapsulating cyclophosphamide also exhibit the ability to attenuate fibrosis by modulating immunity, suppressing inflammation, and reducing collagen synthesis. These findings underscore the value of this dendritic polymer nanogel in the treatment of chronic SSc, indicating its broader potential for clinical applications.
Collapse
Affiliation(s)
- Junjie Lu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Danqing Huang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Rui Liu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Haofang Zhu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China
| | - Dandan Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| |
Collapse
|
2
|
Jendrek ST, Schmelter F, Schinke S, Hackel A, Graßhoff H, Lamprecht P, Humrich JY, Sina C, Müller A, Günther U, Riemekasten G. Metabolomic signature identifies HDL and apolipoproteins as potential biomarker for systemic sclerosis with interstitial lung disease. Respir Med 2024; 234:107825. [PMID: 39357678 DOI: 10.1016/j.rmed.2024.107825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/18/2024] [Accepted: 09/29/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND High-density lipoproteins (HDL) affect endothelial functions such as the expression of endothelial cell adhesion molecules and exert anti-apoptotic/-thrombotic functionalities. Therefore, profound analysis of lipoproteins may unveil biomarkers for (micro-)vasculopathy in systemic sclerosis (SSc) and mortality determining disease manifestations like interstitial lung disease (SSc-ILD). Because nuclear magnetic resonance (NMR) spectroscopy provides a wide range of lipoprotein parameters beyond the capabilities of classical analyses it has been used herein to examine lipoprotein profiles in SSc. METHODS To detect the metabolic and lipidomic profile serum samples from clinically well-characterized SSc patients (n = 100) and age-and sex-matched healthy controls (n = 40) were analyzed by 1H NMR spectroscopy using Bruker's in-vitro diagnostic research (IVDr) protocol. Statistical analyses were performed to validate significant findings and to search for associations between lipoproteins and clinical phenotypes. RESULTS Patients with SSc-ILD and lung fibrosis displayed reduced HDL levels. Furthermore, a reduction in apolipoprotein A1 + A2 and its HDL fractions reflected a distinct lipoprotein profile for SSc-ILD patients. This association was independent of potential clinical confounders for dyslipidemia. Notably, in SSc-ILD HDL levels correlate with FVC (forced vital capacity), DLCO (diffusion capacity of the lungs for carbon monoxide), and the modified Rodnan-Skin-Score. CONCLUSION These results suggest HDL and its lipoproteins may be considered as potential new biomarkers for SSc-ILD. Immune-mediated HDL effects on the endothelium facilitate microvasculopathy - one of the pathophysiological hallmarks in SSc. Therefore, a closer prospective evaluation of the capability of HDL-determination and its lipoproteins regarding a more individualized evaluation of SSc-ILD is warranted.
Collapse
Affiliation(s)
- Sebastian T Jendrek
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany.
| | | | - Susanne Schinke
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Alexander Hackel
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Hanna Graßhoff
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Peter Lamprecht
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Jens Y Humrich
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Christian Sina
- Institute of Nutritional Medicine, University of Lübeck, Lübeck, Germany; Medical Department I, University Hospital Schleswig-Holstein, Lübeck, Germany
| | - Antje Müller
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| | - Ulrich Günther
- Institute of Chemistry and Metabolomics, University of Lübeck, Lübeck, Germany
| | - Gabriela Riemekasten
- Clinic for Rheumatology and Clinical Immunology, University Hospital Schleswig-Holstein, Campus Lübeck, Germany
| |
Collapse
|
3
|
Park JS, Kim C, Choi J, Jeong HY, Moon YM, Kang H, Lee EK, Cho ML, Park SH. MicroRNA-21a-5p inhibition alleviates systemic sclerosis by targeting STAT3 signaling. J Transl Med 2024; 22:323. [PMID: 38561750 PMCID: PMC10983659 DOI: 10.1186/s12967-024-05056-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/02/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND MicroRNA (miRNA)-21-5p participates in various biological processes, including cancer and autoimmune diseases. However, its role in the development of fibrosis in the in vivo model of systemic sclerosis (SSc) has not been reported. This study investigated the effects of miRNA-21a-5p overexpression and inhibition on SSc fibrosis using a bleomycin-induced SSc mouse model. METHODS A murine SSc model was induced by subcutaneously injecting 100 μg bleomycin dissolved in 0.9% NaCl into C57BL/6 mice daily for 5 weeks. On days 14, 21, and 28 from the start of bleomycin injection, 100 μg pre-miRNA-21a-5p or anti-miRNA-21a-5p in 1 mL saline was hydrodynamically injected into the mice. Fibrosis analysis was conducted in lung and skin tissues of SSc mice using hematoxylin and eosin as well as Masson's trichrome staining. Immunohistochemistry was used to examine the expression of inflammatory cytokines, phosphorylated signal transducer and activator of transcription-3 (STAT3) at Y705 or S727, and phosphatase and tensin homologue deleted on chromosome-10 (PTEN) in skin tissues of SSc mice. RESULTS MiRNA-21a-5p overexpression promoted lung fibrosis in bleomycin-induced SSc mice, inducing infiltration of cells expressing TNF-α, IL-1β, IL-6, or IL-17, along with STAT3 phosphorylated cells in the lesional skin. Conversely, anti-miRNA-21a-5p injection improved fibrosis in the lung and skin tissues of SSc mice, reducing the infiltration of cells secreting inflammatory cytokines in the skin tissue. In particular, it decreased STAT3-phosphorylated cell infiltration at Y705 and increased the infiltration of PTEN-expressing cells in the skin tissue of SSc mice. CONCLUSION MiRNA-21a-5p promotes fibrosis in an in vivo murine SSc model, suggesting that its inhibition may be a therapeutic strategy for improving fibrosis in SSc.
Collapse
Affiliation(s)
- Jin-Sil Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, South Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, South Korea
| | - Chongtae Kim
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, South Korea
| | - JeongWon Choi
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, South Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, South Korea
| | - Ha Yeon Jeong
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, South Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, South Korea
| | - Young-Mee Moon
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, South Korea
| | - Hoin Kang
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, South Korea
| | - Eun Kyung Lee
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, South Korea.
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, South Korea.
- Department of Medical Lifescience, College of Medicine, The Catholic University of Korea, Seoul, South Korea.
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, South Korea.
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, South Korea.
| | - Sung-Hwan Park
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-Daero, Seocho-gu, Seoul, 06591, South Korea.
- Divison of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpo-Daero, Seocho-Gu, Seoul, 06591, South Korea.
| |
Collapse
|
4
|
Muruganandam M, Ariza-Hutchinson A, Patel RA, Sibbitt WL. Biomarkers in the Pathogenesis, Diagnosis, and Treatment of Systemic Sclerosis. J Inflamm Res 2023; 16:4633-4660. [PMID: 37868834 PMCID: PMC10590076 DOI: 10.2147/jir.s379815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 09/27/2023] [Indexed: 10/24/2023] Open
Abstract
Systemic sclerosis (SSc) is a complex autoimmune disease characterized by vascular damage, vasoinstability, and decreased perfusion with ischemia, inflammation, and exuberant fibrosis of the skin and internal organs. Biomarkers are analytic indicators of the biological and disease processes within an individual that can be accurately and reproducibly measured. The field of biomarkers in SSc is complex as recent studies have implicated at least 240 pathways and dysregulated proteins in SSc pathogenesis. Anti-nuclear antibodies (ANA) are classical biomarkers with well-described clinical classifications and are present in more than 90% of SSc patients and include anti-centromere, anti-Th/To, anti-RNA polymerase III, and anti-topoisomerase I antibodies. Transforming growth factor-β (TGF-β) is central to the fibrotic process of SSc and is intimately intertwined with other biomarkers. Tyrosine kinases, interferon-1 signaling, IL-6 signaling, endogenous thrombin, peroxisome proliferator-activated receptors (PPARs), lysophosphatidic acid receptors, and amino acid metabolites are new biomarkers with the potential for developing new therapeutic agents. Other biomarkers implicated in SSc-ILD include signal transducer and activator of transcription 4 (STAT4), CD226 (DNAX accessory molecule 1), interferon regulatory factor 5 (IRF5), interleukin-1 receptor-associated kinase-1 (IRAK1), connective tissue growth factor (CTGF), pyrin domain containing 1 (NLRP1), T-cell surface glycoprotein zeta chain (CD3ζ) or CD247, the NLR family, SP-D (surfactant protein), KL-6, leucine-rich α2-glycoprotein-1 (LRG1), CCL19, genetic factors including DRB1 alleles, the interleukins (IL-1, IL-4, IL-6, IL-8, IL-10 IL-13, IL-16, IL-17, IL-18, IL-22, IL-32, and IL-35), the chemokines CCL (2,3,5,13,20,21,23), CXC (8,9,10,11,16), CX3CL1 (fractalkine), and GDF15. Adiponectin (an indicator of PPAR activation) and maresin 1 are reduced in SSc patients. A new trend has been the use of biomarker panels with combined complex multifactor analysis, machine learning, and artificial intelligence to determine disease activity and response to therapy. The present review is an update of the various biomarker molecules, pathways, and receptors involved in the pathology of SSc.
Collapse
Affiliation(s)
- Maheswari Muruganandam
- Department of Internal Medicine, Division of Rheumatology and School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Angie Ariza-Hutchinson
- Department of Internal Medicine, Division of Rheumatology and School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Rosemina A Patel
- Department of Internal Medicine, Division of Rheumatology and School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Wilmer L Sibbitt
- Department of Internal Medicine, Division of Rheumatology and School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|
5
|
Ferreira HB, Melo T, Guerra IMS, Moreira ASP, Laranjeira P, Paiva A, Goracci L, Bonciarelli S, Domingues P, Domingues MR. Whole Blood and Plasma-Based Lipid Profiling Reveals Distinctive Metabolic Changes in Systemic Lupus Erythematosus and Systemic Sclerosis. J Proteome Res 2023; 22:2995-3008. [PMID: 37606915 DOI: 10.1021/acs.jproteome.3c00321] [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: 08/23/2023]
Abstract
Autoimmune diseases (AID), such as systemic lupus erythematosus (SLE) and systemic sclerosis (SS), are complex conditions involving immune system dysregulation. Diagnosis is challenging, requiring biomarkers for improved detection and prediction of relapses. Lipids have emerged as potential biomarkers due to their role in inflammation and immune response. This study uses an untargeted C18 RP-LC-MS lipidomics approach to comprehensively assess changes in lipid profiles in patients with SLE and SS. By analyzing whole blood and plasma, the study aims to simplify the lipidomic analysis, explore cellular-level lipids, and compare lipid signatures of SLE and SS with healthy controls. Our findings showed variations in the lipid profile of SLE and SS. Sphingomyelin and ceramide molecular species showed significant increases in plasma samples from SS patients, suggesting an atherosclerotic profile and potentially serving as lipid biomarkers. Phosphatidylserine species in whole blood from SLE patients exhibited elevated levels supporting previously reported dysregulated processes of cell death and defective clearance of dying cells in this AID. Moreover, decreased phospholipids bearing PUFA were observed, potentially attributed to the degradation of these species through lipid peroxidation processes. Further studies are needed to better understand the role of lipids in the pathological mechanisms underlying SLE and SS.
Collapse
Affiliation(s)
- Helena Beatriz Ferreira
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
- CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago 3810-193 Aveiro, Portugal
| | - Tânia Melo
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
- CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago 3810-193 Aveiro, Portugal
| | - Inês M S Guerra
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
- CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago 3810-193 Aveiro, Portugal
| | - Ana S P Moreira
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Paula Laranjeira
- Unidade de Gestão Operacional em Citometria, Centro Hospitalar e Universitário de Coimbra (CHUC), 3004-561 Coimbra, Portugal
| | - Artur Paiva
- Unidade de Gestão Operacional em Citometria, Centro Hospitalar e Universitário de Coimbra (CHUC), 3004-561 Coimbra, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-370 Coimbra, Portugal
- Ciências Biomédicas Laboratoriais, ESTESC - Coimbra Health School, Instituto Politécnico de Coimbra, 3046-854 Coimbra, Portugal
| | - Laura Goracci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Piazza dell' Università, 1, 06123 Perugia, Italy
| | - Stefano Bonciarelli
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Piazza dell' Università, 1, 06123 Perugia, Italy
| | - Pedro Domingues
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - M Rosário Domingues
- Mass Spectrometry Center, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
- CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago 3810-193 Aveiro, Portugal
| |
Collapse
|
6
|
Huda MN, Borrego EA, Guerena CD, Varela-Ramirez A, Aguilera RJ, Hamadani CM, Tanner EEL, Badruddoza AZM, Agarwal SK, Nurunnabi M. Topical Administration of an Apoptosis Inducer Mitigates Bleomycin-Induced Skin Fibrosis. ACS Pharmacol Transl Sci 2023; 6:829-841. [PMID: 37200808 PMCID: PMC10186622 DOI: 10.1021/acsptsci.3c00039] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Indexed: 05/20/2023]
Abstract
Pathological fibrosis is distinguished from physiological wound healing by persistent myofibroblast activation, suggesting that therapies that induce myofibroblast apoptosis selectively could prevent progression and potentially reverse the established fibrosis, such as for scleroderma (a heterogeneous autoimmune disease characterized by multiorgan fibrosis). Navitoclax (NAVI) is a BCL-2/BCL-xL inhibitor with antifibrotic properties and has been investigated as a potential therapeutic for fibrosis. NAVI makes myofibroblasts particularly vulnerable to apoptosis. However, despite NAVI's significant potency, clinical translation of BCL-2 inhibitors, NAVI in this case, is hindered due to the risk of thrombocytopenia. Therefore, in this work, we utilized a newly developed ionic liquid formulation of NAVI for direct topical application to the skin, thereby avoiding systemic circulation and off-target-mediated side effects. The ionic liquid composed of choline and octanoic acid (COA) at a 1:2 molar ionic ratio increases skin diffusion and transportation of NAVI and maintains their retention within the dermis for a prolonged duration. Topical administration of NAVI-mediated BCL-xL and BCL-2 inhibition results in the transition of myofibroblast to fibroblast and ameliorates pre-existing fibrosis, as demonstrated in a scleroderma mouse model. We have observed a significant reduction of α-SMA and collagen, which are known as fibrosis marker proteins, as a result of the inhibition of anti-apoptotic proteins BCL-2/BCL-xL. Overall, our findings show that COA-assisted topical delivery of NAVI upregulates apoptosis specific to myofibroblasts, with minimal presence of the drug in the systemic circulation, resulting in an accelerated therapeutic effect with no discernible drug-associated toxicity.
Collapse
Affiliation(s)
- Md Nurul Huda
- Department
of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
| | - Edgar A. Borrego
- Department
of Biological Sciences, College of Sciences, University of Texas at El Paso, El Paso, Texas 79956, United States
- Border
Biomedical Research Center, University of
Texas at El Paso, El Paso, Texas 79956, United States
| | - Cristina D. Guerena
- Department
of Biological Sciences, College of Sciences, University of Texas at El Paso, El Paso, Texas 79956, United States
- Border
Biomedical Research Center, University of
Texas at El Paso, El Paso, Texas 79956, United States
| | - Armando Varela-Ramirez
- Department
of Biological Sciences, College of Sciences, University of Texas at El Paso, El Paso, Texas 79956, United States
- Border
Biomedical Research Center, University of
Texas at El Paso, El Paso, Texas 79956, United States
| | - Renato J. Aguilera
- Department
of Biological Sciences, College of Sciences, University of Texas at El Paso, El Paso, Texas 79956, United States
- Border
Biomedical Research Center, University of
Texas at El Paso, El Paso, Texas 79956, United States
| | - Christine M. Hamadani
- Department
of Chemistry & Biochemistry, The University
of Mississippi, University, Mississippi 38677, United States
| | - Eden E. L. Tanner
- Department
of Chemistry & Biochemistry, The University
of Mississippi, University, Mississippi 38677, United States
| | - Abu Zayed Md Badruddoza
- Department
of Chemical and Life Sciences Engineering, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Sandeep K. Agarwal
- Department
of Medicine, Section of Immunology, Allergy and Rheumatology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Md Nurunnabi
- Department
of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
- Border
Biomedical Research Center, University of
Texas at El Paso, El Paso, Texas 79956, United States
- Biomedical Engineering, and Aerospace Center, College of Engineering, University of Texas at El Paso, El Paso, Texas 79956, United States
| |
Collapse
|
7
|
Sanges S, Rice L, Tu L, Valenzi E, Cracowski JL, Montani D, Mantero JC, Ternynck C, Marot G, Bujor AM, Hachulla E, Launay D, Humbert M, Guignabert C, Lafyatis R. Biomarkers of haemodynamic severity of systemic sclerosis-associated pulmonary arterial hypertension by serum proteome analysis. Ann Rheum Dis 2023; 82:365-373. [PMID: 36600187 PMCID: PMC9918672 DOI: 10.1136/ard-2022-223237] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022]
Abstract
OBJECTIVES To mine the serum proteome of patients with systemic sclerosis-associated pulmonary arterial hypertension (SSc-PAH) and to detect biomarkers that may assist in earlier and more effective diagnosis and treatment. METHODS Patients with limited cutaneous SSc, no extensive interstitial lung disease and no PAH-specific therapy were included. They were classified as cases if they had PAH confirmed by right heart catheterisation (RHC) and serum collected on the same day as RHC; and as controls if they had no clinical evidence of PAH. RESULTS Patients were mostly middle-aged females with anticentromere-associated SSc. Among 1129 proteins assessed by a high-throughput proteomic assay (SOMAscan), only 2 were differentially expressed and correlated significantly with pulmonary vascular resistance (PVR) in SSc-PAH patients (n=15): chemerin (ρ=0.62, p=0.01) and SET (ρ=0.62, p=0.01). To validate these results, serum levels of chemerin were measured by ELISA in an independent cohort. Chemerin levels were confirmed to be significantly higher (p=0.01) and correlate with PVR (ρ=0.42, p=0.04) in SSc-PAH patients (n=24). Chemerin mRNA expression was detected in fibroblasts, pulmonary artery smooth muscle cells (PA-SMCs)/pericytes and mesothelial cells in SSc-PAH lungs by single-cell RNA-sequencing. Confocal immunofluorescence revealed increased expression of a chemerin receptor, CMKLR1, on SSc-PAH PA-SMCs. SSc-PAH serum seemed to induce higher PA-SMC proliferation than serum from SSc patients without PAH. This difference appeared neutralised when adding the CMKLR1 inhibitor α-NETA. CONCLUSION Chemerin seems an interesting surrogate biomarker for PVR in SSc-PAH. Increased chemerin serum levels and CMKLR1 expression by PA-SMCs may contribute to SSc-PAH pathogenesis by inducing PA-SMC proliferation.
Collapse
Affiliation(s)
- Sébastien Sanges
- Boston University School of Medicine, E5 Arthritis Center, Boston, Massachusetts, USA
- Univ. Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
- INSERM, Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, Lille, France
- Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), Lille, France
- Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ReCONNET), Lille, France
| | - Lisa Rice
- Boston University School of Medicine, E5 Arthritis Center, Boston, Massachusetts, USA
| | - Ly Tu
- Université Paris Saclay, School of Medicine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hôpital Marie-Lannelongue, Le Plessis-Robinson, France
| | - Eleanor Valenzi
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | | | - David Montani
- Université Paris Saclay, School of Medicine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hôpital Marie-Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Hôpital Bicêtre, Kremlin-Bicêtre, France
| | - Julio C Mantero
- Boston University School of Medicine, E5 Arthritis Center, Boston, Massachusetts, USA
| | - Camille Ternynck
- Univ. Lille, CHU Lille, ULR 2694 - METRICS: Évaluation des technologies de santé et des pratiques médicales, Lille, France
| | - Guillemette Marot
- Univ. Lille, CHU Lille, ULR 2694 - METRICS: Évaluation des technologies de santé et des pratiques médicales, Lille, France
- Inria, MODAL: MOdels for Data Analysis and Learning, Lille, France
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UAR 2014 - US 41 - PLBS, bilille, Lille, France
| | - Andreea M Bujor
- Boston University School of Medicine, E5 Arthritis Center, Boston, Massachusetts, USA
| | - Eric Hachulla
- Univ. Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
- INSERM, Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, Lille, France
- Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), Lille, France
- Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ReCONNET), Lille, France
| | - David Launay
- Univ. Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
- INSERM, Lille, France
- CHU Lille, Département de Médecine Interne et Immunologie Clinique, Lille, France
- Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), Lille, France
- Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases Network (ReCONNET), Lille, France
| | - Marc Humbert
- Université Paris Saclay, School of Medicine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hôpital Marie-Lannelongue, Le Plessis-Robinson, France
- AP-HP, Department of Respiratory and Intensive Care Medicine, Hôpital Bicêtre, Kremlin-Bicêtre, France
| | - Christophe Guignabert
- Université Paris Saclay, School of Medicine, Le Kremlin-Bicêtre, France
- INSERM UMR_S 999, Hôpital Marie-Lannelongue, Le Plessis-Robinson, France
| | - Robert Lafyatis
- Division of Rheumatology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
8
|
The Clinical Significance of Salusins in Systemic Sclerosis-A Cross-Sectional Study. Diagnostics (Basel) 2023; 13:diagnostics13050848. [PMID: 36899991 PMCID: PMC10001236 DOI: 10.3390/diagnostics13050848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/14/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Background: Systemic sclerosis (SSc) is a connective tissue disease manifesting with progressive fibrosis of the skin and internal organs. Its pathogenesis is strictly associated with vascular disfunction and damage. Salusin-α and salusin-β, endogenous peptides regulating secretion of pro-inflammatory cytokines and vascular smooth muscle proliferation, may potentially play a role in SSc pathogenesis. Objectives: The aim of this study was to assess the concentration of salusins in sera of patients with SSc and healthy controls and to evaluate correlations between the salusins levels and selected clinical parameters within the study group. Materials and methods: 48 patients with SSc (44 women; mean age, 56.4, standard deviation, 11.4) and 25 adult healthy volunteers (25 women; mean age, 55.2, standard deviation, 11.2) were enrolled. All patients with SSc were treated with vasodilators and twenty-seven of them (56%) also received immunosuppressive therapy. Results: Circulating salusin-α was significantly elevated in patients with SSc in comparison to healthy controls (U = 350.5, p = 0.004). Patients with SSc receiving immunosuppression had higher serum salusin-α concentrations compared with those without immunosuppressive therapy (U = 176.0, p = 0.026). No correlation was observed between salusins concentrations and skin or internal organ involvement parameters. Conclusions: Salusin-α, a bioactive peptide mitigating the endothelial disfunction, was elevated in patients with systemic sclerosis receiving vasodilators and immunosuppressants. Increased salusin-α concertation may be associated with the initiation of atheroprotective processes in patients with SSc managed pharmacologically, which requires verification in future studies.
Collapse
|
9
|
Centromere defects, chromosome instability, and cGAS-STING activation in systemic sclerosis. Nat Commun 2022; 13:7074. [PMID: 36400785 PMCID: PMC9674829 DOI: 10.1038/s41467-022-34775-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/04/2022] [Indexed: 11/21/2022] Open
Abstract
Centromere defects in Systemic Sclerosis (SSc) have remained unexplored despite the fact that many centromere proteins were discovered in patients with SSc. Here we report that lesion skin fibroblasts from SSc patients show marked alterations in centromeric DNA. SSc fibroblasts also show DNA damage, abnormal chromosome segregation, aneuploidy (only in diffuse cutaneous (dcSSc)) and micronuclei (in all types of SSc), some of which lose centromere identity while retaining centromere DNA sequences. Strikingly, we find cytoplasmic "leaking" of centromere proteins in limited cutaneous SSc (lcSSc) fibroblasts. Cytoplasmic centromere proteins co-localize with antigen presenting MHC Class II molecules, which correlate precisely with the presence of anti-centromere antibodies. CENPA expression and micronuclei formation correlate highly with activation of the cGAS-STING/IFN-β pathway as well as markers of reactive oxygen species (ROS) and fibrosis, ultimately suggesting a link between centromere alterations, chromosome instability, SSc autoimmunity, and fibrosis.
Collapse
|
10
|
Matei AE, Kubánková M, Xu L, Györfi AH, Boxberger E, Soteriou D, Papava M, Prater J, Hong X, Bergmann C, Kräter M, Schett G, Guck J, Distler JHW. Identification of a Distinct Monocyte-Driven Signature in Systemic Sclerosis Using Biophysical Phenotyping of Circulating Immune Cells. Arthritis Rheumatol 2022; 75:768-781. [PMID: 36281753 DOI: 10.1002/art.42394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 09/08/2022] [Accepted: 10/18/2022] [Indexed: 11/11/2022]
Abstract
OBJECTIVE Pathologically activated circulating immune cells, including monocytes, play major roles in systemic sclerosis (SSc). Their functional characterization can provide crucial information with direct clinical relevance. However, tools for the evaluation of pathologic immune cell activation and, in general, of clinical outcomes in SSc are scarce. Biophysical phenotyping (including characterization of cell mechanics and morphology) provides access to a novel, mostly unexplored layer of information regarding pathophysiologic immune cell activation. We hypothesized that the biophysical phenotyping of circulating immune cells, reflecting their pathologic activation, can be used as a clinical tool for the evaluation and risk stratification of patients with SSc. METHODS We performed biophysical phenotyping of circulating immune cells by real-time fluorescence and deformability cytometry (RT-FDC) in 63 SSc patients, 59 rheumatoid arthritis (RA) patients, 28 antineutrophil cytoplasmic antibody-associated vasculitis (AAV) patients, and 22 age- and sex-matched healthy donors. RESULTS We identified a specific signature of biophysical properties of circulating immune cells in SSc patients that was mainly driven by monocytes. Since it is absent in RA and AAV, this signature reflects an SSc-specific monocyte activation rather than general inflammation. The biophysical properties of monocytes indicate current disease activity, the extent of skin or lung fibrosis, and the severity of manifestations of microvascular damage, as well as the risk of disease progression in SSc patients. CONCLUSION Changes in the biophysical properties of circulating immune cells reflect their pathologic activation in SSc patients and are associated with clinical outcomes. As a high-throughput approach that requires minimal preparations, RT-FDC-based biophysical phenotyping of monocytes can serve as a tool for the evaluation and risk stratification of patients with SSc.
Collapse
Affiliation(s)
- Alexandru-Emil Matei
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Markéta Kubánková
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany, and Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Liyan Xu
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Andrea-Hermina Györfi
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Evgenia Boxberger
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Despina Soteriou
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany
| | - Maria Papava
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Julia Prater
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Xuezhi Hong
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christina Bergmann
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Martin Kräter
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany, and Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Georg Schett
- Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jochen Guck
- Max Planck Institute for the Science of Light & Max-Planck-Center für Physik und Medizin, Erlangen, Germany, and Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Jörg H W Distler
- Department of Rheumatology and Hiller Research Unit, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Department of Internal Medicine 3-Rheumatology and Immunology and Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU), and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
11
|
Kim Y, Nam Y, Rim YA, Ju JH. Anti-fibrotic effect of a selective estrogen receptor modulator in systemic sclerosis. Stem Cell Res Ther 2022; 13:303. [PMID: 35841004 PMCID: PMC9284699 DOI: 10.1186/s13287-022-02987-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 06/17/2022] [Indexed: 11/16/2022] Open
Abstract
Background The rarity of systemic sclerosis (SSc) has hampered the development of therapies for this intractable autoimmune disease. Induced pluripotent stem cell (iPSC) can be differentiated into the key disease-affected cells in vitro. The generation of patient-derived iPSCs has opened up possibilities for rare disease modeling. Since these cells can recapitulate the disease phenotypes of the cell in question, they are useful high-throughput platforms for screening for drugs that can reverse these abnormal phenotypes. Methods SSc iPSC was generated from PBMC by Sendai virus. Human iPSC lines from SSc patients were differentiated into dermal fibroblasts and keratinocytes. The iPSC-derived differentiated cells from the SSc patients were used on high-throughput platforms to screen for FDA-approved drugs that could be effective treatments for SSc. Results Skin organoids were generated from these cells exhibited fibrosis that resembled SSc skin. Screening of the 770-FDA-approved drug library showed that the anti-osteoporotic drug raloxifene reduced SSc iPSC-derived fibroblast proliferation and extracellular matrix production and skin fibrosis in organoids and bleomycin-induced SSc-model mice. Conclusions This study reveals that a disease model of systemic sclerosis generated using iPSCs-derived skin organoid is a novel tool for in vitro and in vivo dermatologic research. Since raloxifene and bazedoxifene are well-tolerated anti-osteoporotic drugs, our findings suggest that selective estrogen receptor modulator (SERM)-class drugs could treat SSc fibrosis. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02987-w.
Collapse
Affiliation(s)
- Yena Kim
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea.,YiPSCELL Inc., 47-3, Banpo-dearo 39-gil, Seocho-gu, Seoul, Republic of Korea
| | - Yoojun Nam
- YiPSCELL Inc., 47-3, Banpo-dearo 39-gil, Seocho-gu, Seoul, Republic of Korea
| | - Yeri Alice Rim
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ji Hyeon Ju
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, Seoul, South Korea. .,YiPSCELL Inc., 47-3, Banpo-dearo 39-gil, Seocho-gu, Seoul, Republic of Korea. .,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 137-040, Republic of Korea.
| |
Collapse
|
12
|
Kuley R, Stultz RD, Duvvuri B, Wang T, Fritzler MJ, Hesselstrand R, Nelson JL, Lood C. N-Formyl Methionine Peptide-Mediated Neutrophil Activation in Systemic Sclerosis. Front Immunol 2022; 12:785275. [PMID: 35069556 PMCID: PMC8766990 DOI: 10.3389/fimmu.2021.785275] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022] Open
Abstract
Exaggerated neutrophil activation and formation of neutrophil extracellular traps (NETs) are reported in systemic sclerosis (SSc) but its involvement in SSc pathogenesis is not clear. In the present study we assessed markers of neutrophil activation and NET formation in SSc patients in relation to markers of inflammation and disease phenotype. Factors promoting neutrophil activation in SSc remain largely unknown. Among the neutrophil activating factors, mitochondrial-derived N-formyl methionine (fMet) has been reported in several autoinflammatory conditions. The aim of the current study is to assess whether SSc patients have elevated levels of fMet and the role of fMet in neutrophil-mediated inflammation on SSc pathogenesis. Markers of neutrophil activation (calprotectin, NETs) and levels of fMet were analyzed in plasma from two SSc cohorts (n=80 and n=20, respectively) using ELISA. Neutrophil activation assays were performed in presence or absence of formyl peptide receptor 1 (FPR1) inhibitor cyclosporin H. Elevated levels of calprotectin and NETs were observed in SSc patients as compared to healthy controls (p<0.0001) associating with SSc clinical disease characteristics. Further, SSc patients had elevated levels of circulating fMet as compared to healthy controls (p<0.0001). Consistent with a role for fMet-mediated neutrophil activation, fMet levels correlated with levels of calprotectin and NETs (r=0.34, p=0.002; r=0.29, p<0.01 respectively). Additionally, plasma samples from SSc patients with high levels of fMet induced de novo neutrophil activation through FPR1-dependent mechanisms. Our data for the first time implicates an important role for the mitochondrial component fMet in promoting neutrophil-mediated inflammation in SSc.
Collapse
Affiliation(s)
- Runa Kuley
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, United States
| | - Ryan D Stultz
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, United States
| | - Bhargavi Duvvuri
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, United States
| | - Ting Wang
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, United States
| | - Marvin J Fritzler
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Roger Hesselstrand
- Department of Clinical Sciences Lund University, Section of Rheumatology, Lund, Sweden
| | - J Lee Nelson
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, United States.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Christian Lood
- Department of Medicine, Division of Rheumatology, University of Washington, Seattle, WA, United States
| |
Collapse
|
13
|
Hromadka M, Baxa J, Seidlerova J, Miklik R, Rajdl D, Sudova V, Suchy D, Rokyta R. Myocardial Involvement Detected Using Cardiac Magnetic Resonance Imaging in Patients with Systemic Sclerosis: A Prospective Observational Study. J Clin Med 2021; 10:jcm10225364. [PMID: 34830647 PMCID: PMC8620356 DOI: 10.3390/jcm10225364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/12/2021] [Accepted: 11/15/2021] [Indexed: 11/16/2022] Open
Abstract
Introduction and objectives: Cardiac involvement in systemic sclerosis (SSc) patients affects mortality. Cardiac magnetic resonance (CMR) is capable of detecting structural changes, including diffuse myocardial fibrosis that may develop over time. Our aim was to evaluate myocardial structure and function changes using CMR in patients with SSc without known cardiac disease during a 5-year follow-up and find possible correlations with selected biomarkers. Methods: A total of 25 patients underwent baseline and follow-up CMR examinations according to a pre-specified protocol. Standard biochemistry, five biomarkers (hsTnI, NT-proBNP, galectin-3, sST2, and GDF-15), and disease-specific functional parameters enabling the classification of disease severity were also measured. Results: After five years, no patient suffered from manifest heart disease. Mean extracellular volume (ECV) and T1 mapping values did not change significantly (p ≥ 0.073). However, individual increases in native T1 time and ECV correlated with increased galectin-3 serum levels (r = 0.56; p = 0.0050, and r = 0.71; p = 0.0001, respectively). The progression of skin involvement assessed using the Rodnan skin score and a decrease in the diffusing capacity of the lungs were associated with increased GDF-15 values (r = 0.63; p = 0.0009, and r = −0.51; p = 0.011, respectively). Conclusions: During the 5-year follow-up, there was no new onset of heart disease observed in patients with SSc. However, in some patients, CMR detected progression of sub-clinical myocardial fibrosis that significantly correlated with elevated galectin-3 levels. GDF-15 values were found to be associated with disease severity progression.
Collapse
Affiliation(s)
- Milan Hromadka
- Department of Cardiology, University Hospital and Faculty of Medicine in Pilsen and Faculty Hospital, Charles University, Alej Svobody 80, 304 60 Pilsen, Czech Republic; (M.H.); (R.R.)
| | - Jan Baxa
- Department of Imaging Methods, University Hospital and Faculty of Medicine in Pilsen, Charles University, Alej Svobody 80, 304 60 Pilsen, Czech Republic;
| | - Jitka Seidlerova
- Internal Department II, University Hospital and Faculty of Medicine in Pilsen, Charles University, Edvarda Benese 1128/13, 305 99 Pilsen, Czech Republic;
| | - Roman Miklik
- Department of Cardiology, University Hospital and Faculty of Medicine in Pilsen and Faculty Hospital, Charles University, Alej Svobody 80, 304 60 Pilsen, Czech Republic; (M.H.); (R.R.)
- Correspondence:
| | - Dan Rajdl
- Department of Clinical Biochemistry and Hematology, University Hospital and Faculty of Medicine in Pilsen, Alej Svobody 80, 304 60 Pilsen, Czech Republic; (D.R.); (V.S.)
| | - Vendula Sudova
- Department of Clinical Biochemistry and Hematology, University Hospital and Faculty of Medicine in Pilsen, Alej Svobody 80, 304 60 Pilsen, Czech Republic; (D.R.); (V.S.)
| | - David Suchy
- Department of Clinical Pharmacology, Rheumatology, University Hospital and Faculty of Medicine in Pilsen, Charles University, Edvarda Benese 1128/13, 305 99 Pilsen, Czech Republic;
| | - Richard Rokyta
- Department of Cardiology, University Hospital and Faculty of Medicine in Pilsen and Faculty Hospital, Charles University, Alej Svobody 80, 304 60 Pilsen, Czech Republic; (M.H.); (R.R.)
| |
Collapse
|
14
|
Alkema W, Koenen H, Kersten BE, Kaffa C, Dinnissen JWB, Damoiseaux JGMC, Joosten I, Driessen-Diks S, van der Molen RG, Vonk MC, Smeets RL. Autoantibody profiles in systemic sclerosis; a comparison of diagnostic tests. Autoimmunity 2021; 54:148-155. [PMID: 33818234 DOI: 10.1080/08916934.2021.1907842] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Autoimmune antibody profiling plays a prominent role in both classification and prognosis of systemic sclerosis (SSc). In the last years novel autoantibodies have been discovered and have become available in diagnostic assays. However, standardization in autoimmune serology is lacking, which may have a negative impact on the added value of autoantibodies in diagnosis and prognosis of SSc. In this paper we describe the comparison of commercially available diagnostic assays for the detection of SSc-associated autoantibodies and explored the coexistence of multiple SSc-associated autoantibodies within patients. METHODS Serum samples of 347 patients from the Nijmegen Systemic Sclerosis Cohort were included in this study. All patients fulfilled the ACR/EULAR 2013 classification criteria for SSc and were classified as DcSSc or LcSSc according to the Leroy and Medsger criteria. All samples were evaluated on standard laboratory diagnostic tests for detection of SSc-specific autoantibodies CENPA and CENPB (ACA), Scl-70 (ATA), RNA Polymerase III (rp11/155) (ARA), and SSc-associated autoantibodies Fibrillarin, Th-To, PM-scl75, PM-Scl100, RNP68/A/C, Ku, NOR90, and PDGFR from suppliers EUROIMMUN, D-tek and Thermo Fisher Scientific. RESULTS We found that 79% of the patients was positive for one or more of the SSc autoantibodies. Overall, a high agreement was observed between the diagnostic methods for the SSC-specific autoantibodies listed in the ACR/EULAR criteria (ATA, ACA, and ARA) (Cohen's kappa 0.53-0.97). However, a lower agreement was found for SSc-associated autoantibodies PM-Scl, and Ku, as well as for the SSc-specific autoantibodies fibrillarin and Th-To. Furthermore, the data revealed that the presence of ATA, ARA and ACA is predominantly mutually exclusive, with only a fraction of the patients testing positive for both ATA and ARA. CONCLUSION Our data showed high concordance of prevalent SSc-specific autoantibodies between different diagnostic assays. Further standardisation for low prevalent SSc-specific and SSc-associated autoantibodies is needed.
Collapse
Affiliation(s)
- Wynand Alkema
- Institute of Life Science and Technology, Hanze University of Applied Sciences, Groningen, The Netherlands.,TenWise B.V., Oss, The Netherlands
| | - Hans Koenen
- Laboratory for Medical Immunology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Brigit E Kersten
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Charlotte Kaffa
- Center for Molecular and Biomolecular informatics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jacqueline W B Dinnissen
- Radboudumc Laboratory for Diagnostics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jan G M C Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Irma Joosten
- Laboratory for Medical Immunology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sophie Driessen-Diks
- Laboratory for Medical Immunology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Renate G van der Molen
- Laboratory for Medical Immunology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Madelon C Vonk
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ruben L Smeets
- Laboratory for Medical Immunology, Radboud University Medical Center, Nijmegen, The Netherlands.,Radboudumc Laboratory for Diagnostics, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
15
|
Butt S, Jeppesen JL, Iversen LV, Fenger M, Eugen-Olsen J, Andersson C, Jacobsen S. Association of soluble urokinase plasminogen activator receptor levels with fibrotic and vascular manifestations in systemic sclerosis. PLoS One 2021; 16:e0247256. [PMID: 33617568 PMCID: PMC7899346 DOI: 10.1371/journal.pone.0247256] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 02/03/2021] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE We assessed the association of suPAR (soluble urokinase plasminogen activator receptor) plasma levels with fibrotic and vascular manifestations in patients with systemic sclerosis (SSc). METHODS suPAR plasma levels were measured in 121 consecutive patients with SSc and correlated to pulmonary and vascular features of SSc, including interstitial lung disease as characterized by percentage of predicted CO diffusing capacity (DLco) and forced vital capacity (FVC), pulmonary fibrosis by computed tomography, and pulmonary arterial hypertension, telangiectasias, and digital ulcers. RESULTS Overall, 121 SSc patients (84% females; mean age, 57 ± 12 [range: 22-79] years) were enrolled; 35% had diffuse cutaneous SSc. suPAR plasma levels ranged from 1.3-10.2 [median: 2.9 (p25-p75: 2.3-3.9)] ng/mL. Log(suPAR) levels correlated with DLco (r = -0.41, p <0.0001) and FVC (r = -0.26, p = 0.004), also when adjusted for age, sex, and pulmonary hypertension. A suPAR cut-off level of >2.5 ng/mL showed a sensitivity of 91% for identifying patients with either DLco <50% or FVC < 60% of the predicted values. Similarly, 19 (90%) had a suPAR >2.5 ng/mL among those diagnosed with pulmonary fibrosis vs. 59 (60%) among those who did not (p = 0.008). suPAR values were not associated with vascular manifestations. CONCLUSION suPAR levels strongly correlated with pulmonary involvement in SSc. Future studies should test if suPAR estimation can be used for surveillance of severe pulmonary involvement in SSc.
Collapse
Affiliation(s)
- Sheraz Butt
- Department of Internal Medicine, Amager and Hvidovre University Hospital, Glostrup, Denmark
- * E-mail:
| | - Jørgen L. Jeppesen
- Department of Internal Medicine, Amager and Hvidovre University Hospital, Glostrup, Denmark
| | - Line Vinderslev Iversen
- Department of Dermatology, Bispebjerg University Hospital, Copenhagen, Denmark
- Department of Dermatology and Allergy, Odense University Hospital, Odense, Denmark
| | - Mogens Fenger
- Department of Clinical Biochemistry, Amager and Hvidovre University Hospital, Hvidovre, Denmark
| | - Jesper Eugen-Olsen
- Clinical Research Centre, Amager and Hvidovre University Hospital, Hvidovre, Denmark
| | - Charlotte Andersson
- Department of Cardiology, Herlev-Gentofte University Hospital, Hellerup, Denmark
| | - Søren Jacobsen
- Copenhagen Lupus and Vasculitis Clinic, Centre for Rheumatology and Spine Disease, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
16
|
Soluble Biomarkers for Prediction of Vascular and Gastrointestinal Disease Severity in Patients with Systemic Sclerosis. CURRENT TREATMENT OPTIONS IN RHEUMATOLOGY 2021. [DOI: 10.1007/s40674-021-00171-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
17
|
Abstract
Hydrogels, swellable hydrophilic polymer networks fabricated through chemical cross-linking or physical entanglement are increasingly utilized in various biomedical applications over the past few decades. Hydrogel-based microparticles, dressings and microneedle patches have been explored to achieve safe, sustained and on-demand therapeutic purposes toward numerous skin pathologies, through incorporation of stimuli-responsive moieties and therapeutic agents. More recently, these platforms are expanded to fulfill the diagnostic and monitoring role. Herein, the development of hydrogel technology to achieve diagnosis and monitoring of pathological skin conditions are highlighted, with proteins, nucleic acids, metabolites, and reactive species employed as target biomarkers, among others. The scope of this review includes the characteristics of hydrogel materials, its fabrication procedures, examples of diagnostic studies, as well as discussion pertaining clinical translation of hydrogel systems.
Collapse
|
18
|
Ramírez-Aragón M, Hernández-Sánchez F, Rodríguez-Reyna TS, Buendía-Roldán I, Güitrón-Castillo G, Núñez-Alvarez CA, Hernández-Ramírez DF, Benavides-Suárez SA, Esquinca-González A, Torres-Machorro AL, Mendoza-Milla C. The Transcription Factor SCX is a Potential Serum Biomarker of Fibrotic Diseases. Int J Mol Sci 2020; 21:ijms21145012. [PMID: 32708589 PMCID: PMC7404299 DOI: 10.3390/ijms21145012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/19/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022] Open
Abstract
Fibrosing diseases are causes of morbidity and mortality around the world, and they are characterized by excessive extracellular matrix (ECM) accumulation. The bHLH transcription factor scleraxis (SCX) regulates the synthesis of ECM proteins in heart fibrosis. SCX expression was evaluated in lung fibroblasts and tissue derived from fibrotic disease patients and healthy controls. We also measured SCX in sera from 57 healthy controls, and 56 Idiopathic Pulmonary Fibrosis (IPF), 40 Hypersensitivity Pneumonitis (HP), and 100 Systemic Sclerosis (SSc) patients. We report high SCX expression in fibroblasts and tissue from IPF patients versus controls. High SCX-serum levels were observed in IPF (0.663 ± 0.559 ng/mL, p < 0.01) and SSc (0.611 ± 0.296 ng/mL, p < 0.001), versus controls (0.351 ± 0.207 ng/mL) and HP (0.323 ± 0.323 ng/mL). Serum levels of the SCX heterodimerization partner, TCF3, did not associate with fibrotic illness. IPF patients with severely affected respiratory capacities and late-stage SSc patients presenting anti-topoisomerase I antibodies and interstitial lung disease showed the highest SCX-serum levels. SCX gain-of-function induced the expression of alpha-smooth muscle actin (α-SMA/ACTA2) in fibroblasts when co-overexpressed with TCF3. As late and severe stages of the fibrotic processes correlated with high circulating SCX, we postulate it as a candidate biomarker of fibrosis and a potential therapeutic target.
Collapse
Affiliation(s)
- Miguel Ramírez-Aragón
- Departamento de Investigación en Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Colonia Belisario Domínguez Sección XVI, Alcaldía Tlalpan, Mexico City 14080, Mexico; (M.R.-A.); (I.B.-R.); (G.G.-C.)
- Departamento de Neuropatología Molecular, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de Mexico, Mexico City 04510, Mexico
| | - Fernando Hernández-Sánchez
- Departamento de Investigación en Virología y Micología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Colonia Belisario Domínguez Sección XVI, Alcaldía Tlalpan, Mexico City 14080, Mexico;
| | - Tatiana S. Rodríguez-Reyna
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga 15, Colonia Belisario Domínguez Sección XVI. Alcaldía Tlalpan, Mexico City 14080, Mexico; (T.S.R.-R.); (C.A.N.-A.); (D.F.H.-R.); (S.A.B.-S.); (A.E.-G.)
| | - Ivette Buendía-Roldán
- Departamento de Investigación en Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Colonia Belisario Domínguez Sección XVI, Alcaldía Tlalpan, Mexico City 14080, Mexico; (M.R.-A.); (I.B.-R.); (G.G.-C.)
| | - Gael Güitrón-Castillo
- Departamento de Investigación en Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Colonia Belisario Domínguez Sección XVI, Alcaldía Tlalpan, Mexico City 14080, Mexico; (M.R.-A.); (I.B.-R.); (G.G.-C.)
| | - Carlos A. Núñez-Alvarez
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga 15, Colonia Belisario Domínguez Sección XVI. Alcaldía Tlalpan, Mexico City 14080, Mexico; (T.S.R.-R.); (C.A.N.-A.); (D.F.H.-R.); (S.A.B.-S.); (A.E.-G.)
| | - Diego F. Hernández-Ramírez
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga 15, Colonia Belisario Domínguez Sección XVI. Alcaldía Tlalpan, Mexico City 14080, Mexico; (T.S.R.-R.); (C.A.N.-A.); (D.F.H.-R.); (S.A.B.-S.); (A.E.-G.)
| | - Sergio A. Benavides-Suárez
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga 15, Colonia Belisario Domínguez Sección XVI. Alcaldía Tlalpan, Mexico City 14080, Mexico; (T.S.R.-R.); (C.A.N.-A.); (D.F.H.-R.); (S.A.B.-S.); (A.E.-G.)
| | - Alexia Esquinca-González
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga 15, Colonia Belisario Domínguez Sección XVI. Alcaldía Tlalpan, Mexico City 14080, Mexico; (T.S.R.-R.); (C.A.N.-A.); (D.F.H.-R.); (S.A.B.-S.); (A.E.-G.)
| | - Ana Lilia Torres-Machorro
- Departamento de Investigación en Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Colonia Belisario Domínguez Sección XVI, Alcaldía Tlalpan, Mexico City 14080, Mexico; (M.R.-A.); (I.B.-R.); (G.G.-C.)
- Consejo Nacional de Ciencia y Tecnología and Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Colonia Belisario Domínguez Sección XVI, Alcaldía Tlalpan, Mexico City 14080, Mexico
- Correspondence: (A.L.T.-M.); (C.M.-M.); Tel.: +52-555-487-1700 (ext.5257) (A.L.T.-M. & C.M.-M.)
| | - Criselda Mendoza-Milla
- Departamento de Investigación en Fibrosis Pulmonar, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Calzada de Tlalpan 4502, Colonia Belisario Domínguez Sección XVI, Alcaldía Tlalpan, Mexico City 14080, Mexico; (M.R.-A.); (I.B.-R.); (G.G.-C.)
- Correspondence: (A.L.T.-M.); (C.M.-M.); Tel.: +52-555-487-1700 (ext.5257) (A.L.T.-M. & C.M.-M.)
| |
Collapse
|
19
|
Feng D, Gerarduzzi C. Emerging Roles of Matricellular Proteins in Systemic Sclerosis. Int J Mol Sci 2020; 21:E4776. [PMID: 32640520 PMCID: PMC7369781 DOI: 10.3390/ijms21134776] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/11/2020] [Accepted: 06/13/2020] [Indexed: 02/07/2023] Open
Abstract
Systemic sclerosis is a rare chronic heterogenous disease that involves inflammation and vasculopathy, and converges in end-stage development of multisystem tissue fibrosis. The loss of tight spatial distribution and temporal expression of proteins in the extracellular matrix (ECM) leads to progressive organ stiffening, which is a hallmark of fibrotic disease. A group of nonstructural matrix proteins, known as matricellular proteins (MCPs) are implicated in dysregulated processes that drive fibrosis such as ECM remodeling and various cellular behaviors. Accordingly, MCPs have been described in the context of fibrosis in sclerosis (SSc) as predictive disease biomarkers and regulators of ECM synthesis, with promising therapeutic potential. In this present review, an informative summary of major MCPs is presented highlighting their clear correlations to SSc- fibrosis.
Collapse
Affiliation(s)
- Daniel Feng
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada;
- Centre de recherche de l’Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l’Université de Montréal, Montréal, QC H1T 2M4, Canada
| | - Casimiro Gerarduzzi
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada;
- Centre de recherche de l’Hôpital Maisonneuve-Rosemont, Faculté de Médecine, Centre affilié à l’Université de Montréal, Montréal, QC H1T 2M4, Canada
- Département de Médecine, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| |
Collapse
|
20
|
Smeets RL, Kersten BE, Joosten I, Kaffa C, Alkema W, Koenen HJPM, Vonk MC. Diagnostic profiles for precision medicine in systemic sclerosis; stepping forward from single biomarkers towards pathophysiological panels. Autoimmun Rev 2020; 19:102515. [PMID: 32173517 DOI: 10.1016/j.autrev.2020.102515] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 11/14/2019] [Indexed: 01/07/2023]
Abstract
Systemic sclerosis (SSc) is an autoimmune disease which is characterized by vasculopathy, tissue fibrosis and activation of the innate and adaptive immune system. Clinical features of the disease consists of skin thickening and internal organ involvement. Due to the heterogeneous nature of the disease it is difficult to predict disease progression and complications. Despite the discovery of novel autoantibodies associated with SSc, there is an unmet need for biomarkers for diagnosis, disease progression and response to treatment. To date, the use of single (surrogate) biomarkers for these purposes has been unsuccessful. Combining multiple biomarkers in to predictive panels or ultimately algorithms could be more precise. Given the limited therapeutic options and poor prognosis of many SSc patients, a better understanding of the immune-pathofysiological profiles might aid to an adjusted therapeutic approach. Therefore, we set out to explore immunological fingerprints in various clinically defined forms of SSc. We used multilayer profiling to identify unique immune profiles underlying distinct autoantibody signatures. These immune profiles could fill the unmet need for prognosis and response to therapy in SSc. Here, we present 3 pathophysiological fingerprints in SSc based on the expression of circulating antibodies, vascular markers and immunomodulatory mediators.
Collapse
Affiliation(s)
- Ruben L Smeets
- Radboudumc laboratory for Diagnostics, Radboud university medical center, the Netherlands; Laboratory for Medical Immunology, Radboud university medical center, the Netherlands.
| | - Brigit E Kersten
- Department of Rheumatology, Radboud university medical center, the Netherlands
| | - Irma Joosten
- Laboratory for Medical Immunology, Radboud university medical center, the Netherlands
| | - Charlotte Kaffa
- Center for Molecular and Biomolecular Informatics, Radboud university medical center, the Netherlands
| | - Wynand Alkema
- Center for Molecular and Biomolecular Informatics, Radboud university medical center, the Netherlands
| | - Hans J P M Koenen
- Laboratory for Medical Immunology, Radboud university medical center, the Netherlands
| | - Madelon C Vonk
- Department of Rheumatology, Radboud university medical center, the Netherlands
| |
Collapse
|
21
|
Teodoro WR, de Jesus Queiroz ZA, Dos Santos LA, Catanozi S, Dos Santos Filho A, Bueno C, Vendramini MBG, Fernezlian SDM, Eher EM, Sampaio-Barros PD, Pasoto SG, Lopes FDTQS, Velosa APP, Capelozzi VL. Proposition of a novel animal model of systemic sclerosis induced by type V collagen in C57BL/6 mice that reproduces fibrosis, vasculopathy and autoimmunity. Arthritis Res Ther 2019; 21:278. [PMID: 31829272 PMCID: PMC6907238 DOI: 10.1186/s13075-019-2052-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/05/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Type V collagen (Col V) has the potential to become an autoantigen and has been associated with the pathogenesis of systemic sclerosis (SSc). We characterized serological, functional, and histopathological features of the skin and lung in a novel SSc murine model induced by Col V immunization. METHODS Female C57BL/6 mice (n = 19, IMU-COLV) were subcutaneously immunized with two doses of Col V (125 μg) emulsified in complete Freund adjuvant, followed by two intramuscular boosters. The control group (n = 19) did not receive Col V. After 120 days, we examined the respiratory mechanics, serum autoantibodies, and vascular manifestations of the mice. The skin and lung inflammatory processes and the collagen gene/protein expressions were analyzed. RESULTS Vascular manifestations were characterized by endothelial cell activity and apoptosis, as shown by the increased expression of VEGF, endothelin-1, and caspase-3 in endothelial cells. The IMU-COLV mice presented with increased tissue elastance and a nonspecific interstitial pneumonia (NSIP) histologic pattern in the lung, combined with the thickening of the small and medium intrapulmonary arteries, increased Col V fibers, and increased COL1A1, COL1A2, COL3A1, COL5A1, and COL5A2 gene expression. The skin of the IMU-COLV mice showed thickness, epidermal rectification, decreased papillary dermis, atrophied appendages, and increased collagen, COL5A1, and COL5A2 gene expression. Anti-collagen III and IV and ANA antibodies were detected in the sera of the IMU-COLV mice. CONCLUSION We demonstrated that cutaneous, vascular, and pulmonary remodeling are mimicked in the Col V-induced SSc mouse model, which thus represents a suitable preclinical model to study the mechanisms and therapeutic approaches for SSc.
Collapse
Affiliation(s)
- Walcy Rosolia Teodoro
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, São Paulo, SP, BR, Av. Dr. Arnaldo, 455, sala 3124, Cerqueira César, São Paulo, SP, 01246-903, Brazil.
| | - Zelita Aparecida de Jesus Queiroz
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, São Paulo, SP, BR, Av. Dr. Arnaldo, 455, sala 3124, Cerqueira César, São Paulo, SP, 01246-903, Brazil
| | - Lais Araujo Dos Santos
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, São Paulo, SP, BR, Av. Dr. Arnaldo, 455, sala 3124, Cerqueira César, São Paulo, SP, 01246-903, Brazil
| | - Sergio Catanozi
- Lipid Laboratory of the Endocrinology and Metabology Discipline of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, Sao Paulo, SP, Brazil
| | - Antonio Dos Santos Filho
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, São Paulo, SP, BR, Av. Dr. Arnaldo, 455, sala 3124, Cerqueira César, São Paulo, SP, 01246-903, Brazil
| | - Cleonice Bueno
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, São Paulo, SP, BR, Av. Dr. Arnaldo, 455, sala 3124, Cerqueira César, São Paulo, SP, 01246-903, Brazil
| | - Margarete B G Vendramini
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, São Paulo, SP, BR, Av. Dr. Arnaldo, 455, sala 3124, Cerqueira César, São Paulo, SP, 01246-903, Brazil
| | - Sandra de Morais Fernezlian
- Department of Pathology of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, Sao Paulo, SP, Brazil
| | - Esmeralda M Eher
- Department of Pathology of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, Sao Paulo, SP, Brazil
| | - Percival D Sampaio-Barros
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, São Paulo, SP, BR, Av. Dr. Arnaldo, 455, sala 3124, Cerqueira César, São Paulo, SP, 01246-903, Brazil
| | - Sandra Gofinet Pasoto
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, São Paulo, SP, BR, Av. Dr. Arnaldo, 455, sala 3124, Cerqueira César, São Paulo, SP, 01246-903, Brazil
| | - Fernanda Degobbi T Q S Lopes
- Experimental Therapy Laboratory of the Department of Clinical Medicine of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, Sao Paulo, SP, Brazil
| | - Ana Paula Pereira Velosa
- Rheumatology Division of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, São Paulo, SP, BR, Av. Dr. Arnaldo, 455, sala 3124, Cerqueira César, São Paulo, SP, 01246-903, Brazil
| | - Vera Luiza Capelozzi
- Department of Pathology of the Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, FMUSP, Sao Paulo, SP, Brazil
| |
Collapse
|
22
|
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: 1.7] [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
|
23
|
Wermuth PJ, Piera-Velazquez S, Rosenbloom J, Jimenez SA. Existing and novel biomarkers for precision medicine in systemic sclerosis. Nat Rev Rheumatol 2019; 14:421-432. [PMID: 29789665 DOI: 10.1038/s41584-018-0021-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The discovery and validation of biomarkers resulting from technological advances in the analysis of genomic, transcriptomic, lipidomic and metabolomic pathways involved in the pathogenesis of complex human diseases have led to the development of personalized and rationally designed approaches for the clinical management of such disorders. Although some of these approaches have been applied to systemic sclerosis (SSc), an unmet need remains for validated, non-invasive biomarkers to aid in the diagnosis of SSc, as well as in the assessment of disease progression and response to therapeutic interventions. Advances in global transcriptomic technology over the past 15 years have enabled the assessment of microRNAs that circulate in the blood of patients and the analysis of the macromolecular content of a diverse group of lipid bilayer membrane-enclosed extracellular vesicles, such as exosomes and other microvesicles, which are released by all cells into the extracellular space and circulation. Such advances have provided new opportunities for the discovery of biomarkers in SSc that could potentially be used to improve the design and evaluation of clinical trials and that will undoubtedly enable the development of personalized and individualized medicine for patients with SSc.
Collapse
Affiliation(s)
- Peter J Wermuth
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA.,The Joan and Joel Rosenbloom Center for Fibrosis Research, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sonsoles Piera-Velazquez
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA.,The Joan and Joel Rosenbloom Center for Fibrosis Research, Thomas Jefferson University, Philadelphia, PA, USA
| | - Joel Rosenbloom
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA.,The Joan and Joel Rosenbloom Center for Fibrosis Research, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sergio A Jimenez
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA. .,The Joan and Joel Rosenbloom Center for Fibrosis Research, Thomas Jefferson University, Philadelphia, PA, USA. .,The Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
24
|
Serum YKL-40 and IL-6 levels correlate with ultrasound findings of articular and periarticular involvement in patients with systemic sclerosis. Rheumatol Int 2019; 39:1841-1848. [DOI: 10.1007/s00296-019-04402-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 07/26/2019] [Indexed: 12/16/2022]
|
25
|
Bufei Qingyu Granules Inhibit the Development of Systemic Sclerosis via Notch-1/Jagged-2 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:6709278. [PMID: 31354856 PMCID: PMC6633870 DOI: 10.1155/2019/6709278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 03/20/2019] [Accepted: 03/27/2019] [Indexed: 11/21/2022]
Abstract
Systemic sclerosis (SSc) is a rare chronic autoimmune disorder, mainly characterized by skin sclerosis. In this study, Bufei Qingyu Granules (BQG), a Chinese herbal formula, was used to treat SSc. To better understand the effects and molecular mechanisms of BQG, we successfully established a Bleomycin- (BLM-) induced SSc mouse model, and the mice were treated by BQG. Meanwhile, transcriptomic and bioinformatics analyses were conducted on those samples. As a result, we visually showed that BQG ameliorated the overall health of mice, including body weight, spleen, and thymus index. Thus, it also significantly alleviated inflammation presented by Chemokine (C-X-C motif) ligand 2 (Cxcl2), vasculopathy characterized by α-smooth muscle actin (α-SMA), and fibrotic changes elaborated by not only pathological images, but also the hydroxyproline (HYP) content. After testing by transcriptomic analysis, Cxcl2, Synaptosomal-associated protein 25 (Snap25), and Eukaryotic translation initiation factor 3, and subunit J2 (Eif3j2) which were differentially expressed genes, were verified, so that the data were credible. We further found that BQG could regulate Notch signaling pathway by significantly decreasing both mRNA and protein expression levels of Notch-1 and Jagged-2. Hence, this study demonstrated that BQG could ameliorate the sclerotic skin in mice model involved in inflammation, vascular changes, and fibrosis effects, which was partly mediated by Notch signaling pathway.
Collapse
|
26
|
Gonçalves RSG, Pereira MC, Dantas AT, Almeida ARD, Rego MJBM, Lima EA, Pitta IDR, Duarte ALBP, Pitta MGDR. CCL3, IL-7, IL-13 and IFNγ transcripts are increased in skin's biopsy of systemic sclerosis. Exp Dermatol 2019; 28:1172-1175. [PMID: 31162840 DOI: 10.1111/exd.13982] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 04/14/2019] [Accepted: 05/28/2019] [Indexed: 01/17/2023]
Abstract
Although several cytokines and chemokines have been investigated as possible mediators of fibrosis in systemic sclerosis (SSc), specific correlation between cytokines and organ involvement have not been found yet, and a cytokine profile characteristic of SSc is far to be identified. We studied the profile of antifibrotic and profibrotic transcripts involved in skin of SSc patients. The mRNA expression was detected by fluorescence-based quantitative real-time PCR (qPCR) in skin's biopsies from 14 patients with SSc and 5 healthy controls. PDGF-A, CTGF, CCL3, IL-6, IL-13, IL-7, IFNγ, IL-17, IL-22 and RORc were analysed in these samples. CCL3, IL-7, IL-13 and IFN-γ were more expressed in skin's biopsy of patients with SSc (P = 0.0002, P = 0.0082, P = 0.0243, P = 0.0335, respectively) when compared with healthy controls. We also found a positive correlation between CCL3 and IL-7 transcripts (P = 0.0050 r = 0.7187). Furthermore, we observed that patients with lung involvement had lower expression of PDGF-A (P = 0.0385). We found an increase in IL-7, IFN-γ, CCL3 and IL-13 relative mRNA expressions on the skin's biopsy of patients with SSc, and a positive correlation between IL-7 and CCL3. These molecules are involved in the pathogenesis of SSc, and how their interactions occur should be the subject of further studies.
Collapse
Affiliation(s)
- Rafaela Silva Guimarães Gonçalves
- Department of Rheumatology, Clinical Hospital of Federal University of Pernambuco, Recife, Brazil.,Laboratório de Imunomodulação e Novas Abordagens Terapêuticas, Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, Recife, Brazil
| | - Michelly Cristiny Pereira
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas, Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, Recife, Brazil
| | - Andréa Tavares Dantas
- Department of Rheumatology, Clinical Hospital of Federal University of Pernambuco, Recife, Brazil.,Laboratório de Imunomodulação e Novas Abordagens Terapêuticas, Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, Recife, Brazil
| | - Anderson Rodrigues de Almeida
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas, Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, Recife, Brazil
| | - Moacyr J B M Rego
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas, Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, Recife, Brazil
| | - Emerson Andrade Lima
- Department of Rheumatology, Clinical Hospital of Federal University of Pernambuco, Recife, Brazil
| | - Ivan da Rocha Pitta
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas, Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, Recife, Brazil
| | - Angela Luzia Branco Pinto Duarte
- Department of Rheumatology, Clinical Hospital of Federal University of Pernambuco, Recife, Brazil.,Laboratório de Imunomodulação e Novas Abordagens Terapêuticas, Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, Recife, Brazil
| | - Maira Galdino da Rocha Pitta
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas, Núcleo de Pesquisa em Inovação Terapêutica, Universidade Federal de Pernambuco, Recife, Brazil
| |
Collapse
|
27
|
Elhai M, Hoffmann‐Vold AM, Avouac J, Pezet S, Cauvet A, Leblond A, Fretheim H, Garen T, Kuwana M, Molberg Ø, Allanore Y. Performance of Candidate Serum Biomarkers for Systemic Sclerosis–Associated Interstitial Lung Disease. Arthritis Rheumatol 2019; 71:972-982. [DOI: 10.1002/art.40815] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 12/13/2018] [Indexed: 01/20/2023]
Affiliation(s)
- Muriel Elhai
- INSERM U1016, UMR8104, Cochin Institute, Paris Descartes University, and Cochin Hospital, Paris Descartes University Paris France
| | | | - Jérôme Avouac
- INSERM U1016, UMR8104, Cochin Institute, Paris Descartes University, and Cochin Hospital, Paris Descartes University Paris France
| | - Sonia Pezet
- INSERM U1016, UMR8104, Cochin Institute, Paris Descartes University Paris France
| | - Anne Cauvet
- INSERM U1016, UMR8104, Cochin Institute, Paris Descartes University Paris France
| | - Agathe Leblond
- INSERM U1016, UMR8104, Cochin Institute, Paris Descartes University Paris France
| | - Håvard Fretheim
- Oslo University Hospital, Oslo and Institute of Clinical MedicineUniversity of Oslo Oslo Norway
| | - Torhild Garen
- Oslo University Hospital, Oslo and Institute of Clinical MedicineUniversity of Oslo Oslo Norway
| | | | - Øyvind Molberg
- Oslo University Hospital, Oslo and Institute of Clinical MedicineUniversity of Oslo Oslo Norway
| | - Yannick Allanore
- INSERM U1016, UMR8104, Cochin Institute, Paris Descartes University, and Cochin Hospital, Paris Descartes University Paris France
| |
Collapse
|
28
|
Kim E, Lee HN, Kim YK, Kim GT, So MW, Ahn E, Sohn DH, Lee SG. Increased serum uric acid levels are associated with digital ulcers in patients with systemic sclerosis. Rheumatol Int 2019; 39:255-263. [DOI: 10.1007/s00296-019-04240-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 01/08/2019] [Indexed: 02/07/2023]
|
29
|
Jeon CH. Early Detection of Pulmonary Hypertension in Connective Tissue Disease. JOURNAL OF RHEUMATIC DISEASES 2019. [DOI: 10.4078/jrd.2019.26.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Chan Hong Jeon
- Division of Rheumatology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| |
Collapse
|
30
|
Fibroblast Growth Factor 23 to Alpha-Klotho Index Correlates with Systemic Sclerosis Activity: A Proposal for Novel Disease Activity Marker. J Clin Med 2018; 7:jcm7120558. [PMID: 30562918 PMCID: PMC6306722 DOI: 10.3390/jcm7120558] [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: 11/02/2018] [Revised: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 11/17/2022] Open
Abstract
Systemic sclerosis, a connective tissue disease, is characterized by thickening of the skin, massive fibrosis of internal organs, vasculopathy, and immune system functioning aberration. Recently, vitamin D (VD) deficit, seen almost universally in patients with systemic sclerosis (SSc), has gained much attention. VD metabolism is precisely orchestrated at the level of the kidney by regulators: parathyroid hormone (PTH) and fibroblast growth factor 23 (FGF23) and their receptors with a FGF23 co-receptor—α-Klotho. The aim of this study was to assess the levels of VD, α-Klotho, FGF23 in SSc patients and to find the relationship between those parameters and disease activity. We enrolled 48 SSc patients with a diffuse variant of SSc and 23 sex- and age-matched healthy volunteers that served as the control group (CG). Patients were characterized by lower level of VD in comparison to CG (19.8 (12.6–28.9) vs. 24.5 (21.3–31.5) ng/mL; p < 0.01), significantly reduced levels of iFGF23 (19.3 (12.1–30.5) vs. 73.9 (59.7–110.2) pg/mL p < 0.001), and similar α-Klotho concentrations (1415 ± 557 vs. 1526 ± 397 pg/mL), respective. None of these parameters correlated with the extent of skin involvement (modified Rodnan Skin Score) and disease activity according to Eustar 2017 guidelines. The FGF23/α-Klotho index was significantly reduced in SSc patients (0.013 (0.0081–0.025) vs. 0.055 (0.038–0.095); p < 0.001), and its log10 correlated (r = 0.35; p < 0.001) with disease activity score (Eular2017). Our data showed that the FGF23/α-Klotho index may be considered as a novel, potential marker of systemic sclerosis activity.
Collapse
|
31
|
Hoffmann-Vold AM, Hesselstrand R, Fretheim H, Ueland T, Andreassen AK, Brunborg C, Palchevskiy V, Midtvedt Ø, Garen T, Aukrust P, Belperio JA, Molberg Ø. CCL21 as a Potential Serum Biomarker for Pulmonary Arterial Hypertension in Systemic Sclerosis. Arthritis Rheumatol 2018; 70:1644-1653. [PMID: 29687634 DOI: 10.1002/art.40534] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 04/17/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Systemic sclerosis (SSc) is a major cause of pulmonary arterial hypertension (PAH). Murine models indicate key roles for chemokines CCL19 and CCL21 and their receptor CCR7 in lung inflammation leading to PAH. The objective of this study was to assess the chemokine CCL19-CCL21 axis in patients with SSc-related PAH. METHODS Serum samples obtained from 2 independent prospective SSc cohorts (n = 326), patients with idiopathic PAH (n = 12), and healthy control subjects (n = 100) were analyzed for CCL19/CCL21 levels, by enzyme-linked immunosorbent assay. The levels were defined as either high or low, using the mean + 2 SD value in controls as the cutoff value. Risk stratification at the time of PAH diagnosis and PAH-related events were performed. Descriptive and Cox regression analyses were conducted. RESULTS CCL21 levels were higher in patients with SSc compared with controls and were elevated prior to the diagnosis of PAH. PAH was more frequent in patients with high CCL21 levels (≥0.4 ng/ml) than in those with low CCL21 levels (33.3% versus 5.3% [P < 0.001]). In multivariate analyses, CCL21 was associated with PAH (hazard ratio [HR] 5.1, 95% CI 2.39-10.76 [P < 0.001]) and occurrence of PAH-related events (HR 4.7, 95% CI 2.12-10.46, P < 0.001). Risk stratification at the time of PAH diagnosis alone did not predict PAH-related events. However, when risk at diagnosis was combined with high or low CCL21 level, there was a significant predictive effect (HR 1.3, 95% CI 1.03-1.60 [P = 0.027]). A high CCL21 level was associated with decreased survival (P < 0.001). CONCLUSION CCL21 appears to be a promising marker for predicting the risk of SSc-related PAH and PAH progression. CCL21 may be part of a dysregulated immune pathway linked to the development of lung vascular damage in SSc.
Collapse
Affiliation(s)
| | | | - Håvard Fretheim
- Oslo University Hospital, Rikshospitalet, and University of Oslo, Oslo, Norway
| | - Thor Ueland
- Oslo University Hospital, Rikshospitalet, and University of Oslo, Oslo, Norway
| | | | | | | | | | - Torhild Garen
- Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Pål Aukrust
- Oslo University Hospital, Rikshospitalet, and University of Oslo, Oslo, Norway
| | - John A Belperio
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Øyvind Molberg
- Oslo University Hospital, Rikshospitalet, and University of Oslo, Oslo, Norway
| |
Collapse
|
32
|
Korman B, Alejo R, Sudhakar D, Hinchcliff M, Agrawal R, Varga J, Marangoni RG. The novel adipokine C1q-TNF related protein 9 (CTRP9) is elevated in systemic sclerosis-associated interstitial lung disease. Clin Exp Rheumatol 2018; 36 Suppl 113:184-185. [PMID: 30183591 PMCID: PMC7389309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/30/2018] [Indexed: 06/08/2023]
Affiliation(s)
- Benjamin Korman
- Northwestern Scleroderma Program, Departments of Medicine and Dermatology, Division of Rheumatology, Northwestern University, Chicago, IL, USA
| | - Rafael Alejo
- Northwestern Scleroderma Program, Departments of Medicine and Dermatology, Division of Rheumatology, Northwestern University, Chicago, IL, USA
| | - Deepshika Sudhakar
- Northwestern Scleroderma Program,Departments of Medicine and Dermatology, Division of Rheumatology, Northwestern University, Chicago, IL, USA
| | - Monique Hinchcliff
- Northwestern Scleroderma Program, Departments of Medicine and Dermatology, Division of Rheumatology, and Institute for Public Health and Medicine, Northwestern University, Chicago, IL, USA
| | - Rishi Agrawal
- Department of Radiology, Northwestern University, Chicago, IL, USA
| | - John Varga
- Northwestern Scleroderma Program, Departments of Medicine and Dermatology, Division of Rheumatology, Northwestern University, Chicago, IL, USA
| | - Roberta G Marangoni
- Northwestern Scleroderma Program, Departments of Medicine and Dermatology, Division of Rheumatology, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
33
|
Frech TM, Machin DR, Murtaugh MA, Stoddard GJ, Bloom SI, Phibbs JV, Donato AJ. Implications of endothelial shear stress on systemic sclerosis vasculopathy and treatment. Clin Exp Rheumatol 2018; 36 Suppl 113:175-182. [PMID: 30277867 PMCID: PMC6542469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 07/30/2018] [Indexed: 02/16/2023]
Abstract
There are no Federal Drug Administration approved drugs for the treatment of systemic sclerosis vascular digital ulcers (DU) in the United States, which are thought to be an end-stage result of prolonged ischaemia due to severe, prolonged Raynaud's phenomenon. Most therapeutics for vasodilation used in SSc work different pathways to target the smooth muscle to induce vessel relaxation. Longitudinal studies of vascular function allow insight into the effects of medications used for Raynaud's phenomenon in the SSc patient population. In this review, we discuss vascular tone, the function of the endothelium in SSc, and provide the rationale for longitudinal studies of vascular function and therapeutics that target the endothelial shear stress in addition to vasodilation for treatment and prevention of DU. This review provides the rationale for vasodilatory medication use for treatment of SSc-related DU and justifies access to non-FDA approved medications for this indication.
Collapse
Affiliation(s)
- Tracy M Frech
- University of Utah, Department of Internal Medicine; University of Utah Hospitals and Clinics; and VAMC Salt Lake City, GRECC, Salt Lake City, UT, USA.
| | - Daniel R Machin
- University of Utah, Department of Internal Medicine, Salt Lake City, UT, USA
| | - Maureen A Murtaugh
- University of Utah, Department of Internal Medicine, Salt Lake City, UT, USA
| | - Gregory J Stoddard
- University of Utah, Department of Internal Medicine, Salt Lake City, UT, USA
| | - Samuel I Bloom
- University of Utah, Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Jessica V Phibbs
- University of Utah Hospitals and Clinics, Salt Lake City, UT, USA
| | - Anthony J Donato
- University of Utah, Department of Internal Medicine; University of Utah, Department of Exercise and Sport Science; University of Utah, Dept. of Nutrition and Integrative Physiology, University of Utah; and VAMC Salt Lake City, GRECC, Salt Lake City, USA
| |
Collapse
|
34
|
Computational analysis identifies putative prognostic biomarkers of pathological scarring in skin wounds. J Transl Med 2018; 16:32. [PMID: 29458433 PMCID: PMC5819197 DOI: 10.1186/s12967-018-1406-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 02/12/2018] [Indexed: 01/06/2023] Open
Abstract
Background Pathological scarring in wounds is a prevalent clinical outcome with limited prognostic options. The objective of this study was to investigate whether cellular signaling proteins could be used as prognostic biomarkers of pathological scarring in traumatic skin wounds. Methods We used our previously developed and validated computational model of injury-initiated wound healing to simulate the time courses for platelets, 6 cell types, and 21 proteins involved in the inflammatory and proliferative phases of wound healing. Next, we analysed thousands of simulated wound-healing scenarios to identify those that resulted in pathological (i.e., excessive) scarring. Then, we identified candidate proteins that were elevated (or decreased) at the early stages of wound healing in those simulations and could therefore serve as predictive biomarkers of pathological scarring outcomes. Finally, we performed logistic regression analysis and calculated the area under the receiver operating characteristic curve to quantitatively assess the predictive accuracy of the model-identified putative biomarkers. Results We identified three proteins (interleukin-10, tissue inhibitor of matrix metalloproteinase-1, and fibronectin) whose levels were elevated in pathological scars as early as 2 weeks post-wounding and could predict a pathological scarring outcome occurring 40 days after wounding with 80% accuracy. Conclusion Our method for predicting putative prognostic wound-outcome biomarkers may serve as an effective means to guide the identification of proteins predictive of pathological scarring. Electronic supplementary material The online version of this article (10.1186/s12967-018-1406-x) contains supplementary material, which is available to authorized users.
Collapse
|
35
|
Landeck L, Kneip C, Reischl J, Asadullah K. Biomarkers and personalized medicine: current status and further perspectives with special focus on dermatology. Exp Dermatol 2018; 25:333-9. [PMID: 27167702 DOI: 10.1111/exd.12948] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2016] [Indexed: 01/02/2023]
Abstract
Biomarkers are of increasingly high importance in medicine, particularly in the realm of 'personalized medicine'. They are valuable for predicting prognosis and dose selection. Moreover, they may be helpful in detecting therapeutic and adverse responses and in patient stratification based on efficacy or safety prediction. Thus, biomarkers are essential tools for the selection of appropriate patients for treatment with certain drugs to and enable personalized medicine, that is 'providing the right treatment to the right patient, at the right dose at the right time'. Currently, there are six drugs approved for dermatological indications with recommended or mandatory biomarker testing. Most of them are used to treat melanoma and human immunodeficiency virus infection. In contrast to the few fully validated biomarkers, many exploratory biomarkers and biomarker candidates have potential applications. Prognostic biomarkers are of particular significance for malignant conditions. Similarly, diagnostic biomarkers are important in autoimmune diseases. Disease severity biomarkers are helpful tools in the treatment for inflammatory skin diseases. Identification, qualification and implementation of the different kinds of biomarkers are challenging and frequently necessitate collaborative efforts. This is particularly true for stratification biomarkers that require a companion diagnostic marker that is co-developed with a certain drug. In this article general definitions and requirements for biomarkers as well as for the impact of biomarkers in dermatology are reviewed and opportunities and challenges are discussed.
Collapse
Affiliation(s)
- Lilla Landeck
- Department of Dermatology, Ernst von Bergmann General Hospital Potsdam, Teaching Hospital of Charité, University Medicine Berlin, Berlin, Germany
| | | | - Joachim Reischl
- Bayer Global Drug Discovery, Berlin, Germany.,Astra Zeneca, Personalized Healthcare and Biomarkers, Gothenburg, Sweden
| | - Khusru Asadullah
- Bayer Global Drug Discovery, Berlin, Germany.,Charité, University Medicine Berlin, Berlin, Germany
| |
Collapse
|
36
|
Serum microRNA screening and functional studies reveal miR-483-5p as a potential driver of fibrosis in systemic sclerosis. J Autoimmun 2018; 89:162-170. [PMID: 29371048 DOI: 10.1016/j.jaut.2017.12.015] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 12/21/2017] [Accepted: 12/21/2017] [Indexed: 02/06/2023]
Abstract
OBJECTIVE MicroRNAs (miRNAs) are regulatory molecules, which have been addressed as potential biomarkers and therapeutic targets in rheumatic diseases. Here, we investigated the miRNA signature in the serum of systemic sclerosis (SSc) patients and we further assessed their expression in early stages of the disease. METHODS The levels of 758 miRNAs were evaluated in the serum of 26 SSc patients as compared to 9 healthy controls by using an Openarray platform. Three miRNAs were examined in an additional cohort of 107 SSc patients and 24 healthy donors by single qPCR. MiR-483-5p expression was further analysed in the serum of patients with localized scleroderma (LoS) (n = 22), systemic lupus erythematosus (SLE) (n = 33) and primary Sjögren's syndrome (pSS) (n = 23). The function of miR-483-5p was examined by transfecting miR-483-5p into primary human dermal fibroblasts and pulmonary endothelial cells. RESULTS 30 miRNAs were significantly increased in patients with SSc. Of these, miR-483-5p showed reproducibly higher levels in an independent SSc cohort and was also elevated in patients with preclinical-SSc symptoms (early SSc). Notably, miR-483-5p was not differentially expressed in patients with SLE or pSS, whereas it was up-regulated in LoS, indicating that this miRNA could be involved in the development of skin fibrosis. Consistently, miR-483-5p overexpression in fibroblasts and endothelial cells modulated the expression of fibrosis-related genes. CONCLUSIONS Our findings showed that miR-483-5p is up-regulated in the serum of SSc patients, from the early stages of the disease onwards, and indicated its potential function as a fine regulator of fibrosis in SSc.
Collapse
|
37
|
Hromádka M, Seidlerová J, Suchý D, Rajdl D, Lhotský J, Ludvík J, Rokyta R, Baxa J. Myocardial fibrosis detected by magnetic resonance in systemic sclerosis patients – Relationship with biochemical and echocardiography parameters. Int J Cardiol 2017; 249:448-453. [DOI: 10.1016/j.ijcard.2017.08.072] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 07/13/2017] [Accepted: 08/29/2017] [Indexed: 01/06/2023]
|
38
|
De Marco R, Greco A, Calonghi N, Dattoli SD, Baiula M, Spampinato S, Picchetti P, De Cola L, Anselmi M, Cipriani F, Gentilucci L. Selective detection of α4β1 integrin (VLA-4)-expressing cells using peptide-functionalized nanostructured materials mimicking endothelial surfaces adjacent to inflammatory sites. Biopolymers 2017; 110. [PMID: 29178262 DOI: 10.1002/bip.23081] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/06/2017] [Accepted: 10/18/2017] [Indexed: 01/01/2023]
Abstract
Persistent accumulation of immune cells mediated by α4β1 integrin (VLA-4) is a hallmark of the inflammatory diseases and of chronic inflammation observed in the affected tissues of autoimmune diseases. Aiming at exploring new methods for monitoring the course of the inflammatory processes, we designed the first peptide-functionalized nanostructured devices capable to mimic the high-density multivalency binding between the α4β1 integrin-expressing cells and the ligands overexpressed on the endothelial surfaces, in the proximity of the sites of inflammation. Specifically, we describe the first examples of monolayers constituted by dye-loaded zeolite L crystals, coated with α4β1 integrin peptide ligands, and we analyze the adhesion of model Jurkat cells in comparison to non-α4β1 integrin-expressing cells. In particular, the peptidomimetic diphenylurea-Leu-Asp-Val-diamine allows significant and selective detection of α4β1 integrin-expressing Jurkat cells, after very rapid incubation time, supporting the possible implementation in a diagnostic device capable to detect the desired cells from biological fluids, obtainable from patients in a noninvasive way.
Collapse
Affiliation(s)
- Rossella De Marco
- Department of Chemistry "G. Ciamician", University of Bologna, via Selmi 2, Bologna, 40126, Italy
| | - Arianna Greco
- Department of Chemistry "G. Ciamician", University of Bologna, via Selmi 2, Bologna, 40126, Italy
| | - Natalia Calonghi
- Department of Pharmacy and Biotechnology, University of Bologna, via Irnerio 48, Bologna, 40126, Italy
| | - Samantha D Dattoli
- Department of Pharmacy and Biotechnology, University of Bologna, via Irnerio 48, Bologna, 40126, Italy
| | - Monica Baiula
- Department of Pharmacy and Biotechnology, University of Bologna, via Irnerio 48, Bologna, 40126, Italy
| | - Santi Spampinato
- Department of Pharmacy and Biotechnology, University of Bologna, via Irnerio 48, Bologna, 40126, Italy
| | - Pierre Picchetti
- Institut de science et d'ingénierie supramoléculaires (ISIS), Université de Strasbourg and CNR UMR 7006, 8 Allée Gaspard Monge, Strasbourg, 67000, France
| | - Luisa De Cola
- Institut de science et d'ingénierie supramoléculaires (ISIS), Université de Strasbourg and CNR UMR 7006, 8 Allée Gaspard Monge, Strasbourg, 67000, France
- Institut fűr Nanotechnologie (INT), Karlsruhe Institute of Technology (KIT) - Campus Nord, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, 76344, Germany
| | - Michele Anselmi
- Department of Chemistry "G. Ciamician", University of Bologna, via Selmi 2, Bologna, 40126, Italy
| | - Francesca Cipriani
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 11, Bologna, 40138, Italy
| | - Luca Gentilucci
- Department of Chemistry "G. Ciamician", University of Bologna, via Selmi 2, Bologna, 40126, Italy
| |
Collapse
|
39
|
Expression of TLR7, TLR9, JAK2, and STAT3 genes in peripheral blood mononuclear cells from patients with systemic sclerosis. J Appl Genet 2017; 59:59-66. [DOI: 10.1007/s13353-017-0415-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 11/02/2017] [Accepted: 11/03/2017] [Indexed: 02/04/2023]
|
40
|
Wermuth PJ, Piera-Velazquez S, Jimenez SA. Identification of novel systemic sclerosis biomarkers employing aptamer proteomic analysis. Rheumatology (Oxford) 2017; 57:1698-1706. [DOI: 10.1093/rheumatology/kex404] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Indexed: 12/17/2022] Open
Affiliation(s)
- Peter J Wermuth
- Jefferson Institute of Molecular Medicine and The Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sonsoles Piera-Velazquez
- Jefferson Institute of Molecular Medicine and The Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sergio A Jimenez
- Jefferson Institute of Molecular Medicine and The Scleroderma Center, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
41
|
Affandi AJ, Carvalheiro T, Radstake TRDJ, Marut W. Dendritic cells in systemic sclerosis: Advances from human and mice studies. Immunol Lett 2017; 195:18-29. [PMID: 29126878 DOI: 10.1016/j.imlet.2017.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/05/2017] [Accepted: 11/06/2017] [Indexed: 12/20/2022]
Abstract
Systemic sclerosis (SSc) is a complex heterogeneous fibrotic autoimmune disease with an unknown exact etiology, and characterized by three hallmarks: fibrosis, vasculopathy, and immune dysfunction. Dendritic cells (DCs) are specialized cells in pathogen sensing with high potency of antigen presentation and capable of releasing mediators to shape the immune response. Altered DCs distributions and their impaired functions may account for their role in breaking the immune tolerance and driving inflammation in SSc, and the direct contribution of DCs in promoting endothelial dysfunction and fibrotic process has only begun to be understood. Plasmacytoid dendritic cells in particular have been implicated due to their high production of type I interferon as well as other cytokines and chemokines, including the pro-inflammatory and anti-angiogenic CXCL4. Furthermore, a deeper understanding of human and mouse DC biology has clarified their identification and function in different tissues, and novel DC subsets have only recently been discovered. In this review, we highlight key findings and recent advances exploring DC role in the pathogenesis of SSc and other related autoimmune diseases, and consideration of their potential use as targeted therapy in SSc.
Collapse
Affiliation(s)
- Alsya J Affandi
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Tiago Carvalheiro
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Timothy R D J Radstake
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Wioleta Marut
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands; Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
42
|
Molberg Ø, Hoffmann-Vold AM. Interstitial lung disease in systemic sclerosis: progress in screening and early diagnosis. Curr Opin Rheumatol 2017; 28:613-8. [PMID: 27387267 DOI: 10.1097/bor.0000000000000323] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Interstitial lung disease (ILD) is the major determinant of morbidity and mortality in systemic sclerosis (SSc). In highly selected SSc patients, it was recently shown that stem cell therapy early in the disease course improved survival and reduced the extent of ILD, providing a rationale for early ILD detection strategies in this disease. Here, we review recent progress on ILD screening and early diagnosis in SSc. RECENT FINDINGS Two studies showed that over 60% of unselected SSc cases with ILD by high-resolution computer tomography (HRCT) had normal range pulmonary function tests (PFTs); indicating poor performance of PFTs for ILD screening purposes. Serial, paired HRCT and PFT analyses indicated that screening by HRCT at baseline predicted risk for lung fibrosis development, progression rate of fibrosis and PFT decline. Analyses of circulating biomarkers, like CCL18, and nonradiating lung imaging modalities, like ultrasound and MRI, showed promise as tools for early ILD detection; but further work is needed. SUMMARY Prospective cohort data indicated poor performance of PFT as a stand-alone method for ILD screening. Lung HRCT appeared promising, but radiation is an issue. Promising biomarker data indicate the possibility of new ILD screening algorithms in SSc.
Collapse
Affiliation(s)
- Øyvind Molberg
- aDepartment of Rheumatology, Oslo University Hospital (OUH) bInstitute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | |
Collapse
|
43
|
Galectin-3 is an independent predictor of survival in systemic sclerosis. Int J Cardiol 2016; 233:118-124. [PMID: 28043664 DOI: 10.1016/j.ijcard.2016.12.140] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/17/2016] [Accepted: 12/19/2016] [Indexed: 11/23/2022]
Abstract
BACKGROUND Galectin-3 is a beta-galactoside-binding lectin that may be related to tissue sclerosis or aberrant activation of angiogenesis in systemic sclerosis (SSc). The aim of our study was to determine the associations between galectin-3 levels and patient characteristics, as well as to investigate the long term prognostic value of galectin-3 in a large cohort of SSc patients. METHODS 152 patients with SSc (55±11years, 138 female) were included in our follow-up study. Blood samples and clinical data were collected at baseline. Primary and secondary outcomes were all-cause and cardiovascular mortality, respectively. RESULTSS Galectin-3 levels showed positive correlation with the grade of left ventricular diastolic function (r=0.193; p=0.026), erythrocyte sedimentation rate (r=0.172; p=0.036) and serum level of C-reactive protein (r=0.200; p=0.015) while negative correlation with diffusing capacity for carbon monoxide (r=-0.228; p=0.006), in age, gender and BSA adjusted analyses. During the follow-up of 7.2±2.3years, 35 SSc patients (23%) died. In multivariate Cox regression analyses adjusted for age, gender, BSA, creatinine and NT-proBNP levels, galectin-3 was an independent predictor both of the all-cause mortality (HR: 2.780, 95% CI: 1.320-5.858, p=0.007) and cardiovascular mortality (HR: 3.346, 95% CI: 1.118-10.012, p=0.031). Using receiver-operating characteristic analysis, galectin-3>10.25ng/ml was found to be the best predictor of the all-cause mortality. CONCLUSIONS Our results suggest that galectin-3 is an independent predictor of all-cause and cardiovascular mortality in SSc. Validation studies are required to establish whether galectin-3 may be proposed as simple biomarker for identifying patients with high mortality risk in SSc.
Collapse
|
44
|
|
45
|
Chairta P, Nicolaou P, Christodoulou K. Genomic and genetic studies of systemic sclerosis: A systematic review. Hum Immunol 2016; 78:153-165. [PMID: 27984087 DOI: 10.1016/j.humimm.2016.10.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/27/2016] [Accepted: 10/27/2016] [Indexed: 12/12/2022]
Abstract
Systemic sclerosis is an autoimmune rheumatic disease characterised by fibrosis, vasculopathy and inflammation. The exact aetiology of SSc remains unknown but evidences show that various genetic factors may be involved. This review aimed to assess HLA alleles/non-HLA polymorphisms, microsatellites and chromosomal abnormalities that have thus far been associated with SSc. PubMed, Embase and Scopus databases were searched up to July 29, 2015 using a combination of search-terms. Articles retrieved were evaluated based on set exclusion and inclusion criteria. A total of 150 publications passed the filters. HLA and non-HLA studies showed that particular alleles in the HLA-DRB1, HLA-DQB1, HLA-DQA1, HLA-DPB1 genes and variants in STAT4, IRF5 and CD247 are frequently associated with SSc. Non-HLA genes analysis was performed using the PANTHER and STRING10 databases. PANTHER classification revealed that inflammation mediated by chemokine and cytokine, interleukin and integrin signalling pathways are among the common extracted pathways associated with SSc. STRING10 analysis showed that NFKB1, CSF3R, STAT4, IFNG, PRL and ILs are the main "hubs" of interaction network of the non-HLA genes associated with SSc. This study gathers data of valid genetic factors associated with SSc and discusses the possible interactions of implicated molecules.
Collapse
Affiliation(s)
- Paraskevi Chairta
- Neurogenetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus; Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus
| | - Paschalis Nicolaou
- Neurogenetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus; Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus
| | - Kyproula Christodoulou
- Neurogenetics Department, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus; Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia 2370, Cyprus.
| |
Collapse
|
46
|
Abstract
Systemic sclerosis is a heterogeneous condition characterized by microvascular damage, dysregulation of the immune system, and progressive fibrosis affecting skin and internal organs. Currently, there are no approved disease-modifying therapies, and management mostly involves treatment of organ-specific complications. In recent years, major advances have greatly improved our understanding of the disease process, especially the molecular mechanisms by which fibrosis becomes self-sustaining. We discuss selected aspects of these mechanisms with a focus on those relevant to ongoing efforts to develop disease-modifying therapies. We also discuss advances in identification of patient subtypes, and selected examples of potential disease-modifying therapies in clinical development.
Collapse
|
47
|
Del Papa N, Zaccara E. From mechanisms of action to therapeutic application: A review on current therapeutic approaches and future directions in systemic sclerosis. Best Pract Res Clin Rheumatol 2016; 29:756-69. [PMID: 27107511 DOI: 10.1016/j.berh.2016.02.004] [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: 12/02/2015] [Revised: 02/01/2016] [Indexed: 01/08/2023]
Abstract
Systemic sclerosis (SSc) is one of the most complex connective tissue diseases. Although the pathogenesis of SSc still remains elusive, it is generally accepted that initial vascular injury due to autoimmunity might result in the constitutive activation of fibroblasts and fibrosis. All of these three processes interact and affect one another resulting in a polymorphous spectrum of clinical and pathologic manifestations of SSc. The disease pleomorphism poses numerous difficulties in defining the ideal outcomes to be used in clinical trials. Despite significant progress over the past decades, the clinical management of patients with SSc remains a challenge. Novel therapies are currently being tested in the treatment of SSc and have the potential for modifying the disease process and improving the clinical outcomes. However, the evaluation of the studies is still difficult, due to either the small size of included patients or the different types and phases of the scleroderma disease under scrutiny.
Collapse
Affiliation(s)
- Nicoletta Del Papa
- U.O.C. Day Hospital Reumatologia, Dip. Reumatologia, Ospedale G. Pini, Milan, Italy.
| | - Eleonora Zaccara
- U.O.C. Day Hospital Reumatologia, Dip. Reumatologia, Ospedale G. Pini, Milan, Italy
| |
Collapse
|