1
|
Khanna D, Kramer F, Höfler J, Ghadessi M, Sandner P, Allanore Y, Denton CP, Kuwana M, Matucci-Cerinic M, Pope JE, Atsumi T, Bečvář R, Czirják L, De Langhe E, Hachulla E, Ishii T, Ishikawa O, Johnson SR, Riccieri V, Schiopu E, Silver RM, Smith V, Stagnaro C, Steen V, Stevens W, Szücs G, Truchetet ME, Wosnitza M, Distler O. Biomarker analysis from the phase 2b randomized placebo-controlled trial of riociguat in early diffuse cutaneous systemic sclerosis. Rheumatology (Oxford) 2024; 63:3124-3134. [PMID: 38460548 PMCID: PMC11534119 DOI: 10.1093/rheumatology/keae150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 01/11/2024] [Accepted: 02/07/2024] [Indexed: 03/11/2024] Open
Abstract
OBJECTIVE To examine disease and target engagement biomarkers in the RISE-SSc trial of riociguat in early diffuse cutaneous systemic sclerosis and their potential to predict the response to treatment. METHODS Patients were randomized to riociguat (n = 60) or placebo (n = 61) for 52 weeks. Skin biopsies and plasma/serum samples were obtained at baseline and week 14. Plasma cyclic guanosine monophosphate (cGMP) was assessed using radio-immunoassay. α-Smooth muscle actin (αSMA) and skin thickness were determined by immunohistochemistry, mRNA markers of fibrosis by qRT-PCR in skin biopsies, and serum CXC motif chemokine ligand 4 (CXCL-4) and soluble platelet endothelial cell adhesion molecule-1 (sPECAM-1) by enzyme-linked immunosorbent assay. RESULTS By week 14, cGMP increased by 94 (78)% with riociguat and 10 (39)% with placebo (P < 0.001, riociguat vs placebo). Serum sPECAM-1 and CXCL-4 decreased with riociguat vs placebo (P = 0.004 and P = 0.008, respectively). There were no differences in skin collagen markers between the two groups. Higher baseline serum sPECAM-1 or the detection of αSMA-positive cells in baseline skin biopsies was associated with a larger reduction of modified Rodnan skin score from baseline at week 52 with riociguat vs placebo (interaction P-values 0.004 and 0.02, respectively). CONCLUSION Plasma cGMP increased with riociguat, suggesting engagement with the nitric oxide-soluble guanylate cyclase-cGMP pathway. Riociguat was associated with a significant reduction in sPECAM-1 (an angiogenic biomarker) vs placebo. Elevated sPECAM-1 and the presence of αSMA-positive skin cells may help to identify patients who could benefit from riociguat in terms of skin fibrosis. TRIAL REGISTRATION Clinicaltrials.gov, NCT02283762.
Collapse
Affiliation(s)
- Dinesh Khanna
- Division of Rheumatology, University of Michigan, Ann Arbor, MI, USA
| | - Frank Kramer
- Research and Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany
| | | | - Mercedeh Ghadessi
- Research and Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany
| | - Peter Sandner
- Research and Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany
| | - Yannick Allanore
- Rheumatology A Department, Cochin Hospital, APAP, Paris Descartes University, Paris, France
| | - Christopher P Denton
- Division of Medicine, Centre for Rheumatology, University College London, London, UK
| | - Masataka Kuwana
- Department of Allergy and Rheumatology, Nippon Medical School Graduate School of Medicine, Tokyo, Japan
| | - Marco Matucci-Cerinic
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Firenze, Florence, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital, Milan, Italy
| | - Janet E Pope
- Division of Rheumatology, Schulich School of Medicine, University of Western Ontario, London, ON, Canada
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Radim Bečvář
- Institute of Rheumatology, Department of Rheumatology, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - László Czirják
- Department of Rheumatology and Immunology, Medical School, University of Pécs, Pécs, Hungary
| | - Ellen De Langhe
- Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, Division of Rheumatology, Department of Development and Regeneration, KU Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Eric Hachulla
- Department of Internal Medicine and Clinical Immunology, Referral Centre for Centre for Rare Systemic Autoimmune Diseases North and North-West of France, CHU Lille, University of Lille, Inserm, U1286 - INFINITE—Institute for Translational Research in Inflammation, Lille, France
| | - Tomonori Ishii
- Clinical Research, Innovation and Education Center, Tohoku University, Sendai, Japan
| | - Osamu Ishikawa
- Department of Dermatology, Gunma University Postgraduate School of Medicine, Maebashi, Japan
| | - Sindhu R Johnson
- Division of Rheumatology, Department of Medicine, Toronto Western Hospital, University Health Network, Mount Sinai Hospital, University of Toronto, Toronto Scleroderma Research Program, Toronto, ON, Canada
| | - Valeria Riccieri
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Elena Schiopu
- Division of Rheumatology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Richard M Silver
- Division of Rheumatology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Vanessa Smith
- Department of Internal Medicine, Ghent University, Belgium and Department of Rheumatology, Ghent University Hospital, Belgium, and Unit for Molecular Immunology and Inflammation, VIB Inflammation Research Center, Ghent, Belgium
| | - Chiara Stagnaro
- Rheumatology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Virginia Steen
- Division of Rheumatology, Department of Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Wendy Stevens
- Department of Rheumatology, St Vincent's Hospital Melbourne, Melbourne, VIC, Australia
| | - Gabriella Szücs
- Department of Rheumatology, University of Debrecen, Debrecen, Hungary
| | | | - Melanie Wosnitza
- Research and Development, Pharmaceuticals, Bayer AG, Wuppertal, Germany
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
2
|
Parker MJS, Jee AS, Hansen D, Proudman S, Youssef P, Kenna TJ, Stevens W, Nikpour M, Sahhar J, Corte TJ. Multiple serum biomarkers associate with mortality and interstitial lung disease progression in systemic sclerosis. Rheumatology (Oxford) 2024; 63:2981-2988. [PMID: 38366632 PMCID: PMC11534140 DOI: 10.1093/rheumatology/keae110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 12/12/2023] [Accepted: 12/14/2023] [Indexed: 02/18/2024] Open
Abstract
OBJECTIVES To investigate the prognostic utility of 28 serum biomarkers in systemic sclerosis (SSc), SSc-associated interstitial lung disease (SSc-ILD) and clinically relevant disease subgroups. METHODS Participants with sera, high-resolution CT and lung function within 12 months of baseline were identified from the Australian Scleroderma Cohort Study. Baseline was the time of serum collection. Twenty-seven of the prespecified 28 serum biomarkers were analysed and biomarker associations with mortality and ILD progression were investigated in univariable and multivariable analyses, including within disease subgroups and combined with established risk factors for poorer prognosis in SSc. RESULTS A total of 407 participants were identified, 252 (61.9%) with SSc-ILD. The median (interquartile range) follow-up after biomarker measurement was 6.31 (3.11-9.22) years. Sixteen biomarkers were associated with increased mortality. High levels of VCAM-1 were most strongly associated with mortality [hazard ratio (HR) 3.55; 95% CI 2.37-5.33; P < 0.001]. Five additional biomarkers had an HR >2: SP-D (2.28, 1.57-3.31; P < 0.001), E-selectin (2.19, 1.53-3.14; P < 0.001), IL-6 (2.15, 1.50-3.09; P < 0.001), MMP-3 (2.05, 1.42-2.95; P < 0.001) and ET-1 (2.03, 1.40-2.92; P < 0.001). Eleven biomarkers were independently associated with mortality following adjustment for sex, age and baseline forced vital capacity (FVC%predicted). Three biomarkers were associated with ILD progression at 1-year follow-up: CXCL4 (odds ratio 2.67, 1.46-4.88; P = 0.001), MMP-1 (2.56, 1.43-4.59; P = 0.002) and ET-1 (2.18, 1.24-3.83; P = 0.007). CONCLUSION Multiple biomarkers, especially VCAM-1, E-selectin, SP-D and CXCL4, provide prognostic utility beyond that of established risk factors for patients with SSc.
Collapse
Affiliation(s)
- Matthew James Sinclair Parker
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Rheumatology Department, RPA Institute for Academic Medicine, Royal Prince Alfred Hospital, Sydney, Australia
- NHMRC Centre of Research Excellence in Pulmonary Fibrosis, Sydney, Australia
| | - Adelle S Jee
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- NHMRC Centre of Research Excellence in Pulmonary Fibrosis, Sydney, Australia
- Department of Respiratory Medicine, Royal Prince Alfred Hospital, Sydney, Australia
| | - Dylan Hansen
- Department of Rheumatology, St Vincent’s Hospital, Melbourne, Australia
| | - Susanna Proudman
- Rheumatology Unit, Royal Adelaide Hospital, Adelaide, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, Australia
| | - Peter Youssef
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Rheumatology Department, RPA Institute for Academic Medicine, Royal Prince Alfred Hospital, Sydney, Australia
- Institute for Musculoskeletal Health, Camperdown, Sydney, Australia
| | - Tony J Kenna
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
| | - Wendy Stevens
- Department of Rheumatology, St Vincent’s Hospital, Melbourne, Australia
| | - Mandana Nikpour
- Department of Rheumatology, St Vincent’s Hospital, Melbourne, Australia
- Department of Medicine, The University of Melbourne, Melbourne, Australia
| | - Joanne Sahhar
- Department of Rheumatology, Monash Health, Melbourne, Australia
- Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - Tamera J Corte
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- NHMRC Centre of Research Excellence in Pulmonary Fibrosis, Sydney, Australia
- Department of Respiratory Medicine, Royal Prince Alfred Hospital, Sydney, Australia
| |
Collapse
|
3
|
Ivanova K, Zolovs M, Blennow K, Zetterberg H, Kurjāne N, Ķēniņa V. Polyneuropathy in systemic sclerosis: exploring the causes and biomarkers. Front Med (Lausanne) 2024; 11:1412706. [PMID: 39156689 PMCID: PMC11329926 DOI: 10.3389/fmed.2024.1412706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/25/2024] [Indexed: 08/20/2024] Open
Abstract
Introduction Systemic sclerosis (SSc) is a rare autoimmune disease with multiple organ involvement; however, the contribution of the nervous system (NS) remains relatively understudied. There are no specific data on the role of the autoimmune response and inflammation in the development of peripheral nerve system (PNS) damage in SSc and markers to assess this damage have yet to be identified. Objectives The primary objective of this study was to define the autoimmune mechanisms that lead to neuropathy by identifying antibodies (Abs) that target certain component of the NS or are associated with SSc. The secondary objective was to identify markers of NS damage that correlate with the detection and progression of polyneuropathy (PNP). Methods This study included patients diagnosed with SSc who met ACR/EULAR 2013 classification criteria at two leading Latvian hospitals between January 2016 and December 2021. Patients underwent a nerve conduction study (NCS). The SSc-associated Abs, Abs against myelin-associated glycoprotein (MAG) and anti-ganglioside Abs (GM1, GM2, GD1a, GD1b and GQ1b) were analysed. Potential serum PNS biomarkers-neurofilament light chain (NfL), glial fibrillary acidic protein (GFAP), fibroblast growth factor 21 (FGF21) and growth/differentiation factor 15 (GDF15)-were measured. Results We recruited 103 Caucasian patients diagnosed with SSc. SSc-associated Abs did not differ significantly between patients with and without PNP (p > 0.05). Anti-MAG and anti-ganglioside Abs in patients with PNP did not present a significant increase above the reference range. NfL, GFAP and GDF15 were significantly elevated in the presence of PNP (p < 0.05), with a moderate to high effect size (r = 0.36-0.65). Our regression analysis revealed a strong association between the HAQ-DI score, older age, male gender and the risk of developing PNP. Conclusion The development of PNP in patients with SSc is most likely due to ageing, natural progression and the sequelae of the disease. Several serum biomarkers-NfL, GFAP and GDF15-could be used as relevant diagnostic biomarkers for PNP in patients with SSc. Future studies are warranted to validate the diagnostic efficacy of these biomarkers and to unravel the complex interplay of factors leading to PNP in patients with SSc.
Collapse
Affiliation(s)
- Kristīne Ivanova
- Department of Doctoral Studies, Rīga Stradinš University, Rīga, Latvia
- Department of Rheumatology, Pauls Stradiņš Clinical University Hospital, Rīga, Latvia
| | - Maksims Zolovs
- Statistics Unit, Rīga Stradinš University, Rīga, Latvia
- Institute of Life Sciences and Technology, Daugavpils University, Daugavpils, Latvia
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Paris Brain Institute, ICM, Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
- Neurodegenerative Disorder Research Center, Division of Life Sciences and Medicine, and Department of Neurology, Institute on Aging and Brain Disorders, University of Science and Technology of China and First Affiliated Hospital of USTC, Hefei, China
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
- UK Dementia Research Institute at UCL, London, United Kingdom
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Kowloon, Hong Kong SAR, China
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin–Madison, Madison, WI, United States
| | - Nataļja Kurjāne
- Department of Biology and Microbiology, Rīga Stradinš University, Rīga, Latvia
- Institute of Oncology and Molecular Genetics, Rīga Stradinš University, Rīga, Latvia
- Outpatient Department, Pauls Stradiņš Clinical University Hospital, Rīga, Latvia
| | - Viktorija Ķēniņa
- Department of Biology and Microbiology, Rīga Stradinš University, Rīga, Latvia
- Institute of Oncology and Molecular Genetics, Rīga Stradinš University, Rīga, Latvia
- Department of Neurology, Pauls Stradiņš Clinical University Hospital, Rīga, Latvia
| |
Collapse
|
4
|
San Antonio E, Silván J, Sevilla-Montero J, González-Sánchez E, Muñoz-Callejas A, Sánchez-Abad I, Ramos-Manzano A, Muñoz-Calleja C, González-Álvaro I, Tomero EG, García-Pérez J, García-Vicuña R, Vicente-Rabaneda EF, Castañeda S, Urzainqui A. PSGL-1, ADAM8, and selectins as potential biomarkers in the diagnostic process of systemic lupus erythematosus and systemic sclerosis: an observational study. Front Immunol 2024; 15:1403104. [PMID: 39100683 PMCID: PMC11297358 DOI: 10.3389/fimmu.2024.1403104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/26/2024] [Indexed: 08/06/2024] Open
Abstract
Background Early diagnosis and treatment of Systemic lupus erythematosus (SLE) and Systemic sclerosis (SSc) present significant challenges for clinicians. Although various studies have observed changes in serum levels of selectins between healthy donors and patients with autoimmune diseases, including SLE and SSc, their potential as biomarkers has not been thoroughly explored. We aimed to investigate serum profiles of PSGL-1 (sPSGL-1), ADAM8 (sADAM8) and P-, E- and L-selectins (sP-, sE- and sL-selectins) in defined SLE and SSc patient cohorts to identify disease-associated molecular patterns. Methods We collected blood samples from 64 SLE patients, 58 SSc patients, and 81 healthy donors (HD). Levels of sPSGL-1, sADAM8 and selectins were analyzed by ELISA and leukocyte membrane expression of L-selectin and ADAM8 by flow cytometry. Results Compared to HD, SLE and SSc patients exhibited elevated sE-selectin and reduced sL-selectin levels. Additionally, SLE patients exhibited elevated sPSGL-1 and sADAM8 levels. Compared to SSc, SLE patients had decreased sL-selectin and increased sADAM8 levels. Furthermore, L-selectin membrane expression was lower in SLE and SSc leukocytes than in HD leukocytes, and ADAM8 membrane expression was lower in SLE neutrophils compared to SSc neutrophils. These alterations associated with some clinical characteristics of each disease. Using logistic regression analysis, the sL-selectin/sADAM8 ratio in SLE, and a combination of sL-selectin/sE-selectin and sE-selectin/sPSGL-1 ratios in SSc were identified and cross-validated as potential serum markers to discriminate these patients from HD. Compared to available diagnostic biomarkers for each disease, both sL-selectin/sADAM8 ratio for SLE and combined ratios for SSc provided higher sensitivity (98% SLE and and 67% SSc correctly classified patients). Importantly, the sADAM8/% ADAM8(+) neutrophils ratio discriminated between SSc and SLE patients with the same sensitivity and specificity than current disease-specific biomarkers. Conclusion SLE and SSc present specific profiles of sPSGL-1, sE-, sL-selectins, sADAM8 and neutrophil membrane expression which are potentially relevant to their pathogenesis and might aid in their early diagnosis.
Collapse
Affiliation(s)
- Esther San Antonio
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Javier Silván
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Javier Sevilla-Montero
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Elena González-Sánchez
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Antonio Muñoz-Callejas
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
- Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, Madrid, Spain
| | - Inés Sánchez-Abad
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Alejandra Ramos-Manzano
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Autónoma of Madrid, Madrid, Spain
| | - Cecilia Muñoz-Calleja
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Autónoma of Madrid, Madrid, Spain
| | - Isidoro González-Álvaro
- Rheumatology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Eva G. Tomero
- Rheumatology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Javier García-Pérez
- Pulmonology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Rosario García-Vicuña
- Medicine Department, School of Medicine, Universidad Autónoma of Madrid, Madrid, Spain
- Rheumatology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Esther F. Vicente-Rabaneda
- Medicine Department, School of Medicine, Universidad Autónoma of Madrid, Madrid, Spain
- Rheumatology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Santos Castañeda
- Rheumatology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| | - Ana Urzainqui
- Immunology Department, Fundacion para la Investigacion Biomedica (FIB)-Hospital Universitario de La Princesa, Instituto de Investigacion Sanitaria (IIS)-Princesa, Madrid, Spain
| |
Collapse
|
5
|
Apostolo D, D’Onghia D, Nerviani A, Ghirardi GM, Sola D, Perazzi M, Tonello S, Colangelo D, Sainaghi PP, Bellan M. Could Gas6/TAM Axis Provide Valuable Insights into the Pathogenesis of Systemic Sclerosis? Curr Issues Mol Biol 2024; 46:7486-7504. [PMID: 39057085 PMCID: PMC11275301 DOI: 10.3390/cimb46070444] [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: 06/19/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disorder characterized by microvascular injury, extracellular matrix deposition, autoimmunity, inflammation, and fibrosis. The clinical complexity and high heterogeneity of the disease make the discovery of potential therapeutic targets difficult. However, the recent progress in the comprehension of its pathogenesis is encouraging. Growth Arrest-Specific 6 (Gas6) and Tyro3, Axl, and MerTK (TAM) receptors are involved in multiple biological processes, including modulation of the immune response, phagocytosis, apoptosis, fibrosis, inflammation, cancer development, and autoimmune disorders. In the present manuscript, we review the current evidence regarding SSc pathogenesis and the role of the Gas6/TAM system in several human diseases, suggesting its likely contribution in SSc and highlighting areas where further research is necessary to fully comprehend the role of TAM receptors in this condition. Indeed, understanding the involvement of TAM receptors in SSc, which is currently unknown, could provide valuable insights for novel potential therapeutic targets.
Collapse
Affiliation(s)
- Daria Apostolo
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (D.D.); (D.S.); (M.P.); (S.T.); (P.P.S.); (M.B.)
- Centre for Experimental Medicine and Rheumatology, Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK;
| | - Davide D’Onghia
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (D.D.); (D.S.); (M.P.); (S.T.); (P.P.S.); (M.B.)
| | - Alessandra Nerviani
- Centre for Experimental Medicine and Rheumatology, Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK;
| | - Giulia Maria Ghirardi
- Centre for Experimental Medicine and Rheumatology, Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK;
| | - Daniele Sola
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (D.D.); (D.S.); (M.P.); (S.T.); (P.P.S.); (M.B.)
- IRCCS Istituto Auxologico Italiano, UO General Medicine, 28824 Oggebbio, Italy
| | - Mattia Perazzi
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (D.D.); (D.S.); (M.P.); (S.T.); (P.P.S.); (M.B.)
- Internal Medicine and Rheumatology Unit, A.O.U. Maggiore della Carità, 28100 Novara, Italy
| | - Stelvio Tonello
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (D.D.); (D.S.); (M.P.); (S.T.); (P.P.S.); (M.B.)
| | - Donato Colangelo
- Department of Health Sciences, Pharmacology, University of Piemonte Orientale (UPO), 28100 Novara, Italy;
| | - Pier Paolo Sainaghi
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (D.D.); (D.S.); (M.P.); (S.T.); (P.P.S.); (M.B.)
- Internal Medicine and Rheumatology Unit, A.O.U. Maggiore della Carità, 28100 Novara, Italy
- Center on Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Mattia Bellan
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (D.D.); (D.S.); (M.P.); (S.T.); (P.P.S.); (M.B.)
- Internal Medicine and Rheumatology Unit, A.O.U. Maggiore della Carità, 28100 Novara, Italy
- Center on Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
6
|
Ferrigno I, Verzellesi L, Ottone M, Bonacini M, Rossi A, Besutti G, Bonelli E, Colla R, Facciolongo N, Teopompi E, Massari M, Mancuso P, Ferrari AM, Pattacini P, Trojani V, Bertolini M, Botti A, Zerbini A, Giorgi Rossi P, Iori M, Salvarani C, Croci S. CCL18, CHI3L1, ANG2, IL-6 systemic levels are associated with the extent of lung damage and radiomic features in SARS-CoV-2 infection. Inflamm Res 2024:10.1007/s00011-024-01852-1. [PMID: 38308760 DOI: 10.1007/s00011-024-01852-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/17/2024] [Accepted: 01/21/2024] [Indexed: 02/05/2024] Open
Abstract
OBJECTIVE AND DESIGN We aimed to identify cytokines whose concentrations are related to lung damage, radiomic features, and clinical outcomes in COVID-19 patients. MATERIAL OR SUBJECTS Two hundred twenty-six patients with SARS-CoV-2 infection and chest computed tomography (CT) images were enrolled. METHODS CCL18, CHI3L1/YKL-40, GAL3, ANG2, IP-10, IL-10, TNFα, IL-6, soluble gp130, soluble IL-6R were quantified in plasma samples using Luminex assays. The Mann-Whitney U test, the Kruskal-Wallis test, correlation and regression analyses were performed. Mediation analyses were used to investigate the possible causal relationships between cytokines, lung damage, and outcomes. AVIEW lung cancer screening software, pyradiomics, and XGBoost classifier were used for radiomic feature analyses. RESULTS CCL18, CHI3L1, and ANG2 systemic levels mainly reflected the extent of lung injury. Increased levels of every cytokine, but particularly of IL-6, were associated with the three outcomes: hospitalization, mechanical ventilation, and death. Soluble IL-6R showed a slight protective effect on death. The effect of age on COVID-19 outcomes was partially mediated by cytokine levels, while CT scores considerably mediated the effect of cytokine levels on outcomes. Radiomic-feature-based models confirmed the association between lung imaging characteristics and CCL18 and CHI3L1. CONCLUSION Data suggest a causal link between cytokines (risk factor), lung damage (mediator), and COVID-19 outcomes.
Collapse
Affiliation(s)
- Ilaria Ferrigno
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Laura Verzellesi
- Unit of Medical Physics, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Marta Ottone
- Unit of Epidemiology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Martina Bonacini
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessandro Rossi
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Giulia Besutti
- Unit of Radiology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Department of Surgery, Medicine, Dentistry and Morphological Sciences With Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Efrem Bonelli
- Unit of Radiology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
- Clinical Chemistry and Endocrinology Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Rossana Colla
- Clinical Chemistry and Endocrinology Laboratory, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Nicola Facciolongo
- Unit of Respiratory Diseases, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Elisabetta Teopompi
- Multidisciplinary Internal Medicine Unit, Guastalla Hospital, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Marco Massari
- Unit of Infectious Diseases, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Pamela Mancuso
- Unit of Epidemiology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Anna Maria Ferrari
- Department of Emergency, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Pierpaolo Pattacini
- Unit of Radiology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Valeria Trojani
- Unit of Medical Physics, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Marco Bertolini
- Unit of Medical Physics, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Andrea Botti
- Unit of Medical Physics, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Alessandro Zerbini
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Paolo Giorgi Rossi
- Unit of Epidemiology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Mauro Iori
- Unit of Medical Physics, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Carlo Salvarani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences With Interest in Transplant, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Rheumatology, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Stefania Croci
- Unit of Clinical Immunology, Allergy and Advanced Biotechnologies, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| |
Collapse
|
7
|
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
|
8
|
Di Maggio G, Confalonieri P, Salton F, Trotta L, Ruggero L, Kodric M, Geri P, Hughes M, Bellan M, Gilio M, Lerda S, Baratella E, Confalonieri M, Mondini L, Ruaro B. Biomarkers in Systemic Sclerosis: An Overview. Curr Issues Mol Biol 2023; 45:7775-7802. [PMID: 37886934 PMCID: PMC10604992 DOI: 10.3390/cimb45100490] [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: 09/02/2023] [Revised: 09/19/2023] [Accepted: 09/22/2023] [Indexed: 10/28/2023] Open
Abstract
Systemic sclerosis (SSc) is a complex autoimmune disease characterized by significant fibrosis of the skin and internal organs, with the main involvement of the lungs, kidneys, heart, esophagus, and intestines. SSc is also characterized by macro- and microvascular damage with reduced peripheral blood perfusion. Several studies have reported more than 240 pathways and numerous dysregulation proteins, giving insight into how the field of biomarkers in SSc is still extremely complex and evolving. Antinuclear antibodies (ANA) are present in more than 90% of SSc patients, and anti-centromere and anti-topoisomerase I antibodies are considered classic biomarkers with precise clinical features. Recent studies have reported that trans-forming growth factor β (TGF-β) plays a central role in the fibrotic process. In addition, interferon regulatory factor 5 (IRF5), interleukin receptor-associated kinase-1 (IRAK-1), connective tissue growth factor (CTGF), transducer and activator of transcription signal 4 (STAT4), pyrin-containing domain 1 (NLRP1), as well as genetic factors, including DRB1 alleles, are implicated in SSc damage. Several interleukins (e.g., IL-1, IL-6, IL-10, IL-17, IL-22, and IL-35) and chemokines (e.g., CCL 2, 5, 23, and CXC 9, 10, 16) are elevated in SSc. While adiponectin and maresin 1 are reduced in patients with SSc, biomarkers are important in research but will be increasingly so in the diagnosis and therapeutic approach to SSc. This review aims to present and highlight the various biomarker molecules, pathways, and receptors involved in the pathology of SSc.
Collapse
Affiliation(s)
- Giuseppe Di Maggio
- Pulmonology Unit, Department of Medical Surgical and Healt Sciencies, Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy; (G.D.M.); (M.K.); (P.G.); (L.M.)
| | - Paola Confalonieri
- Pulmonology Unit, Department of Medical Surgical and Healt Sciencies, Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy; (G.D.M.); (M.K.); (P.G.); (L.M.)
| | - Francesco Salton
- Pulmonology Unit, Department of Medical Surgical and Healt Sciencies, Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy; (G.D.M.); (M.K.); (P.G.); (L.M.)
| | - Liliana Trotta
- Pulmonology Unit, Department of Medical Surgical and Healt Sciencies, Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy; (G.D.M.); (M.K.); (P.G.); (L.M.)
| | - Luca Ruggero
- Pulmonology Unit, Department of Medical Surgical and Healt Sciencies, Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy; (G.D.M.); (M.K.); (P.G.); (L.M.)
| | - Metka Kodric
- Pulmonology Unit, Department of Medical Surgical and Healt Sciencies, Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy; (G.D.M.); (M.K.); (P.G.); (L.M.)
| | - Pietro Geri
- Pulmonology Unit, Department of Medical Surgical and Healt Sciencies, Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy; (G.D.M.); (M.K.); (P.G.); (L.M.)
| | - Michael Hughes
- Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester & Salford Royal NHS Foundation Trust, Manchester M6 8HD, UK;
| | - Mattia Bellan
- Department of Translational Medicine, Università del Piemonte Orientale (UPO), 28100 Novara, Italy
- Center for Autoimmune and Allergic Disease (CAAD), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
- Department of Medicine, Azienda Ospedaliero–Universitaria, Maggiore della Carità, 28100 Novara, Italy
| | - Michele Gilio
- Infectious Disease Unit, San Carlo Hospital, 85100 Potenza, Italy
| | - Selene Lerda
- Graduate School, University of Milan, 20149 Milano, Italy
| | - Elisa Baratella
- Department of Radiology, Cattinara Hospital, University of Trieste, 34149 Trieste, Italy
| | - Marco Confalonieri
- Pulmonology Unit, Department of Medical Surgical and Healt Sciencies, Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy; (G.D.M.); (M.K.); (P.G.); (L.M.)
| | - Lucrezia Mondini
- Pulmonology Unit, Department of Medical Surgical and Healt Sciencies, Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy; (G.D.M.); (M.K.); (P.G.); (L.M.)
| | - Barbara Ruaro
- Pulmonology Unit, Department of Medical Surgical and Healt Sciencies, Hospital of Cattinara, University of Trieste, 34149 Trieste, Italy; (G.D.M.); (M.K.); (P.G.); (L.M.)
| |
Collapse
|
9
|
Fioretto BS, Rosa I, Matucci-Cerinic M, Romano E, Manetti M. Current Trends in Vascular Biomarkers for Systemic Sclerosis: A Narrative Review. Int J Mol Sci 2023; 24:ijms24044097. [PMID: 36835506 PMCID: PMC9965592 DOI: 10.3390/ijms24044097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Systemic sclerosis (SSc, scleroderma) is a multifaceted rare connective tissue disease whose pathogenesis is dominated by immune dysregulation, small vessel vasculopathy, impaired angiogenesis, and both cutaneous and visceral fibrosis. Microvascular impairment represents the initial event of the disease, preceding fibrosis by months or years and accounting for the main disabling and/or life-threatening clinical manifestations, including telangiectasias, pitting scars, periungual microvascular abnormalities (e.g., giant capillaries, hemorrhages, avascular areas, ramified/bushy capillaries) clinically detectable by nailfold videocapillaroscopy, ischemic digital ulcers, pulmonary arterial hypertension, and scleroderma renal crisis. Despite a variety of available treatment options, treatment of SSc-related vascular disease remains problematic, even considering SSc etherogenity and the quite narrow therapeutic window. In this context, plenty of studies have highlighted the great usefulness in clinical practice of vascular biomarkers allowing clinicians to assess the evolution of the pathological process affecting the vessels, as well as to predict the prognosis and the response to therapy. The current narrative review provides an up-to-date overview of the main candidate vascular biomarkers that have been proposed for SSc, focusing on their main reported associations with characteristic clinical vascular features of the disease.
Collapse
Affiliation(s)
- Bianca Saveria Fioretto
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Irene Rosa
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Marco Matucci-Cerinic
- Section of Internal Medicine, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Hospital, 20132 Milan, Italy
| | - Eloisa Romano
- Section of Internal Medicine, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Mirko Manetti
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Imaging Platform, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
- Correspondence:
| |
Collapse
|
10
|
Fernández-Lázaro D, Iglesias-Lázaro M, Garrosa E, Rodríguez-García S, Jerves Donoso D, Gutiérrez-Abejón E, Jorge-Finnigan C. Impact of Innovative Treatment Using Biological Drugs for the Modulation of Diffuse Cutaneous Systemic Sclerosis: A Systematic Review. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59020247. [PMID: 36837449 PMCID: PMC9967997 DOI: 10.3390/medicina59020247] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023]
Abstract
Scleroderma or systemic sclerosis (SSc) is an autoimmune disease affecting the connective tissue, characterized by fibrosis of the skin and internal organs. There is currently no curative treatment available, so therapeutic action is aimed at a symptomatic treatment of the affected organs. The development of biotechnology has made it possible to implement certain biological drugs that could represent a window of opportunity to modulate the evolution and symptomatology of scleroderma with greater efficacy and less toxicity than conventional treatments. This study aimed to review the current evidence critically and systematically on the effects of biological drugs on the pulmonary function, skin disease, and health status of patients afflicted by diffuse cutaneous systemic sclerosis (dcSSc). Three electronic databases (Pubmed, Dialnet, and Cochrane Library Plus) were systematically searched until the cut-off date of October 2022. The review was conducted following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and included original articles in English and Spanish with a controlled trial design, comparing biological drug treatments (tocilizumab, belimumab, riociguat, abatacept, and romilkimab) with a control group. The methodological quality of the studies was assessed using the McMaster quantitative form and the PEDro scale. A total of 383 studies were identified, 6 of them met the established criteria and were included in the present systematic review. A total of 426 patients treated with tocilizumab, belimumab, riociguat, abatacept, and romilkimab were included. The results showed substantial non-significant (p < 0.05) improvement trends after treatment with the biological drugs included in this review for the modified Rodnan Scale Value, Forced Vital Capacity, and Carbon Monoxide Diffusion Test; however, no benefits were shown on the Health Assessment Questionnaire-Disability Index when compared to the control group. Biological drugs, therefore, maybe a new therapeutic strategy for dcSSc and could be recommended as an additional and/or adjunctive treatment that promotes anti-fibrotic activity. This review could further define the clinical rationale for the use of biologics in the treatment of dcSSc and could provide key details on the study protocol, design, and outcome reporting.
Collapse
Affiliation(s)
- Diego Fernández-Lázaro
- Department of Cellular Biology, Genetics, Histology and Pharmacology, Faculty of Health Sciences, University of Valladolid, Campus of Soria, 42004 Soria, Spain
- Neurobiology Research Group, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain
- Correspondence: (D.F.-L.); (E.G.)
| | - María Iglesias-Lázaro
- Department of Cellular Biology, Genetics, Histology and Pharmacology, Faculty of Health Sciences, University of Valladolid, Campus of Soria, 42004 Soria, Spain
| | - Evelina Garrosa
- Neurobiology Research Group, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain
- Correspondence: (D.F.-L.); (E.G.)
| | - Saray Rodríguez-García
- Department of Medicine, Faculty of Health Sciences, University of Valladolid, Campus of Soria, 42003 Soria, Spain
- Internal Medicine Department of Soria University Assistance Complex (CAUSO), Santa Bárbara Hospital, Castile and Leon Health (SACyL), 42005 Soria, Spain
| | - David Jerves Donoso
- Pneumology Department of Soria University Assistance Complex (CAUSO), Santa Bárbara Hospital, Castile and Leon Health (SACyL), 42003 Soria, Spain
- Department of Anatomy and Radiology, Faculty of Health Sciences, University of Valladolid, Campus of Soria, 42004 Soria, Spain
| | - Eduardo Gutiérrez-Abejón
- Pharmacological Big Data Laboratory, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain
- Pharmacy Directorate, Castile and Leon Health Council, 47007 Valladolid, Spain
| | - Conrado Jorge-Finnigan
- Department of Anatomy and Radiology, Faculty of Health Sciences, University of Valladolid, Campus of Soria, 42004 Soria, Spain
- Dermatology Department of Soria University Assistance Complex (CAUSO), Santa Bárbara Hospital, Castile and Leon Health (SACyL), 42005 Soria, Spain
| |
Collapse
|
11
|
Stec A, Maciejewska M, Paralusz-Stec K, Michalska M, Giebułtowicz J, Rudnicka L, Sikora M. The Gut Microbial Metabolite Trimethylamine N-Oxide is Linked to Specific Complications of Systemic Sclerosis. J Inflamm Res 2023; 16:1895-1904. [PMID: 37152867 PMCID: PMC10162098 DOI: 10.2147/jir.s409489] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/27/2023] [Indexed: 05/09/2023] Open
Abstract
Background Systemic sclerosis (SSc) is a rare immune-mediated connective tissue disease characterized by fibrosis of the skin and internal organs, whose pathogenesis is not fully understood. Recent studies have revealed dysbiosis in patients with systemic sclerosis and have indicated the possible role of the microbiota and its metabolites in the pathogenesis of the disease. Trimethylamine N-oxide (TMAO) is a compound produced by dysbiotic microbiota observed at higher concentrations in several autoimmune diseases. Objective To determine concentrations of the bacteria-derived metabolite TMAO in patients with systemic sclerosis and to assess possible correlation between TMAO and a specific manifestation of the disease. Patients and Methods The study included 63 patients with SSc and 47 matched control subjects. The concentration of TMAO was measured with high-performance liquid chromatography. Results Plasma TMAO level was significantly increased in patients with SSc (283.0 [188.5-367.5] ng/mL versus 205.5 [101.0-318.0] ng/mL; p < 0.01). An increased concentration of TMAO was observed in patients with concomitant interstitial lung disease (ILD) (302.0 ng/mL [212.0-385.5] ng/mL versus 204.0 [135.5-292.0] ng/mL; p < 0.01) and esophageal dysmotility (289.75 [213.75-387.5] ng/mL versus 209.5 ng/mL [141.5-315.0] ng/mL; p < 0.05) compared to patients without these complications. Furthermore, TMAO concentration exhibited significant correlation with markers of heart involvement (left ventricle ejection fraction, NT-proBNP), marker of ILD severity and Scleroderma Clinical Trials Consortium Damage Index. Conclusion The concentration of TMAO, gut microbiota-associated metabolite, is increased in systemic sclerosis, particularly in patients with advanced organ involvement. This is the first study evaluating plasma TMAO in systemic sclerosis. Bacterial metabolites may be a link between dysbiosis and organ involvement in the course of the disease. Modulation of gut bacterial-derived metabolites may represent a new therapeutic approach in the management of systemic sclerosis.
Collapse
Affiliation(s)
- Albert Stec
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | | | | | - Milena Michalska
- Department of General, Vascular and Transplant Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Joanna Giebułtowicz
- Department of Bioanalysis and Drugs Analysis, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | - Mariusz Sikora
- National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
- Correspondence: Mariusz Sikora, National Institute of Geriatrics, Rheumatology and Rehabilitation, Spartańska 1, Warsaw, 02-637, Poland, Tel +48 22 670 91 00, Fax +48 22 844 77 97, Email
| |
Collapse
|
12
|
ABO Blood Groups in Systemic Sclerosis: Distribution and Association with This Disease's Characteristics. J Clin Med 2022; 12:jcm12010148. [PMID: 36614946 PMCID: PMC9821217 DOI: 10.3390/jcm12010148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease associated with endothelial activation and fibrosis. Non-O blood group patients carry an increased risk of thrombosis, fibrosis and autoimmune diseases. The aim of our work was to evaluate the distribution of ABO groups in SSc patients and their association with the disease's characteristics. ABO groups were determined in 504 SSc patients (with 131 completed by a genotypic analysis). The distribution of ABO groups and their diplotypes in SSc patients was comparable to that of the general population, except for haplotypes O1 and B (65.6% vs. 61.6% and 8.8% vs. 5.8% in SSc patients vs. the general population, respectively, p = 0.01). The frequency of interstitial lung disease, pulmonary hypertension, calcinosis, digital ulcers, digestive diseases and venous thrombosis, and the Medsger score, were higher in non-O than in O-SSc patients, although they did not display statistical significance. Patients in the non-O group had higher levels of inflammation and endothelial activation biomarkers. In conclusion, the ABO blood group distribution of SSc patients did not differ significantly from that of the general population, but non-O blood groups were associated with inflammation and endothelial activation, and with a non-significant higher frequency of pulmonary and vascular complications in SSc.
Collapse
|
13
|
Li K, Wang Q, Lv Q, Guo K, Han L, Duan P, Deng Y, Bian H. Wenyang Huazhuo Tongluo formula alleviates pulmonary vascular injury and downregulates HIF-1α in bleomycin-induced systemic sclerosis mouse model. BMC Complement Med Ther 2022; 22:167. [PMID: 35733188 PMCID: PMC9215020 DOI: 10.1186/s12906-022-03651-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 06/08/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Vascular damage, autoimmune abnormalities, and fibrosis are the three pathological features of systemic sclerosis (SSc).However, pulmonary vascular damage is the main factor affecting the progression and prognosis of SSc. The main purpose of this study was to explore the molecular mechanism of Wenyang Huazhuo Tongluo Formula in alleviating pulmonary vascular injury in bleomycin-induced SSc mouse model.
Methods
Masson staining and H&E staining were used to analyze the degree of pulmonary vascular fibrosis and the infiltration of leukocyte cells in lung tissue ofbleomycin-induced SSc mouse models treated with saline (BLM group), Wenyang Huazhuo Tongluo Formula (WYHZTL group) and HIF-1α inhibitor KC7F2 (KC7F2 group). Blood vessel exudation was determined by analyzing the cell number and albumin concentration in bronchoalveolar lavage fluid using a cell counter and ELISA assay, respectively. The degree of vascular injury was assessed by measuring the expression levels of vWF, E-selectin, ICAM-1, VCAM-1, VE-cadherin and claudin-5 in serum and pulmonary vascular endothelial cells using ELISA and immunofluorescence staining. Finally, the effect of Wenyang Huazhuo Tongluo Formula on the expression of HIF-1α was detected using immunofluorescence staining.
Results
Wenyang Huazhuo Tongluo Formula and KC7F2 significantly inhibited bleomycin-induced pulmonary vascular fibrosis and the level of perivascular inflammatory cell infiltration. The number of cells and the concentration of albumin were significantly reduced in the bronchoalveolar lavage fluid of the WYHZTL group and KC7F2 group compared with the BLM group. In addition, treatment with Wenyang Huazhuo Tongluo Formula and KC7F2 significantly downregulated the expression levels of vWF, E-selectin, ICAM-1, VCAM-1 and HIF-1α, but upregulated the expression of VE-cadherin and claudin-5 in serum and pulmonary vascular endothelial cells, compared with treatment with saline.
Conclusions
This study reveals that Wenyang Huazhuo Tongluo Formula plays a new role in the treatment of SSc by alleviating pulmonary vascular damage. Furthermore, we found that Wenyang Huazhuo Tongluo Formula alleviates pulmonary vascular injury and inhibits HIF-1α expression.
Collapse
|
14
|
MiR-27a as a diagnostic biomarker and potential therapeutic target in systemic sclerosis. Sci Rep 2022; 12:18932. [PMID: 36344812 PMCID: PMC9640682 DOI: 10.1038/s41598-022-23723-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Systemic sclerosis (SSc) or scleroderma is a multiorgan rheumatoid disease characterized by skin tightening or organ dysfunction due to fibrosis, vascular damage, and autoimmunity. No specific cause has been discovered for this illness, and hence no effective treatment exists for it. On the other hand, due to the lack of diagnostic biomarkers capable of effectively and specifically differentiating the patients, early diagnosis has not been possible. Due to their potent regulatory roles in molecular pathways, microRNAs are among the novel candidates for the diagnosis and treatment of diseases like SSc. MiR-27a is a microRNA known for its role in the pathogenesis of fibrosis and cancer, both of which employ similar signaling pathways; hence we hypothesized that Mir-27a could be dysregulated in the blood of individuals affected by SSc and it might be useful in the diagnosis or treatment of this disease. Blood was collected from 60 SSc patients (30 limited and 30 diffuse) diagnosed by a rheumatologist according to ACR/AULAR criteria; following RNA isolation and cDNA synthesis; real-time qPCR was performed on the samples using Taq-Man probes and data were analyzed by the ΔΔCT method. Also, potential targets of miR-27a were evaluated using bioinformatics. It was revealed that miR-27a was significantly down-regulated in SSc patients in comparison to healthy individuals, but there was no difference in miR-27 expression between limited and diffused SSc patients. Besides, miR-27a was found to target several contributing factors to SSc. It seems that miR-27a has a protective role in SSc, and its downregulation could result in the disease's onset. Based on bioinformatics analyses, it is speculated that miR-27a likely targets factors contributing to the pathogenesis of SSc, which are elevated upon the downregulation of miR-27a; hence, miR-27a mimics could be considered as potential therapeutic agents for the treatment of SSc in future studies. Since no difference was observed between limited and diffuse patient groups, it is unlikely that this microRNA has a role in disease progression. According to ROC analysis of qPCR data, miR-27a could be employed as a valuable diagnostic biomarker for SSc.
Collapse
|
15
|
Bălănescu A, Băicuș C, Bălănescu E, Bălănescu P. Circulatory cytokeratin 17, marginal zone B1 protein and leucine-rich α2-glycoprotein-1 as biomarkers for disease severity and fibrosis in systemic sclerosis patients. Biochem Med (Zagreb) 2022; 32:030707. [PMID: 36277429 PMCID: PMC9562799 DOI: 10.11613/bm.2022.030707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/20/2022] [Indexed: 11/24/2022] Open
Abstract
Introduction Systemic sclerosis (Ssc) is a multiorgan debilitating autoimmune disease that associates the triad: vascular involvement, tissue fibrosis and profound immune response alterations. Numerous previous studies focused on identification of candidate proteomic Ssc biomarkers using mass-spectrometry techniques and a large number of candidate Ssc biomarkers emerged. These biomarkers must firstly be confirmed in independent patient groups. The aim of the present study was to investigate the association of cytokeratin 17 (CK17), marginal zone B1 protein (MZB1) and leucine-rich α2-glycoprotein-1 (LRG1) with clinical and biological Ssc characteristics. Material and methods Serum CK17, MZB1 and LRG1 were assessed in samples of the available Ssc biobank comprising of samples from 53 Ssc patients and 26 matched age and gender controls. Results Circulatory CK17, LRG1 and MZB1 concentrations were increased in Ssc patients. Cytokeratin 17 is independently associated with Ssc disease activity. Patients with pulmonary fibrosis expressed higher LRG1 and MZB1 concentrations. Serum MZB1 concentrations were also associated with extensive skin fibrosis. Conclusions Serum CK17, MZB1 and LRG1 were confirmed biomarkers for Ssc. LRG1 seems a good biomarker for pulmonary fibrosis, while MZB1 is a good biomarker for extensive skin fibrosis. CK17 proved to be independently associated with Ssc disease severity, higher CK17 values being protective for a more active disease.
Collapse
Affiliation(s)
- Anca Bălănescu
- Pediatrics Department, University of Medicine and Pharmacy Carol Davila, Bucharest, Romania
| | - Cristian Băicuș
- Internal Medicine Department, University of Medicine and Pharmacy Carol Davila, Bucharest, Romania
| | - Eugenia Bălănescu
- Clinical Immunology Laboratory CDPC, Colentina Clinical Hospital, Bucharest, Romania
| | - Paul Bălănescu
- Internal Medicine Department, University of Medicine and Pharmacy Carol Davila, Bucharest, Romania
| |
Collapse
|
16
|
Kondo M, Suzuki T, Kawano Y, Kojima S, Miyashiro M, Matsumoto A, Kania G, Błyszczuk P, Ross RL, Mulipa P, Del Galdo F, Zhang Y, Distler JHW. Dersimelagon, a novel oral melanocortin 1 receptor agonist, demonstrates disease-modifying effects in preclinical models of systemic sclerosis. Arthritis Res Ther 2022; 24:210. [PMID: 36050717 PMCID: PMC9434962 DOI: 10.1186/s13075-022-02899-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Background Activation of melanocortin 1 receptor (MC1R) is known to exert broad anti-inflammatory and anti-fibrotic effects. The purpose of this study is to investigate the potential of dersimelagon, a novel oral MC1R agonist, as a therapeutic agent for systemic sclerosis (SSc). Methods The effects of dersimelagon phosphoric acid (MT-7117) on skin fibrosis and lung inflammation were evaluated in bleomycin (BLM)-induced SSc murine models that were optimized for prophylactic and therapeutic evaluation. Microarray-based gene expression analysis and serum protein profiling were performed in the BLM-induced SSc models. The effect of MT-7117 on transforming growth factor-β (TGF-β)-induced activation of human dermal fibroblasts was evaluated in vitro. Immunohistochemical analyses of MC1R expression in the skin of SSc patients were performed. Results Prophylactic treatment with MT-7117 (≥ 0.3 mg/kg/day p.o.) significantly inhibited skin fibrosis and lung inflammation, and therapeutic treatment with MT-7117 (≥ 3 mg/kg/day p.o.) significantly suppressed the development of skin fibrosis in the BLM-induced SSc models. Gene array analysis demonstrated that MT-7117 exerts an anti-inflammatory effect via suppression of the activation of inflammatory cells and inflammation-related signals; additionally, vascular dysfunction was extracted as the pathology targeted by MT-7117. Serum protein profiling revealed that multiple SSc-related biomarkers including P-selectin, osteoprotegerin, cystatin C, growth and differentiation factor-15, and S100A9 were suppressed by MT-7117. MT-7117 inhibited the activation of human dermal fibroblasts by suppressing TGF-β-induced ACTA2 (encoding α-smooth muscle actin) mRNA elevation. MC1R was expressed by monocytes/macrophages, neutrophils, blood vessels (endothelial cells), fibroblasts, and epidermis (keratinocytes) in the skin of SSc patients, suggesting that these MC1R-positive cells could be targets for MT-7117. Conclusions MT-7117 demonstrates disease-modifying effects in preclinical models of SSc. Investigations of its mechanism of action and target expression analyses indicate that MT-7117 exerts its positive effect by affecting inflammation, vascular dysfunction, and fibrosis, which are all key pathologies of SSc. The results of the present study suggest that MT-7117 is a potential therapeutic agent for SSc. A phase 2 clinical trial investigating the efficacy and tolerability of MT-7117 in patients with early, progressive diffuse cutaneous SSc is currently in progress. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02899-3.
Collapse
Affiliation(s)
- Masahiro Kondo
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa, 227-0033, Japan.
| | - Tsuyoshi Suzuki
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa, 227-0033, Japan
| | - Yuko Kawano
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa, 227-0033, Japan
| | - Shinji Kojima
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa, 227-0033, Japan
| | - Masahiko Miyashiro
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa, 227-0033, Japan
| | - Atsuhiro Matsumoto
- Sohyaku. Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa, 227-0033, Japan
| | - Gabriela Kania
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Wagistr. 14, 8952, Schlieren, Switzerland
| | - Przemysław Błyszczuk
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, Wagistr. 14, 8952, Schlieren, Switzerland
| | - Rebecca L Ross
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, LS9 7TF, UK.,NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds, UK
| | - Panji Mulipa
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, LS9 7TF, UK.,NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds, UK
| | - Francesco Del Galdo
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, LS9 7TF, UK.,NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds, UK
| | - Yun Zhang
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| | - Jörg H W Distler
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
17
|
Chikhoune L, Brousseau T, Morell-Dubois S, Farhat MM, Maillard H, Ledoult E, Lambert M, Yelnik C, Sanges S, Sobanski V, Hachulla E, Launay D. Association between Routine Laboratory Parameters and the Severity and Progression of Systemic Sclerosis. J Clin Med 2022; 11:jcm11175087. [PMID: 36079017 PMCID: PMC9457158 DOI: 10.3390/jcm11175087] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Systemic sclerosis (SSc) is a heterogeneous connective tissue disease with a high mortality and morbidity rate. Identification of biomarkers that can predict the evolution of SSc is a key factor in the management of patients. The aim of this study was to assess the association of routine laboratory parameters, widely used in practice and easily available, with the severity and progression of SSc. (2) Methods: In this retrospective monocentric cohort study, 372 SSc patients were included. We gathered clinical and laboratory data including routine laboratory parameters: C-reactive-protein (CRP), erythrocyte sedimentation rate (ESR), complete blood count, serum sodium and potassium levels, creatinin, urea, ferritin, albumin, uric acid, N-terminal pro-brain natriuretic peptide (NTproBNP), serum protein electrophoresis, and liver enzymes. Associations between these routine laboratory parameters and clinical presentation and outcome were assessed. (3) Results: Median (interquartile range) age was 59.0 (50.0; 68.0) years. White blood cell, monocyte, and neutrophil absolute counts were significantly higher in patients with diffuse cutaneous SSc and with interstitial lung disease (ILD) (p < 0.001). CRP was significantly higher in patients with ILD (p < 0.001). Hemoglobin and ferritin were significantly lower in patients with pulmonary hypertension (PH) including pulmonary arterial hypertension and ILD associated PH (p = 0.016 and 0.046, respectively). Uric acid and NT pro BNP were significantly higher in patients with PH (<0.001). Monocyte count was associated with ILD progression over time. (4) Conclusions: Overall, our study highlights the association of routine laboratory parameters used in current practice with the severity and progression of SSc.
Collapse
Affiliation(s)
- Liticia Chikhoune
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
| | - Thierry Brousseau
- CHU Lille, Service de Biochimie Automatisée Protéines, F-59000 Lille, France
| | - Sandrine Morell-Dubois
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
| | - Meryem Maud Farhat
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
- U1286—INFINITE—Institute for Translational Research in Inflammation, Université de Lille, F-59000 Lille, France
- Inserm, F-59000 Lille, France
| | - Helene Maillard
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
| | - Emmanuel Ledoult
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
- U1286—INFINITE—Institute for Translational Research in Inflammation, Université de Lille, F-59000 Lille, France
- Inserm, F-59000 Lille, France
| | - Marc Lambert
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
| | - Cecile Yelnik
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
| | - Sebastien Sanges
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
- U1286—INFINITE—Institute for Translational Research in Inflammation, Université de Lille, F-59000 Lille, France
- Inserm, F-59000 Lille, France
| | - Vincent Sobanski
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
- U1286—INFINITE—Institute for Translational Research in Inflammation, Université de Lille, F-59000 Lille, France
- Inserm, F-59000 Lille, France
| | - Eric Hachulla
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
- U1286—INFINITE—Institute for Translational Research in Inflammation, Université de Lille, F-59000 Lille, France
- Inserm, F-59000 Lille, France
| | - David Launay
- CHU Lille, Service de Médecine Interne et Immunologie Clinique, Centre de Référence des Maladies Auto-Immunes Systémiques Rares du Nord et Nord-Ouest de France (CeRAINO), F-59000 Lille, France
- U1286—INFINITE—Institute for Translational Research in Inflammation, Université de Lille, F-59000 Lille, France
- Inserm, F-59000 Lille, France
- Correspondence: ; Tel.: +33-3-2044-4433
| |
Collapse
|
18
|
Cotton T, Fritzler MJ, Choi MY, Zheng B, Niaki OZ, Pineau CA, Lukusa L, Bernatsky S. Serologic phenotypes distinguish systemic lupus erythematosus patients developing interstitial lung disease and/or myositis. Lupus 2022; 31:1477-1484. [PMID: 36018314 PMCID: PMC9580032 DOI: 10.1177/09612033221122158] [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] [Indexed: 11/15/2022]
Abstract
Objectives To determine if serologic phenotypes could be identified in systemic lupus erythematosus patients developing interstitial lung disease (ILD) and/or myositis. Methods Adult SLE patients (without myositis/ILD at baseline) had annual assessments and serum sampling between 2000 and 2017. New-onset ILD was identified using the SDI pulmonary fibrosis item. New-onset myositis was identified using the SLICC Damage Index muscle atrophy/weakness item, the SLEDAI-2K item for myositis, and annual creatinine kinase testing. Chart review confirmed ILD/myositis cases and randomly sampled SLE patients from baseline formed our sub-cohort (N = 72). Cases and sub-cohort were compared regarding myositis-related biomarkers at baseline and at a randomly selected follow-up between baseline and end of observation (date of ILD/myositis diagnosis or Dec. 31, 2017). Descriptive analyses and hazards ratios (HRs) were generated for ILD/myositis incidence, focusing on baseline serology and adjusting for sex, race/ethnicity, age at SLE diagnosis, and SLE duration. Results Fourteen SLE patients developed ILD (N = 9), myositis (N = 3), and/or both (N = 2). Thirteen of those (92.9%) developing ILD/myositis had at least one biomarker at baseline, versus 47 (65.3%) SLE patients who never developed myositis/ILD. The most common biomarkers in myositis/ILD were KL-6, anti-Ro52, and anti-Ku. Baseline biomarkers tended to remain positive in follow-up. In multivariate Cox regressions, SLE patients had higher risk of developing myositis/ILD with elevated baseline KL-6 (adjusted hazard ratio 3.66; 95% confidence interval 1.01, 13.3). When updating biomarkers over time, we also saw correlations between anti-Smith and ILD/myositis. Conclusions Baseline myositis-related biomarkers were highly associated with ILD/myositis incidence. This is the first identification of biomarker phenotypes with ILD/myositis risk in SLE.
Collapse
Affiliation(s)
- Thaisa Cotton
- Department of Medicine, 5620McGill University, Montreal, QC, Canada
| | - Marvin J Fritzler
- Department of Medicine, Cumming School of Medicine, 70401University of Calgary, Calgary, AB, Canada
| | - May Y Choi
- Department of Medicine, Cumming School of Medicine, 70401University of Calgary, Calgary, AB, Canada
| | - Boyang Zheng
- Department of Medicine, 5620McGill University, Montreal, QC, Canada.,Division of Rheumatology, 5620McGill UniversityHealth Centre, Montreal, QC, Canada
| | | | - Christian A Pineau
- Department of Medicine, 5620McGill University, Montreal, QC, Canada.,Division of Rheumatology, 5620McGill UniversityHealth Centre, Montreal, QC, Canada
| | - Luck Lukusa
- Division of Rheumatology, 5620McGill UniversityHealth Centre, Montreal, QC, Canada
| | - Sasha Bernatsky
- Department of Medicine, 5620McGill University, Montreal, QC, Canada.,Division of Rheumatology, 5620McGill UniversityHealth Centre, Montreal, QC, Canada
| |
Collapse
|
19
|
Maritati F, Provenzano M, Lerario S, Corradetti V, Bini C, Busutti M, Grandinetti V, Cuna V, La Manna G, Comai G. Kidney transplantation in systemic sclerosis: Advances in graft, disease, and patient outcome. Front Immunol 2022; 13:878736. [PMID: 35958558 PMCID: PMC9360313 DOI: 10.3389/fimmu.2022.878736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 06/30/2022] [Indexed: 11/30/2022] Open
Abstract
Systemic sclerosis (SSc) is an immune-mediated rheumatic disease characterized by vascular abnormalities, tissue fibrosis, and inflammation. Renal disease occurring in patients with SSc may have a variable clinicopathological picture. However, the most specific renal condition associated with this disease is the scleroderma renal crisis (SRC), characterized by acute onset of renal failure and severe hypertension. SRC develops in about 20% of cases of SSc, especially in those patients with diffuse cutaneous disease. The prognosis of this condition is often negative, with a rapid progression to end-stage renal disease (ESRD). The advent of the antihypertensive angiotensin-converting enzyme inhibitors in 1980 was associated with a significant improvement in patients’ survival and recovery of renal function. However, the prognosis of these patients can still be improved. The dialytic condition is associated with early death, and mortality is significantly higher than among patients undergoing renal replacement therapy (RRT) due to other conditions. Patients with SRC who show no signs of renal functional recovery despite timely blood pressure control are candidates for kidney transplantation (KT). In this review, we reported the most recent advances in KT in patients with ESRD due to SSc, with a particular overview of the risk of disease recurrence after transplantation and the evolution of other disease manifestations.
Collapse
|
20
|
Kowalska-Kępczyńska A. Systemic Scleroderma-Definition, Clinical Picture and Laboratory Diagnostics. J Clin Med 2022; 11:2299. [PMID: 35566425 PMCID: PMC9100749 DOI: 10.3390/jcm11092299] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/08/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023] Open
Abstract
(1) Background: Scleroderma (Sc) is a rare connective tissue disease classified as an autoimmune disorder. The pathogenesis of this disease is not fully understood. (2) Methods: This article reviews the literature on systemic scleroderma (SSc). A review of available scientific articles was conducted using the PubMed database with a time range of January 1985 to December 2021. (3) Results and Conclusions: The article is a review of information on epidemiology, criteria for diagnosis, pathogenesis, a variety of clinical pictures and the possibility of laboratory diagnostic in the diagnosis and monitoring of systemic scleroderma.
Collapse
Affiliation(s)
- Anna Kowalska-Kępczyńska
- Department of Biochemical Diagnostics, Chair of Laboratory Diagnostics, Medical University of Lublin, 20-081 Lublin, Poland
| |
Collapse
|
21
|
Cardoneanu A, Burlui AM, Macovei LA, Bratoiu I, Richter P, Rezus E. Targeting Systemic Sclerosis from Pathogenic Mechanisms to Clinical Manifestations: Why IL-6? Biomedicines 2022; 10:biomedicines10020318. [PMID: 35203527 PMCID: PMC8869570 DOI: 10.3390/biomedicines10020318] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 12/04/2022] Open
Abstract
Systemic sclerosis (SS) is a chronic autoimmune disorder, which has both cutaneous and systemic clinical manifestations. The disease pathogenesis includes a triad of manifestations, such as vasculopathy, autoimmunity, and fibrosis. Interleukin-6 (IL-6) has a special role in SS development, both in vascular damage and in the development of fibrosis. In the early stages, IL-6 participates in vascular endothelial activation and apoptosis, leading to the release of damage-associated molecular patterns (DAMPs), which maintain inflammation and autoimmunity. Moreover, IL-6 plays an important role in the development of fibrotic changes by mediating the transformation of fibroblasts into myofibroblasts. All of these are associated with disabling clinical manifestations, such as skin thickening, pulmonary fibrosis, pulmonary arterial hypertension (PAH), heart failure, and dysphagia. Tocilizumab is a humanized monoclonal antibody that inhibits IL-6 by binding to the specific receptor, thus preventing its proinflammatory and fibrotic actions. Anti-IL-6 therapy with Tocilizumab is a new hope for SS patients, with data from clinical trials supporting the favorable effect, especially on skin and lung damage.
Collapse
Affiliation(s)
- Anca Cardoneanu
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.M.B.); (L.A.M.); (I.B.); (P.R.); (E.R.)
- Rehabilitation Hospital, 700661 Iasi, Romania
- Correspondence:
| | - Alexandra Maria Burlui
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.M.B.); (L.A.M.); (I.B.); (P.R.); (E.R.)
- Rehabilitation Hospital, 700661 Iasi, Romania
| | - Luana Andreea Macovei
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.M.B.); (L.A.M.); (I.B.); (P.R.); (E.R.)
- Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ioana Bratoiu
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.M.B.); (L.A.M.); (I.B.); (P.R.); (E.R.)
- Rehabilitation Hospital, 700661 Iasi, Romania
| | - Patricia Richter
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.M.B.); (L.A.M.); (I.B.); (P.R.); (E.R.)
- Rehabilitation Hospital, 700661 Iasi, Romania
| | - Elena Rezus
- Department of Rheumatology, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.M.B.); (L.A.M.); (I.B.); (P.R.); (E.R.)
- Rehabilitation Hospital, 700661 Iasi, Romania
| |
Collapse
|
22
|
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: 1.0] [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
|
23
|
Biomarkers in systemic sclerosis: mechanistic insights into pathogenesis and treatment. Curr Opin Rheumatol 2021; 33:480-485. [PMID: 34420004 DOI: 10.1097/bor.0000000000000827] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE OF REVIEW Systemic sclerosis (SSc) is heterogenous on molecular, cellular, tissue, and clinical levels. Although many biomarkers have been described in clinical studies, few have been rigorously mapped to specific molecular pathways, tissue pathologies, and clinical manifestations. A focused assessment of peripheral blood levels of C-C Motif Chemokine Ligand-18 (CCL18) and periostin illustrates how biomarkers can link molecular mediators to clinical outcomes. RECENT FINDINGS CCL18 is produced by pulmonary macrophages in response to type 2 cytokines and IL6. Elevated serum CCL18 is associated with interstitial lung disease (ILD) in SSc patients and is prognostic for ILD progression. It is pharmacologically modulated by IL6 inhibition, and associated with stabilization of lung function decline but not with improvements in skin fibrosis. Periostin is produced by dermal fibroblasts in SSc in response to type 2 cytokines and transforming growth factor-beta. Elevated serum periostin is associated with cutaneous disease in SSc patients but not ILD. Other cell- and tissue-specific biomarkers detectable in peripheral blood and informative with respect to SSc pathogenesis include KL-6 and SP-D in lung epithelium, osteopontin in lung macrophages, and cartilage oligomeric matrix protein in dermal fibroblasts. SUMMARY Blood biomarkers related to specific molecular mediators, cell types, and tissues of origin can help to link therapeutic targets to treatable traits in SSc.
Collapse
|