1
|
Liang CM, Lee W, Chou CC, Tung H, Chen HC, Chen HM, Lee WJ, Chen YM. Nailfold capillary measurements correlated to NOTCH3 R544C mutation in preclinical CADASIL patients. J Neurol Sci 2024; 462:123109. [PMID: 38941707 DOI: 10.1016/j.jns.2024.123109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/23/2024] [Accepted: 06/22/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL) is a hereditary disease caused by NOTCH3 mutation. Nailfold capillaroscopy is a non-invasive technique typically used for rheumatic diseases. It has potential in other conditions linked to vascular pathology. However, capillaroscopy in CADASIL has not been explored. This study aims to investigate whether capillaroscopy measurements can correlate with brain vascular changes in preclinical CADASIL patients, specifically those with NOTCH3 mutation. METHODS This study included 69 participants from the Taiwan Precision Medicine Initiative (TPMI) dataset who visited Taichung Veterans General Hospital from January to December 2022. All individuals underwent genetic studies, brain imaging and nailfold capillaroscopy. The Mann-Whitney U test was used to compare results of brain imaging between carriers and controls. It was also used to compare measurements in nailfold capillaroscopy within each group. Spearman Rank Correlation Analysis was used to explore the relationship between capillary measurements and brain MRI results. RESULTS White matter hyperintensities (WMH) expression was positively correlated with capillary dimension and negatively correlated with density. Our results presented that R544C carriers exhibited a diffuse increase in WMH (p < 0.001) and a global reduction in gray matter volume but preserved in specific areas. The white matter lesion scores in all brain regions were higher in the mutation carriers than the controls. (p < 0.001). CONCLUSION This research highlights the association of nailfold capillaroscopy findings with white matter lesions in preclinical CADASIL patients. Capillaroscopy guides an effective screening strategy in individuals with NOTCH3 mutations.
Collapse
Affiliation(s)
- Chun-Min Liang
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Wei Lee
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chien-Chih Chou
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Ophthalmology, Taichung Veterans General Hospital, Taichung, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsin Tung
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan; Center of Faculty Development, Taichung Veterans General Hospital, Taichung, Taiwan; Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hung-Chieh Chen
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Neuroradiology, Department of Radiology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Hsian-Min Chen
- Center for Quantitative Imaging in Medicine, Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Wei-Ju Lee
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan; Neurological Institute, Taichung Veterans General Hospital, Taichung, Taiwan; Dementia Center, Taichung Veterans General Hospital, Taichung, Taiwan; Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Ming Chen
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan; Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan; Institute of Biomedical Science and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan; Precision Medicine Research Center, College of Medicine, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
2
|
Prajjwal P, Marsool MDM, Yadav V, Kanagala RSD, Reddy YB, John J, Lam JR, Karra N, Amiri B, Islam MU, Nithya V, Marsool ADM, Gadam S, Vora N, Hussin OA. Neurological, cardiac, musculoskeletal, and renal manifestations of scleroderma along with insights into its genetics, pathophysiology, diagnostic, and therapeutic updates. Health Sci Rep 2024; 7:e2072. [PMID: 38660003 PMCID: PMC11040569 DOI: 10.1002/hsr2.2072] [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: 07/24/2023] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/26/2024] Open
Abstract
Background Scleroderma, also referred to as systemic sclerosis, is a multifaceted autoimmune condition characterized by abnormal fibrosis and impaired vascular function. Pathologically, it encompasses the persistent presence of inflammation, abnormal collagen buildup, and restructuring of blood vessels in various organs, resulting in a wide range of clinical symptoms. This review incorporates the most recent scientific literature on scleroderma, with a particular emphasis on its pathophysiology, clinical manifestations, diagnostic approaches, and treatment options. Methodology A comprehensive investigation was carried out on numerous databases, such as PubMed, MEDLINE, Scopus, Web of Science, and Google Scholar, to collect pertinent studies covering diverse facets of scleroderma research. Results Scleroderma presents with a range of systemic manifestations, such as interstitial lung disease, gastrointestinal dysmotility, Raynaud's phenomenon, pulmonary arterial hypertension, renal complications, neurological symptoms, and cardiac abnormalities. Serological markers, such as antinuclear antibodies, anti-centromere antibodies, and anti-topoisomerase antibodies, are important for classifying diseases and predicting their outcomes. Discussion The precise identification of scleroderma is crucial for promptly and correctly implementing effective treatment plans. Treatment approaches aim to improve symptoms, reduce complications, and slow down the progression of the disease. An integrated approach that combines pharmacological agents, including immunosuppressants, endothelin receptor antagonists, and prostanoids, with nonpharmacological interventions such as physical and occupational therapy is essential for maximizing patient care. Conclusion Through the clarification of existing gaps in knowledge and identification of emerging trends, our goal is to improve the accuracy of diagnosis, enhance the effectiveness of therapeutic interventions, and ultimately enhance the overall quality of life for individuals suffering from scleroderma. Ongoing cooperation and creative research are necessary to advance the field and achieve improved patient outcomes and new therapeutic discoveries.
Collapse
Affiliation(s)
| | | | - Vikas Yadav
- Department of Internal MedicinePt. B. D. S. Postgraduate Institute of Medical SciencesRohtakIndia
| | | | | | - Jobby John
- Department of Internal MedicineDr. Somervell Memorial CSI Medical College and HospitalNeyyāttinkaraIndia
| | - Justin Riley Lam
- Department of Internal MedicineCebu Institute of MedicineCebuPhilippines
| | - Nanditha Karra
- Department of Internal MedicineOsmania Medical CollegeHyderabadTelanganaIndia
| | - Bita Amiri
- Cardiovascular Research CenterTabriz University of Medical SciencesTabrizIran
| | - Moiz Ul Islam
- Department of Internal MedicinePunjab Medical CollegeFaisalabadPakistan
| | - Venkatesh Nithya
- Department of Internal MedicineS. D. Asfendiyarov Kazakh National Medical UniversityAlmatyKazakhstan
| | | | | | | | - Omniat Amir Hussin
- Department of MedicineAlmanhal University Academy of ScienceKhartoumSudan
| |
Collapse
|
3
|
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
|
4
|
Truchetet ME, Hughes M. Ulcères digitaux de la sclérodermie. REVUE DU RHUMATISME 2023; 90:640-647. [DOI: 10.1016/j.rhum.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
5
|
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
|
6
|
Ataş F, Kaya M, Ayhan Z, Ozkan O, Birlik M. Evaluation of choroidal vascularity index in systemic sclerosis patients. Photodiagnosis Photodyn Ther 2023; 41:103297. [PMID: 36682429 DOI: 10.1016/j.pdpdt.2023.103297] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/07/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023]
Abstract
OBJECTIVE To investigate the comparison of choroidal vascularity index (CVI) between patients with systemic sclerosis and healthy individuals. METHODS This study was designed prospective non-randomized cross-sectional study. Eighty-six eyes (43 patients) with limited cutaneous systemic sclerosis (lcSSc), 60 eyes (30 patients) with diffuse cutaneous systemic sclerosis (dcSSc) and 60 eyes (30 subjects) of age-and sex-matched healthy individuals were recruited. Subfoveal choroidal thickness, CVI and modified Rodnan skin score (mRSS) were evaluated. Enhanced depth imaging- optical coherence tomography scans were binarized using Niblack's autolocal threshold and CVI was determined as the luminal choroidal area/total choroidal area ratio. RESULTS The mean CCT values were 268.00±68.59 µm, 286.90±70.88 µm, 321.73±94.13 µm in lcSSc group, dcSSc group and control group, respectively. The mean CVI was 61.84±2.84% in lcSSc group, 54.62±5.84% in dcSSc group and 62.41±4.13% in control group (p=0.001). The mean CVI of the SSc patients was 58.91±5.58 and there was significant difference between control group (p<0.001). The mean mRSS was 3.84±2.50 in lcSSc group and 14.07±6.81 in dcSSc group (p<0.001). There was a statistically significant negative correlation between mRSS and CVI (r=-0.448, p<0.001) CONCLUSION: Choroidal vascularity index provides valuable information to monitor the disease progression and lower CVI values seem to be related to the disease severity in patients with systemic sclerosis.
Collapse
Affiliation(s)
- Ferdane Ataş
- Department of Ophthalmology, Çerkezköy State Hospital, Tekirdağ, Turkey.
| | - Mahmut Kaya
- Department of Ophthalmology, Dokuz Eylul University, Izmir, Turkey
| | - Ziya Ayhan
- Department of Ophthalmology, Dokuz Eylul University, Izmir, Turkey
| | - Ozlem Ozkan
- Department of Ophthalmology, Dokuz Eylul University, Izmir, Turkey
| | - Merih Birlik
- Department of Rheumatology, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
7
|
Frech TM, Frech M, Saknite I, O'Connell KA, Ghosh S, Baba J, Tkaczyk ER. Novel therapies and innovation for systemic sclerosis skin ulceration. Best Pract Res Clin Rheumatol 2022; 36:101813. [PMID: 36609122 PMCID: PMC11671032 DOI: 10.1016/j.berh.2022.101813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Skin ulceration is an important cause of morbidity in systemic sclerosis and can occur at anytime during disease progression. Incident disease cohorts are important for understanding whether skin ulceration represents active vasculopathy versus resultant damage. Biomarkers for skin ulcer pathogenesis, both serum and imaging, are under investigation to elucidate the functional consequences of the structural abnormalities. Novel therapeutics for the treatment of vasculopathy benefit from reliable biomarkers able to predict the disease evolution remains an important unmet need. Nonetheless, a diagnostic approach that captures early skin ulceration and treatments that restore vascular and immune homeostasis is critical for effective systemic sclerosis (SSc) vasculopathy management.
Collapse
Affiliation(s)
- Tracy M Frech
- US Department of Veterans Affairs, Tennessee Valley Healthcare System, Dermatology Service and Research Service, Nashville, TN, USA; Vanderbilt University Medical Center, Department of Medicine, Division of Rheumatology and Immunology, Nashville, TN, USA.
| | | | - Inga Saknite
- Vanderbilt University Medical Center, Department of Dermatology, Nashville, TN, USA; University of Latvia, Institute of Atomic Physics and Spectroscopy, Biophotonics Laboratory, Riga, Latvia
| | - Katie A O'Connell
- Vanderbilt University Medical Center, Department of Dermatology, Nashville, TN, USA
| | - Shramana Ghosh
- US Department of Veterans Affairs, Tennessee Valley Healthcare System, Dermatology Service and Research Service, Nashville, TN, USA; Vanderbilt University Medical Center, Department of Dermatology, Nashville, TN, USA
| | - Justin Baba
- Vanderbilt Biophotonics Center, Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Eric R Tkaczyk
- US Department of Veterans Affairs, Tennessee Valley Healthcare System, Dermatology Service and Research Service, Nashville, TN, USA; Vanderbilt University Medical Center, Department of Dermatology, Nashville, TN, USA; Vanderbilt Biophotonics Center, Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Vanderbilt-Ingram Cancer Center, Nashville, TN, USA
| |
Collapse
|
8
|
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
|
9
|
Colasanti T, Stefanantoni K, Fantini C, Corinaldesi C, Vasile M, Marampon F, Di Luigi L, Antinozzi C, Sgrò P, Lenzi A, Riccieri V, Crescioli C. The Prostacyclin Analogue Iloprost Modulates CXCL10 in Systemic Sclerosis. Int J Mol Sci 2022; 23:ijms231710150. [PMID: 36077548 PMCID: PMC9456348 DOI: 10.3390/ijms231710150] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
The prostacyclin analogue iloprost is used to treat vascular alterations and digital ulcers, the early derangements manifesting in systemic sclerosis (SSc), an autoimmune disease leading to skin and organ fibrosis. Bioindicator(s) of SSc onset and progress are still lacking and the therapeutic approach remains a challenge. The T helper 1 (Th1) chemokine interferon (IFN)γ-induced protein 10 (IP-10/CXCL10) associates with disease progression and worse prognosis. Endothelial cells and fibroblasts, under Th1-dominance, release CXCL10, further enhancing SSc’s detrimental status. We analyzed the effect of iloprost on CXCL10 in endothelial cells, dermal fibroblasts, and in the serum of SSc patients. Human endothelial cells and dermal fibroblasts activated with IFNγ/Tumor Necrosis Factor (TNF)α, with/without iloprost, were investigated for CXCL10 secretion/expression and for intracellular signaling cascade underlying chemokine release (Signal Transducer and Activator of Transcription 1, STAT1; Nuclear Factor kappa-light-chain-enhancer of activated B cells, NF-kB; c-Jun NH2-terminal kinase, JNK: Phosphatidyl-Inositol 3-kinase (PI3K)/protein kinase B, AKT; Extracellular signal-Regulated Kinase 1/2, ERK1/2). CXCL10 was quantified in sera from 25 patients taking iloprost, satisfying the American College of Rheumatology (ACR)/European Alliance of Associations for Rheumatology (EULAR) 2013 classification criteria for SSc, and in sera from 20 SSc sex/age-matched subjects without therapy, previously collected. In human endothelial cells and fibroblasts, iloprost targeted CXCL10, almost preventing IFNγ/TNFα-dependent cascade activation in endothelial cells. In SSc subjects taking iloprost, serum CXCL10 was lower. These in vitro and in vivo data suggest a potential role of iloprost to limit CXCL10 at local vascular/dermal and systemic levels in SSc and warrant further translational research aimed to ameliorate SSc understanding/management.
Collapse
Affiliation(s)
- Tania Colasanti
- Rheumatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155-00161 Rome, Italy
| | - Katia Stefanantoni
- Rheumatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155-00161 Rome, Italy
| | - Cristina Fantini
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis, 006-00135 Rome, Italy
| | - Clarissa Corinaldesi
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis, 006-00135 Rome, Italy
- Institute for Cancer Genetics, Columbia University, New York, NY 10027, USA
| | - Massimiliano Vasile
- Rheumatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155-00161 Rome, Italy
| | - Francesco Marampon
- Department of Radiotherapy, Sapienza University of Rome, Viale del Policlinico, 155-00161 Rome, Italy
| | - Luigi Di Luigi
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis, 006-00135 Rome, Italy
| | - Cristina Antinozzi
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis, 006-00135 Rome, Italy
| | - Paolo Sgrò
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis, 006-00135 Rome, Italy
| | - Andrea Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Viale del Policlinico, 155-00161 Rome, Italy
| | - Valeria Riccieri
- Rheumatology Unit, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico, 155-00161 Rome, Italy
- Correspondence: (V.R.); (C.C.); Tel.: +39-06-49974641 (V.R.); +39-06-36733395 (C.C.)
| | - Clara Crescioli
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis, 006-00135 Rome, Italy
- Correspondence: (V.R.); (C.C.); Tel.: +39-06-49974641 (V.R.); +39-06-36733395 (C.C.)
| |
Collapse
|
10
|
Romano E, Rosa I, Fioretto BS, Matucci-Cerinic M, Manetti M. Circulating Neurovascular Guidance Molecules and Their Relationship with Peripheral Microvascular Impairment in Systemic Sclerosis. LIFE (BASEL, SWITZERLAND) 2022; 12:life12071056. [PMID: 35888144 PMCID: PMC9316343 DOI: 10.3390/life12071056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/06/2022] [Accepted: 07/14/2022] [Indexed: 11/16/2022]
Abstract
Systemic sclerosis (SSc, scleroderma) is a complex connective tissue disease whose earliest clinical manifestations are microvascular tone dysregulation and peripheral microcirculatory abnormalities. Following previous evidence of an association between circulating neurovascular guidance molecules and SSc disturbed angiogenesis, here, we measured the levels of soluble neuropilin 1 (sNRP1), semaphorin 3E (Sema3E), and Slit2 by enzyme-linked immunosorbent assay in serum samples from a large case series of 166 SSc patients vs. 110 healthy controls. We focused on their possible correlation with vascular disease clinical features and applied logistic regression analysis to determine which of them could better reflect disease activity and severity. Our results demonstrate that, in SSc: (i) sNRP1 is significantly decreased, with lower sNRP1 serum levels correlating with the severity of nailfold videocapillaroscopy (NVC) abnormalities and the presence of ischemic digital ulcers (DUs); (ii) both Sema3E and Slit2 are increased, with Sema3E better reflecting early NVC abnormalities; and (iii) higher Sema3E correlates with the absence of DUs, while augmented Slit2 associates with the presence of DUs. Receiver operator characteristics curve analysis revealed that both circulating sNRP1 and Sema3E show a moderate diagnostic accuracy. Moreover, logistic regression analysis allowed to identify sNRP1 and Sema3E as more suitable independent biomarkers reflecting the activity and severity of SSc-related peripheral microvasculopathy.
Collapse
Affiliation(s)
- Eloisa Romano
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (E.R.); (B.S.F.); (M.M.-C.)
| | - Irene Rosa
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy;
| | - Bianca Saveria Fioretto
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (E.R.); (B.S.F.); (M.M.-C.)
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy;
| | - Marco Matucci-Cerinic
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (E.R.); (B.S.F.); (M.M.-C.)
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Hospital, 20132 Milan, Italy
| | - Mirko Manetti
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy;
- Correspondence: ; Tel.: +39-055-275-8073
| |
Collapse
|
11
|
Colic J, Pruner I, Damjanov N, Pekmezovic T, Sefik-Bukilica M, Antovic A. Impaired Fibrinolysis Is Linked With Digital Vasculopathy and Onset of New Digital Ulcers in Systemic Sclerosis. J Rheumatol 2022; 49:598-606. [PMID: 35169064 DOI: 10.3899/jrheum.210931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2022] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To assess thrombin generation, fibrin formation, and structure together with the fibrinolytic status in patients with systemic sclerosis (SSc) in relation to the occurrence of digital ulcers (DUs) during the course of disease. METHODS We studied variables of endothelial dysfunction, thrombin generation, overall hemostatic potential, and fibrin clot turbidity in plasma from 58 patients with SSc (39 with DU history and 19 DU-naïve) and 46 matched healthy controls (HCs). Fibrin structure was visualized using scanning electron microscopy (SEM). Finally, 39 patients with a history of DUs were followed for 1.5 years and the predictive value of all investigated markers for new DU onset was explored. RESULTS Significantly enhanced endogenous thrombin potential (ETP) and prolonged clot lysis time (CLT) were found in patients with DUs compared to HCs. CLT was prolonged in patients with DUs compared to those without, showing good validity in identifying DUs with an area under the curve of 0.7 (95% CI 0.6-0.8). The levels of ETP and intercellular adhesion molecule 1 were independently associated with CLT. Over the follow-up period, 20 patients developed new DUs. CLT was prolonged (P < 0.001) in patients with new DU episodes, especially those with recurrent DUs. Regression analysis showed that the Raynaud phenomenon visual analog scale and CLT were predictors of new DUs (OR 1.1, 95% CI 1.0-1.1 and OR 1.2, 95% CI 1.1-1.3, respectively). SEM confirmed denser fibrin clots in patients with new DUs. CONCLUSION Our results suggest that impaired fibrinolysis might have an emerging role in underlying digital vasculopathy and its progression in SSc.
Collapse
Affiliation(s)
- Jelena Colic
- J. Colic, MD, PhDc, Institute of Rheumatology, Belgrade, Serbia;
| | - Iva Pruner
- I. Pruner, PhD, Karolinska Institutet, Department of Molecular Medicine and Surgery, Stockholm, Sweden
| | - Nemanja Damjanov
- N. Damjanov, MD, PhD, M. Sefik-Bukilica, MD, PhD, Institute of Rheumatology, Belgrade, Serbia School of Medicine University of Belgrade, Belgrade, Serbia
| | - Tatjana Pekmezovic
- T. Pekmezovic, MD, PhD, Institute of Epidemiology, School of Medicine University of Belgrade, Belgrade, Serbia
| | - Mirjana Sefik-Bukilica
- N. Damjanov, MD, PhD, M. Sefik-Bukilica, MD, PhD, Institute of Rheumatology, Belgrade, Serbia School of Medicine University of Belgrade, Belgrade, Serbia
| | - Aleksandra Antovic
- A. Antovic, MD, PhD, Karolinska Institutet, Department of Medicine Division Solna of Rheumatology, Karolinska University Hospital, Rheumatology, Stockholm, Sweden
| |
Collapse
|
12
|
Manetti M, Rosa I, Fioretto BS, Matucci-Cerinic M, Romano E. Decreased Serum Levels of SIRT1 and SIRT3 Correlate with Severity of Skin and Lung Fibrosis and Peripheral Microvasculopathy in Systemic Sclerosis. J Clin Med 2022; 11:1362. [PMID: 35268452 PMCID: PMC8910971 DOI: 10.3390/jcm11051362] [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: 01/26/2022] [Revised: 02/22/2022] [Accepted: 02/28/2022] [Indexed: 02/01/2023] Open
Abstract
Systemic sclerosis (SSc, scleroderma) is a severe autoimmune connective tissue disease characterized by widespread peripheral microvasculopathy, and progressive cutaneous and visceral fibrosis, leading to significant organ dysfunction. Sirtuins (SIRTs) are a family of NAD-dependent protein deacetylases with pleiotropic effects on a variety of biological processes, including metabolism, cell survival, and aging. In the last decades, increasing studies have explored the contribution of SIRTs to the pathogenesis of SSc, highlighting a significant antifibrotic effect of both SIRT1 and SIRT3. On these bases, the aim of this study was to measure circulating SIRT1 and SIRT3 levels by enzyme-linked immune-sorbent assay in a well-characterized cohort of SSc patients (n = 80) and healthy controls (n = 71), focusing on their possible association with disease clinical features, and their potential as biomarkers reflecting SSc activity and severity. Significantly decreased serum levels of both SIRT1 and SIRT3 were found in SSc patients compared to controls. In SSc, the reduction in circulating SIRT1 and SIRT3 associated with a greater extent of cutaneous fibrosis, presence of interstitial lung disease, and worse pulmonary function. Serum SIRT1 and SIRT3 decrease also correlated with the severity of nailfold microvascular damage, with SIRT3 levels being additionally related to the occurrence of digital ulcers. The levels of these two proteins showed a direct correlation with one another in the circulation of SSc patients. Of the two SIRTs, serum SIRT3 was found to better reflect disease activity and severity in a logistic regression analysis model. Our findings suggest that serum SIRT1 and SIRT3 may represent novel potential biomarkers of increased risk for a more severe, life-threatening SSc disease course.
Collapse
Affiliation(s)
- Mirko Manetti
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (I.R.); (B.S.F.)
| | - Irene Rosa
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (I.R.); (B.S.F.)
| | - Bianca Saveria Fioretto
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (I.R.); (B.S.F.)
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (M.M.-C.); (E.R.)
| | - Marco Matucci-Cerinic
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (M.M.-C.); (E.R.)
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Hospital, 20132 Milan, Italy
| | - Eloisa Romano
- Division of Rheumatology, Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (M.M.-C.); (E.R.)
| |
Collapse
|
13
|
CİZMECİOGLU A, TEZCAN D, HAKBİLEN S, YİLMAZ S. Prolonged Capillary Refill Time Highlights Early Performing of Nailfold Capillaroscopy in Patients with Systemic Sclerosis. KONURALP TIP DERGISI 2022. [DOI: 10.18521/ktd.1050110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
14
|
Non-HLA Antibodies in Hand Transplant Recipients Are Connected to Multiple Acute Rejection Episodes and Endothelial Activation. J Clin Med 2022; 11:jcm11030833. [PMID: 35160284 PMCID: PMC8837026 DOI: 10.3390/jcm11030833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 02/01/2023] Open
Abstract
The role of anti-HLA antibodies in transplant rejection is well-known but the injury associated with non-HLA antibodies is now widely discussed. The aim of our study was to investigate a role of non-HLA antibodies in hand allografts rejection. The study was performed on six patients after hand transplantation. The control group consisted of: 12 kidney transplant recipients and 12 healthy volunteers. The following non-HLA antibodies were tested: antibody against angiotensin II type 1 receptor (AT1R-Ab), antibody against endothelin-1 type-A-receptor (ETAR-Ab), antibody against protease-activated receptor 1 (PAR-1-Ab) and anti-VEGF-A antibody (VEGF-A-Ab). Chosen proinflammatory cytokines (Il-1, IL-6, IFNγ) were used to evaluate the post-transplant humoral response. Laboratory markers of endothelial activation (VEGF, sICAM, vWF) were used to assess potential vasculopathy. The patient with the highest number of acute rejections had both positive non-HLA antibodies: AT1R-Ab and ETAR-Ab. The same patient had the highest VEGF-A-Ab and very high PAR1-Ab. All patients after hand transplantation had high levels of laboratory markers of endothelial activation. The existence of non-HLA antibodies together with multiple acute rejections observed in patient after hand transplantation should stimulate to look for potential role of non-HLA antibodies in humoral injury in vascular composite allotransplantation.
Collapse
|
15
|
Wienke J, Mertens JS, Garcia S, Lim J, Wijngaarde CA, Yeo JG, Meyer A, van den Hoogen LL, Tekstra J, Hoogendijk JE, Otten HG, Fritsch-Stork RDE, de Jager W, Seyger MMB, Thurlings RM, de Jong EMGJ, van der Kooi AJ, van der Pol WL, Arkachaisri T, Radstake TRDJ, van Royen-Kerkhof A, van Wijk F. Biomarker profiles of endothelial activation and dysfunction in rare systemic autoimmune diseases: implications for cardiovascular risk. Rheumatology (Oxford) 2021; 60:785-801. [PMID: 32810267 DOI: 10.1093/rheumatology/keaa270] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/19/2020] [Indexed: 01/08/2023] Open
Abstract
OBJECTIVES Vasculopathy is an important hallmark of systemic chronic inflammatory connective tissue diseases (CICTD) and is associated with increased cardiovascular risk. We investigated disease-specific biomarker profiles associated with endothelial dysfunction, angiogenic homeostasis and (tissue) inflammation, and their relation to disease activity in rare CICTD. METHODS A total of 38 serum proteins associated with endothelial (dys)function and inflammation were measured by multiplex-immunoassay in treatment-naive patients with localized scleroderma (LoS, 30), eosinophilic fasciitis (EF, 8) or (juvenile) dermatomyositis (34), 119 (follow-up) samples during treatment, and 65 controls. Data were analysed by unsupervised clustering, Spearman correlations, non-parametric t test and ANOVA. RESULTS The systemic CICTD, EF and dermatomyositis, had distinct biomarker profiles, with 'signature' markers galectin-9 (dermatomyositis) and CCL4, CCL18, CXCL9, fetuin, fibronectin, galectin-1 and TSP-1 (EF). In LoS, CCL18, CXCL9 and CXCL10 were subtly increased. Furthermore, dermatomyositis and EF shared upregulation of markers related to interferon (CCL2, CXCL10), endothelial activation (VCAM-1), inhibition of angiogenesis (angiopoietin-2, sVEGFR-1) and inflammation/leucocyte chemo-attraction (CCL19, CXCL13, IL-18, YKL-40), as well as disturbance of the Angiopoietin-Tie receptor system and VEGF-VEGFR system. These profiles were related to disease activity, and largely normalized during treatment. However, a subgroup of CICTD patients showed continued elevation of CXCL10, CXCL13, galectin-9, IL-18, TNFR2, VCAM-1, and/or YKL-40 during clinically inactive disease, possibly indicating subclinical interferon-driven inflammation and/or endothelial dysfunction. CONCLUSION CICTD-specific biomarker profiles revealed an anti-angiogenic, interferon-driven environment during active disease, with incomplete normalization under treatment. This warrants further investigation into monitoring of vascular biomarkers during clinical follow-up, or targeted interventions to minimize cardiovascular risk in the long term.
Collapse
Affiliation(s)
- Judith Wienke
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jorre S Mertens
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands.,Department of Dermatology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Samuel Garcia
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Johan Lim
- Department of Neurology, Amsterdam University Medical Centre, University of Amsterdam, Neuroscience Institute, Amsterdam, Netherlands
| | - Camiel A Wijngaarde
- Department of Neurology and Neurosurgery, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Joo Guan Yeo
- Rheumatology and Immunology Service, Department of Paediatric Subspecialties, KK Women's and Children's Hospital and Duke-NUS Medical School, Duke, NUS, Singapore.,Translational Immunology Institute, SingHealth-Academic Medical Centre, Duke, NUS, Singapore
| | - Alain Meyer
- Service de Physiologie et d'Explorations Fonctionnelles, Centre, de Référence des, Maladies Autoimmunes Rares, Rhumatologie, Institut de Physiologie, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, France
| | - Lucas L van den Hoogen
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Janneke Tekstra
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Jessica E Hoogendijk
- Department of Neurology and Neurosurgery, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Henny G Otten
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Ruth D E Fritsch-Stork
- Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands.,Sigmund Freud Private University, Vienna, Austria, Vienna, Austria.,Medizinische Abteilung Hanusch Krankenhaus und Ludwig Boltzmann Institut für Osteologie, Vienna, Austria
| | - Wilco de Jager
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Marieke M B Seyger
- Department of Dermatology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Rogier M Thurlings
- Department of Rheumatic Diseases, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Elke M G J de Jong
- Department of Dermatology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Anneke J van der Kooi
- Department of Neurology, Amsterdam University Medical Centre, University of Amsterdam, Neuroscience Institute, Amsterdam, Netherlands
| | - W Ludo van der Pol
- Department of Neurology and Neurosurgery, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Thaschawee Arkachaisri
- Rheumatology and Immunology Service, Department of Paediatric Subspecialties, KK Women's and Children's Hospital and Duke-NUS Medical School, Duke, NUS, Singapore
| | - Timothy R D J Radstake
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Rheumatology and Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Annet van Royen-Kerkhof
- Paediatric Rheumatology and Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Femke van Wijk
- Centre for Translational Immunology, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
16
|
Evaluation of Ocular Surface and Meibomian Glands in Patients With Scleroderma. Cornea 2020; 40:977-981. [PMID: 33079918 DOI: 10.1097/ico.0000000000002551] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/15/2020] [Indexed: 11/26/2022]
Abstract
PURPOSE To evaluate the prevalence of dry eye and meibomian gland dysfunction in patients with scleroderma. METHODS A total of 32 patients with scleroderma (study group) and 31 healthy subjects (control group) were enrolled. Besides routine ophthalmologic evaluation, Schirmer 1 test, tear break-up time, tear osmolarity, ocular surface staining with fluorescein (Oxford score), and Ocular Surface Disease Index (OSDI) score, meibomian gland dysfunction evaluation (foamy tears, telangiectasia of the eyelid, eyelid contour abnormalities, and meibomian plugs), and meibography were performed. RESULTS Sixty-four eyes of 32 patients with scleroderma and 62 eyes of 31 healthy individuals were evaluated. Mean ages of the study group was 48.34 ± 9.73 years (21-62 years) and of the control group was 45.84 ± 4.42 years (38-54 years) (P = 0.067). Mean duration of systemic sclerosis or scleroderma in study group was 9.78 ± 7.40 years (1-30 years). Mean tear break-up time was shorter in study group than that in the control group (P < 0.0005). No statistical differences were detected for osmolarity, OSDI score, and Schirmer 1 values between groups (P = 0.051, P = 0.053, and P = 0.358, respectively). The prevalence of grade 1 and higher Oxford score was higher in the study group (P < 0.0005). Upper meiboscores of grade 1 and higher were found to be more common in the study group than those in the control group (P = 0.036). The presence of foamy tears and telangiectasia of the eyelids were significantly higher in the study group (P = 0.002 and P = 0.002, respectively). OSDI score was the only significantly correlated data with disease duration (Spearman ρ coefficient = 0.396, P = 0.001). CONCLUSIONS Evaporative type dry eye is more common in patients with scleroderma than the healthy population.
Collapse
|
17
|
Mecoli CA, Shah AA, Boin F, Wigley FM, Hummers LK. The Utility of Plasma Vascular Biomarkers in Systemic Sclerosis: A Prospective Longitudinal Analysis. Arthritis Rheumatol 2020; 72:1341-1349. [PMID: 32200572 DOI: 10.1002/art.41265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 03/17/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE In cross-sectional studies, pulmonary hypertension (PH) and ischemic digital lesions are 2 scleroderma vascular outcomes associated with abnormalities in biomarkers of angiogenesis. The clinical usefulness of these biomarkers is unknown, in part due to lack of data on longitudinal measurement. This prospective longitudinal study was undertaken to evaluate vascular biomarker measurements in patients with systemic sclerosis (SSc) over time. METHODS We conducted a prospective cohort study of 300 patients with SSc who were followed up for at least a 5-year period and lacked evidence of PH and/or active ischemic digital lesions at enrollment. Levels of hepatocyte growth factor (HGF), soluble Flt-1 (sFlt-1), soluble endoglin, endostatin, and placental growth factor (PLGF) were obtained at multiple time points and assessed for their ability to predict the development of PH/ischemic digital lesions. Hazard ratios (HRs) with 95% confidence intervals (95% CIs) were calculated. RESULTS Forty-six patients (15%) developed PH and 69 patients (23%) developed an ischemic digital lesion. In time-to-event analyses, the following 3 biomarkers measured at cohort entry were found to be significantly associated with the development of PH: HGF (HR 1.99 [95% CI 1.24-3.17], P = 0.004), sFlt-1 (HR 3.04 [95% CI 1.29-7.14], P = 0.011), and PLGF (HR 2.74 [95% CI 1.32-5.69], P = 0.007). As time approaching PH diagnosis decreased, there was no corresponding increase in any biomarker level. Upon converting each continuous vascular biomarker into a binary variable, a dose-response relationship was observed for the number of elevated biomarkers at cohort entry and the risk of developing PH. With each additional elevated biomarker at cohort entry, there was a 78% increase in the risk of developing PH (HR 1.78 [95% CI 1.2-2.6], P = 0.004). CONCLUSION These data suggest that molecules involved in angiogenesis reflect vascular perturbation, and that elevations in these biomarkers at first encounter can indicate patients who are at risk of PH development.
Collapse
Affiliation(s)
| | - Ami A Shah
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | | | - Laura K Hummers
- Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
18
|
Systemic Sclerosis Serum Significantly Impairs the Multi-Step Lymphangiogenic Process: in Vitro Evidence. Int J Mol Sci 2019; 20:ijms20246189. [PMID: 31817940 PMCID: PMC6940874 DOI: 10.3390/ijms20246189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/22/2019] [Accepted: 12/06/2019] [Indexed: 01/29/2023] Open
Abstract
In systemic sclerosis (SSc), the possible involvement of lymphatic microcirculation and lymphangiogenesis has traditionally been overshadowed by the greater emphasis placed on dysfunctional blood vascular system and angiogenesis. In the present in vitro study, we explore for the first time whether the SSc microenvironment may interfere with lymphangiogenesis, a complex, multi-step process in which lymphatic microvascular endothelial cells (LMVECs) sprout, migrate, and proliferate to generate new lymphatic capillaries. Normal human adult dermal LMVECs from three donors were treated with serum from SSc patients (n = 8), serum from healthy individuals (n = 8), or recombinant human vascular endothelial growth factor (VEGF)-C as a positive control for lymphangiogenesis. Cell proliferation, Boyden chamber Matrigel chemoinvasion, wound healing capacity, and lymphatic capillary morphogenesis on Geltrex were assayed. VEGF-C serum levels were measured by enzyme-linked immunosorbent assay. Gene and protein expression levels of the lymphangiogenic orchestrators VEGF receptor-3 (VEGFR-3)/Flt-4 and neuropilin-2 (NRP-2) were determined by real-time PCR and Western blotting, respectively. Conditioning with SSc serum significantly inhibited LMVEC proliferation, Matrigel invasion, and wound healing capacity with respect to healthy serum. The ability of LMVECs to form lymphatic tubes on Geltrex was also severely compromised in the presence of SSc serum. VEGF-C levels were comparable in SSc and healthy sera. Treatment with SSc serum resulted in a significant downregulation of both VEGFR-3/Flt-4 and NRP-2 mRNA and protein levels. In SSc, the pathologic environment severely hampers every lymphangiogenesis step, likely through the reduction of pro-lymphangiogenic VEGFR-3/NRP-2 co-receptor signaling. The impairment of the lymphangiogenic process opens a new scenario underlying SSc vascular pathophysiology, which is worth investigating further.
Collapse
|
19
|
Th1- and Th17-Related Cytokines in Venous and Arterial Blood of Sclerodermic Patients with and without Digital Ulcers. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7908793. [PMID: 31687398 PMCID: PMC6800960 DOI: 10.1155/2019/7908793] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/10/2019] [Accepted: 08/30/2019] [Indexed: 01/01/2023]
Abstract
The earliest clinical manifestation of SSc is usually Raynaud's phenomenon, a small-arteries vasospasm driven by vascular tone dysregulation and microcirculatory abnormalities, resulting in digital ulcers (DU) in up to 50% of patients. Many cytokines as well as growth factors have been shown to play a role in promoting vascular smooth muscle cell proliferation and fibroblast activation, leading to ischemic damage as well as skin fibrosis. We aim to investigate a possible difference in venous and arterial blood levels of many cytokines (Th1- and Th17-related), GM-CSF, and endothelin-1 (ET1) in patients with and without DU. In the same patients, the correlations between capillary damage, evaluated by nailfold videocapillaroscopy (NVC), extension of skin fibrosis, calculated by modified Rodnan skin score (mRSS), and cytokines, ET-1, and GM-CSF levels were also measured. Patients with DU showed venous levels of IL-1β (p=0.024), IL-6 (p=0.012), IL-22(p=0.006), and TGF-β (p=0.046) significantly higher compared to arterial levels and arterial levels of GM-CSF and TNF-alpha significantly higher compared to venous levels (p < 0.001). NVC abnormalities were correlated with arterial TNFa and venous IL22, IL23, and IL17 levels and negatively correlated with venous ET-1 levels, whereas mRSS showed a negative correlation with IL-21(ρ = -0.427, p=0.050). The increased Th17-cytokine levels in venous compared to arterial blood of patients with DU suggest local cytokine production on ulcer site. The higher TNFa and GM-CSF levels in arterial blood of DU patients support the attempt to mitigate the hypoxic damage, and the correlation between Th17-cytokines, mRSS, NVC, and ET1 agrees with the potent profibrotic stimulus at the onset of the disease, which decreases as the SSc progresses.
Collapse
|
20
|
Multiple External Root Resorption of Teeth as a New Manifestation of Systemic Sclerosis-A Cross-Sectional Study in Japan. J Clin Med 2019; 8:jcm8101628. [PMID: 31590327 PMCID: PMC6832113 DOI: 10.3390/jcm8101628] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Multiple external root resorption (MERR) has been reported in systemic sclerosis (SSc) patients in Japan and Spain. To establish whether MERR is a new manifestation, we investigated the prevalence of MERR and systemic and oral manifestations to be associated with MERR in patients with SSc. METHODS Root resorption was detected by dental X-rays, panoramagraphy or cone beam computed tomography (CBCT). The prevalence of systemic and oral manifestations was examined by rheumatologists and dentists, respectively. Autoantibodies were investigated using laboratory tests. RESULTS MERR was detected in four out of the 41 patients (9.8%) who participated in the present study. The prevalence of digital ulcers was significantly higher in patients with MERR (MERR vs non-MERR, 75% vs 16.2%, p < 0.05), whereas that of other systemic manifestations was not. The prevalence of face skin sclerosis (100% vs 10.8%, p < 0.01), calcinosis at the facial region (75% vs 0%, p < 0.01), limited mouth opening (75% vs 18.9% p < 0.05), temporomandibular disorder symptoms (50% vs 2.7%, p < 0.05), and tongue rigidity (75% vs 2.7%, p < 0.05) was significantly higher in patients with MERR. CONCLUSION SSc patients with MERR had highly homogenous maxillofacial manifestations. Further clinical and basic studies are needed to elucidate the mechanisms underlying MERR in SSc patients.
Collapse
|
21
|
Szucs G, Szekanecz Z, Aszalos Z, Gesztelyi R, Zsuga J, Szodoray P, Kemeny-Beke A. A Wide Spectrum of Ocular Manifestations Signify Patients with Systemic Sclerosis. Ocul Immunol Inflamm 2019; 29:81-89. [PMID: 31577461 DOI: 10.1080/09273948.2019.1657467] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Objectives: Systemic sclerosis (SSc) is a rare, chronic connective tissue disease involving multiple organ systems, including the eye. We evaluated the detailed clinical ocular manifestations of outpatients with SSc.Methods: Demographics, disease duration and subtype, nailfold capillaroscopy (NFC) patterns and laboratory parameters encompassing the autoantibody profile of 51 SSc patients were evaluated, and a general ocular examination was performed for each participant.Results: Twenty-nine patients (56.86%) had eyelid skin alterations, 26 (50.98%) had retinal abnormalities, 26 (50.98%) had cataracts, 8 (15.69%) had conjunctival changes, 7 (13.73%) had iris abnormalities, 33 (64.71%) suffered from dry eye disease (DED), and 11 (21.57%) suffered from glaucoma. Significant positive correlations were found between NFC data and both tear breakup time and Ocular Surface Disease Index test values.Conclusions: Eyelid skin abnormalities, DED and retinal abnormalities are among the most common SSc-related ocular involvements. Diverse ophthalmic findings are attributed to the heterogeneity of SSc.
Collapse
Affiliation(s)
- Gabriella Szucs
- Department of Rheumatology, Faculty of Medicine, Institute of Medicine, Debrecen, Hungary
| | - Zoltan Szekanecz
- Department of Rheumatology, Faculty of Medicine, Institute of Medicine, Debrecen, Hungary
| | - Zsuzsa Aszalos
- Department of Immunology, Institute of Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Rudolf Gesztelyi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit Zsuga
- Department of Health Systems Management and Quality Management for Health Care, Faculty of Public Health, University of Debrecen, Debrecen, Hungary
| | - Peter Szodoray
- Department of Immunology, Rikshospitalet, Oslo University Hospital, Oslo, Norway
| | - Adam Kemeny-Beke
- Department of Ophthalmology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
22
|
Nagaraja V, Spino C, Bush E, Tsou PS, Domsic RT, Lafyatis R, Frech T, Gordon JK, Steen VD, Khanna D. A multicenter randomized, double-blind, placebo-controlled pilot study to assess the efficacy and safety of riociguat in systemic sclerosis-associated digital ulcers. Arthritis Res Ther 2019; 21:202. [PMID: 31481106 PMCID: PMC6724329 DOI: 10.1186/s13075-019-1979-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/16/2019] [Indexed: 11/10/2022] Open
Abstract
Background To determine the effect of riociguat, an oral, selective soluble guanylate cyclase stimulator, on the net digital ulcer (DU) burden in systemic sclerosis (SSc). Methods Participants with SSc-related active or painful indeterminate DUs were recruited in a multicenter, double-blind, randomized, placebo-controlled, proof-of-concept trial. Eligible participants were required to have at least one visible, active ischemic DU or painful indeterminate DU at screening, located at or distal to the proximal interphalangeal joint and that developed or worsened within 8 weeks prior to screening. Participants were randomized 1:1 to placebo or riociguat in individualized doses (maximum of 2.5 mg three times daily) during an 8-week titration period, followed by an 8-week stable dosing period. This was followed by an optional 16-week open-label extension phase for participants with active DU/reoccurrence of DUs within 1 month of the end of the main treatment phase. The primary endpoint was the change from baseline to week 16 in net ulcer burden (NUB), analyzed using ANCOVA. Other endpoints included plasma biomarkers and proportion of participants with treatment-emergent adverse events (AEs). Results Seventeen participants (eight placebo, nine riociguat) were randomized at five centers. Six participants in each group transitioned to the open-label extension. Baseline characteristics were comparable between the treatment groups, except participants randomized to placebo were older and had longer disease duration (p < 0.05). At baseline, the mean (SD) NUB was 2.5 (2.0) in the placebo and 2.4 (1.4) in the riociguat. No significant treatment difference was observed in the change from baseline to 16 weeks in NUB (adjusted mean treatment difference − 0.24, 95% CI (− 1.46, 0.99), p = 0.70). Four participants experienced five serious AE (four in riociguat and one in placebo); none was considered related to study medication. Statistically significant elevation of cGMP was observed at 16 weeks in the riociguat group (p = 0.05); no other biomarkers showed significant changes. In the open-label extension, participants in the riociguat-riociguat arm had complete healing of their DUs. Conclusion In participants with SSc-DU, treatment with riociguat did not reduce the number of DU net burden compared with placebo at 16 weeks. Open-label extension suggests that longer duration is needed to promote DU healing, which needs to be confirmed in a new trial. Trial registration ClinicalTrials.gov, NCT02915835. Registered on September 27, 2016. Electronic supplementary material The online version of this article (10.1186/s13075-019-1979-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vivek Nagaraja
- Division of Rheumatology/Department of Internal Medicine, University of Michigan Scleroderma Program, Suite 7C27, 300 North Ingalls Street, SPC 5422, Ann Arbor, MI, 48109, USA
| | - Cathie Spino
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Erica Bush
- Division of Rheumatology/Department of Internal Medicine, University of Michigan Scleroderma Program, Suite 7C27, 300 North Ingalls Street, SPC 5422, Ann Arbor, MI, 48109, USA
| | - Pei-Suen Tsou
- Division of Rheumatology/Department of Internal Medicine, University of Michigan Scleroderma Program, Suite 7C27, 300 North Ingalls Street, SPC 5422, Ann Arbor, MI, 48109, USA
| | - Robyn T Domsic
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Robert Lafyatis
- Division of Rheumatology and Clinical Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Tracy Frech
- Division of Rheumatology, University of Utah, Salt Lake City, UT, USA
| | - Jessica K Gordon
- Division of Rheumatology, Hospital of Special Surgery, New York, NY, USA
| | - Virginia D Steen
- Division of Rheumatology, Georgetown University Medical Center, Washington, DC, USA
| | - Dinesh Khanna
- Division of Rheumatology/Department of Internal Medicine, University of Michigan Scleroderma Program, Suite 7C27, 300 North Ingalls Street, SPC 5422, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
23
|
Mise au point sur les sclérodermies très précoces et précoces. Rev Med Interne 2019; 40:517-522. [DOI: 10.1016/j.revmed.2019.03.327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/11/2019] [Accepted: 03/21/2019] [Indexed: 01/29/2023]
|
24
|
Michalska-Jakubus M, Cutolo M, Smith V, Krasowska D. Imbalanced serum levels of Ang1, Ang2 and VEGF in systemic sclerosis: Integrated effects on microvascular reactivity. Microvasc Res 2019; 125:103881. [PMID: 31075243 DOI: 10.1016/j.mvr.2019.103881] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 02/03/2019] [Accepted: 05/04/2019] [Indexed: 11/30/2022]
Abstract
INTRODUCTION AND AIM Microangiopathy is a hallmark of systemic sclerosis (SSc). It is a progressive process from an early inflammatory and proangiogenic environment to insufficient microvascular repair with loss of microvessels. The exact underlying mechanisms remain ill-defined. Aim of the study was to investigate whether imbalanced angiopoietins/VEGF serum profile should be related in SSc to the altered microvascular reactivity characterized by aberrant angiogenesis and avascularity. MATERIALS AND METHODS Serum levels of Angiopoietin-1 (Ang1), Angiopoietin-2 (Ang2) and VEGF were measured by ELISA in 47 SSc patients and 27 healthy controls. Microvascular alterations were assessed by nailfold videocapillaroscopy (NVC). RESULTS Serum concentrations of Ang1 were significantly lower [mean (S.D.): 21516.04 (11,441.035) pg/ml], and Ang2 significantly increased [25,89.55 (934.225) pg/ml] in SSc as compared with the control group [Ang1: 28,457.08 (10,431.905) pg/ml; Ang2: 1556.23 (481.255) pg/ml, p < 0.01, respectively], whereas VEGF did not differ significantly. The ratios of Ang1/Ang2 and Ang1/VEGF were significantly lower in SSc patients (8.346 ± 4.523 and 95.17 ± 75.0, respectively) than in healthy subjects (17.612 ± 6.731 p < 0.000001 and 183.11 ± 137.73; p = 0.004]. Formation of giant capillaries with vascular leakage and collapse was associated with significant increase in VEGF and concomitant Ang1 deficiency. Capillary loss was related to significant increase in VEGF with respect to those with preserved capillary number (395.12 ± 256.27 pg/mL vs. 254.80 ± 213.61 pg/mL) whereas elevated Ang2 levels induced more advanced capillary damage as indicated by the presence of the "Late" NVC pattern. CONCLUSIONS We found that serum levels of Ang1, Ang2 and VEGF are differentially expressed in SSc and altered Ang1/Ang2 profile might underlay the aberrant angiogenesis in SSc despite increase in VEGF. For the first time we identified that significant deficiency of Ang1 might be involved in early capillary enlargement, followed by collapse and lack of stable newly-formed vessels in VEGF-enriched environment, whereas Ang2 levels seem to increase later in disease progression and advanced microvascular damage ("Late" NVC pattern).
Collapse
Affiliation(s)
- Małgorzata Michalska-Jakubus
- Department of Dermatology, Venereology and Paediatric Dermatology, Medical University of Lublin, Lublin, Poland.
| | - Maurizio Cutolo
- Research Laboratory and Academic Division of Clinical Rheumatology, Department of Internal Medicine, University of Genoa, Genoa, Italy.
| | - Vanessa Smith
- Faculty of Internal Medicine, Ghent University, Belgium.
| | - Dorota Krasowska
- Department of Dermatology, Venereology and Paediatric Dermatology, Medical University of Lublin, Lublin, Poland.
| |
Collapse
|
25
|
Bertolazzi C, Gallegos-Nava S, Villarreal-Treviño AV, Alfaro-Rodriguez A, Clavijo-Cornejo D, Gutierrez M. The current role of capillaroscopy in vasculitides. Clin Rheumatol 2019; 38:2299-2307. [DOI: 10.1007/s10067-018-4399-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/03/2018] [Accepted: 12/09/2018] [Indexed: 12/16/2022]
|
26
|
The correlation between serum E-selectin levels and soluble interleukin-2 receptors with relation to disease activity in localized scleroderma. Postepy Dermatol Alergol 2018; 35:614-619. [PMID: 30618531 PMCID: PMC6320493 DOI: 10.5114/ada.2018.77613] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/02/2018] [Indexed: 02/02/2023] Open
Abstract
Introduction Scleroderma is a chronic connective tissue disease resulting in fibrosis. Aim The aim of the study was to determine the connection between sE-selectin and sIL-2R and the severity of skin lesions in various subtypes of LoS. Evaluation of disease severity, the location of skin lesions, the duration of symptoms and disease activity were assessed in relation to the three different LoS subtypes in patients with localized scleroderma. Material and methods The study included 42 patients with localized scleroderma and the control group consisted of 41 healthy subjects. All patients in the LoS study group had a confirmed diagnosis via skin biopsy and underwent serology testing for sE-selectin and sIL-2R concentrations by enzyme-linked immunosorbent assay (ELISA). Results Significantly higher levels of sE-selectin and sIL-2R were observed in the LoS study group when compared with the control group (p < 0.001). The analysis showed a result close to statistical significance (p = 0.058) between sE-selectin concentration during the time of active disease in the LoS study group. The highest concentrations of sE-selectin and sIL-2R were observed in patients with the generalized subtype of LoS. A positive, statistically significant, curvilinear relationship was shown amid the modified Localized Skin Severity Index (mLoSSI) and sE-selectin and sIL-2R concentrations in the LoS study group. Conclusions Concentrations of the circulating form of sE-selectin appear to be an adequate marker of the endothelial function, positively correlating with the severity of the disease. The proven correlation of sIL-2R concentrations with the severity of the disease indicates that it is a valuable prognostic factor for predicting the impending course of the disease.
Collapse
|
27
|
Romano E, Manetti M, Rosa I, Fioretto BS, Ibba-Manneschi L, Matucci-Cerinic M, Guiducci S. Slit2/Robo4 axis may contribute to endothelial cell dysfunction and angiogenesis disturbance in systemic sclerosis. Ann Rheum Dis 2018; 77:1665-1674. [PMID: 30021803 DOI: 10.1136/annrheumdis-2018-213239] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/11/2018] [Accepted: 07/04/2018] [Indexed: 01/07/2023]
Abstract
OBJECTIVE In systemic sclerosis (SSc), early microvascular injury is followed by impaired angiogenesis and peripheral capillary loss. Here, we investigated the possible contribution of the neurovascular guidance molecule Slit2 and its Roundabout (Robo) receptors to SSc-related endothelial cell dysfunction. METHODS Circulating Slit2 levels were measured in patients with SSc and healthy controls. Slit2, Robo1 and Robo4 expression was investigated in SSc and healthy skin biopsies and explanted dermal microvascular endothelial cells (MVECs). Slit2/Robo4 function in MVEC angiogenesis was studied by cell viability, wound healing and capillary-like tube formation assays. RESULTS Circulating Slit2 was significantly increased in either SSc or patients with a very early diagnosis of SSc (VEDOSS) compared with controls. Interestingly, serum Slit2 levels were raised in patients with VEDOSS with nailfold videocapillaroscopy (NVC) abnormalities, while they were similar in VEDOSS with normal NVC and controls. In SSc, Slit2 and Robo4 expression was upregulated in clinically affected skin and explanted MVECs in respect to controls. The angiogenic performance of healthy MVECs was significantly reduced after challenge with recombinant human Slit2 or SSc sera. These inhibitory effects were significantly attenuated when SSc sera were preincubated with an anti-Slit2 blocking antibody. In vitro angiogenesis was severely compromised in SSc-MVECs and could be significantly ameliorated by Slit2 neutralisation or ROBO4 gene silencing. Slit2/Robo4 axis interfered with angiogenesis through the inhibition of Src kinase phosphorylation. CONCLUSIONS In SSc, increased circulating levels of Slit2 and activation of the Slit2/Robo4 antiangiogenic axis may contribute to peripheral microangiopathy since the very early phase of the disease.
Collapse
Affiliation(s)
- Eloisa Romano
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Scleroderma Unit, Azienda Ospedaliero-Universitaria Careggi (AOUC), Florence, Italy
| | - Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Irene Rosa
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Scleroderma Unit, Azienda Ospedaliero-Universitaria Careggi (AOUC), Florence, Italy
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Bianca Saveria Fioretto
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Scleroderma Unit, Azienda Ospedaliero-Universitaria Careggi (AOUC), Florence, Italy
| | - Lidia Ibba-Manneschi
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Scleroderma Unit, Azienda Ospedaliero-Universitaria Careggi (AOUC), Florence, Italy
| | - Serena Guiducci
- Department of Experimental and Clinical Medicine, Division of Rheumatology, University of Florence, Scleroderma Unit, Azienda Ospedaliero-Universitaria Careggi (AOUC), Florence, Italy
| |
Collapse
|
28
|
Benyamine A, Magalon J, Sabatier F, Lyonnet L, Robert S, Dumoulin C, Morange S, Mazodier K, Kaplanski G, Reynaud-Gaubert M, Rossi P, Dignat-George F, Granel B, Paul P. Natural Killer Cells Exhibit a Peculiar Phenotypic Profile in Systemic Sclerosis and Are Potent Inducers of Endothelial Microparticles Release. Front Immunol 2018; 9:1665. [PMID: 30072999 PMCID: PMC6058015 DOI: 10.3389/fimmu.2018.01665] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/04/2018] [Indexed: 12/22/2022] Open
Abstract
The pathophysiology of systemic sclerosis (SSc) involves early endothelial and immune activation, both preceding the onset of fibrosis. We previously identified soluble fractalkine and circulating endothelial microparticles (EMPs) as biomarkers of endothelial inflammatory activation in SSc. Fractalkine plays a dual role as a membrane-bound adhesion molecule expressed in inflamed endothelial cells (ECs) and as a chemokine involved in the recruitment, transmigration, and cytotoxic activation of immune cells that express CX3CR1, the receptor of fractalkine, namely CD8 and γδ T cells and natural killer (NK) cells. We aimed to quantify circulating cytotoxic immune cells and their expression of CX3CR1. We further investigated the expression profile of NK cells chemokine receptors and activation markers and the potential of NK cells to induce EC activation in SSc. We performed a monocentric study (NCT 02636127) enrolling 15 SSc patients [15 females, median age of 55 years (39–63), 11 limited cutaneous form and 4 diffuse] and 15 healthy controls. Serum fractalkine levels were significantly increased in SSc patients. Circulating CD8 T cells numbers were decreased in SSc patients with no difference in their CX3CR1 expression. Circulating γδ T cells and NK cells numbers were preserved. CX3CR1 expression in CD8 and γδ T cells did not differ between SSc patients and controls. The percentage and level of CX3CR1 expression in NK cells were significantly lowered in SSc patients. Percentages of CXCR4, NKG2D, CD69-expressing NK cells, and their expression levels were decreased in NK cells. Conversely, CD16 level expression and percentages of CD16+ NK cells were preserved. The exposure of human microvascular dermic EC line (HMVEC-d) to peripheral blood mononuclear cells resulted in similar NK cells degranulation activity in SSc patients and controls. We further showed that NK cells purified from the blood of SSc patients induced enhanced release of EMPs than NK cells from controls. This study evidenced a peculiar NK cells phenotype in SSc characterized by decreased chemokine and activation receptors expression, that might reflect NK cells involvement in the pathogenic process. It also highlighted the role of NK cells as a potent mechanism inducing endothelial activation through enhanced EMPs release.
Collapse
Affiliation(s)
- Audrey Benyamine
- Internal Medicine Department, Pôle MINC, Hôpital Nord, Assistance Publique Hôpitaux de Marseille (AP-HM), Marseille, France.,Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France
| | - Jérémy Magalon
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France.,Cell Therapy Unit, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France
| | - Florence Sabatier
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France.,Cell Therapy Unit, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France.,Hematology and Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France.,Centre d'Investigation clinique (CIC), Hôpital de la Conception, AP-HM, Marseille, France
| | - Luc Lyonnet
- Hematology and Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| | | | - Chloé Dumoulin
- Cell Therapy Unit, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France.,Hematology and Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Sophie Morange
- Centre d'Investigation clinique (CIC), Hôpital de la Conception, AP-HM, Marseille, France
| | - Karin Mazodier
- Internal Medicine and Clinical Immunology Department, Pôle MINC, Hôpital de la Conception, AP-HM, Marseille, France
| | - Gilles Kaplanski
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France.,Internal Medicine and Clinical Immunology Department, Pôle MINC, Hôpital de la Conception, AP-HM, Marseille, France
| | | | - Pascal Rossi
- Internal Medicine Department, Pôle MINC, Hôpital Nord, Assistance Publique Hôpitaux de Marseille (AP-HM), Marseille, France.,Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France
| | - Françoise Dignat-George
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France.,Hematology and Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| | - Brigitte Granel
- Internal Medicine Department, Pôle MINC, Hôpital Nord, Assistance Publique Hôpitaux de Marseille (AP-HM), Marseille, France.,Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France
| | - Pascale Paul
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France.,Cell Therapy Unit, Hôpital de la Conception, AP-HM, INSERM CIC BT 1409, Marseille, France.,Hematology and Vascular Biology Department, Hôpital de la Conception, AP-HM, Marseille, France
| |
Collapse
|
29
|
Hearing loss in patients with scleroderma: associations with clinical manifestations and capillaroscopy. Clin Rheumatol 2018; 37:2439-2446. [PMID: 29860565 DOI: 10.1007/s10067-018-4162-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/18/2018] [Accepted: 05/28/2018] [Indexed: 10/14/2022]
Abstract
Systemic sclerosis is a multi-systemic disease with widespread small-vessel vasculopathy and fibrosis. Involvement of the middle and inner ear and hearing loss has been reported as an uncommon manifestation of scleroderma in some studies. In this study, we evaluated hearing problems in scleroderma patients and determined its association with clinical manifestations and capillaroscopy. We evaluated 54 patients with scleroderma referred to Hafez Hospital clinic of scleroderma related to Shiraz University of Medical Science; they fulfilled the LeRoy and ACR/EULAR criteria for scleroderma. Control group consisted of 60 normal individuals. All clinical manifestations, nail fold capillaroscopy, pure tone audiometry, speech reception threshold, and speech audiometry were recorded during evaluation. Subjective hearing loss and objective hearing loss were seen in 10 and 36 patients of the case group (18.5%, 66.7%) and 6 and 10 of the control group (10%, 28.3%) (P values 0.03, < 0.001). Sensorineural hearing loss, abnormal pure tone audiometry, and abnormal speech reception threshold were more common in scleroderma patients compared to the control group (P values of < 0.001, < 0.001, and < 0.001). There was no correlation between objective hearing loss and type of scleroderma, duration of disease, skin score, interstitial lung disease, digital ulcer, gastrointestinal involvement, or nail fold capillaroscopy patterns (all P values > 0.05). In our study, subjective and objective hearing loss were higher in patients with scleroderma compared to the control group and also sensorineural hearing loss, abnormal pure tone audiometry, and abnormal speech reception threshold. There was no correlation between objective hearing loss and clinical manifestations or capillaroscopy findings.
Collapse
|
30
|
Xu J, Rosen M, Luffman CI, Law C, Laloo A, Rosen S, Faulkner-Jones BE. The Convergence of Vasculopathy and Vasculitis: Computer Mapping Analysis of 2 Renal Biopsies in a Patient with both Systemic Sclerosis and ANCA-Related Vasculitis. Case Rep Nephrol Dial 2018; 8:35-44. [PMID: 29692989 PMCID: PMC5903098 DOI: 10.1159/000487262] [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: 11/21/2017] [Accepted: 01/28/2018] [Indexed: 11/19/2022] Open
Abstract
Scleroderma vasculopathy and ANCA (antineutrophil cytoplasmic antibodies)-associated glomerulonephritis have rarely been reported to occur simultaneously in one patient. Herein, we report a patient who presented with a classic constellation of clinical and laboratory findings of systemic scleroderma and was subsequently found to be positive for p-ANCA. Two renal biopsies, performed 5 months apart, demonstrated typical changes of the two entities in both acute and "healed" phases, which were analyzed by computer mapping techniques. The two renal biopsies were serially sectioned and stained routinely, and with CD31 and CD34 as endothelial markers. The slides were digitized, aligned and analyzed. Each glomerular tuft was sequentially studied in terms of total area (µm2) and each biopsy was individually profiled. All arterial vessels were sequentially studied with whole vessel and luminal areas delineated and ratios calculated. The initial biopsy contained 32 glomeruli almost all with extensive fibrinoid necrosis and destruction of the capillary network. The arterial vessels (interlobular and arcuate) showed intimal edema with luminal occlusion. CD31/CD34 stains showed variable endothelial intactness but demonstrated the luminal size shifts. The second biopsy had 37 glomeruli that were either segmentally or globally sclerotic with no active changes. The vessels were now normally patent. Each glomerular tuft and arterial vessel in both biopsies was analyzed as a serial section histogram documenting these changes. These studies depict the rare occurrence of two entities together, the scleroderma kidney vasculopathy and the glomerulonephritis of ANCA-associated vasculitis syndrome both in an acute and healing phase, profiled by computer mapping techniques.
Collapse
Affiliation(s)
- Jia Xu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Milan Rosen
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Christina I Luffman
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Anita Laloo
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Seymour Rosen
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Beverly E Faulkner-Jones
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
31
|
Gigante A, Navarini L, Margiotta D, Barbano B, Afeltra A, Rosato E. In systemic sclerosis, microvascular and hands digital arteries damage correlates with serum levels of endostatin. Microcirculation 2018. [DOI: 10.1111/micc.12449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Antonietta Gigante
- Department of Clinical Medicine; Scleroderma Unit; Sapienza University of Rome; Rome Italy
| | - Luca Navarini
- Department of Immuno-Rheumatology Unit; Campus Bio-Medico University of Rome; Rome Italy
| | - Domenico Margiotta
- Department of Immuno-Rheumatology Unit; Campus Bio-Medico University of Rome; Rome Italy
| | - Biagio Barbano
- Department of Clinical Medicine; Scleroderma Unit; Sapienza University of Rome; Rome Italy
| | - Antonella Afeltra
- Department of Immuno-Rheumatology Unit; Campus Bio-Medico University of Rome; Rome Italy
| | - Edoardo Rosato
- Department of Clinical Medicine; Scleroderma Unit; Sapienza University of Rome; Rome Italy
| |
Collapse
|
32
|
Cutolo M, Melsens K, Wijnant S, Ingegnoli F, Thevissen K, De Keyser F, Decuman S, Müller-Ladner U, Piette Y, Riccieri V, Ughi N, Vandecasteele E, Vanhaecke A, Smith V. Nailfold capillaroscopy in systemic lupus erythematosus: A systematic review and critical appraisal. Autoimmun Rev 2018; 17:344-352. [PMID: 29427827 DOI: 10.1016/j.autrev.2017.11.025] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 11/02/2017] [Indexed: 01/03/2023]
Abstract
Nailfold capillaroscopy is an easy, non-invasive technique to assess microvascular involvement in rheumatic diseases. Multiple studies describe capillaroscopic changes in systemic lupus erythematosus (SLE), including a wide range of non-specific findings. On behalf of the European League Against Rheumatism (EULAR) study group on microcirculation in rheumatic diseases, a systematic review was done to obtain all original research studies (in English) in which SLE patients had capillaroscopy. Forty such studies are identified. This article firstly provides a résumé of the results of these studies according to capillaroscopic parameters (density, dimensions, morphology, haemorrhages), semi-quantitative assessment and qualitative assessment of capillaroscopy in SLE patients. Secondly, the correlations between capillaroscopic parameters in SLE patients and clinical and laboratory parameters (including auto-immune parameters) are outlined. The following capillaroscopic parameters are found to be significantly more prevalent in SLE patients compared to healthy controls: tortuous capillaries, abnormal morphology and haemorrhages. Hairpin-shaped capillaries are significantly less prevalent than in healthy persons. The semi-quantitatively determined nailfold capillaroscopic score (NFC score) in SLE patients is also higher than in healthy controls. Several correlations between clinical and laboratory parameters and capillaroscopic parameters are identified in the review. Disease activity is correlated with NFC score in seven studies, with abnormal morphology (i.e. "meandering") in one study and with haemorrhages in one study. Frequent attacks of Raynaud's phenomenon (RP) and gangrene are significantly correlated with dilated capillaries. In two studies a possible correlation between anti-SSA antibodies and lower density of capillaries is withheld. About other immune parameters conflicting results are found. In one study a significant negative correlation is found between 24-hour proteinuria and abnormal morphology (i.e. "meandering"). For the first time, an overview of the nailfold capillaroscopic changes that have been described in SLE and their correlations with clinical and laboratory findings is given. Further large-scale research on the identification of capillaroscopic changes in SLE and their correlations with standardised clinical and laboratory parameters, is ongoing at the EULAR study group on microcirculation in rheumatic diseases.
Collapse
Affiliation(s)
- Maurizio Cutolo
- Research Laboratory and Academic Unit of Clinical Rheumatology, Department of Internal Medicine, University of Genoa, Via Balbi 5, Genoa, Italy.
| | - Karin Melsens
- Department of Rheumatology, Ghent University Hospital, C. Heymanslaan 10, Ghent, Belgium; Department of Internal Medicine, Ghent University, C. Heymanslaan 10, Ghent, Belgium.
| | - Sara Wijnant
- Department of Internal Medicine, Ghent University, C. Heymanslaan 10, Ghent, Belgium.
| | - Francesca Ingegnoli
- Division of Rheumatology, ASST Gaetano Pini, Department of Clinical Sciences and Community Health, University of Milan, Piazza Cardinal Andrea Ferrari 1, Milan, Italy.
| | - Kristof Thevissen
- Department of Rheumatology, Ghent University Hospital, C. Heymanslaan 10, Ghent, Belgium.
| | - Filip De Keyser
- Department of Rheumatology, Ghent University Hospital, C. Heymanslaan 10, Ghent, Belgium; Department of Internal Medicine, Ghent University, C. Heymanslaan 10, Ghent, Belgium.
| | - Saskia Decuman
- Department of Internal Medicine, Ghent University, C. Heymanslaan 10, Ghent, Belgium.
| | - Ulf Müller-Ladner
- Rheumatology and Clinical Immunology, University of Giessen/Kerckhoff-Klinik, Benekestraße 2-8, 61231 Bad Nauheim, Germany.
| | - Yves Piette
- Department of Rheumatology, Ghent University Hospital, C. Heymanslaan 10, Ghent, Belgium.
| | - Valeria Riccieri
- Department of Internal Medicine and Clinical Specialities, Sapienza University, Piazzale Aldo Moro 5, Rome, Italy.
| | - Nicola Ughi
- Division of Rheumatology, ASST Gaetano Pini, Department of Clinical Sciences and Community Health, University of Milan, Piazza Cardinal Andrea Ferrari 1, Milan, Italy
| | - Els Vandecasteele
- Department of Cardiology, Ghent University Hospital, C. Heymanslaan 10, Ghent, Belgium; Department of Internal Medicine, Ghent University, C. Heymanslaan 10, Ghent, Belgium.
| | - Amber Vanhaecke
- Department of Rheumatology, Ghent University Hospital, C. Heymanslaan 10, Ghent, Belgium; Department of Internal Medicine, Ghent University, C. Heymanslaan 10, Ghent, Belgium.
| | - Vanessa Smith
- Department of Rheumatology, Ghent University Hospital, C. Heymanslaan 10, Ghent, Belgium; Department of Internal Medicine, Ghent University, C. Heymanslaan 10, Ghent, Belgium.
| |
Collapse
|
33
|
Allanore Y, Distler O, Matucci-Cerinic M, Denton CP. Review: Defining a Unified Vascular Phenotype in Systemic Sclerosis. Arthritis Rheumatol 2018; 70:162-170. [DOI: 10.1002/art.40377] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/10/2017] [Indexed: 02/06/2023]
Affiliation(s)
- Yannick Allanore
- Cochin Hospital; INSERM U1016; Paris Descartes University; Paris France
| | | | | | | |
Collapse
|
34
|
Markusse IM, Meijs J, de Boer B, Bakker JA, Schippers HPC, Schouffoer AA, Ajmone Marsan N, Kroft LJM, Ninaber MK, Huizinga TWJ, de Vries-Bouwstra JK. Predicting cardiopulmonary involvement in patients with systemic sclerosis: complementary value of nailfold videocapillaroscopy patterns and disease-specific autoantibodies. Rheumatology (Oxford) 2017; 56:1081-1088. [PMID: 27940596 DOI: 10.1093/rheumatology/kew402] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Indexed: 11/13/2022] Open
Abstract
Objective To evaluate the prevalence of anti-extractable nuclear antigen (anti-ENA) antibodies in Dutch SSc patients and the predictive power of the combination of specific anti-ENA antibodies and nailfold videocapillaroscopy (NVC) patterns to improve identification of patients with high risk for cardiopulmonary involvement. Methods A total of 287 patients (79%) from the Leiden SSc-Cohort had data available on NVC-pattern (no SSc-specific, early, active, late) and anti-ENA antibodies. Associations between anti-ENA/NVC combinations with cardiopulmonary parameters were explored using logistic regression. Results Prevalence of ACA was 37%, anti-Scl-70 24%, anti-RNP 9%, anti-RNAPIII 5%, anti-fibrillarin 4%, anti-Pm/Scl 3%, anti-Th/To 0.3% and anti-Ku 1.4%. NVC showed a SSc-specific pattern in 88%: 10% early, 42% active and 36% late. The prevalence of different NVC patterns was equally distributed among specific anti-ENA antibodies, except for the absence of early pattern in anti-RNP positive patients. Fifty-one percent had interstitial lung disease (ILD), 59% had decreased diffusion capacity for carbon monoxide and 16% systolic pulmonary artery pressure >35 mmHg (sPAP↑). Regardless of ENA-subtype, NVC-pattern showed a stable association with presence of ILD or sPAP↑. For ILD, the odds ratios (ORs) were 1.3-1.4 ( P < 0.05 for analyses with anti-RNAPIII, anti-RNP). For diffusion capacity for carbon monoxide, the OR was 1.5 ( P < 0.05 for analyses with ACA, anti-Scl-70, anti-RNAPIII, anti-RNP). For sPAP↑, the ORs were 2.2-2.4 ( P < 0.05 for analyses with anti-RNAPIII, anti-RNP). Conclusion In Dutch SSc patients, all SSc-specific auto-antibodies were found, with ACA and anti-Scl-70 being the most prevalent. Strikingly, the association between NVC-pattern and heart/lung involvement was independent of specific anti-ENA antibodies, which might indicate microangiopathy is an important cause of organ involvement.
Collapse
Affiliation(s)
| | | | | | - Jaap A Bakker
- Department of Clinical Chemistry and Laboratory Medicine
| | | | | | | | | | - Maarten K Ninaber
- Department of Pulmonology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | |
Collapse
|
35
|
Measurement of Serum Klotho in Systemic Sclerosis. DISEASE MARKERS 2017; 2017:9545930. [PMID: 28912623 PMCID: PMC5585626 DOI: 10.1155/2017/9545930] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 07/03/2017] [Accepted: 07/12/2017] [Indexed: 01/05/2023]
Abstract
Background The aim of our study was to evaluate the serum concentration of klotho in a cohort of systemic sclerosis (SSc) patients compared to that of healthy controls and to correlate its levels with the degree and the kind of organ involvement. Methods Blood samples obtained from both patients and controls were collected and analysed by an ELISA test for the determination of human soluble klotho. Scleroderma patients were evaluated for disease activity through clinical, laboratory, and instrumental assessment. Results Our cohort consisted of 81 SSc patients (74 females, mean age 63.9 ± 13.1 years) and 136 healthy controls (78 females, mean age 50.5 ± 10.7 years). When matched for age, serum klotho concentration significantly differed between controls and patients (p < 0.001). However, in SSc patients, we did not find any significant association between serum klotho and clinical, laboratory, and instrumental findings. Lower serum levels of klotho were detected in 4 patients who were anticitrullinated peptide antibody (ACPA) positive (p = 0.005). Conclusions Our data show a lower concentration of klotho in the serum of SSc patients compared to that of healthy controls, without any significant association with clinical manifestations and laboratory and instrumental findings. The association between serum klotho and ACPA positivity requires further investigation.
Collapse
|
36
|
Manetti M, Pratesi S, Romano E, Bellando-Randone S, Rosa I, Guiducci S, Fioretto BS, Ibba-Manneschi L, Maggi E, Matucci-Cerinic M. Angiogenic T cell expansion correlates with severity of peripheral vascular damage in systemic sclerosis. PLoS One 2017; 12:e0183102. [PMID: 28797111 PMCID: PMC5552290 DOI: 10.1371/journal.pone.0183102] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 07/29/2017] [Indexed: 11/23/2022] Open
Abstract
The mechanisms underlying endothelial cell injury and defective vascular repair in systemic sclerosis (SSc) remain unclear. Since the recently discovered angiogenic T cells (Tang) may have an important role in the repair of damaged endothelium, this study aimed to analyze the Tang population in relation to disease-related peripheral vascular features in SSc patients. Tang (CD3+CD31+CXCR4+) were quantified by flow cytometry in peripheral blood samples from 39 SSc patients and 18 healthy controls (HC). Circulating levels of the CXCR4 ligand stromal cell-derived factor (SDF)-1α and proangiogenic factors were assessed in paired serum samples by immunoassay. Serial skin sections from SSc patients and HC were subjected to CD3/CD31 and CD3/CXCR4 double immunofluorescence. Circulating Tang were significantly increased in SSc patients with digital ulcers (DU) compared either with SSc patients without DU or with HC. Tang levels were significantly higher in SSc patients with late nailfold videocapillaroscopy (NVC) pattern than in those with early/active NVC patterns and in HC. No difference in circulating Tang was found when comparing either SSc patients without DU or patients with early/active NVC patterns and HC. In SSc peripheral blood, Tang percentage was inversely correlated to levels of SDF-1α and CD34+CD133+VEGFR-2+ endothelial progenitor cells (EPC), and positively correlated to levels of vascular endothelial growth factor and matrix metalloproteinase-9. Tang were frequently detected in SSc dermal perivascular inflammatory infiltrates. In summary, our findings demonstrate for the first time that Tang cells are selectively expanded in the circulation of SSc patients displaying severe peripheral vascular complications like DU. In SSc, Tang may represent a potentially useful biomarker reflecting peripheral vascular damage severity. Tang expansion may be an ineffective attempt to compensate the need for increased angiogenesis and EPC function. Further studies are required to clarify the function of Tang cells and investigate the mechanisms responsible for their change in SSc.
Collapse
Affiliation(s)
- Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
- * E-mail:
| | - Sara Pratesi
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy
| | - Eloisa Romano
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy
| | - Silvia Bellando-Randone
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy
| | - Irene Rosa
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy
| | - Serena Guiducci
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy
| | - Bianca Saveria Fioretto
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy
| | - Lidia Ibba-Manneschi
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Enrico Maggi
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Section of Internal Medicine, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy
| |
Collapse
|
37
|
Soulaidopoulos S, Triantafyllidou E, Garyfallos A, Kitas GD, Dimitroulas T. The role of nailfold capillaroscopy in the assessment of internal organ involvement in systemic sclerosis: A critical review. Autoimmun Rev 2017; 16:787-795. [DOI: 10.1016/j.autrev.2017.05.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 05/01/2017] [Indexed: 10/19/2022]
|
38
|
Matsushita T, Takehara K. An update on biomarker discovery and use in systemic sclerosis. Expert Rev Mol Diagn 2017; 17:823-833. [DOI: 10.1080/14737159.2017.1356722] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Takashi Matsushita
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Kazuhiko Takehara
- Department of Dermatology, Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
39
|
Chora I, Romano E, Manetti M, Mazzotta C, Costa R, Machado V, Cortez A, Bruni C, Lepri G, Guiducci S, De Paulis A, Soares R, Matucci-Cerinic M. Evidence for a Derangement of the Microvascular System in Patients with a Very Early Diagnosis of Systemic Sclerosis. J Rheumatol 2017; 44:1190-1197. [PMID: 28507177 DOI: 10.3899/jrheum.160791] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2017] [Indexed: 01/03/2023]
Abstract
OBJECTIVE To investigate whether patients with a very early diagnosis of systemic sclerosis (VEDOSS) may already present circulating markers and in vitro signs of microvascular dysfunction. METHODS Serum samples were obtained from 55 patients with systemic sclerosis (SSc), 25 patients with VEDOSS, and 55 matched healthy controls (HC). Serum levels of pan-vascular endothelial growth factor (VEGF) and soluble neuropilin-1 (sNRP-1) were measured by ELISA. Human dermal microvascular endothelial cells (H-MVEC) were cultured and stimulated with SSc, VEDOSS, and HC sera. Protein expression of NRP-1 was analyzed by Western blotting, cell proliferation by 5'-bromodeoxyuridine assay, migration capacity by wound-healing assay, and capillary-like tube formation by Matrigel assay. RESULTS Serum levels of pan-VEGF were increased in patients with VEDOSS and SSc versus HC (p = 0.05 and p = 0.003, respectively). Serum levels of sNRP-1 were significantly reduced in patients with VEDOSS and SSc compared with controls (p = 0.012 and p = 0.027, respectively). NRP-1 expression was decreased in H-MVEC stimulated with VEDOSS sera (p < 0.001 vs HC). Proliferation was reduced in H-MVEC stimulated either with VEDOSS or SSc sera in comparison with HC sera (p = 0.015 and p = 0.043, respectively). Wound healing was compromised in H-MVEC stimulated with VEDOSS and SSc sera versus HC sera (p < 0.01 for both). Capillarogenesis was decreased in H-MVEC stimulated with VEDOSS sera (p < 0.01) and SSc sera (p < 0.001) compared with cells stimulated with HC sera. CONCLUSION Similar to patients with SSc, patients with VEDOSS already present biological signs of endothelial dysfunction. Our data demonstrate that VEDOSS sera significantly modify endothelial cell behavior and impair the angiogenic potential of the microvascular system.
Collapse
Affiliation(s)
- Inês Chora
- From the Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro; I3S, Instituto de Investigação e Inovação em Saúde, University of Porto; Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; Nobre Laboratory, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence; Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence; Department of Translational Medical Sciences, Centre for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy. .,I. Chora, MD, Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro, and I3S, University of Porto; E. Romano, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; M. Manetti, PhD, Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence; C. Mazzotta, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; R. Costa, MSc, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; V. Machado, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; A. Cortez, MSc, Nobre Laboratory, Faculty of Medicine, University of Porto; C. Bruni, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; G. Lepri, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; S. Guiducci, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; A. De Paulis, MD, Department of Translational Medical Sciences, CISI, University of Naples Federico II; R. Soares, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; M. Matucci-Cerinic, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence.
| | - Eloisa Romano
- From the Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro; I3S, Instituto de Investigação e Inovação em Saúde, University of Porto; Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; Nobre Laboratory, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence; Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence; Department of Translational Medical Sciences, Centre for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,I. Chora, MD, Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro, and I3S, University of Porto; E. Romano, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; M. Manetti, PhD, Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence; C. Mazzotta, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; R. Costa, MSc, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; V. Machado, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; A. Cortez, MSc, Nobre Laboratory, Faculty of Medicine, University of Porto; C. Bruni, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; G. Lepri, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; S. Guiducci, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; A. De Paulis, MD, Department of Translational Medical Sciences, CISI, University of Naples Federico II; R. Soares, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; M. Matucci-Cerinic, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence
| | - Mirko Manetti
- From the Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro; I3S, Instituto de Investigação e Inovação em Saúde, University of Porto; Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; Nobre Laboratory, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence; Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence; Department of Translational Medical Sciences, Centre for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,I. Chora, MD, Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro, and I3S, University of Porto; E. Romano, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; M. Manetti, PhD, Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence; C. Mazzotta, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; R. Costa, MSc, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; V. Machado, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; A. Cortez, MSc, Nobre Laboratory, Faculty of Medicine, University of Porto; C. Bruni, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; G. Lepri, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; S. Guiducci, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; A. De Paulis, MD, Department of Translational Medical Sciences, CISI, University of Naples Federico II; R. Soares, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; M. Matucci-Cerinic, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence
| | - Celestina Mazzotta
- From the Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro; I3S, Instituto de Investigação e Inovação em Saúde, University of Porto; Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; Nobre Laboratory, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence; Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence; Department of Translational Medical Sciences, Centre for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,I. Chora, MD, Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro, and I3S, University of Porto; E. Romano, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; M. Manetti, PhD, Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence; C. Mazzotta, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; R. Costa, MSc, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; V. Machado, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; A. Cortez, MSc, Nobre Laboratory, Faculty of Medicine, University of Porto; C. Bruni, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; G. Lepri, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; S. Guiducci, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; A. De Paulis, MD, Department of Translational Medical Sciences, CISI, University of Naples Federico II; R. Soares, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; M. Matucci-Cerinic, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence
| | - Raquel Costa
- From the Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro; I3S, Instituto de Investigação e Inovação em Saúde, University of Porto; Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; Nobre Laboratory, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence; Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence; Department of Translational Medical Sciences, Centre for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,I. Chora, MD, Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro, and I3S, University of Porto; E. Romano, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; M. Manetti, PhD, Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence; C. Mazzotta, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; R. Costa, MSc, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; V. Machado, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; A. Cortez, MSc, Nobre Laboratory, Faculty of Medicine, University of Porto; C. Bruni, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; G. Lepri, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; S. Guiducci, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; A. De Paulis, MD, Department of Translational Medical Sciences, CISI, University of Naples Federico II; R. Soares, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; M. Matucci-Cerinic, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence
| | - Vera Machado
- From the Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro; I3S, Instituto de Investigação e Inovação em Saúde, University of Porto; Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; Nobre Laboratory, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence; Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence; Department of Translational Medical Sciences, Centre for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,I. Chora, MD, Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro, and I3S, University of Porto; E. Romano, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; M. Manetti, PhD, Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence; C. Mazzotta, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; R. Costa, MSc, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; V. Machado, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; A. Cortez, MSc, Nobre Laboratory, Faculty of Medicine, University of Porto; C. Bruni, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; G. Lepri, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; S. Guiducci, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; A. De Paulis, MD, Department of Translational Medical Sciences, CISI, University of Naples Federico II; R. Soares, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; M. Matucci-Cerinic, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence
| | - Alice Cortez
- From the Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro; I3S, Instituto de Investigação e Inovação em Saúde, University of Porto; Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; Nobre Laboratory, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence; Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence; Department of Translational Medical Sciences, Centre for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,I. Chora, MD, Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro, and I3S, University of Porto; E. Romano, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; M. Manetti, PhD, Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence; C. Mazzotta, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; R. Costa, MSc, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; V. Machado, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; A. Cortez, MSc, Nobre Laboratory, Faculty of Medicine, University of Porto; C. Bruni, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; G. Lepri, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; S. Guiducci, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; A. De Paulis, MD, Department of Translational Medical Sciences, CISI, University of Naples Federico II; R. Soares, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; M. Matucci-Cerinic, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence
| | - Cosimo Bruni
- From the Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro; I3S, Instituto de Investigação e Inovação em Saúde, University of Porto; Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; Nobre Laboratory, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence; Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence; Department of Translational Medical Sciences, Centre for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,I. Chora, MD, Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro, and I3S, University of Porto; E. Romano, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; M. Manetti, PhD, Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence; C. Mazzotta, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; R. Costa, MSc, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; V. Machado, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; A. Cortez, MSc, Nobre Laboratory, Faculty of Medicine, University of Porto; C. Bruni, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; G. Lepri, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; S. Guiducci, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; A. De Paulis, MD, Department of Translational Medical Sciences, CISI, University of Naples Federico II; R. Soares, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; M. Matucci-Cerinic, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence
| | - Gemma Lepri
- From the Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro; I3S, Instituto de Investigação e Inovação em Saúde, University of Porto; Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; Nobre Laboratory, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence; Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence; Department of Translational Medical Sciences, Centre for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,I. Chora, MD, Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro, and I3S, University of Porto; E. Romano, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; M. Manetti, PhD, Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence; C. Mazzotta, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; R. Costa, MSc, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; V. Machado, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; A. Cortez, MSc, Nobre Laboratory, Faculty of Medicine, University of Porto; C. Bruni, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; G. Lepri, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; S. Guiducci, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; A. De Paulis, MD, Department of Translational Medical Sciences, CISI, University of Naples Federico II; R. Soares, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; M. Matucci-Cerinic, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence
| | - Serena Guiducci
- From the Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro; I3S, Instituto de Investigação e Inovação em Saúde, University of Porto; Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; Nobre Laboratory, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence; Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence; Department of Translational Medical Sciences, Centre for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,I. Chora, MD, Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro, and I3S, University of Porto; E. Romano, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; M. Manetti, PhD, Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence; C. Mazzotta, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; R. Costa, MSc, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; V. Machado, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; A. Cortez, MSc, Nobre Laboratory, Faculty of Medicine, University of Porto; C. Bruni, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; G. Lepri, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; S. Guiducci, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; A. De Paulis, MD, Department of Translational Medical Sciences, CISI, University of Naples Federico II; R. Soares, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; M. Matucci-Cerinic, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence
| | - Amato De Paulis
- From the Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro; I3S, Instituto de Investigação e Inovação em Saúde, University of Porto; Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; Nobre Laboratory, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence; Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence; Department of Translational Medical Sciences, Centre for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,I. Chora, MD, Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro, and I3S, University of Porto; E. Romano, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; M. Manetti, PhD, Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence; C. Mazzotta, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; R. Costa, MSc, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; V. Machado, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; A. Cortez, MSc, Nobre Laboratory, Faculty of Medicine, University of Porto; C. Bruni, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; G. Lepri, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; S. Guiducci, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; A. De Paulis, MD, Department of Translational Medical Sciences, CISI, University of Naples Federico II; R. Soares, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; M. Matucci-Cerinic, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence
| | - Raquel Soares
- From the Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro; I3S, Instituto de Investigação e Inovação em Saúde, University of Porto; Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; Nobre Laboratory, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence; Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence; Department of Translational Medical Sciences, Centre for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,I. Chora, MD, Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro, and I3S, University of Porto; E. Romano, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; M. Manetti, PhD, Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence; C. Mazzotta, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; R. Costa, MSc, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; V. Machado, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; A. Cortez, MSc, Nobre Laboratory, Faculty of Medicine, University of Porto; C. Bruni, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; G. Lepri, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; S. Guiducci, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; A. De Paulis, MD, Department of Translational Medical Sciences, CISI, University of Naples Federico II; R. Soares, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; M. Matucci-Cerinic, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence
| | - Marco Matucci-Cerinic
- From the Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro; I3S, Instituto de Investigação e Inovação em Saúde, University of Porto; Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; Nobre Laboratory, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence; Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence; Department of Translational Medical Sciences, Centre for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,I. Chora, MD, Department of Internal Medicine, São João Hospital Center, Al Prof Hernâni Monteiro, and I3S, University of Porto; E. Romano, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; M. Manetti, PhD, Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence; C. Mazzotta, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; R. Costa, MSc, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; V. Machado, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; A. Cortez, MSc, Nobre Laboratory, Faculty of Medicine, University of Porto; C. Bruni, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; G. Lepri, MD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; S. Guiducci, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence; A. De Paulis, MD, Department of Translational Medical Sciences, CISI, University of Naples Federico II; R. Soares, PhD, I3S, University of Porto, and Departamento de Biomedicina, Unidade de Bioquímica, Faculty of Medicine, University of Porto; M. Matucci-Cerinic, MD, PhD, Department of Experimental and Clinical Medicine, Division of Rheumatology, AOUC, University of Florence
| |
Collapse
|
40
|
Marou E, Liaskos C, Simopoulou T, Efthymiou G, Dardiotis E, Katsiari C, Scheper T, Meyer W, Hadjigeorgiou G, Bogdanos DP, Sakkas LI. Human cytomegalovirus (HCMV) UL44 and UL57 specific antibody responses in anti-HCMV-positive patients with systemic sclerosis. Clin Rheumatol 2017; 36:863-869. [PMID: 28124759 DOI: 10.1007/s10067-017-3553-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/17/2016] [Accepted: 01/17/2017] [Indexed: 02/06/2023]
Abstract
The role of human cytomegalovirus (HCMV) has been postulated as a trigger of systemic sclerosis (SSc). The aim of the study was to assess the prevalence of antibodies against HCMV UL44 and UL57 antigens not tested in the past. Sixty SSc patients, 40 multiple sclerosis and 17 normal controls (NCs), all anti-HCMV positive, were tested by immunoblotting. Reactivity to HCMV antigens, expressed as arbitrary units (AUs), was assessed for correlation with clinical and immunological parameters, including types of SSc-related autoantibodies. Anti-UL44 and anti-UL57 HCMV antibodies were present in 3/60 (5%) and 58/60 (96.7%) SSc patients, respectively (p < 0.001). Anti-UL57 antibodies were present in 35/40 (87.5%) MS patients and 16/17 (94.1%) NCs (SSc vs MS, MS vs NC, p = ns). Strong (50-75 AU) and very strong (75-100 AU) anti-UL57 immunoreactivity was found in 24 (41.4%) and 22 (37.9%) SSc patients, respectively (p = ns). Dilution experiments showed anti-UL57 antibody persistence in up to 1/5000. Overall, there was no difference in the frequency or the magnitude of anti-UL57 immunoreactivity between diffuse cutaneous systemic sclerosis and limited cutaneous systemic sclerosis patients (96.67 vs 96.67%; 65.45 ± 20.19 vs 64.31 ± 21.11 AU, p > 0.05) but strong anti-UL57 reactivity were more frequent in SSc compared to NCs (p = 0.007). Anti-UL57 reactivity was not inhibited by SSc-specific autoantigens. Anti-UL57 seropositivity did not correlate with demographic, clinical or immunological features of SSc. Anti-HCMV UL57 antibodies are universally present in anti-HCMV-positive patients with SSc, while those against UL44 are rarely seen. Because anti-UL57 lack disease specificity and are not involved in cross-reactive responses, their immunopathogenetic potential is to be questioned.
Collapse
Affiliation(s)
- Emmanouela Marou
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis, 40500, Larissa, Thessaly, Greece
- Cellular Immunotherapy and Molecular Immunodiagnostics, Biomedical Section, Centre for Research and Technology-Hellas (CERTH)-Institute for Research and Technology-Thessaly (IRETETH), 41222, Larissa, Greece
| | - Christos Liaskos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis, 40500, Larissa, Thessaly, Greece
- Cellular Immunotherapy and Molecular Immunodiagnostics, Biomedical Section, Centre for Research and Technology-Hellas (CERTH)-Institute for Research and Technology-Thessaly (IRETETH), 41222, Larissa, Greece
| | - Theodora Simopoulou
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis, 40500, Larissa, Thessaly, Greece
| | - Georgios Efthymiou
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis, 40500, Larissa, Thessaly, Greece
- Cellular Immunotherapy and Molecular Immunodiagnostics, Biomedical Section, Centre for Research and Technology-Hellas (CERTH)-Institute for Research and Technology-Thessaly (IRETETH), 41222, Larissa, Greece
| | - Efthymios Dardiotis
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University General Hospital of Larissa, University of Thessaly, 40500, Larissa, Greece
| | - Christina Katsiari
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis, 40500, Larissa, Thessaly, Greece
| | - Thomas Scheper
- Institute of Immunology, Euroimmun, 23560, Lübeck, Germany
| | - Wolfgang Meyer
- Institute of Immunology, Euroimmun, 23560, Lübeck, Germany
| | - Georgios Hadjigeorgiou
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University General Hospital of Larissa, University of Thessaly, 40500, Larissa, Greece
| | - Dimitrios P Bogdanos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis, 40500, Larissa, Thessaly, Greece
- Cellular Immunotherapy and Molecular Immunodiagnostics, Biomedical Section, Centre for Research and Technology-Hellas (CERTH)-Institute for Research and Technology-Thessaly (IRETETH), 41222, Larissa, Greece
| | - Lazaros I Sakkas
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Viopolis, 40500, Larissa, Thessaly, Greece.
| |
Collapse
|
41
|
Recent advances in the pathogenesis and management of Raynaud's phenomenon and digital ulcers. Curr Opin Rheumatol 2016; 28:577-85. [DOI: 10.1097/bor.0000000000000332] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Capillaroscopic findings and vascular biomarkers in systemic sclerosis: Association of low CD40L levels with late scleroderma pattern. Microvasc Res 2016; 108:17-21. [DOI: 10.1016/j.mvr.2016.07.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 07/02/2016] [Indexed: 11/21/2022]
|
43
|
|
44
|
Romano E, Chora I, Manetti M, Mazzotta C, Rosa I, Bellando-Randone S, Blagojevic J, Soares R, Avouac J, Allanore Y, Ibba-Manneschi L, Matucci-Cerinic M, Guiducci S. Decreased expression of neuropilin-1 as a novel key factor contributing to peripheral microvasculopathy and defective angiogenesis in systemic sclerosis. Ann Rheum Dis 2016; 75:1541-9. [PMID: 26359450 DOI: 10.1136/annrheumdis-2015-207483] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 08/20/2015] [Indexed: 01/13/2023]
Abstract
OBJECTIVES In systemic sclerosis (SSc), vascular involvement is characterised by vascular endothelial growth factor (VEGF)-A/VEGF receptor (VEGFR) system disturbances. Neuropilin-1 (NRP1), a receptor for both class-3 semaphorins (Sema3s) and VEGF-A, is required for optimal VEGF-A/VEGFR-2 signalling. Here, we investigated the possible involvement of Sema3A/NRP1 axis in SSc. METHODS Circulating Sema3A and soluble NRP1 (sNRP1) were measured in patients with SSc and controls. NRP1 and Sema3A expression in skin biopsies was evaluated by immunofluorescence and western blotting. NRP1 expression was assessed in SSc and healthy dermal microvascular endothelial cells (SSc-MVECs and H-MVECs), and in SSc and control endothelial progenitor cell (EPC)-derived endothelial cells (ECs). The possible impact of transcription factor Friend leukaemia integration 1 (Fli1) deficiency on endothelial NRP1 expression was investigated by gene silencing. The binding of Fli1 to NRP1 gene promoter was evaluated using chromatin immunoprecipitation. Capillary morphogenesis was performed on Matrigel. RESULTS Decreased sNRP1 levels in SSc were associated with active and late nailfold videocapillaroscopy patterns and digital ulcers. No difference in Sema3A was found between patients and controls. NRP1 was significantly decreased in SSc-MVECs both ex vivo and in vitro. NRP1 and Fli1 significantly decreased in H-MVECs challenged with SSc sera, while they were not different in SSc and control EPC-derived ECs. Fli1 occupied the NRP1 gene promoter and Fli1 gene silencing reduced NRP1 expression in H-MVECs. NRP1 gene silencing in H-MVECs resulted in a significantly impaired angiogenic capacity comparable to that of cells treated with SSc sera. CONCLUSION In SSc, NRP1 deficiency may be an additional factor in the perturbed VEGF-A/VEGFR-2 system contributing to peripheral microvasculopathy and defective angiogenesis.
Collapse
Affiliation(s)
- Eloisa Romano
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy
| | - Inês Chora
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy Department of Internal Medicine, São João Hospital Center, Porto, Portugal
| | - Mirko Manetti
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Celestina Mazzotta
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy
| | - Irene Rosa
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Silvia Bellando-Randone
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy
| | - Jelena Blagojevic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy
| | - Raquel Soares
- Department of Biochemistry, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Jerôme Avouac
- Cochin Institute, Paris Descartes University, INSERM U1016 and CNRS UMR8104, Paris, France
| | - Yannick Allanore
- Cochin Institute, Paris Descartes University, INSERM U1016 and CNRS UMR8104, Paris, France
| | - Lidia Ibba-Manneschi
- Department of Experimental and Clinical Medicine, Section of Anatomy and Histology, University of Florence, Florence, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy
| | - Serena Guiducci
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Azienda Ospedaliero-Universitaria Careggi (AOUC), University of Florence, Florence, Italy
| |
Collapse
|
45
|
Cestelli V, Manfredi A, Sebastiani M, Praino E, Cannarile F, Giuggioli D, Ferri C. Effect of treatment with iloprost with or without bosentan on nailfold videocapillaroscopic alterations in patients with systemic sclerosis. Mod Rheumatol 2016; 27:110-114. [PMID: 27310203 DOI: 10.1080/14397595.2016.1192761] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Vascular involvement plays a decisive role in systemic sclerosis (SSc) pathogenesis; it is responsible for some important clinical manifestations of the disease such as Raynaud's phenomenon and digital ulcers (DU). Bosentan, a dual receptor endothelin antagonist, and iloprost, often in combination therapy, seems to be able to interfere with the scleroderma microangiopathy. OBJECTIVES Aim of the study was to evaluate the effect of bosentan and iloprost on scleroderma microangiopathy, analyzed by means of capillaroscopic skin ulcer risk index (CSURI), in SSc patients treated for the prevention of DU. METHODS Nailfold videocapillaroscopy (NVC) was performed in 95 SSc patients, treated with iloprost alone (group 1) or combination therapy with iloprost and bosentan (group 2), at baseline and after one year. In all patients CSURI was calculated according to the formula "diameter × number of megacapillaries/(total number of capillaries)2": in addition, total number of capillaries, giant capillaries, micro-hemorrhages, disorganization of the vascular array, and ramified capillaries were evaluated by means of a semiquantitative score. RESULTS After 12 months, we observed a reduction of the number of giant capillaries in both groups, while an increase of ramified capillaries was recorded only in group 2. CSURI improved slightly in group 2 without statistical significance; on the contrary, in group 1 a significant worsening was recorded (p ≤ 0.001). CONCLUSIONS Our study confirms the effectiveness of bosentan, in combination with iloprost, in SSc microangiopathy observed to NVC. Moreover, the observed findings further support the role of CSURI in the evaluation and monitoring of SSc microangiopathy.
Collapse
Affiliation(s)
| | - Andreina Manfredi
- a Rheumatology Unit , University of Modena and Reggio Emilia , Italy
| | - Marco Sebastiani
- a Rheumatology Unit , University of Modena and Reggio Emilia , Italy
| | | | | | - Dilia Giuggioli
- a Rheumatology Unit , University of Modena and Reggio Emilia , Italy
| | - Clodoveo Ferri
- a Rheumatology Unit , University of Modena and Reggio Emilia , Italy
| |
Collapse
|
46
|
Maria ATJ, Maumus M, Le Quellec A, Jorgensen C, Noël D, Guilpain P. Adipose-Derived Mesenchymal Stem Cells in Autoimmune Disorders: State of the Art and Perspectives for Systemic Sclerosis. Clin Rev Allergy Immunol 2016; 52:234-259. [DOI: 10.1007/s12016-016-8552-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
47
|
Huang XL, Wu GC, Wang YJ, Yang XK, Yang GJ, Tao JH, Duan Y, Yan JW, Li XP, Ye DQ, Wang J. Association of interleukin-1 family cytokines single nucleotide polymorphisms with susceptibility to systemic sclerosis: an independent case–control study and a meta-analysis. Immunol Res 2016; 64:1041-52. [DOI: 10.1007/s12026-016-8797-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
48
|
Ligon C, Hummers LK. Biomarkers in Scleroderma: Progressing from Association to Clinical Utility. Curr Rheumatol Rep 2016; 18:17. [DOI: 10.1007/s11926-016-0565-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
49
|
Ariani A, Aiello M, Silva M, Alfieri V, Bonati E, Lumetti F, Delsante G, Sverzellati N, Chetta A. Quantitative CT indexes are significantly associated with exercise oxygen desaturation in interstitial lung disease related to systemic sclerosis. CLINICAL RESPIRATORY JOURNAL 2016; 11:983-989. [DOI: 10.1111/crj.12451] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 11/25/2015] [Accepted: 01/09/2016] [Indexed: 11/29/2022]
Affiliation(s)
- Alarico Ariani
- Department of Medicine, Internal Medicine and Rheumatology Unit; Azienda Ospedaliero-Universitaria di Parma; Parma Italy
| | - Marina Aiello
- Department of Clinical & Experimental Medicine, Respiratory Disease and Lung Function Unit; University of Parma; Parma Italy
| | - Mario Silva
- Department of Clinical Sciences; Section of Radiology, University of Parma; Parma Italy
| | - Veronica Alfieri
- Department of Clinical & Experimental Medicine, Respiratory Disease and Lung Function Unit; University of Parma; Parma Italy
| | - Elisa Bonati
- Department of Clinical & Experimental Medicine, Respiratory Disease and Lung Function Unit; University of Parma; Parma Italy
| | - Federica Lumetti
- Department of Medicine, Internal Medicine and Rheumatology Unit; Azienda Ospedaliero-Universitaria di Parma; Parma Italy
| | - Giovanni Delsante
- Department of Medicine, Internal Medicine and Rheumatology Unit; Azienda Ospedaliero-Universitaria di Parma; Parma Italy
| | - Nicola Sverzellati
- Department of Clinical Sciences; Section of Radiology, University of Parma; Parma Italy
| | - Alfredo Chetta
- Department of Clinical & Experimental Medicine, Respiratory Disease and Lung Function Unit; University of Parma; Parma Italy
| |
Collapse
|
50
|
Perosa F, Prete M, Di Lernia G, Ostuni C, Favoino E, Valentini G. Anti-centromere protein A antibodies in systemic sclerosis: Significance and origin. Autoimmun Rev 2015; 15:102-9. [PMID: 26455561 DOI: 10.1016/j.autrev.2015.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 10/04/2015] [Indexed: 02/03/2023]
Abstract
Systemic sclerosis (SSc) is systemic, autoimmune, connective tissue disorder characterized by vascular abnormalities, collagen deposition (fibrosis), and the production of autoantibodies to nuclear proteins. About 20%-40% of patients have antibodies to centromere protein (CENP)-A or -B. Despite the known association of anti-CENP antibodies with certain clinical features of SSc, the role of these antibodies in SSc physiopathology is still poorly understood. To better understand the clinical significance and origin of these antibodies, we and others have been studying the epitopic motifs (amino acid contact sites) on CENP-A with the aim of determining whether other proteins can prime or be targeted by them. Here, we review published and ongoing studies aimed at defining the fine specificity and origin of anti-CENP-A antibodies. We describe progress made in identifying the CENP-A epitopic motif amino acids, and the discovery of one of these motifs in forkhead box protein E3 (FOXE-3), a transcription factor previously studied only for its role in the development of lens fiber cells. Moreover, we discuss preliminary evidence for a possible role of FOXE-3 in SSc pathogenesis and for the association of different subsets of anti-CENP-A antibodies, heterogeneously expressed among SSc patients, with some clinical correlates.
Collapse
Affiliation(s)
- Federico Perosa
- Department of Biomedical Sciences and Human Oncology (DIMO), Section of Systemic Rheumatic and Autoimmune Diseases, University of Bari Medical School, Bari, Italy.
| | - Marcella Prete
- Department of Biomedical Sciences and Human Oncology (DIMO), Section of Internal Medicine, University of Bari Medical School, Bari, Italy
| | - Giuseppe Di Lernia
- Department of Biomedical Sciences and Human Oncology (DIMO), Section of Systemic Rheumatic and Autoimmune Diseases, University of Bari Medical School, Bari, Italy
| | - Carmela Ostuni
- Department of Biomedical Sciences and Human Oncology (DIMO), Section of Clinical Oncology, University of Bari Medical School, Bari, Italy
| | - Elvira Favoino
- Department of Biomedical Sciences and Human Oncology (DIMO), Section of Systemic Rheumatic and Autoimmune Diseases, University of Bari Medical School, Bari, Italy
| | - Gabriele Valentini
- Department of Clinical and Experimental Internal Medicine "F. Magrassi, A. Lanzara", Rheumatology Section, Second University of Naples, Naples, Italy
| |
Collapse
|