1
|
Kowalska-Kępczyńska A, Mleczko M, Komajda K, Michalska-Jakubus M, Krasowska D, Korpysz M. Extended Inflammation Parameters (EIP) as Markers of Inflammation in Systemic Sclerosis. Int J Inflam 2024; 2024:3786206. [PMID: 39364215 PMCID: PMC11449563 DOI: 10.1155/2024/3786206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/16/2024] [Indexed: 10/05/2024] Open
Abstract
Background Systemic sclerosis (SSc) is an autoimmune disease characterized by inflammation, progressive vasculopathy, and fibrosis of skin and internal organs. The aim of the study was to evaluate extended inflammatory parameters (EIP) in patients with SSc in comparison to the control group of healthy subjects. Methods A total of 28 patients with SSc and 29 healthy controls (HCs) were included in the study. The following EIP parameters were analyzed: neutrophil reactive intensity (NEUT-RI), neutrophil granularity intensity (NEUT-GI), antibody-synthesizing lymphocytes (AS-LYMP), and reactive lymphocytes (RE-LYMP). Results Patients with SSc showed significantly higher values of parameters determining neutrophil reactivity and neutrophil granularity when compared to HCs (respectively, 49.16 FI vs. 44.33 FI, p < 0.001, and 152.01 SI vs. 147.51 SI, p < 0.001). Moreover, patients with SSc had higher absolute numbers of RE-LYMP than HCs (0.69 × 103/µl vs. 0.04 × 103/µl, p < 0.001). Importantly, significant correlations between the RE-LYMP and either IL-6 (R = 0.447, p < 0.001) or ESR (R = 0.532, p < 0.001) were found among patients with SSc. Conclusions Changes in NEUT-RI, NEUT-GI, and RE-LYMP levels positively correlate with inflammation in SSc and, thus, could potentially be used as an additional reliable inflammatory biomarker to assess inflammation in this disease.
Collapse
Affiliation(s)
- Anna Kowalska-Kępczyńska
- Department of Biochemical DiagnosticsChair of Laboratory DiagnosticsMedical University of Lublin, al. Solidarności 8, Lublin 20-081, Poland
| | - Mateusz Mleczko
- Department of DermatologyVenereology and Pediatric DermatologyMedical University of Lublin, ul. Staszica 11, Lublin 20-081, Poland
| | - Kamila Komajda
- Laboratory of Forensic ToxicologyMedical University of Lublin, ul. Jaczewskiego 8b, Lublin 20-080, Poland
| | - Małgorzata Michalska-Jakubus
- Department of DermatologyVenereology and Pediatric DermatologyMedical University of Lublin, ul. Staszica 11, Lublin 20-081, Poland
| | - Dorota Krasowska
- Department of DermatologyVenereology and Pediatric DermatologyMedical University of Lublin, ul. Staszica 11, Lublin 20-081, Poland
| | - Maciej Korpysz
- Department of Biochemical DiagnosticsChair of Laboratory DiagnosticsMedical University of Lublin, al. Solidarności 8, Lublin 20-081, Poland
| |
Collapse
|
2
|
Naik A, Stratton RJ, Leask A. Digital ulcers associated with scleroderma: A major unmet medical need. Wound Repair Regen 2024. [PMID: 39323322 DOI: 10.1111/wrr.13224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
Scleroderma or systemic sclerosis (SSc)-associated digital ischaemic complications, such as digital ulcers (SSc-DUs), appear relatively early during the disease course and are a major burden with substantial deterioration of quality of life. Expert rheumatologist and wound specialists have defined a DU; however, international application of the definition is still disorganised. Appearance of SSc-DUs is secondary to the onset of Raynaud's phenomenon and as a consequence, recommended first-line of treatment mainly includes vasodilators; however, many DUs are refractory to this treatment. Despite important practical issues, such as a lack of well-characterised SSc-wound healing animal model, significant efforts are needed to mechanistically understand the pathogenesis of SSc-DUs for developing clinically targetable disease modifying therapies.
Collapse
Affiliation(s)
- Angha Naik
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Richard J Stratton
- Centre for Rheumatology and Connective Tissue Disease, University College London (Royal Free Campus), London, UK
| | - Andrew Leask
- College of Dentistry, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
3
|
Shen S, Hu M, Peng Y, Zheng Y, Zhang R. Research Progress in pathogenesis of connective tissue disease-associated interstitial lung disease from the perspective of pulmonary cells. Autoimmun Rev 2024; 23:103600. [PMID: 39151642 DOI: 10.1016/j.autrev.2024.103600] [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: 04/14/2024] [Revised: 07/16/2024] [Accepted: 08/10/2024] [Indexed: 08/19/2024]
Abstract
The lungs are a principal factor in the increased morbidity and mortality observed in patients with Connective Tissue Disease (CTD), frequently presenting as CTD-associated Interstitial Lung Disease (ILD). Currently, there is a lack of comprehensive descriptions of the pulmonary cells implicated in the development of CTD-ILD. This review leverages the Human Lung Cell Atlas (HLCA) and spatial multi-omics atlases to discuss the advancements in research on the pathogenesis of CTD-ILD from a pulmonary cell perspective. This facilitates a more precise localization of disease sites and a more systematic consideration of disease progression, supporting further mechanistic studies and targeted therapies.
Collapse
Affiliation(s)
- Shuyi Shen
- Department of Rheumatology, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Ming Hu
- Department of Rheumatology, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Yi Peng
- Department of Rheumatology, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Yi Zheng
- Department of Rheumatology, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Rong Zhang
- Department of Rheumatology, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| |
Collapse
|
4
|
Romano E, Rosa I, Fioretto BS, Manetti M. Recent Insights into Cellular and Molecular Mechanisms of Defective Angiogenesis in Systemic Sclerosis. Biomedicines 2024; 12:1331. [PMID: 38927538 PMCID: PMC11201654 DOI: 10.3390/biomedicines12061331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
In systemic sclerosis (SSc, or scleroderma), defective angiogenesis, clinically manifesting with abnormal capillary architecture and severe capillary reduction, represents a hallmark of early-stage disease, usually preceding the onset of tissue fibrosis, and is caused by several cellular and molecular mechanisms affecting microvascular endothelial cells with different outcomes. Indeed, once damaged, endothelial cells can be dysfunctionally activated, thus becoming unable to undergo angiogenesis and promoting perivascular inflammation. They can also undergo apoptosis, transdifferentiate into profibrotic myofibroblasts, or acquire a senescence-associated secretory phenotype characterized by the release of exosomes and several profibrotic and proinflammatory mediators. In this narrative review, we aimed to give a comprehensive overview of recent studies dealing with the cellular and molecular mechanisms underlying SSc defective angiogenesis and the related endothelial cell dysfunctions, mainly the endothelial-to-mesenchymal transition process. We also discussed potential novel vascular treatment strategies able to restore the angiogenic process and reduce the endothelial-to-mesenchymal transition in this complex disease.
Collapse
Affiliation(s)
- Eloisa Romano
- Section of Internal Medicine, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy;
| | - Irene Rosa
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 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, Largo Brambilla 3, 50134 Florence, Italy; (I.R.); (B.S.F.)
| | - Mirko Manetti
- Section of Anatomy and Histology, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy; (I.R.); (B.S.F.)
- Imaging Platform, Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy
| |
Collapse
|
5
|
Liu X, Hua L, Chu J, Zhou W, Jiang F, Wang L, Xu F, Liu M, Shi J, Xue G. Endothelial dysfunction and disease severity in COVID-19: Insights from circulating Tang cell counts as a potential biomarker. Int Immunopharmacol 2024; 130:111788. [PMID: 38447419 DOI: 10.1016/j.intimp.2024.111788] [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: 01/07/2024] [Revised: 02/15/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024]
Abstract
BACKGROUND AND AIM Endothelial dysfunction is a common risk factor of severe COVID-19. Angiogenic T cells (Tang cells) play a critical role in repairing endothelial injury; however, their changes and potential roles in COVID-19 remain unclear. We aimed to assess Tang cell counts in patients with COVID-19 and evaluate their association with disease severity and prognosis. METHODS Circulating Tang cell populations in patients with COVID-19 and healthy controls were quantified using flow cytometry. Demographic and routine laboratory data were recorded. RESULTS The Tang cell count decreased significantly with increasing disease severity and were lowest in fatal cases. Additionally, the Tang cell count was significantly decreased in patients with comorbid cardiovascular disease or hypertension. Tang cell counts were negatively correlated with inflammatory markers, kidney and myocardial injury markers, coagulation dysfunction indicators, and viral load and positively correlated with oxidative stress markers, nutritional markers, and lymphocytes. Receiver operating characteristic curves confirmed that Tang cell count could serve as a potential biomarker for predicting disease severity and patient mortality. CONCLUSIONS Circulating Tang cell count is significantly reduced in patients with COVID-19 and is correlated with disease severity and prognosis. The Tang cell count is an important potential biomarker for COVID-19 clinical management. Additionally, these findings provide insight into the pathological features of COVID-19 endothelial injury and provide new directions for treatment.
Collapse
Affiliation(s)
- Xiaofeng Liu
- Department of Clinical Laboratory, Jiujiang No.1 People's Hospital, Jiujiang, 332000, PR China
| | - Lin Hua
- Department of Clinical Laboratory, Jiujiang No.1 People's Hospital, Jiujiang, 332000, PR China
| | - Jinshen Chu
- Department of Clinical Laboratory, Jiujiang No.1 People's Hospital, Jiujiang, 332000, PR China
| | - Wei Zhou
- Department of Clinical Laboratory, Jiujiang No.1 People's Hospital, Jiujiang, 332000, PR China
| | - Fangtinghui Jiang
- Department of Clinical Laboratory, Jiujiang No.1 People's Hospital, Jiujiang, 332000, PR China
| | - Lu Wang
- Department of Clinical Laboratory, Jiujiang No.1 People's Hospital, Jiujiang, 332000, PR China
| | - Fanglin Xu
- Department of Intensive Care Medicine, Jiujiang No.1 People's Hospital, Jiujiang, 332000, PR China
| | - Mingjiao Liu
- Department of Intensive Care Medicine, Jiujiang No.1 People's Hospital, Jiujiang, 332000, PR China
| | - Jianbang Shi
- Department of Respiratory Medicine, Jiujiang No.1 People's Hospital, Jiujiang, 332000, PR China
| | - Guohui Xue
- Department of Clinical Laboratory, Jiujiang No.1 People's Hospital, Jiujiang, 332000, PR China.
| |
Collapse
|
6
|
Guilhem A, Ciudad M, Aubriot-Lorton MH, Greigert H, Cladière C, Leguy-Seguin V, Audia S, Samson M, Bonnotte B. Pro-angiogenic changes of T-helper lymphocytes in hereditary hemorrhagic telangiectasia. Front Immunol 2023; 14:1321182. [PMID: 38143764 PMCID: PMC10748412 DOI: 10.3389/fimmu.2023.1321182] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/24/2023] [Indexed: 12/26/2023] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is a rare inherited disease due to heterozygous loss-of-function mutations on the BMP9/10 pathway (ENG, ACVRL1 or MADH4 mainly). HHT endothelial cells are prone to lose their quiescence, leading to progressive appearance of numerous telangiectases on skin and mucosa (complicated by epistaxis and anemia), and to larger arteriovenous malformations in lungs, liver and brain. HHT is also associated with T lymphocyte abnormalities, which are currently poorly understood. We quantified by flow-cytometry the main T lymphocyte circulating subsets in 40 HHT patients and 20 matched healthy controls. Immunostaining was done on 2 HHT skin telangiectases. Disruptions in T lymphocyte homeostasis was observed, characterized by increases in subsets known to promote angiogenesis: Th2 (1.38% vs 1.15%, p=0.021), Th17 (0.32% vs 0.22%, p=0.019 2) and Treg (4.94% vs 3.51%, p= 0.027). T angiogenic lymphocytes (Tang), defined as CD3+CD31+CXCR4+ T cells, were at similar levels in both groups, but the proportion of VEGF-A+ Tang after stimulation was higher in the HHT group compared to controls (68.2% vs 44.9%, p=0.012). The global HHT T lymphopenia predominantly affected the effector memory T-helper cells (200 vs 270 cells/mm3, p=0.017), and the lymphocytic infiltrate around HHT telangiectases consisted of memory T-helper cells. The Th17 circulating subset was positively correlated with the monthly epistaxis duration (r coefficient: +0,431, p=0.042), prospectively assessed. HHT T-helper lymphocytes are affected by several pro-angiogenic changes, potentially resulting from their recruitment by abnormal endothelial cells. They could constitute a biologically relevant source of VEGF-A and a valuable therapeutic target in HHT.
Collapse
Affiliation(s)
- Alexandre Guilhem
- Service de Médecine Interne et Immunologie Clinique, Centre de compétence maladie de Rendu-Osler, Centre Hospitalo-Universitaire Dijon Bourgogne, Dijon, France
- Université de Bourgogne, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Dijon, France
| | - Marion Ciudad
- Université de Bourgogne, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Dijon, France
| | | | - Hélène Greigert
- Service de Médecine Interne et Immunologie Clinique, Centre de compétence maladie de Rendu-Osler, Centre Hospitalo-Universitaire Dijon Bourgogne, Dijon, France
| | - Claudie Cladière
- Université de Bourgogne, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Dijon, France
| | - Vanessa Leguy-Seguin
- Service de Médecine Interne et Immunologie Clinique, Centre de compétence maladie de Rendu-Osler, Centre Hospitalo-Universitaire Dijon Bourgogne, Dijon, France
| | - Sylvain Audia
- Service de Médecine Interne et Immunologie Clinique, Centre de compétence maladie de Rendu-Osler, Centre Hospitalo-Universitaire Dijon Bourgogne, Dijon, France
- Université de Bourgogne, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Dijon, France
| | - Maxime Samson
- Service de Médecine Interne et Immunologie Clinique, Centre de compétence maladie de Rendu-Osler, Centre Hospitalo-Universitaire Dijon Bourgogne, Dijon, France
- Université de Bourgogne, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Dijon, France
| | - Bernard Bonnotte
- Service de Médecine Interne et Immunologie Clinique, Centre de compétence maladie de Rendu-Osler, Centre Hospitalo-Universitaire Dijon Bourgogne, Dijon, France
- Université de Bourgogne, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Dijon, France
| |
Collapse
|
7
|
Feugray G, Miranda S, Le Cam Duchez V, Bellien J, Billoir P. Endothelial Progenitor Cells in Autoimmune Disorders. Stem Cell Rev Rep 2023; 19:2597-2611. [PMID: 37676423 DOI: 10.1007/s12015-023-10617-y] [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: 08/26/2023] [Indexed: 09/08/2023]
Abstract
Circulating endothelial progenitor cells (EPCs) were first described in 1997 by Asahara et al. as "putative endothelial cells" from human peripheral blood. The study of endothelial progenitors is also intensifying in several pathologies associated with endothelial damage, including diabetes, myocardial infarction, sepsis, pulmonary arterial hypertension, obstructive bronchopneumopathy and transplantation. EPCs have been studied in several autoimmune diseases with endothelial involvement such as systemic lupus erythematosus, thrombotic thrombocytopenic purpura, antineutrophil cytoplasmic antibodies, vasculitis, rheumatoid arthritis, Goujerot-Sjögren and antiphospholipid syndrome. Factors involved in endothelial damage are due to overexpression of pro-inflammatory cytokines and/or autoantibodies. Management of these pathologies, particularly the long-term use of glucocorticoids and methotrexate, promote atherosclerosis. A lack of standardized assessment of the number and function of EPCs represents a serious challenge for the use of EPCs as prognostic markers of cardiovascular diseases (CVD). The objective of this review was to describe EPCs, their properties and their involvement in several autoimmune diseases.
Collapse
Affiliation(s)
- Guillaume Feugray
- UNIROUEN, INSERM U1096 EnVI, CHU Rouen, Department of General Biochemistry, Normandie University, F-76000, Rouen, France
| | - Sébastien Miranda
- UNIROUEN, INSERM U1096, CHU Rouen. Department of Internal Medicine, Normandie University, Rouen, France
| | | | - Jérémy Bellien
- UNIROUEN, INSERM U1096 EnVI, CHU Rouen, Department of Pharmacology, Normandie University, F-76000, Rouen, France
| | - Paul Billoir
- UNIROUEN, INSERM U1096, CHU Rouen. Department of Internal Medicine, Normandie University, Rouen, France.
- Normandy Univ, U1096, Rouen University Hospital, Vascular Hemostasis Unit, Rouen, France.
| |
Collapse
|
8
|
Ko J, Noviani M, Chellamuthu VR, Albani S, Low AHL. The Pathogenesis of Systemic Sclerosis: The Origin of Fibrosis and Interlink with Vasculopathy and Autoimmunity. Int J Mol Sci 2023; 24:14287. [PMID: 37762589 PMCID: PMC10532389 DOI: 10.3390/ijms241814287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease associated with increased mortality and poor morbidity, impairing the quality of life in patients. Whilst we know that SSc affects multiple organs via vasculopathy, inflammation, and fibrosis, its exact pathophysiology remains elusive. Microvascular injury and vasculopathy are the initial pathological features of the disease. Clinically, the vasculopathy in SSc is manifested as Raynaud's phenomenon (reversible vasospasm in reaction to the cold or emotional stress) and digital ulcers due to ischemic injury. There are several reports that medications for vasculopathy, such as bosentan and soluble guanylate cyclase (sGC) modulators, improve not only vasculopathy but also dermal fibrosis, suggesting that vasculopathy is important in SSc. Although vasculopathy is an important initial step of the pathogenesis for SSc, it is still unclear how vasculopathy is related to inflammation and fibrosis. In this review, we focused on the clinical evidence for vasculopathy, the major cellular players for the pathogenesis, including pericytes, adipocytes, endothelial cells (ECs), and myofibroblasts, and their signaling pathway to elucidate the relationship among vasculopathy, inflammation, and fibrosis in SSc.
Collapse
Affiliation(s)
- Junsuk Ko
- Duke-National University of Singapore Medical School, Singapore 169857, Singapore; (J.K.); (M.N.); (S.A.)
| | - Maria Noviani
- Duke-National University of Singapore Medical School, Singapore 169857, Singapore; (J.K.); (M.N.); (S.A.)
- Department of Rheumatology and Immunology, Singapore General Hospital, Singapore 169608, Singapore
- Translational Immunology Institute, SingHealth Duke-National University of Singapore Academic Medical Centre, Singapore 169856, Singapore;
| | - Vasuki Ranjani Chellamuthu
- Translational Immunology Institute, SingHealth Duke-National University of Singapore Academic Medical Centre, Singapore 169856, Singapore;
| | - Salvatore Albani
- Duke-National University of Singapore Medical School, Singapore 169857, Singapore; (J.K.); (M.N.); (S.A.)
- Translational Immunology Institute, SingHealth Duke-National University of Singapore Academic Medical Centre, Singapore 169856, Singapore;
| | - Andrea Hsiu Ling Low
- Duke-National University of Singapore Medical School, Singapore 169857, Singapore; (J.K.); (M.N.); (S.A.)
- Department of Rheumatology and Immunology, Singapore General Hospital, Singapore 169608, Singapore
| |
Collapse
|
9
|
Lozano Í, Bangueses R, Rodríguez I, Pevida M, Rodríguez-Aguilar R, Rodríguez D, Espasandín-Arias M, Llames S, Meana Á, Suárez A, Rodríguez-Carrio J. In-stent restenosis is associated with proliferative skin healing and specific immune and endothelial cell profiles: results from the RACHEL trial. Front Immunol 2023; 14:1138247. [PMID: 37325628 PMCID: PMC10265483 DOI: 10.3389/fimmu.2023.1138247] [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: 01/05/2023] [Accepted: 05/22/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction In-stent restenosis (ISR) is a major challenge in interventional cardiology. Both ISR and excessive skin healing are aberrant hyperplasic responses, which may be functionally related. However, the cellular component underlying ISR remains unclear, especially regarding vascular homeostasis. Recent evidence suggest that novel immune cell populations may be involved in vascular repair and damage, but their role in ISR has not been explored. The aims of this study is to analyze (i) the association between ISR and skin healing outcomes, and (ii) the alterations in vascular homeostasis mediators in ISR in univariate and integrative analyses. Methods 30 patients with ≥1 previous stent implantation with restenosis and 30 patients with ≥1 stent without restenosis both confirmed in a second angiogram were recruited. Cellular mediators were quantified in peripheral blood by flow cytometry. Skin healing outcomes were analyzed after two consecutive biopsies. Results Hypertrophic skin healing was more frequent in ISR patients (36.7%) compared to those ISR-free (16.7%). Patients with ISR were more likely to develop hypertrophic skin healing patterns (OR 4.334 [95% CI 1.044-18.073], p=0.033), even after correcting for confounders. ISR was associated with decreased circulating angiogenic T-cells (p=0.005) and endothelial progenitor cells (p<0.001), whereas CD4+CD28null and detached endothelial cells counts were higher (p<0.0001 and p=0.006, respectively) compared to their ISR-free counterparts. No differences in the frequency of monocyte subsets were found, although Angiotensin-Converting Enzyme expression was increased (non-classical: p<0.001; and intermediate: p<0.0001) in ISR. Despite no differences were noted in Low-Density Granulocytes, a relative increase in the CD16- compartment was observed in ISR (p=0.004). An unsupervised cluster analysis revealed the presence of three profiles with different clinical severity, unrelated to stent types or traditional risk factors. Conclusion ISR is linked to excessive skin healing and profound alterations in cellular populations related to vascular repair and endothelial damage. Distinct cellular profiles can be distinguished within ISR, suggesting that different alterations may uncover different ISR clinical phenotypes.
Collapse
Affiliation(s)
- Íñigo Lozano
- Department of Cardiology, Hospital Universitario Cabueñes, Gijón, Asturias, Spain
- Cardiac Pathology Research Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Roi Bangueses
- Department of Cardiology, Hospital Universitario Cabueñes, Gijón, Asturias, Spain
| | - Isabel Rodríguez
- Cardiac Pathology Research Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
| | - Marta Pevida
- Blood Tansfusion Center and Tissue Bank of Asturias, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Oviedo, Asturias, Spain
- Grupo de Investigación en Oftalmología, Ciencias de la Visión y Terapias Avanzadas (GOVITA), Instituto de Salud del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, Oviedo, Asturias, Spain
| | - Raúl Rodríguez-Aguilar
- Department of Pathology Anatomy, Hospital Universitario Cabueñes, Gijón, Asturias, Spain
| | - Diana Rodríguez
- Department of Pathology Anatomy, Hospital Universitario Cabueñes, Gijón, Asturias, Spain
| | | | - Sara Llames
- Blood Tansfusion Center and Tissue Bank of Asturias, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Oviedo, Asturias, Spain
- Grupo de Investigación en Oftalmología, Ciencias de la Visión y Terapias Avanzadas (GOVITA), Instituto de Salud del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Fundación Jiménez Díaz, Madrid, Spain
| | - Álvaro Meana
- Blood Tansfusion Center and Tissue Bank of Asturias, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Oviedo, Asturias, Spain
- Grupo de Investigación en Oftalmología, Ciencias de la Visión y Terapias Avanzadas (GOVITA), Instituto de Salud del Principado de Asturias (ISPA), Oviedo, Asturias, Spain
- Instituto Universitario Fernández-Vega, Fundación de Investigación Oftalmológica, Oviedo, Asturias, Spain
| | - Ana Suárez
- Area of Immunology, Department of Functional Biology, University of Oviedo, Oviedo, Asturias, Spain
- Grupo de Investigación Básica y Traslacional en Enfermedades Inflamatorias, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Asturias, Spain
| | - Javier Rodríguez-Carrio
- Area of Immunology, Department of Functional Biology, University of Oviedo, Oviedo, Asturias, Spain
- Grupo de Investigación Básica y Traslacional en Enfermedades Inflamatorias, Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Asturias, Spain
| |
Collapse
|
10
|
Kul A, Ozturk N, Kurt AK, Arslan Y. Detection of Angiogenic T Cells and Endothelial Progenitor Cells in Behçet Disease and Determination of Their Relationship with Disease Activity. Life (Basel) 2023; 13:1259. [PMID: 37374042 DOI: 10.3390/life13061259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/18/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Angiogenic T (Tang) cells and endothelial progenitor cells (EPCs) play a role in maintaining vascular integrity and repair. This study considers the association between them and Behçet disease (BD) and disease activity. Fifty patients with BD and forty-five age- and gender-matched healthy controls were included in the study. The participants' demographic, clinical, and laboratory characteristics were recorded, and their blood Tang cell and EPC counts were determined. Fifty patients were diagnosed with BD, consisting of 24 females and 26 males. The blood Tang cell (3.5 ± 1.2 cells/μL in patients, 4 ± 0.9 cells/μL in controls, p = 0.046)) and EPC (2.9 ± 0.9 cells/μL in patients, 3.7 ± 1 cells/μL in controls, p = 0.001) counts were significantly lower for the patient group with BD than for the control group. The blood Tang cell (42.5 ± 4.9% in active patients, 48.9 ± 7.9% in inactive patients, p = 0.001) and EPC (35.5 ± 6.4% in active patients, 41.2 ± 6.3% in inactive patients, p = 0.004) levels were lower for the patient group with active BD than for the inactive patient group. A weak positive correlation was present between the blood Tang cell and EPC percentage values in BD (r: 0.318, p = 0.002). It was determined that Tang cell and EPC counts are lower in BD, and these reductions become more profound with increasing disease activity. This situation may prevent the development of a sufficient immune response against a disease with a course of chronic inflammation or may trigger the formation of autoreactive immunity. A reduction in Tang cells and EPCs may serve as a marker or predictor of vascular damage in BD patients and represents the progression of vascular injury.
Collapse
Affiliation(s)
- Ayhan Kul
- Faculty of Medicine, Physical Medicine and Rehabilitation, Ataturk University, Erzurum 25240, Turkey
| | - Nurinnisa Ozturk
- Faculty of Medicine, Medical Biochemistry, Ataturk University, Erzurum 25240, Turkey
| | - Asli Koseoglu Kurt
- Faculty of Medicine, Medical Biochemistry, Ataturk University, Erzurum 25240, Turkey
| | - Yasar Arslan
- Faculty of Medicine, Physical Medicine and Rehabilitation, Ataturk University, Erzurum 25240, Turkey
| |
Collapse
|
11
|
Laranjeira P, dos Santos F, Salvador MJ, Simões IN, Cardoso CMP, Silva BM, Henriques-Antunes H, Corte-Real L, Couceiro S, Monteiro F, Santos C, Santiago T, da Silva JAP, Paiva A. Umbilical-Cord-Derived Mesenchymal Stromal Cells Modulate 26 Out of 41 T Cell Subsets from Systemic Sclerosis Patients. Biomedicines 2023; 11:1329. [PMID: 37239000 PMCID: PMC10215673 DOI: 10.3390/biomedicines11051329] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/20/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Systemic sclerosis (SSc) is an immune-mediated disease wherein T cells are particularly implicated, presenting a poor prognosis and limited therapeutic options. Thus, mesenchymal-stem/stromal-cell (MSC)-based therapies can be of great benefit to SSc patients given their immunomodulatory, anti-fibrotic, and pro-angiogenic potential, which is associated with low toxicity. In this study, peripheral blood mononuclear cells from healthy individuals (HC, n = 6) and SSc patients (n = 9) were co-cultured with MSCs in order to assess how MSCs affected the activation and polarization of 58 different T cell subsets, including Th1, Th17, and Treg. It was found that MSCs downregulated the activation of 26 out of the 41 T cell subsets identified within CD4+, CD8+, CD4+CD8+, CD4-CD8-, and γδ T cells in SSc patients (HC: 29/42) and affected the polarization of 13 out of 58 T cell subsets in SSc patients (HC: 22/64). Interestingly, SSc patients displayed some T cell subsets with an increased activation status and MSCs were able to downregulate all of them. This study provides a wide-ranging perspective of how MSCs affect T cells, including minor subsets. The ability to inhibit the activation and modulate the polarization of several T cell subsets, including those implicated in SSc's pathogenesis, further supports the potential of MSC-based therapies to regulate T cells in a disease whose onset/development may be due to immune system's malfunction.
Collapse
Affiliation(s)
- Paula Laranjeira
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal;
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Francisco dos Santos
- Stemlab S.A., Famicord Group, 3060-197 Cantanhede, Portugal; (F.d.S.); (I.N.S.); (C.M.P.C.); (H.H.-A.); (L.C.-R.); (S.C.); (F.M.); (C.S.)
| | - Maria João Salvador
- Rheumatology Department, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal; (M.J.S.); (T.S.)
| | - Irina N. Simões
- Stemlab S.A., Famicord Group, 3060-197 Cantanhede, Portugal; (F.d.S.); (I.N.S.); (C.M.P.C.); (H.H.-A.); (L.C.-R.); (S.C.); (F.M.); (C.S.)
| | - Carla M. P. Cardoso
- Stemlab S.A., Famicord Group, 3060-197 Cantanhede, Portugal; (F.d.S.); (I.N.S.); (C.M.P.C.); (H.H.-A.); (L.C.-R.); (S.C.); (F.M.); (C.S.)
| | - Bárbara M. Silva
- Algarve Biomedical Center (ABC), Universidade do Algarve, 8005-139 Faro, Portugal;
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, 8005-139 Faro, Portugal
- Doctoral Program in Biomedical Sciences, Faculty of Medicine and Biomedical Sciences, Universidade do Algarve, 8005-139 Faro, Portugal
| | - Helena Henriques-Antunes
- Stemlab S.A., Famicord Group, 3060-197 Cantanhede, Portugal; (F.d.S.); (I.N.S.); (C.M.P.C.); (H.H.-A.); (L.C.-R.); (S.C.); (F.M.); (C.S.)
| | - Luísa Corte-Real
- Stemlab S.A., Famicord Group, 3060-197 Cantanhede, Portugal; (F.d.S.); (I.N.S.); (C.M.P.C.); (H.H.-A.); (L.C.-R.); (S.C.); (F.M.); (C.S.)
| | - Sofia Couceiro
- Stemlab S.A., Famicord Group, 3060-197 Cantanhede, Portugal; (F.d.S.); (I.N.S.); (C.M.P.C.); (H.H.-A.); (L.C.-R.); (S.C.); (F.M.); (C.S.)
| | - Filipa Monteiro
- Stemlab S.A., Famicord Group, 3060-197 Cantanhede, Portugal; (F.d.S.); (I.N.S.); (C.M.P.C.); (H.H.-A.); (L.C.-R.); (S.C.); (F.M.); (C.S.)
| | - Carolina Santos
- Stemlab S.A., Famicord Group, 3060-197 Cantanhede, Portugal; (F.d.S.); (I.N.S.); (C.M.P.C.); (H.H.-A.); (L.C.-R.); (S.C.); (F.M.); (C.S.)
| | - Tânia Santiago
- Rheumatology Department, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal; (M.J.S.); (T.S.)
| | - José A. P. da Silva
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Rheumatology Department, Hospitais da Universidade de Coimbra, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal; (M.J.S.); (T.S.)
| | - Artur Paiva
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra, 3000-075 Coimbra, Portugal;
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal;
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Instituto Politécnico de Coimbra, ESTESC-Coimbra Health School, Ciências Biomédicas Laboratoriais, 3046-854 Coimbra, Portugal
| |
Collapse
|
12
|
Ravarotto V, Bertoldi G, Rigato M, Pagnin E, Gobbi L, Davis PA, Calò LA. Tracing angiotensin II's yin-yang effects on cardiovascular-renal pathophysiology. Minerva Med 2023; 114:56-67. [PMID: 34180640 DOI: 10.23736/s0026-4806.21.07440-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Adverse changes in cardiovascular and renal systems are major contributors to overall morbidity and mortality. Human cardiovascular and renal systems exhibit a complex network of positive and negative feedback that is reflected in the control of vascular tone via angiotensin II (Ang II) based signaling. This review will examine in some depth, the multiple components and processes that control the status and reflect the health of these various cardiovascular and renal systems, such as pathways associated to monomeric G proteins, RhoA/Rho kinase system and ERK, oxidative stress and NO balance. It will specifically emphasize the "yin-yang" nature of Ang II signaling by comparing and contrasting the effects and activity of various systems, pathways and components found in hypertension to those found in Gitelman's and Bartter's syndromes (GS/BS), two rare autosomal recessive tubulopathies characterized by electrolytic imbalance, metabolic alkalosis, sodium wasting and prominent activation of the renin-angiotensin-aldosterone system. Notwithstanding the activation of the renin-angiotensin-aldosterone system, GS/BS are normo-hypotensive and protected from cardiovascular-renal remodeling and therefore can be considered the mirror image, the opposite of hypertension.
Collapse
Affiliation(s)
- Verdiana Ravarotto
- Unit of Nephrology, Dialysis and Transplantation, Department of Medicine, University of Padua, Padua, Italy
| | - Giovanni Bertoldi
- Unit of Nephrology, Dialysis and Transplantation, Department of Medicine, University of Padua, Padua, Italy
| | - Matteo Rigato
- Unit of Nephrology, Dialysis and Transplantation, Department of Medicine, University of Padua, Padua, Italy
| | - Elisa Pagnin
- Unit of Nephrology, Dialysis and Transplantation, Department of Medicine, University of Padua, Padua, Italy
| | - Laura Gobbi
- Unit of Nephrology, Dialysis and Transplantation, Department of Medicine, University of Padua, Padua, Italy
| | - Paul A Davis
- Department of Nutrition, University of California at Davis, Davis, CA, USA
| | - Lorenzo A Calò
- Unit of Nephrology, Dialysis and Transplantation, Department of Medicine, University of Padua, Padua, Italy -
| |
Collapse
|
13
|
Murdaca G, Noberasco G, Olobardi D, Ogliastro M, Sibilio R, Sambuceti G, Balzano R, Sticchi L, Icardi G, Orsi A. Systemic sclerosis and vaccinations: a three-year register-based cohort study about vaccination rate and uptake from Liguria referral center, northwest Italy. Hum Vaccin Immunother 2022; 18:2025732. [PMID: 35258440 PMCID: PMC8993060 DOI: 10.1080/21645515.2022.2025732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Patients with diffused Systemic Sclerosis (dSSc) are more subject to severe respiratory complications with higher rates of intensive care unit (ICU) admission. Vaccination represents the most effective means of prevention and care for frail patients, such as SSc patients, preventing infections, reducing mortality and morbidity, and granting a better quality of life. Both vaccinations against seasonal influenza and Streptococcus pneumoniae are currently recommended by the European League Against Rheumatism (EULAR) guidelines on vaccination. The aim of this study is to give an updated analysis on S. pneumoniae and seasonal influenza vaccination coverage in a cohort of 91 patients with SSc and to investigate demographic and clinical variables significantly related to vaccine acceptance. The correlation between vaccine administration and other factors was investigated using a binomial logistic regression to evaluate the adjusted odds ratio (aOR). The patients followed up in this study reached higher percentages than the general population, passing the 75% target for both influenza and anti-pneumococcal vaccinations and reaching for influenza vaccine coverage rates of 83.8% for subjects undergoing immunosuppressive therapies and 88.9% for elderly subjects. For the latter group, it is important to emphasize the strong correlation between older age groups and vaccination acceptance.
Collapse
Affiliation(s)
- Giuseppe Murdaca
- Departments of Internal Medicine, University of Genova, Genova, Italy
| | - Giovanni Noberasco
- Department of Health Sciences, Vaccines and Clinical Trials Unit, University of Genova, Genova, Italy
| | - Dario Olobardi
- Department of Health Sciences, Vaccines and Clinical Trials Unit, University of Genova, Genova, Italy
| | - Matilde Ogliastro
- Department of Health Sciences, Vaccines and Clinical Trials Unit, University of Genova, Genova, Italy
| | - Raffaella Sibilio
- Department of Health Sciences, Vaccines and Clinical Trials Unit, University of Genova, Genova, Italy
| | - Giacomo Sambuceti
- Departments of Internal Medicine, University of Genova, Genova, Italy
| | - Riccardo Balzano
- Departments of Internal Medicine, University of Genova, Genova, Italy
| | - Laura Sticchi
- Department of Health Sciences, Vaccines and Clinical Trials Unit, University of Genova, Genova, Italy.,Hygiene Unit, "Ospedale Policlinico San Martino IRCCS", Genova, Italy
| | - Giancarlo Icardi
- Department of Health Sciences, Vaccines and Clinical Trials Unit, University of Genova, Genova, Italy.,Hygiene Unit, "Ospedale Policlinico San Martino IRCCS", Genova, Italy
| | - Andrea Orsi
- Department of Health Sciences, Vaccines and Clinical Trials Unit, University of Genova, Genova, Italy.,Hygiene Unit, "Ospedale Policlinico San Martino IRCCS", Genova, Italy
| |
Collapse
|
14
|
Jin W, Zheng Y, Zhu P. T cell abnormalities in systemic sclerosis. Autoimmun Rev 2022; 21:103185. [PMID: 36031049 DOI: 10.1016/j.autrev.2022.103185] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 11/02/2022]
Abstract
Systemic sclerosis (SSc) is an autoimmune disease with a poor prognosis. To date, the pathogenesis of SSc is still unclear; moreover, its pathological conditions include microvascular damage, inflammation, and immune abnormalities. Different types of T cells may cause vasculitis and fibrosis in SSc by means of up- and down-regulation of cell surface molecules, abnormal release of pro-fibrotic or pro-inflammatory cytokines and direct contact with fibroblasts. These T cells, which are mainly CD4 + T cells, include the subtypes, T follicular helper (Tfh) cells, regulatory T Cells (Treg), interleukin-17 (IL-17)-producing Th17 cells, CD4+ cytotoxic T lymphocytes (CTLs), and angiogenic T (Tang) cells. In addition to the Th1/Th2 imbalance, which has long been established, there is also a Th17/Treg imbalance in SSc. This imbalance may be closely related to the abnormal immune status of SSc. There is mounting evidence that suggest T cell abnormalities may be crucial to the pathogenesis of SSc. In terms of treatment, existing therapies that target T cells, such as immunosuppressive therapy (tacrolimus), Janus kinase(JAK) inhibitors, and biologics(abatacept), have had some success. Other non-drug therapies, including Mesenchymal stem cells (MSCs), have extensive and complex mechanisms of action actually including T cell regulation. Based on the current evidence, we believe that the study of T cells will further our understanding of the pathogenesis of SSc, and may lead to more targeted treatment optionsfor patients with SSc.
Collapse
Affiliation(s)
- Wei Jin
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | - Yan Zheng
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China; National Translational Science Center for Molecular Medicine, Xi'an, PR China
| | - Ping Zhu
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China; National Translational Science Center for Molecular Medicine, Xi'an, PR China.
| |
Collapse
|
15
|
Díaz-Flores L, Gutiérrez R, García MP, González-Gómez M, Díaz-Flores L, Carrasco JL, Madrid JF, Rodríguez Bello A. Comparison of the Behavior of Perivascular Cells (Pericytes and CD34+ Stromal Cell/Telocytes) in Sprouting and Intussusceptive Angiogenesis. Int J Mol Sci 2022; 23:ijms23169010. [PMID: 36012273 PMCID: PMC9409369 DOI: 10.3390/ijms23169010] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Perivascular cells in the pericytic microvasculature, pericytes and CD34+ stromal cells/telocytes (CD34+SCs/TCs), have an important role in angiogenesis. We compare the behavior of these cells depending on whether the growth of endothelial cells (ECs) from the pre-existing microvasculature is toward the interstitium with vascular bud and neovessel formation (sprouting angiogenesis) or toward the vascular lumen with intravascular pillar development and vessel division (intussusceptive angiogenesis). Detachment from the vascular wall, mobilization, proliferation, recruitment, and differentiation of pericytes and CD34+SCs/TCs, as well as associated changes in vessel permeability and functionality, and modifications of the extracellular matrix are more intense, longer lasting over time, and with a greater energy cost in sprouting angiogenesis than in intussusceptive angiogenesis, in which some of the aforementioned events do not occur or are compensated for by others (e.g., sparse EC and pericyte proliferation by cell elongation and thinning). The governing mechanisms involve cell-cell contacts (e.g., peg-and-socket junctions between pericytes and ECs), multiple autocrine and paracrine signaling molecules and pathways (e.g., vascular endothelial growth factor, platelet-derived growth factor, angiopoietins, transforming growth factor B, ephrins, semaphorins, and metalloproteinases), and other factors (e.g., hypoxia, vascular patency, and blood flow). Pericytes participate in vessel development, stabilization, maturation and regression in sprouting angiogenesis, and in interstitial tissue structure formation of the pillar core in intussusceptive angiogenesis. In sprouting angiogenesis, proliferating perivascular CD34+SCs/TCs are an important source of stromal cells during repair through granulation tissue formation and of cancer-associated fibroblasts (CAFs) in tumors. Conversely, CD34+SCs/TCs have less participation as precursor cells in intussusceptive angiogenesis. The dysfunction of these mechanisms is involved in several diseases, including neoplasms, with therapeutic implications.
Collapse
Affiliation(s)
- Lucio Díaz-Flores
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38071 Tenerife, Spain
- Correspondence: ; Tel.: +34-922-319317; Fax: +34-922-319279
| | - Ricardo Gutiérrez
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38071 Tenerife, Spain
| | - Maria Pino García
- Department of Pathology, Eurofins Megalab–Hospiten Hospitals, 38100 Tenerife, Spain
| | - Miriam González-Gómez
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38071 Tenerife, Spain
- Instituto de Tecnologías Biomédicas de Canarias, University of La Laguna, 38071 Tenerife, Spain
| | - Lucio Díaz-Flores
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38071 Tenerife, Spain
| | - Jose Luis Carrasco
- Department of Basic Medical Sciences, Faculty of Medicine, University of La Laguna, 38071 Tenerife, Spain
| | - Juan Francisco Madrid
- Department of Cell Biology and Histology, School of Medicine, Campus of International Excellence “Campus Mare Nostrum”, IMIB-Arrixaca, University of Murcia, 30120 Murcia, Spain
| | - Aixa Rodríguez Bello
- Department of Bioquímica, Microbiología, Biología Celular y Genética, University of La Laguna, 38071 Tenerife, Spain
| |
Collapse
|
16
|
Erratico S, Belicchi M, Meregalli M, Di Silvestre D, Tripodi L, De Palma A, Jones R, Ferrari E, Porretti L, Trombetta E, Merlo GR, Mauri P, Torrente Y. Effective high-throughput isolation of enriched platelets and circulating pro-angiogenic cells to accelerate skin-wound healing. Cell Mol Life Sci 2022; 79:259. [PMID: 35474498 PMCID: PMC9042989 DOI: 10.1007/s00018-022-04284-4] [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: 01/27/2022] [Revised: 04/01/2022] [Accepted: 04/01/2022] [Indexed: 11/30/2022]
Abstract
Delayed wound healing and chronic skin lesions represent a major health problem. Over the past years, growth factors mediated by platelet-rich plasma (PRP) and cell-based therapies were developed as effective and affordable treatment able to improve wound healing capacity. We have advanced existing concepts to develop a highly efficient high-throughput protocol with proven application for the isolation of PRP and pro-angiogenic cells (AngioPRP). This protocol outlines the effectiveness of AngioPRP in promoting the critical healing process including wound closure, re-epithelialization, granulation tissue growth, and blood vessel regeneration. We coupled this effect with normalization of mechanical properties of rescued mouse wounds, which is sustained by a correct arrangement of elastin and collagen fibers. Proteomic analysis of treated wounds demonstrated a fingerprint of AngioPRP based on the up-regulation of detoxification pathway of glutathione metabolism, correlated to a decrease in inflammatory response. Overall, these results have enabled us to provide a framework for how AngioPRP supports wound healing, opening avenues for further clinical advances.
Collapse
Affiliation(s)
| | - Marzia Belicchi
- Unit of Neurology, Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Universitá degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, via Francesco Sforza 35, 20122, Milan, Italy
| | - Mirella Meregalli
- Unit of Neurology, Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Universitá degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, via Francesco Sforza 35, 20122, Milan, Italy
| | - Dario Di Silvestre
- Institute of Technologies in Biomedicine, National Research Council (ITB-CNR), Via Fratelli Cervi, 93, Segrate, 20090, Milan, Italy
| | - Luana Tripodi
- Novystem Spa, viale Piave 21, 20129, Milan, Italy.,Unit of Neurology, Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Universitá degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, via Francesco Sforza 35, 20122, Milan, Italy
| | - Antonella De Palma
- Institute of Technologies in Biomedicine, National Research Council (ITB-CNR), Via Fratelli Cervi, 93, Segrate, 20090, Milan, Italy
| | - Rebecca Jones
- Department of Molecular Biotechnology and Health Science, University of Torino, Via Nizza 52, 10126, Turin, Italy
| | - Emanuele Ferrari
- Institute of Technologies in Biomedicine, National Research Council (ITB-CNR), Via Fratelli Cervi, 93, Segrate, 20090, Milan, Italy
| | - Laura Porretti
- Flow Cytometry Service, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122, Milan, Italy
| | - Elena Trombetta
- Flow Cytometry Service, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, 20122, Milan, Italy
| | - Giorgio R Merlo
- Department of Molecular Biotechnology and Health Science, University of Torino, Via Nizza 52, 10126, Turin, Italy
| | - Pierluigi Mauri
- Institute of Technologies in Biomedicine, National Research Council (ITB-CNR), Via Fratelli Cervi, 93, Segrate, 20090, Milan, Italy
| | - Yvan Torrente
- Unit of Neurology, Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Universitá degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, via Francesco Sforza 35, 20122, Milan, Italy.
| |
Collapse
|
17
|
Moschetti L, Piantoni S, Vizzardi E, Sciatti E, Riccardi M, Franceschini F, Cavazzana I. Endothelial Dysfunction in Systemic Lupus Erythematosus and Systemic Sclerosis: A Common Trigger for Different Microvascular Diseases. Front Med (Lausanne) 2022; 9:849086. [PMID: 35462989 PMCID: PMC9023861 DOI: 10.3389/fmed.2022.849086] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
This review describes the complex interplay between inflammation, vasculopathy and fibrosis that involve the heart and peripheral small vessels, leading to endothelial stiffness, vascular damage, and early aging in patients with systemic lupus erythematosus and systemic sclerosis, which represents two different models of vascular dysfunction among systemic autoimmune diseases. In fact, despite the fact that diagnostic methods and therapies have been significantly improved in the last years, affected patients show an excess of cardiovascular mortality if compared with the general population. In addition, we provide a complete overview on the new techniques which are used for the evaluation of endothelial dysfunction in a preclinical phase, which could represent a new approach in the assessment of cardiovascular risk in these patients.
Collapse
Affiliation(s)
- Liala Moschetti
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Silvia Piantoni
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- *Correspondence: Silvia Piantoni,
| | - Enrico Vizzardi
- Cardiology Unit, ASST Spedali Civili of Brescia, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | | | - Mauro Riccardi
- Cardiology Unit, ASST Spedali Civili of Brescia, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Franco Franceschini
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Ilaria Cavazzana
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
18
|
Angiogenic T Cells: Potential Biomarkers for the Early Diagnosis of Interstitial Lung Disease in Autoimmune Diseases? Biomedicines 2022; 10:biomedicines10040851. [PMID: 35453601 PMCID: PMC9026324 DOI: 10.3390/biomedicines10040851] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/02/2022] [Accepted: 04/04/2022] [Indexed: 02/06/2023] Open
Abstract
(1) Background: We explored, for the first time, the contribution of angiogenic T cells (TAng) in interstitial lung disease associated to autoimmune disease (AD-ILD+) as potential biomarkers of the disease, evaluating their role in the underlying vasculopathy and lung fibrosis. Additionally, the relationship of TAng with clinical manifestations and cellular and molecular endothelial dysfunction-related biomarkers was assessed. (2) Methods: We included 57 AD-ILD+ patients (21 with rheumatoid arthritis (RA)-ILD+, 21 with systemic sclerosis (SSc)-ILD+ and 15 with other AD-ILD+) and three comparative groups: 45 AD-ILD− patients (25 RA-ILD− and 20 SSc-ILD−); 21 idiopathic pulmonary fibrosis (IPF) patients; 21 healthy controls (HC). TAng were considered as CD3+CD184+CD31+ by flow cytometry. (3) Results: A similar TAng frequency was found between AD-ILD+ and IPF, being in both cases lower than that observed in AD-ILD− and HC. A lower TAng frequency was associated with negative Scl-70 status and lower FEV1/FVC ratio in SSc-ILD+, as well as with men in RA-ILD+ and non-specific interstitial pneumonia radiological pattern in other AD-ILD+. No relationship between TAng and endothelial progenitor cells, endothelial cells and vascular endothelial growth factor gene expression and protein levels was disclosed. (4) Conclusions: Our findings suggest TAng as potential biomarkers for the early diagnosis of ILD in AD.
Collapse
|
19
|
Rodríguez-Carrio J, Carrillo-López N, Ulloa C, Martín-Carro B, Rodríguez-Suárez C, Naves-Díaz M, Sánchez-Álvarez E, Rodríguez-García M, Arcidiacono MV, Fernández-Mariño B, Cannata-Andía JB, Suárez A, Dusso AS. Novel Immune Cell Subsets Exhibit Different Associations With Vascular Outcomes in Chronic Kidney Disease Patients-Identifying Potential Biomarkers. Front Med (Lausanne) 2021; 8:618286. [PMID: 34113627 PMCID: PMC8185045 DOI: 10.3389/fmed.2021.618286] [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: 10/16/2020] [Accepted: 04/07/2021] [Indexed: 11/17/2022] Open
Abstract
Background and Aims: Alterations in novel immune cell subsets, such as angiogenic T cells (Tang), senescent T cells (CD4+CD28null), and monocyte subsets are associated with impaired vascular homeostasis in several inflammatory conditions. However, mediators underlying vascular deterioration in chronic kidney disease (CKD) are poorly characterized. This study assessed their role in the vascular deterioration of CKD using a broad spectrum of surrogate markers ranging from altered functionality to overt calcification. Methods: Tang (CD3+CD31+CXCR4+), CD4+CD28null cells, and monocytes [CD14/CD16 subsets and angiotensin-converting enzyme (ACE) expression] were measured in peripheral blood by flow cytometry in 33 CKD stage 5 patients undergoing peritoneal dialysis (CKD5-PD) and 15 healthy controls (HCs). Analyses were replicated in a hemodialysis cohort. Vascular surrogate markers (including adventitial vasa vasorum, pulse wave velocity, intima-media thickness, and vascular calcification) were assessed by appropriate imaging methods. Results: In CKD5-PD, decreased Tang levels (p < 0.001) were unrelated to clinical features or traditional cardiovascular (CV) risk factors but correlated negatively with troponin T levels (r = −0.550, p = 0.003). Instead, CD4+CD28null frequency was increased (p < 0.001), especially in those with vascular calcifications. Quantitative and qualitative differences were also observed within the monocyte pool, a shift toward CD16+ subsets and ACE expression being found in CKD. Equivalent results were observed in the replication cohort. Each subset associated distinctly with adverse vascular outcomes in univariate and multivariate analyses: while Tang depletion was linked to poor vascular function and subclinical atherosclerosis, increases in CD4+CD28null were associated with overt vascular thickening and calcification. Monocytes were not independently associated with vascular outcomes in CKD patients. Conclusions: Novel T cell and monocyte subsets are altered in CKD. Altered T-cell subpopulations, but not monocytes, exhibited distinct associations with different vascular outcomes in CKD. Tang are emerging biomarkers of subclinical vascular deterioration in CKD.
Collapse
Affiliation(s)
- Javier Rodríguez-Carrio
- Area of Immunology, Department of Functional Biology, University of Oviedo, Oviedo, Spain.,Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Natalia Carrillo-López
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,REDinREN-ISCIII, Oviedo, Spain
| | - Catalina Ulloa
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain.,Department of Nephrology, Hospital Universitario de Cabueñes, Gijón, Spain
| | - Beatriz Martín-Carro
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,REDinREN-ISCIII, Oviedo, Spain
| | | | - Manuel Naves-Díaz
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,REDinREN-ISCIII, Oviedo, Spain
| | - Emilio Sánchez-Álvarez
- REDinREN-ISCIII, Oviedo, Spain.,Department of Nephrology, Hospital Universitario de Cabueñes, Gijón, Spain
| | - Minerva Rodríguez-García
- REDinREN-ISCIII, Oviedo, Spain.,Division of Nephrology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | | | - Jorge B Cannata-Andía
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,REDinREN-ISCIII, Oviedo, Spain.,Department of Medicine, University of Oviedo, Oviedo, Spain
| | - Ana Suárez
- Area of Immunology, Department of Functional Biology, University of Oviedo, Oviedo, Spain.,Department of Medicine, University of Oviedo, Oviedo, Spain
| | - Adriana S Dusso
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,REDinREN-ISCIII, Oviedo, Spain.,Department of Medicine, University of Oviedo, Oviedo, Spain
| |
Collapse
|
20
|
Menzel L, Höpken UE, Rehm A. Angiogenesis in Lymph Nodes Is a Critical Regulator of Immune Response and Lymphoma Growth. Front Immunol 2020; 11:591741. [PMID: 33343570 PMCID: PMC7744479 DOI: 10.3389/fimmu.2020.591741] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor-induced remodeling of the microenvironment in lymph nodes (LNs) includes the formation of blood vessels, which goes beyond the regulation of metabolism, and shaping a survival niche for tumor cells. In contrast to solid tumors, which primarily rely on neo-angiogenesis, hematopoietic malignancies usually grow within pre-vascularized autochthonous niches in secondary lymphatic organs or the bone marrow. The mechanisms of vascular remodeling in expanding LNs during infection-induced responses have been studied in more detail; in contrast, insights into the conditions of lymphoma growth and lodging remain enigmatic. Based on previous murine studies and clinical trials in human, we conclude that there is not a universal LN-specific angiogenic program applicable. Instead, signaling pathways that are tightly connected to autochthonous and infiltrating cell types contribute variably to LN vascular expansion. Inflammation related angiogenesis within LNs relies on dendritic cell derived pro-inflammatory cytokines stimulating vascular endothelial growth factor-A (VEGF-A) expression in fibroblastic reticular cells, which in turn triggers vessel growth. In high-grade B cell lymphoma, angiogenesis correlates with poor prognosis. Lymphoma cells immigrate and grow in LNs and provide pro-angiogenic growth factors themselves. In contrast to infectious stimuli that impact on LN vasculature, they do not trigger the typical inflammatory and hypoxia-related stroma-remodeling cascade. Blood vessels in LNs are unique in selective recruitment of lymphocytes via high endothelial venules (HEVs). The dissemination routes of neoplastic lymphocytes are usually disease stage dependent. Early seeding via the blood stream requires the expression of the homeostatic chemokine receptor CCR7 and of L-selectin, both cooperate to facilitate transmigration of tumor and also of protective tumor-reactive lymphocytes via HEV structures. In this view, the HEV route is not only relevant for lymphoma cell homing, but also for a continuous immunosurveillance. We envision that HEV functional and structural alterations during lymphomagenesis are not only key to vascular remodeling, but also impact on tumor cell accessibility when targeted by T cell-mediated immunotherapies.
Collapse
Affiliation(s)
- Lutz Menzel
- Translational Tumor Immunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Uta E. Höpken
- Microenvironmental Regulation in Autoimmunity and Cancer, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Armin Rehm
- Translational Tumor Immunology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
21
|
Ríos-Ríos WDJ, Sosa-Luis SA, Torres-Aguilar H. T Cells Subsets in the Immunopathology and Treatment of Sjogren's Syndrome. Biomolecules 2020; 10:E1539. [PMID: 33187265 PMCID: PMC7698113 DOI: 10.3390/biom10111539] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 02/06/2023] Open
Abstract
Sjogren´s syndrome (SS) is an autoimmune disease whose pathogenesis is characterized by an exacerbated T cell infiltration in exocrine glands, markedly associated to the inflammatory and detrimental features as well as the disease progression. Several helper T cell subsets sequentially converge at different stages of the ailment, becoming involved in specific pathologic roles. Initially, their activated phenotype endows them with high migratory properties and increased pro-inflammatory cytokine secretion in target tissues. Later, the accumulation of immunomodulatory T cells-derived factors, such as IL-17, IFN-γ, or IL-21, preserve the inflammatory environment. These effects favor strong B cell activation, instigating an extrafollicular antibody response in ectopic lymphoid structures mediated by T follicular helper cells (Tfh) and leading to disease progression. Additionally, the memory effector phenotype of CD8+ T cells present in SS patients suggests that the presence of auto-antigen restricted CD8+ T cells might trigger time-dependent and specific immune responses. Regarding the protective roles of traditional regulatory T cells (Treg), uncertain evidence shows decrease or invariable numbers of circulating and infiltrating cells. Nevertheless, an emerging Treg subset named follicular regulatory T cells (Tfr) seems to play a critical protective role owing to their deficiency that enhances SS development. In this review, the authors summarize the current knowledge of T cells subsets contribution to the SS immunopathology, focusing on the cellular and biomolecular properties allowing them to infiltrate and to harm target tissues, and that simultaneously make them key therapeutic targets for SS treatment.
Collapse
Affiliation(s)
- William de Jesús Ríos-Ríos
- Department of Clinical Immunology Research of Biochemical Sciences Faculty, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca City 68120, Mexico;
| | - Sorely Adelina Sosa-Luis
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City 07360, Mexico;
| | - Honorio Torres-Aguilar
- Department of Clinical Immunology Research of Biochemical Sciences Faculty, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca City 68120, Mexico;
| |
Collapse
|
22
|
Bortoluzzi A, Chighizola CB, Fredi M, Raschi E, Bodio C, Privitera D, Gonelli A, Silvagni E, Govoni M, Cavazzana I, Airò P, Meroni PL, Tincani A, Franceschini F, Piantoni S, Casciano F. The IMMENSE Study: The Interplay Between iMMune and ENdothelial Cells in Mediating Cardiovascular Risk in Systemic Lupus Erythematosus. Front Immunol 2020; 11:572876. [PMID: 33193356 PMCID: PMC7658008 DOI: 10.3389/fimmu.2020.572876] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/25/2020] [Indexed: 12/12/2022] Open
Abstract
Patients with systemic lupus erythematosus (SLE) have a significant increase in cardiovascular (CV) risk although they display a preserved number of circulating angiogenic CD3+CD31+CXCR4+ T cells (Tang), a subpopulation of T cells which promotes repair of damaged endothelium. This happens due to the concomitant expansion of a Tang subset with immunosenescent features, such as the loss of CD28. Therefore, the aim of this study was to elucidate the interplay between Tang subpopulations and endothelial cells in a group of young SLE patients without previous cardiovascular events. Twenty SLE female patients and 10 healthy controls (HCs) were recruited. Flow cytometric analysis of endothelial progenitor cells (EPCs) and Tang subsets were performed and serum levels of interleukin (IL)-6, -8, matrix metalloproteinase (MMP)-9 and interferon (IFN)-γ were measured. Human umbilical vein endothelial cells (HUVECs) proliferation and pro-inflammatory phenotype in response to subjects' serum stimulation were also evaluated. Results showed that the percentage of Tang and EPC subsets was reduced in SLE patients compared with HCs, with a marked increase of senescent CD28null cells among Tang subset. SLE disease activity index-2000 (SLEDAI-2K) was inversed related to Tang cells percentage. Furthermore, IL-8 serum levels were directly correlated with the percentage of Tang and inversely related to the CD28null Tang subsets. We indirectly evaluated the role of the Tang subset on the endothelium upon stimulation with serum from subjects with a low percentage of Tang CD3+ cells in HUVECs. HUVECs displayed pro-inflammatory phenotype with up-regulation of mRNA for IL-6, intercellular adhesion molecule (ICAM)-1 and endothelial leukocyte adhesion molecule (ELAM)-1. Cell proliferation rate was directly related to IL-8 serum levels and EPC percentage. In highly selected young SLE patients without previous CV events, we found that the deterioration of Tang compartment is an early event in disease course, preceding the development of an overt cardiovascular disease and potentially mediated by SLE-specific mechanisms. The overcome of the CD28null subset exerts detrimental role over the Tang phenotype, where Tang could exert an anti-inflammatory effect on endothelial cells and might orchestrate via IL-8 the function of EPCs, ultimately modulating endothelial proliferation rate.
Collapse
Affiliation(s)
- Alessandra Bortoluzzi
- Rheumatology Unit, Department of Medical Sciences, University of Ferrara and Azienda Ospedaliero-Universitaria Sant’Anna, Cona, Italy
| | - Cecilia Beatrice Chighizola
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Micaela Fredi
- Rheumatology and Clinical Immunology Unit, Department of Clinical and Experimental Sciences, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Elena Raschi
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Caterina Bodio
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Daniela Privitera
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Arianna Gonelli
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| | - Ettore Silvagni
- Rheumatology Unit, Department of Medical Sciences, University of Ferrara and Azienda Ospedaliero-Universitaria Sant’Anna, Cona, Italy
| | - Marcello Govoni
- Rheumatology Unit, Department of Medical Sciences, University of Ferrara and Azienda Ospedaliero-Universitaria Sant’Anna, Cona, Italy
| | - Ilaria Cavazzana
- Rheumatology and Clinical Immunology Unit, Department of Clinical and Experimental Sciences, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Paolo Airò
- Rheumatology and Clinical Immunology Unit, Department of Clinical and Experimental Sciences, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Pier Luigi Meroni
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Angela Tincani
- Rheumatology and Clinical Immunology Unit, Department of Clinical and Experimental Sciences, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Franco Franceschini
- Rheumatology and Clinical Immunology Unit, Department of Clinical and Experimental Sciences, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Silvia Piantoni
- Rheumatology and Clinical Immunology Unit, Department of Clinical and Experimental Sciences, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Fabio Casciano
- Department of Morphology, Surgery and Experimental Medicine and LTTA Centre, University of Ferrara, Ferrara, Italy
| |
Collapse
|
23
|
Murdaca G, Noberasco G, Battaglini A, Vassallo C, Giusti F, Greco M, Schiavi C, Sticchi L, Icardi G, Orsi A. Systemic Sclerosis and Vaccinations: A Register-Based Cohort Study about Seasonal Influenza and Streptococcus pneumoniae Vaccination Rate and Uptake from Liguria Regional Center, Northwest Italy. Vaccines (Basel) 2020; 8:vaccines8020204. [PMID: 32354027 PMCID: PMC7349884 DOI: 10.3390/vaccines8020204] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/31/2022] Open
Abstract
Systemic sclerosis (SSc) is the connective tissue disease with the highest mortality and patients with chronic inflammatory immune-mediated diseases are at high risk of acquiring infections as they are often treated with immunosuppressive or biological drugs. This study, conducted among the patients followed by our clinical immunology, part of the Internal Medicine Department in the Ospedale Policlinico San Martino, Genoa, northwest Italy, has set itself the primary objective of analyzing the vaccine uptake and the vaccination coverage against both seasonal influenza and S. pneumoniae in a cohort of patients with SSc. We evaluated the influenza and pneumococcal vaccination rate among various subgroups of patients and the source of the recommendation for vaccination. We evaluated the vaccination rate changes between the two years considered in our study. We also calculated a binomial logistic regression between vaccination acceptance and clinical and demographics characteristics of the patients to evaluate the adjusted odds ratio (OR) of each factor on vaccination. The vaccination coverage that resulted was significantly higher than in other similar studies. Age over 65 years old, interstitial lung disease, and ongoing immunosuppressive therapy were significantly related with acceptance to both vaccinations using univariate analyses, but the multivariate logistic regression found a significant correlation only with the age and therapy factors.
Collapse
Affiliation(s)
- Giuseppe Murdaca
- Departments of Internal Medicine, Scleroderma Unit, Clinical Immunology Unit, University of Genova, 16132 Genova, Italy; (C.V.); (F.G.); (M.G.); (C.S.)
- Correspondence: ; Tel.: +39-0105554676
| | - Giovanni Noberasco
- Department of Health Sciences, Vaccines and Clinical Trials Unit, University of Genova, 16132 Genova, Italy; (G.N.); (A.B.); (L.S.); (G.I.); (A.O.)
| | - Alberto Battaglini
- Department of Health Sciences, Vaccines and Clinical Trials Unit, University of Genova, 16132 Genova, Italy; (G.N.); (A.B.); (L.S.); (G.I.); (A.O.)
| | - Chiara Vassallo
- Departments of Internal Medicine, Scleroderma Unit, Clinical Immunology Unit, University of Genova, 16132 Genova, Italy; (C.V.); (F.G.); (M.G.); (C.S.)
| | - Francesca Giusti
- Departments of Internal Medicine, Scleroderma Unit, Clinical Immunology Unit, University of Genova, 16132 Genova, Italy; (C.V.); (F.G.); (M.G.); (C.S.)
| | - Monica Greco
- Departments of Internal Medicine, Scleroderma Unit, Clinical Immunology Unit, University of Genova, 16132 Genova, Italy; (C.V.); (F.G.); (M.G.); (C.S.)
| | - Chiara Schiavi
- Departments of Internal Medicine, Scleroderma Unit, Clinical Immunology Unit, University of Genova, 16132 Genova, Italy; (C.V.); (F.G.); (M.G.); (C.S.)
| | - Laura Sticchi
- Department of Health Sciences, Vaccines and Clinical Trials Unit, University of Genova, 16132 Genova, Italy; (G.N.); (A.B.); (L.S.); (G.I.); (A.O.)
- Hygiene Unit, “Ospedale Policlinico San Martino IRCCS”, 16132 Genova, Italy
| | - Giancarlo Icardi
- Department of Health Sciences, Vaccines and Clinical Trials Unit, University of Genova, 16132 Genova, Italy; (G.N.); (A.B.); (L.S.); (G.I.); (A.O.)
- Hygiene Unit, “Ospedale Policlinico San Martino IRCCS”, 16132 Genova, Italy
| | - Andrea Orsi
- Department of Health Sciences, Vaccines and Clinical Trials Unit, University of Genova, 16132 Genova, Italy; (G.N.); (A.B.); (L.S.); (G.I.); (A.O.)
- Hygiene Unit, “Ospedale Policlinico San Martino IRCCS”, 16132 Genova, Italy
| |
Collapse
|
24
|
Tsai CY, Hsieh SC, Wu TH, Li KJ, Shen CY, Liao HT, Wu CH, Kuo YM, Lu CS, Yu CL. Pathogenic Roles of Autoantibodies and Aberrant Epigenetic Regulation of Immune and Connective Tissue Cells in the Tissue Fibrosis of Patients with Systemic Sclerosis. Int J Mol Sci 2020; 21:ijms21093069. [PMID: 32349208 PMCID: PMC7246753 DOI: 10.3390/ijms21093069] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/19/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Systemic sclerosis (SSc) is a multi-system autoimmune disease with tissue fibrosis prominent in the skin and lung. In this review, we briefly describe the autoimmune features (mainly autoantibody production and cytokine profiles) and the potential pathogenic contributors including genetic/epigenetic predisposition, and environmental factors. We look in detail at the cellular and molecular bases underlying tissue-fibrosis which include trans-differentiation of fibroblasts (FBs) to myofibroblasts (MFBs). We also state comprehensively the pro-inflammatory and pro-fibrotic cytokines relevant to MFB trans-differentiation, vasculopathy-associated autoantibodies, and fibrosis-regulating microRNAs in SSc. It is conceivable that tissue fibrosis is mainly mediated by an excessive production of TGF-β, the master regulator, from the skewed Th2 cells, macrophages, fibroblasts, myofibroblasts, and keratinocytes. After binding with TGF-β receptors on MFB, the downstream Wnt/β-catenin triggers canonical Smad 2/3 and non-canonical Smad 4 signaling pathways to transcribe collagen genes. Subsequently, excessive collagen fiber synthesis and accumulation as well as tissue fibrosis ensue. In the later part of this review, we discuss limited data relevant to the role of long non-coding RNAs (lncRNAs) in tissue-fibrosis in SSc. It is expected that these lncRNAs may become the useful biomarkers and therapeutic targets for SSc in the future. The prospective investigations in the development of novel epigenetic modifiers are also suggested.
Collapse
Affiliation(s)
- Chang-Youh Tsai
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital & National Yang-Ming University, #201 Sec. 2, Shih-Pai Road, Taipei 11217, Taiwan;
- Correspondence: (C.-Y.T.); (C.-L.Y.); Fax: +886-2-28717483 (C.-Y.T.); +886-2-23957801 (C.-L.Y.)
| | - Song-Chou Hsieh
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
| | - Tsai-Hung Wu
- Division of Nephrology, Taipei Veterans General Hospital & National Yang-Ming University, #201 Sec. 2, Shih-Pai Road, Taipei 11217, Taiwan;
| | - Ko-Jen Li
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
| | - Chieh-Yu Shen
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Hsien-Tzung Liao
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital & National Yang-Ming University, #201 Sec. 2, Shih-Pai Road, Taipei 11217, Taiwan;
| | - Cheng-Han Wu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Yu-Min Kuo
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Cheng-Shiun Lu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
- Institute of Clinical Medicine, National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan
| | - Chia-Li Yu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, #7 Chung-Shan South Road, Taipei 10002, Taiwan; (S.-C.H.); (K.-J.L.); (C.-Y.S.); (C.-H.W.); (Y.-M.K.); (C.-S.L.)
- Correspondence: (C.-Y.T.); (C.-L.Y.); Fax: +886-2-28717483 (C.-Y.T.); +886-2-23957801 (C.-L.Y.)
| |
Collapse
|
25
|
Lv T, Yang F, Zhang K, Lv M, Zhang Y, Zhu P. The risk of circulating angiogenic T cells and subsets in patients with systemic sclerosis. Int Immunopharmacol 2020; 81:106282. [PMID: 32066116 DOI: 10.1016/j.intimp.2020.106282] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/19/2020] [Accepted: 02/02/2020] [Indexed: 01/11/2023]
Abstract
To ascertain the number and percentage of angiogenic T (Tang) cell subsets by flow cytometry in systemic sclerosis (SSc), and their relation with specific clinical features. Thirty SSc patients and 15 healthy controls (HCs) were included. Luminex was performed to analyze the levels of interleukin (IL)-17A, vascular endothelial growth factor (VEGF), tumor necrosis factor-α, and vascular cell adhesion molecule (VCAM). The ratio of circulating CD3 + CD31 + CXCR4 + T (CD3 + Tang) cells and CD8+ CD31 + CXCR4 + T (CD8+ Tang) cells in SSc patients was enlarger than in HCs, while CD4 + CD31 + CXCR4 + T cells (CD4 + Tang) exhibited no difference between SSc patients and HCs. The number and percentage of Tang cells were higher in SSc patients with pulmonary artery hypertension (PAH) than in non-PAH SSc patients and HCs. The ratios of Tang cell subsets in nucleolar pattern-positive SSc patients were markedly raised as compared with their negative ones and HCs. Additionally, the percentage of circulating CD3 + Tang cells was positively associated with VEGF serum levels in SSc patients. Meanwhile, the rate of CD8+ tang cells might have been emphatically corresponded to VEGF and VCAM serum levels in SSc patients. These results imply that the increase in Tang cells in peripheral blood are associated with immunoregulatory disturbances in SSc patients.
Collapse
Affiliation(s)
- Tingting Lv
- Department of Clinical Immunology, PLA Specialized Research Institute of Rheumatology & Immunology, Xijing Hospital, Fourth Military Medical University, No. 127 Changle West Road, Xi'an 710032, Shaanxi, China; Institute of Rheumatology, Tangdu Hospital, Fourth Military Medical University, No. 1 Xinsi Road, Xi'an 710038, Shaanxi, China
| | - Fengfan Yang
- Department of Clinical Immunology, PLA Specialized Research Institute of Rheumatology & Immunology, Xijing Hospital, Fourth Military Medical University, No. 127 Changle West Road, Xi'an 710032, Shaanxi, China
| | - Kui Zhang
- Department of Clinical Immunology, PLA Specialized Research Institute of Rheumatology & Immunology, Xijing Hospital, Fourth Military Medical University, No. 127 Changle West Road, Xi'an 710032, Shaanxi, China
| | - Minghua Lv
- Department of Clinical Immunology, PLA Specialized Research Institute of Rheumatology & Immunology, Xijing Hospital, Fourth Military Medical University, No. 127 Changle West Road, Xi'an 710032, Shaanxi, China
| | - Yan Zhang
- Institute of Rheumatology, Tangdu Hospital, Fourth Military Medical University, No. 1 Xinsi Road, Xi'an 710038, Shaanxi, China.
| | - Ping Zhu
- Department of Clinical Immunology, PLA Specialized Research Institute of Rheumatology & Immunology, Xijing Hospital, Fourth Military Medical University, No. 127 Changle West Road, Xi'an 710032, Shaanxi, China; National Translational Science Center for Molecular Medicine, Fourth Military Medical University, No. 169 Changle West Road, Xi'an 710032, Shaanxi, China.
| |
Collapse
|
26
|
Sciatti E, Cavazzana I, Vizzardi E, Bonadei I, Fredi M, Taraborelli M, Ferizi R, Metra M, Tincani A, Franceschini F. Systemic Lupus Erythematosus and Endothelial Dysfunction: A Close Relationship. Curr Rheumatol Rev 2020; 15:177-188. [PMID: 30474532 DOI: 10.2174/1573397115666181126105318] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/04/2018] [Accepted: 11/14/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND Accelerated atherosclerosis, responsible for premature cardiovascular disease, has been estimated to develop or progress in 10% of systemic lupus erythematosus (SLE) patients each year and to be 6-fold more frequent in SLE compared with the general population. The mechanisms underlying accelerated atherosclerosis in SLE are complex and involve classical and "non-classical" cardiovascular risk factors. Subclinical and disseminated atherosclerosis is associated with endothelial dysfunction and arterial stiffness. OBJECTIVE The aim of this review is to analyze the association between SLE and endothelial dysfunction. RESULTS AND CONCLUSION Different mechanisms have been proposed to explain the prevalence of endothelial dysfunction in SLE, which are briefly reported in this review: impaired clearance of apoptotic cells, oxidative stress markers, B cell activation with different circulating autoantibodies, different subtypes of T lymphocytes, cytokine cascade. Several studies and meta-analyses show a significant trend towards a prevalence of subclinical accelerated atherosclerosis in patients with SLE compared with healthy controls, since childhood. Based on general considerations, we suggest a multidisciplinary management to assess endothelial dysfunction at the diagnosis of the disease and to periodically search for and treat the traditional cardiovascular risk factors. Prospective studies are needed to confirm the benefits of this management.
Collapse
Affiliation(s)
- Edoardo Sciatti
- Cardiology Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Ilaria Cavazzana
- Rheumatology and Clinical Immunolgy Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Enrico Vizzardi
- Cardiology Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Ivano Bonadei
- Cardiology Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Micaela Fredi
- Rheumatology and Clinical Immunolgy Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Mara Taraborelli
- Internal Medicine Unit, ASST Franciacorta, Chiari, Brescia, Italy
| | - Romina Ferizi
- Cardiology Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Marco Metra
- Cardiology Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Angela Tincani
- Rheumatology and Clinical Immunolgy Unit, University and ASST Spedali Civili, Brescia, Italy
| | - Franco Franceschini
- Rheumatology and Clinical Immunolgy Unit, University and ASST Spedali Civili, Brescia, Italy
| |
Collapse
|
27
|
Zhao P, Miao J, Zhang K, Yu Z, Lv M, Xu Y, Fu X, Han Q, Zhu P. CD147 participates in the activation function of circulating angiogenic T cells in patients with rheumatoid arthritis. Clin Rheumatol 2019; 38:2621-2628. [DOI: 10.1007/s10067-019-04584-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/16/2019] [Accepted: 04/29/2019] [Indexed: 12/22/2022]
|
28
|
Cutolo M, Soldano S, Smith V. Pathophysiology of systemic sclerosis: current understanding and new insights. Expert Rev Clin Immunol 2019; 15:753-764. [PMID: 31046487 DOI: 10.1080/1744666x.2019.1614915] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Systemic sclerosis (SSc) is a complex autoimmune connective tissue disease characterized by chronic and progressive tissue and organ fibrosis with broad patient-to-patient variability. Some risk factors are known and include combination of persistent Raynaud's phenomenon, steroid hormone imbalance, selected chemicals, thermal, or other injuries. Endogenous and/or exogenous environmental trigger/risk factors promote epigenetic mechanisms in genetically primed subjects. Disease pathogenesis presents early microvascular changes with endothelial cell dysfunction, followed by the activation of mechanisms promoting their transition into myofibroblasts. A complex autoimmune response, involving innate and adaptive immunity with specific/functional autoantibody production, characterizes the disease. Progressive fibrosis and ischemia involve skin and visceral organs resulting in their irreversible damage/failure. Progenitor circulating cells (monocytes, fibrocytes), together with growth factors and cytokines participate in disease diffusion and evolution. Epigenetic, vascular and immunologic mechanisms implicated in systemic fibrosis, represent major targets for incoming disease modifying therapeutic approaches. Areas covered: This review discusses current understanding and new insights of SSc pathogenesis, through an overview of the most relevant advancements to present aspects and mechanisms involved in disease pathogenesis. Expert opinion: Considering SSc intricacy/heterogeneity, early combination therapy with vasodilators, immunosuppressive and antifibrotic drugs should successfully downregulate the disease progression, especially if started from the beginning.
Collapse
Affiliation(s)
- Maurizio Cutolo
- a Research Laboratory and Academic Unit of Clinical Rheumatology, Department of Internal Medicine , University of Genova, IRCCS San Martino Polyclinic Hospital Genova , Genova , Italy
| | - Stefano Soldano
- a Research Laboratory and Academic Unit of Clinical Rheumatology, Department of Internal Medicine , University of Genova, IRCCS San Martino Polyclinic Hospital Genova , Genova , Italy
| | - Vanessa Smith
- b Department of Internal Medicine , Ghent University , Ghent , Belgium.,c Department of Rheumatology , Ghent University Hospital , Ghent , Belgium.,d Unit for Molecular Immunology and Inflammation , VIB Inflammation Research Center (IRC) , Ghent , Belgium
| |
Collapse
|
29
|
Autologous hematopoietic stem cell transplantation in systemic sclerosis induces long-lasting changes in B cell homeostasis toward an anti-inflammatory B cell cytokine pattern. Arthritis Res Ther 2019; 21:106. [PMID: 31036055 PMCID: PMC6489316 DOI: 10.1186/s13075-019-1889-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/02/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Autologous hematopoietic stem cell transplantation (aHSCT) is performed in patients with aggressive forms of systemic sclerosis (SSc). The profile of B cell reconstitution after aHSCT is not fully understood. The aim of this study was to investigate changes of B cell subsets and cytokine production of B cells in patients with SSc after aHSCT. METHODS Peripheral blood of six patients with SSc was collected at defined intervals up to 16 months after aHSCT. Immunophenotyping was performed, and B cell function was determined by measuring cytokine secretion in supernatants of stimulated B cell cultures. RESULTS Within 1 month after aHSCT, a peak in the percentage of CD38++/CD10+/IgD+ transitional B cells and CD38++/CD27++/IgD- plasmablasts was detected. Long-term changes persisted up to 14 months after aHSCT and showed an increased percentage of total B cells; the absolute B cell number did not change significantly. Within the B cell compartment, an increased CD27/IgD+ naïve B cell percentage was found whereas decreased percentages of CD27+/IgD+ pre-switched memory, CD27+/IgD- post-switched memory, and CD27-/IgD- double-negative B cells were seen after aHSCT. Cytokine secretion in B cell cultures showed significantly increased IL-10 concentrations 13 to 16 months after aHSCT. CONCLUSION A changed composition of the B cell compartment is present for up to 14 months after aHSCT indicating positive persisting effects of aHSCT on B cell homeostasis. The cytokine secretion profile of B cells changes in the long term and shows an increased production of the immune regulatory cytokine IL-10 after aHSCT. These findings might promote the clinical improvements after aHSCT in SSc patients.
Collapse
|
30
|
Cavazzana I, Piantoni S, Sciatti E, Fredi M, Taraborelli M, Bonadei I, Airò P, Metra M, Tincani A, Franceschini F, Vizzardi E. Relationship between endothelial dysfunction, videocapillaroscopy and circulating CD3+CD31+CXCR4+ lymphocytes in systemic lupus erythematosus without cardiovascular risk factors. Lupus 2019; 28:210-216. [PMID: 30608206 DOI: 10.1177/0961203318821161] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE The objective of this paper is to analyse whether digital capillary morphology, analysed by nailfold videocapillaroscopy (NVC), and the number of circulating CD3 + CD31 + CXCR4 + lymphocytes (angiogenic T cells) could be markers of endothelial dysfunction (ED) in systemic lupus erythematosus (SLE) without cardiovascular disease (CVD) and CV risk factors. METHODS Nineteen consecutive SLE patients, according to Systemic Lupus International Collaborating Clinics Classification Criteria, with a disease duration less than five years, low disease activity, without CVD and CV risk factors (diabetes, chronic renal disease, uncontrolled systemic arterial hypertension, smoking, hypercholesterolemia, obesity), statin or beta-blocker use were enrolled. Each patient and sex- and age-matched healthy control (HC) underwent Doppler echocardiogram, an endothelial function study by peripheral arterial tonometry technique, NVC and peripheral blood immunophenotyping. RESULTS SLE ED+ more frequently showed NVC abnormalities compared with HCs ( p < 0.0001) in terms of minor alterations ( p = 0.017), lower capillary numbers ( p = 0.0035) and major alterations. SLE ED + showed a higher rate of CD3 + CD31 + CXCR4 + lymphocytes compared with SLE ED- and with HCs. NVC + SLE showed a significantly reduced rate of total CD3 + cells, but a higher rate and absolute number of CD3 + CD31 + CXCR4 + , compared with NVC- SLE. CONCLUSION NVC alterations are frequent in SLE without any CV risk factors and CVD. They are associated with ED and increased circulating CD3 + CD31 + CXCR4 + lymphocytes. These findings demonstrate a clear microvascular perturbation in patients with short disease duration, low disease activity and no CV risk factors.
Collapse
Affiliation(s)
- I Cavazzana
- 1 Rheumatology and Clinical Immunology Unit, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - S Piantoni
- 1 Rheumatology and Clinical Immunology Unit, ASST-Spedali Civili of Brescia, Brescia, Italy.,2 Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - E Sciatti
- 3 Cardiology Unit, ASST-Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - M Fredi
- 1 Rheumatology and Clinical Immunology Unit, ASST-Spedali Civili of Brescia, Brescia, Italy.,2 Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - M Taraborelli
- 4 Internal Medicine Unit, ASST Franciacorta, Chiari (Brescia), Italy
| | - I Bonadei
- 3 Cardiology Unit, ASST-Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - P Airò
- 1 Rheumatology and Clinical Immunology Unit, ASST-Spedali Civili of Brescia, Brescia, Italy
| | - M Metra
- 3 Cardiology Unit, ASST-Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - A Tincani
- 1 Rheumatology and Clinical Immunology Unit, ASST-Spedali Civili of Brescia, Brescia, Italy.,2 Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - F Franceschini
- 1 Rheumatology and Clinical Immunology Unit, ASST-Spedali Civili of Brescia, Brescia, Italy.,2 Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - E Vizzardi
- 3 Cardiology Unit, ASST-Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| |
Collapse
|
31
|
Brown M, O'Reilly S. The immunopathogenesis of fibrosis in systemic sclerosis. Clin Exp Immunol 2018; 195:310-321. [PMID: 30430560 DOI: 10.1111/cei.13238] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2018] [Indexed: 02/07/2023] Open
Abstract
Systemic sclerosis (SSc) is an idiopathic systemic autoimmune disease. It is characterized by a triad of hallmarks: immune dysfunction, fibrosis and vasculopathy. Immune dysfunction in SSc is characterized by the activation and recruitment of immune cells and the production of autoantibodies and cytokines. How immune abnormalities link the fibrosis and vasculopathy in SSc is poorly understood. A plethora of immune cell types are implicated in the immunopathogenesis of SSc, including T cells, B cells, dendritic cells, mast cells and macrophages. How these different cell types interact to contribute to SSc is complicated, and can involve cell-to-cell interactions and communication via cytokines, including transforming growth factor (TGF)-β, interleukin (IL)-6 and IL-4. We will attempt to review significant and recent research demonstrating the importance of immune cell regulation in the immunopathogenesis of SSc with a particular focus on fibrosis.
Collapse
Affiliation(s)
- M Brown
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - S O'Reilly
- Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| |
Collapse
|
32
|
|
33
|
Zhao P, Miao J, Zhang K, Lv M, Han Q, Zhu P. Circulating Angiogenic T Cells Are Increased in Lupus Nephritis Patients. Med Sci Monit 2018; 24:5384-5390. [PMID: 30072685 PMCID: PMC6087034 DOI: 10.12659/msm.908406] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 03/15/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Patients with systemic lupus erythematosus (SLE), especially with lupus nephritis (LN), undergo vascular damage and repair during the course of the disease. Since the recently identified angiogenic T cells (Tang) are involved in endothelial repair coupled with endothelial progenitor cells (EPCs), this study investigated the circulating Tang cells in LN patients and their potential correlations with disease features. MATERIAL AND METHODS Circulating Tang cells and EPCs were assessed by flow cytometry in peripheral blood samples from 67 SLE patients; of these, 32 had LN and 30 were matched healthy controls (HCs). The plasma levels of interleukin IL-17, IL-8, and vascular endothelial growth factor (VEGF) were quantified by immunoassays. RESULTS The percentage of circulating Tang cells in LN patients was significantly increased as compared to the non-LN patients and HCs, and they were positively correlated with the level of EPC and VEGF. Additionally, circulating Tang cell percentages were positively correlated with the extent of proteinuria in LN patients. CONCLUSIONS The increased levels of circulating Tang cells in LN patients might play a role in the balance of endothelium dysfunction in these patients.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, P.R. China
- Department of Hematology and Rheumatology, 3201 Hospital, Xi’an Jiaotong University Health Science Center, Hanzhong, Shaanxi, P.R. China
| | - Jinlin Miao
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, P.R. China
| | - Kui Zhang
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, P.R. China
| | - Minghua Lv
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, P.R. China
| | - Qing Han
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, P.R. China
| | - Ping Zhu
- Department of Clinical Immunology, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi, P.R. China
| |
Collapse
|
34
|
Del Papa N, Pignataro F. The Role of Endothelial Progenitors in the Repair of Vascular Damage in Systemic Sclerosis. Front Immunol 2018; 9:1383. [PMID: 29967618 PMCID: PMC6015881 DOI: 10.3389/fimmu.2018.01383] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 06/04/2018] [Indexed: 01/17/2023] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disease characterized by a complex pathological process where the main scenario is represented by progressive loss of microvascular bed, with the consequent progressive fibrotic changes in involved organ and tissues. Although most aspects of vascular injury in scleroderma are poorly understood, recent data suggest that the scleroderma impairment of neovascularization could be related to both angiogenesis and vasculogenesis failure. Particularly, compensatory angiogenesis does not occur normally in spite of an important increase in many angiogenic factors either in SSc skin or serum. Besides insufficient angiogenesis, the contribution of defective vasculogenesis to SSc vasculopathy has been extensively studied. Over the last decades, our understanding of the processes responsible for the formation of new vessels after tissue ischemia has increased. In the past, adult neovascularization was thought to depend mainly on angiogenesis (a process by which new vessels are formed by the proliferation and migration of mature endothelial cells). More recently, increased evidence suggests that stem cells mobilize from the bone marrow into the peripheral blood (PB), differentiate in circulating endothelial progenitors (EPCs), and home to site of ischemia to contribute to de novo vessel formation. Significant advances have been made in understanding the biology of EPCs, and molecular mechanisms regulating EPC function. Autologous EPCs now are becoming a novel treatment option for therapeutic vascularization and vascular repair, mainly in ischemic diseases. However, different diseases, such as cardiovascular diseases, diabetes, and peripheral artery ischemia are related to EPC dysfunction. Several studies have shown that EPCs can be detected in the PB of patients with SSc and are impaired in their function. Based on an online literature search (PubMed, EMBASE, and Web of Science, last updated December 2017) using keywords related to “endothelial progenitor cells” and “Systemic Sclerosis,” “scleroderma vasculopathy,” “angiogenesis,” “vasculogenesis,” this review gives an overview on the large body of data of current research in this issue, including controversies over the identity and functions of EPCs, their meaning as biomarker of SSc microangiopathy and their clinical potency.
Collapse
|