1
|
Rossides M, Kullberg S, Arkema EV. History and Familial Aggregation of Immune-Mediated Diseases in Sarcoidosis: A Register-Based Case-Control-Family Study. Chest 2024; 166:1082-1092. [PMID: 38857779 DOI: 10.1016/j.chest.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/03/2024] [Accepted: 05/06/2024] [Indexed: 06/12/2024] Open
Abstract
BACKGROUND An autoimmune component in the cause of sarcoidosis has long been debated, but population-based data on the clustering of immune-mediated diseases (IMDs) and sarcoidosis in individuals and families suggestive of shared cause are limited. RESEARCH QUESTION Do patients with a history of IMDs have a higher risk of sarcoidosis and do IMDs cluster in families with sarcoidosis? STUDY DESIGN AND METHODS We conducted a case-control-family study (2001-2020). Patients with sarcoidosis (N = 14,146) were identified in the Swedish National Patient Register using a previously validated definition (≥ 2 International Classification of Diseases [ICD]-coded inpatient or outpatient visits). At diagnosis, patients were matched to up to 10 control participants from the general population (N = 118,478) for birth year, sex, and residential location. Patients, control participants, and their first-degree relatives (FDRs; Multi-Generation Register) were ascertained for IMDs by means of ICD codes in the Patient Register (1968-2020). Conditional logistic regression was used to estimate ORs and 95% CIs of sarcoidosis associated with a history of IMDs in patients and control participants and in FDRs. RESULTS Patients with sarcoidosis exhibited a higher prevalence of IMDs compared with control participants (7.7% vs 4.7%), especially connective tissue diseases, cytopenia, and celiac disease. Familial aggregation was observed across IMDs; the strongest association was with celiac disease (OR, 2.09; 95% CI, 1.22-3.58), followed by cytopenia (OR, 1.88; 95% CI, 0.97-3.65), thyroiditis (OR, 1.72; 95% CI, 1.14-2.60), skin psoriasis (OR, 1.70; 95% CI, 1.34-2.15), inflammatory bowel disease (OR, 1.53; 95% CI, 1.14-2.03), immune-mediated arthritis (OR, 1.49; 95% CI, 1.20-1.85), and connective tissue disease (OR, 1.39; 95% CI, 1.00-1.93). INTERPRETATION This study showed that IMDs confer a higher risk of sarcoidosis and they aggregate in families with sarcoidosis, signaling a shared cause between IMDs and sarcoidosis. Our findings warrant further evaluation of shared genetic mechanisms.
Collapse
Affiliation(s)
- Marios Rossides
- Unit of Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Theme Inflammation and Ageing, Karolinska University Hospital, Stockholm, Sweden.
| | - Susanna Kullberg
- Division of Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Department of Respiratory Medicine and Allergy, Theme Inflammation and Ageing, Karolinska University Hospital, Stockholm, Sweden
| | - Elizabeth V Arkema
- Clinical Epidemiology Division, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
2
|
Buso H, Discardi C, Bez P, Muscianisi F, Ceccato J, Milito C, Firinu D, Landini N, Jones MG, Felice C, Rattazzi M, Scarpa R, Cinetto F. Sarcoidosis versus Granulomatous and Lymphocytic Interstitial Lung Disease in Common Variable Immunodeficiency: A Comparative Review. Biomedicines 2024; 12:1503. [PMID: 39062076 PMCID: PMC11275071 DOI: 10.3390/biomedicines12071503] [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: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Sarcoidosis and Granulomatous and Lymphocytic Interstitial Lung Diseases (GLILD) are two rare entities primarily characterised by the development of Interstitial Lung Disease (ILD) in the context of systemic immune dysregulation. These two conditions partially share the immunological background and pathologic findings, with granuloma as the main common feature. In this narrative review, we performed a careful comparison between sarcoidosis and GLILD, with an overview of their main similarities and differences, starting from a clinical perspective and ending with a deeper look at the immunopathogenesis and possible target therapies. Sarcoidosis occurs in immunocompetent individuals, whereas GLILD occurs in patients affected by common variable immunodeficiency (CVID). Moreover, peculiar extrapulmonary manifestations and radiological and histological features may help distinguish the two diseases. Despite that, common pathogenetic pathways have been suggested and both these disorders can cause progressive impairment of lung function and variable systemic granulomatous and non-granulomatous complications, leading to significant morbidity, reduced quality of life, and survival. Due to the rarity of these conditions and the extreme clinical variability, there are still many open questions concerning their pathogenesis, natural history, and optimal management. However, if studied in parallel, these two entities might benefit from each other, leading to a better understanding of their pathogenesis and to more tailored treatment approaches.
Collapse
Affiliation(s)
- Helena Buso
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| | - Claudia Discardi
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| | - Patrick Bez
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| | - Francesco Muscianisi
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| | - Jessica Ceccato
- Haematology and Clinical Immunology Unit, Department of Medicine (DIMED), University of Padova, 35124 Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), 35131 Padova, Italy
| | - Cinzia Milito
- Department of Molecular Medicine, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Davide Firinu
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Nicholas Landini
- Department of Radiological, Oncological and Pathological Sciences, Policlinico Umberto I Hospital, “Sapienza” University of Rome, 00161 Rome, Italy
| | - Mark G. Jones
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 YD, UK;
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, Southampton SO16 6YD, UK
| | - Carla Felice
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| | - Marcello Rattazzi
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| | - Riccardo Scarpa
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| | - Francesco Cinetto
- Rare Diseases Referral Center, Internal Medicine 1, Department of Medicine (DIMED), AULSS2 Marca Trevigiana, Ca’ Foncello Hospital, University of Padova, 35124 Padova, Italy (C.F.); (M.R.); (R.S.); (F.C.)
| |
Collapse
|
3
|
Jiménez-Gómez G, Campos-Caro A, García-Núñez A, Gallardo-García A, Molina-Hidalgo A, León-Jiménez A. Analysis of Immune Cell Subsets in Peripheral Blood from Patients with Engineered Stone Silica-Induced Lung Inflammation. Int J Mol Sci 2024; 25:5722. [PMID: 38891910 PMCID: PMC11171478 DOI: 10.3390/ijms25115722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/11/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Silicosis caused by engineered stone (ES-silicosis) is an emerging worldwide issue characterized by inflammation and fibrosis in the lungs. To our knowledge, only a few reports have investigated leukocyte/lymphocyte subsets in ES-silicosis patients. The present study was designed to explore the proportions of the main lymphocyte subsets in ES-silicosis patients stratified into two groups, one with simple silicosis (SS) and the other with a more advanced state of the disease, defined as progressive massive fibrosis (PMF). The proportions of B (memory and plasmablasts) cells, T (helper, cytotoxic, regulatory) cells, and natural killer (NK) (regulatory and cytotoxic) cells were investigated by multiparameter flow cytometry in 91 ES-silicosis patients (53 SS patients and 38 PMF patients) and 22 healthy controls (HC). Although the total number of leukocytes did not differ between the groups studied, lymphopenia was observed in patients compared to healthy controls. Compared with those in healthy controls, the proportions of memory B cells, naïve helper T cells, and the CD4+/CD8+ T cells' ratio in the peripheral blood of patients with silicosis were significantly decreased, while the percentages of plasma cells, memory helper T cells, and regulatory T cells were significantly increased. For the NK cell subsets, no significant differences were found between the groups studied. These results revealed altered cellular immune processes in the peripheral blood of patients with ES-silicosis and provided further insight into silicosis pathogenesis.
Collapse
Affiliation(s)
- Gema Jiménez-Gómez
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), 11009 Cadiz, Spain; (G.J.-G.); (A.G.-N.); (A.M.-H.); (A.L.-J.)
- Research Unit, Puerta del Mar University Hospital, 11009 Cadiz, Spain
| | - Antonio Campos-Caro
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), 11009 Cadiz, Spain; (G.J.-G.); (A.G.-N.); (A.M.-H.); (A.L.-J.)
- Genetics Area, Biomedicine, Biotechnology and Public Health Department, School of Marine and Environmental Sciences, University of Cadiz, 11510 Cadiz, Spain
| | - Alejandro García-Núñez
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), 11009 Cadiz, Spain; (G.J.-G.); (A.G.-N.); (A.M.-H.); (A.L.-J.)
- Research Unit, Puerta del Mar University Hospital, 11009 Cadiz, Spain
| | | | - Antonio Molina-Hidalgo
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), 11009 Cadiz, Spain; (G.J.-G.); (A.G.-N.); (A.M.-H.); (A.L.-J.)
- Pulmonology Department, Puerta del Mar University Hospital, 11009 Cadiz, Spain
| | - Antonio León-Jiménez
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), 11009 Cadiz, Spain; (G.J.-G.); (A.G.-N.); (A.M.-H.); (A.L.-J.)
- Pulmonology Department, Puerta del Mar University Hospital, 11009 Cadiz, Spain
| |
Collapse
|
4
|
Miedema J, Cinetto F, Smed-Sörensen A, Spagnolo P. The immunopathogenesis of sarcoidosis. J Autoimmun 2024:103247. [PMID: 38734536 DOI: 10.1016/j.jaut.2024.103247] [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: 02/27/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Sarcoidosis is a granulomatous multiorgan disease, thought to result from exposure to yet unidentified antigens in genetically susceptible individuals. The exaggerated inflammatory response that leads to granuloma formation is highly complex and involves the innate and adaptive immune system. Consecutive immunological studies using advanced technology have increased our understanding of aberrantly activated immune cells, mediators and pathways that influence the formation, maintenance and resolution of granulomas. Over the years, it has become increasingly clear that disease immunopathogenesis can only be understood if the clinical heterogeneity of sarcoidosis is taken into consideration, along with the distribution of immune cells in peripheral blood and involved organs. Most studies offer an immunological snapshot during disease course, while the cellular composition of both the circulation and tissue microenvironment may change over time. Despite these challenges, novel insights on the role of the immune system are continuously published, thus bringing the field forward. This review highlights current knowledge on the innate and adaptive immune responses involved in sarcoidosis pathogenesis, as well as the pathways involved in non-resolving disease and fibrosis development. Additionally, we describe proposed immunological mechanisms responsible for drug-induced sarcoid like reactions. Although many aspects of disease immunopathogenesis remain to be unraveled, the identification of crucial immune reactions in sarcoidosis may help identify new treatment targets. We therefore also discuss potential therapies and future strategies based on the latest immunological findings.
Collapse
Affiliation(s)
- Jelle Miedema
- Department of Pulmonary Medicine, Center of Expertise for Interstitial Lung Disease, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Francesco Cinetto
- Rare Diseases Referral Center, Internal Medicine 1, Ca' Foncello Hospital, AULSS2 Marca Trevigiana, Italy; Department of Medicine - DIMED, University of Padova, Padova, Italy.
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden.
| | - Paolo Spagnolo
- Respiratory Disease Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy.
| |
Collapse
|
5
|
Miedema JR, de Jong LJ, Kahlmann V, Bergen IM, Broos CE, Wijsenbeek MS, Hendriks RW, Corneth OBJ. Increased proportions of circulating PD-1 + CD4 + memory T cells and PD-1 + regulatory T cells associate with good response to prednisone in pulmonary sarcoidosis. Respir Res 2024; 25:196. [PMID: 38715030 PMCID: PMC11075187 DOI: 10.1186/s12931-024-02833-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND The treatment response to corticosteroids in patients with sarcoidosis is highly variable. CD4+ T cells are central in sarcoid pathogenesis and their phenotype in peripheral blood (PB) associates with disease course. We hypothesized that the phenotype of circulating T cells in patients with sarcoidosis may correlate with the response to prednisone treatment. Therefore, we aimed to correlate frequencies and phenotypes of circulating T cells at baseline with the pulmonary function response at 3 and 12 months during prednisone treatment in patients with pulmonary sarcoidosis. METHODS We used multi-color flow cytometry to quantify activation marker expression on PB T cell populations in 22 treatment-naïve patients and 21 healthy controls (HCs). Pulmonary function tests at baseline, 3 and 12 months were used to measure treatment effect. RESULTS Patients with sarcoidosis showed an absolute forced vital capacity (FVC) increase of 14.2% predicted (± 10.6, p < 0.0001) between baseline and 3 months. Good response to prednisone (defined as absolute FVC increase of ≥ 10% predicted) was observed in 12 patients. CD4+ memory T cells and regulatory T cells from patients with sarcoidosis displayed an aberrant phenotype at baseline, compared to HCs. Good responders at 3 months had significantly increased baseline proportions of PD-1+CD4+ memory T cells and PD-1+ regulatory T cells, compared to poor responders and HCs. Moreover, decreased fractions of CD25+ cells and increased fractions of PD-1+ cells within the CD4+ memory T cell population correlated with ≥ 10% FVC increase at 12 months. During treatment, the aberrantly activated phenotype of memory and regulatory T cells reversed. CONCLUSIONS Increased proportions of circulating PD-1+CD4+ memory T cells and PD-1+ regulatory T cells and decreased proportions of CD25+CD4+ memory T cells associate with good FVC response to prednisone in pulmonary sarcoidosis, representing promising new blood biomarkers for prednisone efficacy. TRIAL REGISTRATION NL44805.078.13.
Collapse
Affiliation(s)
- Jelle R Miedema
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands.
| | - Lieke J de Jong
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| | - Vivienne Kahlmann
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| | - Ingrid M Bergen
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| | - Caroline E Broos
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| | - Marlies S Wijsenbeek
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| | - Odilia B J Corneth
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Doctor Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| |
Collapse
|
6
|
Chen C, Luo N, Dai F, Zhou W, Wu X, Zhang J. Advance in pathogenesis of sarcoidosis: Triggers and progression. Heliyon 2024; 10:e27612. [PMID: 38486783 PMCID: PMC10938127 DOI: 10.1016/j.heliyon.2024.e27612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 03/02/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024] Open
Abstract
Sarcoidosis, a multisystemic immune disease, significantly impacts patients' quality of life. The complexity and diversity of its pathogenesis, coupled with limited comprehensive research, had hampered both diagnosis and treatment, resulting in an unsatisfactory prognosis for many patients. In recent years, the research had made surprising progress in the triggers of sarcoidosis (genetic inheritance, infection and environmental factors) and the abnormal regulations on immunity during the formation of granuloma. This review consolidated the latest findings on sarcoidosis research, providing a systematic exploration of advanced studies on triggers, immune-related regulatory mechanisms, and clinical applications. By synthesizing previous discoveries, we aimed to offer valuable insights for future research directions and the development of clinical diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Cong Chen
- Department of Thoracic Surgery and Institute of Thoracic Oncology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, 610097, China
| | - Nanzhi Luo
- Department of Thoracic Surgery and Institute of Thoracic Oncology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, 610097, China
| | - Fuqiang Dai
- Department of Thoracic Surgery and Institute of Thoracic Oncology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, 610097, China
- Department of Thoracic Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Wenjing Zhou
- Department of Thoracic Surgery and Institute of Thoracic Oncology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, 610097, China
| | - Xiaoqing Wu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, 610097, China
| | - Jian Zhang
- Department of Thoracic Surgery and Institute of Thoracic Oncology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, 610097, China
| |
Collapse
|
7
|
Weeratunga P, Moller DR, Ho LP. Immune mechanisms of granuloma formation in sarcoidosis and tuberculosis. J Clin Invest 2024; 134:e175264. [PMID: 38165044 PMCID: PMC10760966 DOI: 10.1172/jci175264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024] Open
Abstract
Sarcoidosis is a complex immune-mediated disease characterized by clusters of immune cells called granulomas. Despite major steps in understanding the cause of this disease, many questions remain. In this Review, we perform a mechanistic interrogation of the immune activities that contribute to granuloma formation in sarcoidosis and compare these processes with its closest mimic, tuberculosis, highlighting shared and divergent immune activities. We examine how Mycobacterium tuberculosis is sensed by the immune system; how the granuloma is initiated, formed, and perpetuated in tuberculosis compared with sarcoidosis; and the role of major innate and adaptive immune cells in shaping these processes. Finally, we draw these findings together around several recent high-resolution studies of the granuloma in situ that utilized the latest advances in single-cell technology combined with spatial methods to analyze plausible disease mechanisms. We conclude with an overall view of granuloma formation in sarcoidosis.
Collapse
Affiliation(s)
- Praveen Weeratunga
- MRC Translational Immunology Discovery Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Ling-Pei Ho
- MRC Translational Immunology Discovery Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
8
|
Patterson KC, Miller WT, Hancock WW, Akimova T. FOXP3+ regulatory T cells are associated with the severity and prognosis of sarcoidosis. Front Immunol 2023; 14:1301991. [PMID: 38173720 PMCID: PMC10761433 DOI: 10.3389/fimmu.2023.1301991] [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: 09/25/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024] Open
Abstract
Rationale Sarcoidosis is an inflammatory granulomatous disease of unknown etiology with predominant lung involvement. Organ involvement and disease severity, as well as the nature of immune alterations, vary among patients leading to a range of clinical phenotypes and outcomes. Our objective was to evaluate the association of disease course and immune responses in pulmonary sarcoidosis. Methods In this prospective cohort study of 30 subjects, most of whom were followed for one year, we evaluated 14 inflammatory markers in plasma, 13 Treg/T cell flow cytometry markers and 8 parameters of FOXP3+ Treg biology, including suppressive function, epigenetic features and stability. Results We identified a set of 13 immunological parameters that differ in sarcoidosis subjects in comparison with healthy donors. Five of those were inversely correlated with suppressive function of Tregs in sarcoidosis, and six (TNFα, TNFR I and II, sCD25, Ki-67 and number of Tregs) were particularly upregulated or increased in subjects with thoracic lymphadenopathy. Treg suppressive function was significantly lower in patients with thoracic lymphadenopathy, and in patients with higher burdens of pulmonary and systemic symptoms. A combination of five inflammatory markers, Ki-67 expression, Treg function, and lung diffusion capacity evaluated at study entry predicted need for therapy at one year follow-up in 90% of cases. Conclusion Tregs may suppress ongoing inflammation at local and systemic levels, and TNFα, TNFR I and II, sCD25 and Ki-67 emerge as attractive biomarkers for in vivo sarcoid inflammatory activity.
Collapse
Affiliation(s)
- Karen C. Patterson
- Division of Pulmonary, Allergy, and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Wallace T. Miller
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Wayne W. Hancock
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, and Biesecker Center for Pediatric Liver Diseases, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Tatiana Akimova
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, and Biesecker Center for Pediatric Liver Diseases, Children’s Hospital of Philadelphia and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
9
|
Kullberg S, Grunewald J, Eklund A. Lymphopenia and high Ki-67 expression in peripheral blood CD4+ and CD8+ T cells associate with progressive sarcoidosis. BMJ Open Respir Res 2023; 10:e001551. [PMID: 38097354 PMCID: PMC10729051 DOI: 10.1136/bmjresp-2022-001551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 11/30/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Early identification of patients at risk for progressive sarcoidosis may improve intervention. High bronchoalveolar lavage fluid (BALF) lymphocytes and peripheral blood (PB) lymphopenia are associated with worse prognosis. The mechanisms behind are not disentangled, and to date, it is not possible to predict disease course with certainty. OBJECTIVES Insight into the frequency of T regulatory cells (Tregs), proliferating CD4+ and CD8+ T cells in BALF and PB in clinically well-characterised patients, may provide clues to mechanisms behind differences in disease course. METHODS Nineteen treatment-naïve patients with newly diagnosed sarcoidosis were assessed with BAL and PB samples at diagnosis. From the majority, repeated PB samples were collected over a year after diagnosis. The patients were followed for a median of 3 years and clinical parameters were used to classify patients into resolving, chronic progressive and chronic stable disease. Lymphocyte counts, frequency of Tregs defined as forkhead box protein 3+ (FoxP3+) CD4+T cells, and proliferating CD4+ and CD8+ T cells assessed with Ki-67 were analysed. RESULTS Eleven patients disclosed a chronic stable, and eight a progressive disease course, no one resolved during the study period. In PB, lower number of lymphocytes associated with chronic progressive disease, an increased frequency of Ki-67+CD4+ and CD8+ T cells, and a tendency towards higher percentage of FoxP3+CD4+ T cells compared with chronic stable patients. CONCLUSION A reduction of PB lymphocytes despite increased proliferation of CD4+and CD8+ T cells was observed in patients with chronic active compared with chronic stable sarcoidosis, indicating an increased PB lymphocyte turn-over in patients with deteriorating disease. Measurement of PB Tregs, Ki-67+CD4+ and Ki-67+CD8+ T cells may help in predicting sarcoidosis disease course.
Collapse
Affiliation(s)
- Susanna Kullberg
- Respiratory Medicine Division, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Respiratory Medicine, Theme Inflammation and Ageing, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Grunewald
- Respiratory Medicine Division, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Respiratory Medicine, Theme Inflammation and Ageing, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Eklund
- Respiratory Medicine Division, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Respiratory Medicine, Theme Inflammation and Ageing, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
10
|
Miedema JR, de Jong LJ, van Uden D, Bergen IM, Kool M, Broos CE, Kahlmann V, Wijsenbeek MS, Hendriks RW, Corneth OBJ. Circulating T cells in sarcoidosis have an aberrantly activated phenotype that correlates with disease outcome. J Autoimmun 2023:103120. [PMID: 37863732 DOI: 10.1016/j.jaut.2023.103120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/22/2023]
Abstract
RATIONALE Disease course in sarcoidosis is highly variable. Bronchoalveolar lavage fluid and mediastinal lymph nodes show accumulation of activated T cells with a T-helper (Th)17.1 signature, which correlates with non-resolving sarcoidosis. We hypothesize that the peripheral blood (PB) T cell phenotype may correlate with outcome. OBJECTIVES To compare frequencies, phenotypes and function of circulating T cell populations in sarcoidosis patients with healthy controls (HCs) and correlate these parameters with outcome. METHODS We used multi-color flow cytometry to quantify activation marker expression on PB T cell subsets in treatment-naïve patients and HCs. The disease course was determined after 2-year follow-up. Cytokine production was measured after T cell stimulation in vitro. MEASUREMENTS AND MAIN RESULTS We observed significant differences between patients and HCs in several T cell populations, including CD8+ and CD4+ T cells, Th1/Th17 subsets, CD4+ T memory stem cells, regulatory T cells (Tregs) and γδ T cells. Decreased frequencies of CD4+ T cells and increased frequencies of Tregs and CD8+ γδ T cells correlated with worse outcome. Naïve CD4+ T cells displayed an activated phenotype with increased CD25 expression in patients with active chronic disease at 2-year follow-up. A distinctive Treg phenotype with increased expression of CD25, CTLA4, CD69, PD-1 and CD95 correlated with chronic sarcoidosis. Upon stimulation, both naïve and memory T cells displayed a different cytokine profile in sarcoidosis compared to HCs. CONCLUSIONS Circulating T cell subpopulations of sarcoidosis patients display phenotypic abnormalities that correlate with disease outcome, supporting a critical role of aberrant T cell activation in sarcoidosis pathogenesis.
Collapse
Affiliation(s)
- Jelle R Miedema
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Lieke J de Jong
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Denise van Uden
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ingrid M Bergen
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Mirjam Kool
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands; Danone Nutricia Research, Center of Excellence Immunology, Utrecht, the Netherlands
| | - Caroline E Broos
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Vivienne Kahlmann
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marlies S Wijsenbeek
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Odilia B J Corneth
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
11
|
Zhang H, Jiang D, Zhu L, Zhou G, Xie B, Cui Y, Costabel U, Dai H. Imbalanced distribution of regulatory T cells and Th17.1 cells in the peripheral blood and BALF of sarcoidosis patients: relationship to disease activity and the fibrotic radiographic phenotype. Front Immunol 2023; 14:1185443. [PMID: 37520566 PMCID: PMC10374842 DOI: 10.3389/fimmu.2023.1185443] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/14/2023] [Indexed: 08/01/2023] Open
Abstract
Rationale Sarcoidosis is a granulomatous interstitial lung disease involving a complex interplay among different cluster of differentiation 4 (CD4+) thymus cell (T-cell) subsets. Originally described as a type 1 T-helper (Th1) inflammatory disease, recent evidence suggests that both effector and regulatory T-cell subgroups play a critical role in sarcoidosis, but this remains controversial. Objectives We aimed to investigate the distribution of CD4+ T-cell subpopulations in sarcoidosis patients and its potential associations with clinical disease activity and a radiographic fibrotic phenotype. Methods We measured the frequencies of regulatory T cells (Tregs), Th1, Th17, and Th17.1 cells in the peripheral blood and/or bronchoalveolar lavage fluid (BALF) of 62 sarcoidosis patients, 66 idiopathic pulmonary fibrosis (IPF) patients, and 41 healthy volunteers using flow cytometry. We also measured the changes in these T-cell subpopulations in the blood at the follow-up visits of 11 sarcoidosis patients. Measurements and results An increased percentage of Tregs was observed in the peripheral blood of sarcoidosis patients, with a positive association to disease activity and a fibrotic radiographic phenotype. We found a higher frequency of Tregs, a lower proportion of Th17.1 cells, and a lower ratio of Th17.1 cells to total Tregs in the peripheral blood of both active and fibrotic sarcoidosis patients, compared with IPF patients or healthy donors. In contrast, a lower frequency of Tregs and a higher proportion of Th17.1 cells was found in the BALF of sarcoidosis patients than in that of IPF patients. There was an imbalance of Tregs and Th17.1 cells between the peripheral blood and BALF in sarcoidosis patients. Following immunoregulatory therapy, the proportion of circulating Tregs in sarcoidosis patients decreased. Conclusion A higher proportion of Tregs in the peripheral blood of sarcoidosis patients was related to disease activity, fibrotic phenotype, and the need for immunoregulatory therapy. The imbalanced distribution of Tregs and Th17.1 cells in patients' peripheral blood and BALF suggests that the lung microenvironment has an effect on the immunological pathogenesis of sarcoidosis. Therefore, further studies on the functional analysis of Tregs and Th17.1 cells in sarcoidosis patients are warranted.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences Peking Union Medical University, Beijing, China
- Department of Pulmonary and Critical Care Medicine, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Dingyuan Jiang
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences Peking Union Medical University, Beijing, China
| | - Lili Zhu
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences Peking Union Medical University, Beijing, China
| | - Guowu Zhou
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences Peking Union Medical University, Beijing, China
| | - Bingbing Xie
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences Peking Union Medical University, Beijing, China
| | - Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ulrich Costabel
- Center for Interstitial and Rare Lung Diseases, Pneumology Department, Ruhrlandklinik, University Hospital, University of Duisburg-Essen, Essen, Germany
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine, China-Japan Friendship Hospital, National Center for Respiratory Medicine; National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences Peking Union Medical University, Beijing, China
| |
Collapse
|
12
|
Kudryavtsev I, Zinchenko Y, Starshinova A, Serebriakova M, Malkova A, Akisheva T, Kudlay D, Glushkova A, Yablonskiy P, Shoenfeld Y. Circulating Regulatory T Cell Subsets in Patients with Sarcoidosis. Diagnostics (Basel) 2023; 13:1378. [PMID: 37189479 PMCID: PMC10137313 DOI: 10.3390/diagnostics13081378] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/03/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
Over recent years, many researchers have supported the autoimmune theory of sarcoidosis. The presence of uncontrolled inflammatory response on local and system levels in patients with sarcoidosis did not define that the immunoregulatory mechanisms could be affected. The aim of this study was to evaluate the distribution and the disturbance circulating Treg cell subsets in the peripheral blood in patients with sarcoidosis. MATERIALS AND METHODS A prospective comparative study was performed in 2016-2018 (34 patients with sarcoidosis (men (67.6%), women (32.3%)) were examined). Healthy subjects-the control group (n = 40). The diagnosis of pulmonary sarcoidosis was performed according to the standard criteria. We used two ten-color combinations of antibodies for Treg immunophenotyping. The first one contained CD39-FITC, CD127-PE, CCR4-PE/Dazzle™ 594, CD25-PC5.5, CD161-PC7, CD4-APC, CD8-APC-AF700, CD3-APC/Cy7, HLA-DR-PacBlue, and CD45 RA-BV 510™, while the second consisted of CXCR3-Alexa Fluor 488, CD25-РЕ, CXCR5-РЕ/Dazzle™ 594, CCR4-PerСP/Сy5.5, CCR6-РЕ/Cy7, CD4-АPC, CD8 АPC-AF700, CD3-АPC/Cy7, CCR7-BV 421, and CD45 RA-BV 510. The flow cytometry data were analyzed by using Kaluza software v2.3. A statistical analysis was performed with Statistica 7.0 and GraphPad Prism 8 software packages. RESULTS OF THE STUDY Primarily, we found that patients with sarcoidosis had decreased absolute numbers of Treg cells in circulation. We noted that the level of CCR7-expressing Tregs decreased in patients with sarcoidosis vs. the control group (65.55% (60.08; 70.60) vs. 76.93% (69.59; 79.86) with p < 0.001). We noticed that the relative numbers of CD45RA-CCR7+ Tregs decreased in patients with sarcoidosis (27.11% vs. 35.43%, p < 0.001), while the frequency of CD45 RA-CCR7- and CD45RA+ CCR7- Tregs increased compared to the control group (33.3% vs. 22.73% and 0.76% vs. 0.51% with p < 0.001 and p = 0.028, respectively). CXCR3-expressing Treg cell subsets-Th1-like CCR60078CXCR3+ Tregs and Th17.1-like CCR6+ CXCR3+ Tregs-significantly increased in patients with sarcoidosis vs. the control group (14.4% vs. 10.5% with p < 0.01 and 27.9% vs. 22.8% with p < 0.01, respectively). Furthermore, the levels of peripheral blood EM Th17-like Tregs significantly decreased in the sarcoidosis group vs. the control group (36.38% vs. 46.70% with p < 0.001). Finally, we found that CXCR5 expression was increased in CM Tregs cell subsets in patients with sarcoidosis. CONCLUSIONS Our data indicated a decrease in circulating Tregs absolute numbers and several alterations in Treg cell subsets. Moreover, our results highlight the presence of increased levels of CM CXCR5+ follicular Tregs in the periphery that could be linked with the imbalance of follicular Th cell subsets and alterations in B cell, based on the immune response. The balance between the two functionally distinct Treg cell populations-Th1-like and Th17-like Tregs-could be used in sarcoidosis diagnosis and the determination of prognosis and disease outcomes. Furthermore, we want to declare that analysis of Treg numbers of phenotypes could fully characterize their functional activity in peripherally inflamed tissues.
Collapse
Affiliation(s)
- Igor Kudryavtsev
- Department of Immunology, Institution of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Yulia Zinchenko
- Phthisiopulmonology Department, St. Petersburg Research Institute of Phthisiopulmonology, 194064 St. Petersburg, Russia
| | - Anna Starshinova
- Almazov National Medical Research Centre, 197341 St. Petersburg, Russia
| | - Maria Serebriakova
- Department of Immunology, Institution of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Anna Malkova
- Laboratory of the Mosaic of Autoimmunity, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Tatiana Akisheva
- Department of Immunology, Institution of Experimental Medicine, 197376 St. Petersburg, Russia
| | - Dmitriy Kudlay
- Department of Pharmacology, Sechenov First Moscow State Medical University, 119992 Moscow, Russia
- Institute of Immunology, 115552 Moscow, Russia
| | - Anzhela Glushkova
- Bekhterev National Research Medical Center for Psychiatry and Neurology, 19201 St. Petersburg, Russia
| | - Piotr Yablonskiy
- Phthisiopulmonology Department, St. Petersburg Research Institute of Phthisiopulmonology, 194064 St. Petersburg, Russia
- Laboratory of the Mosaic of Autoimmunity, St. Petersburg State University, 199034 St. Petersburg, Russia
| | - Yehuda Shoenfeld
- Laboratory of the Mosaic of Autoimmunity, St. Petersburg State University, 199034 St. Petersburg, Russia
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel Hashomer 5265601, Israel
| |
Collapse
|
13
|
Ascoli C, Schott CA, Huang Y, Turturice BA, Wang W, Ecanow N, Sweiss NJ, Perkins DL, Finn PW. Altered transcription factor targeting is associated with differential peripheral blood mononuclear cell proportions in sarcoidosis. Front Immunol 2022; 13:848759. [PMID: 36311769 PMCID: PMC9608777 DOI: 10.3389/fimmu.2022.848759] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 08/01/2022] [Indexed: 11/13/2022] Open
Abstract
IntroductionIn sarcoidosis, peripheral lymphopenia and anergy have been associated with increased inflammation and maladaptive immune activity, likely promoting development of chronic and progressive disease. However, the molecular mechanisms that lead to reduced lymphocyte proportions, particularly CD4+ T-cells, have not been fully elucidated. We posit that paradoxical peripheral lymphopenia is characterized by a dysregulated transcriptomic network associated with cell function and fate that results from altered transcription factor targeting activity.MethodsMessenger RNA-sequencing (mRNA-seq) was performed on peripheral blood mononuclear cells (PBMCs) from ACCESS study subjects with sarcoidosis and matched controls and findings validated on a sarcoidosis case-control cohort and a sarcoidosis case series. Preserved PBMC transcriptomic networks between case-control cohorts were assessed to establish cellular associations with gene modules and define regulatory targeting involved in sarcoidosis immune dysregulation utilizing weighted gene co-expression network analysis and differential transcription factor involvement analysis. Network centrality measures identified master transcriptional regulators of subnetworks related to cell proliferation and death. Predictive models of differential PBMC proportions constructed from ACCESS target gene expression corroborated the relationship between aberrant transcription factor regulatory activity and imputed and clinical PBMC populations in the validation cohorts.ResultsWe identified two unique and preserved gene modules significantly associated with sarcoidosis immune dysregulation. Strikingly, increased expression of a monocyte-driven, and not a lymphocyte-driven, gene module related to innate immunity and cell death was the best predictor of peripheral CD4+ T-cell proportions. Within the gene network of this monocyte-driven module, TLE3 and CBX8 were determined to be master regulators of the cell death subnetwork. A core gene signature of differentially over-expressed target genes of TLE3 and CBX8 involved in cellular communication and immune response regulation accurately predicted imputed and clinical monocyte expansion and CD4+ T-cell depletion.ConclusionsAltered transcriptional regulation associated with aberrant gene expression of a monocyte-driven transcriptional network likely influences lymphocyte function and survival. Although further investigation is warranted, this indicates that crosstalk between hyperactive monocytes and lymphocytes may instigate peripheral lymphopenia and underlie sarcoidosis immune dysregulation and pathogenesis. Future therapies selectively targeting master regulators, or their targets, may mitigate dysregulated immune processes in sarcoidosis and disease progression.
Collapse
Affiliation(s)
- Christian Ascoli
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Cody A. Schott
- University of Illinois at Chicago College of Medicine, Chicago, IL, United States
| | - Yue Huang
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | | | - Wangfei Wang
- Department of Bioengineering, University of Illinois at Chicago College of Engineering and Medicine, Chicago, IL, United States
| | - Naomi Ecanow
- University of Illinois at Chicago College of Medicine, Chicago, IL, United States
| | - Nadera J. Sweiss
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Division of Rheumatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - David L. Perkins
- Division of Nephrology, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Patricia W. Finn
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- *Correspondence: Patricia W. Finn,
| |
Collapse
|
14
|
McKee AS, Atif SM, Falta MT, Fontenot AP. Innate and Adaptive Immunity in Noninfectious Granulomatous Lung Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1835-1843. [PMID: 35418504 PMCID: PMC9106315 DOI: 10.4049/jimmunol.2101159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/08/2022] [Indexed: 11/19/2022]
Abstract
Sarcoidosis and chronic beryllium disease are noninfectious lung diseases that are characterized by the presence of noncaseating granulomatous inflammation. Chronic beryllium disease is caused by occupational exposure to beryllium containing particles, whereas the etiology of sarcoidosis is not known. Genetic susceptibility for both diseases is associated with particular MHC class II alleles, and CD4+ T cells are implicated in their pathogenesis. The innate immune system plays a critical role in the initiation of pathogenic CD4+ T cell responses as well as the transition to active lung disease and disease progression. In this review, we highlight recent insights into Ag recognition in chronic beryllium disease and sarcoidosis. In addition, we discuss the current understanding of the dynamic interactions between the innate and adaptive immune systems and their impact on disease pathogenesis.
Collapse
Affiliation(s)
- Amy S McKee
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO; and
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Shaikh M Atif
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO; and
| | - Michael T Falta
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO; and
| | - Andrew P Fontenot
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO; and
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
15
|
Liu A, Sharma L, Yan X, Dela Cruz CS, Herzog EL, Ryu C. Emerging insights in sarcoidosis: moving forward through reverse translational research. Am J Physiol Lung Cell Mol Physiol 2022; 322:L518-L525. [PMID: 35196896 PMCID: PMC8957321 DOI: 10.1152/ajplung.00266.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 12/13/2021] [Accepted: 02/22/2022] [Indexed: 11/22/2022] Open
Abstract
Sarcoidosis is a chronic granulomatous disease of unknown etiology that primarily affects the lungs. The development of stage IV or fibrotic lung disease accounts for a significant proportion of the morbidity and mortality attributable to sarcoidosis. Further investigation into the active mechanisms of disease pathogenesis and fibrogenesis might illuminate fundamental mediators of injury and repair while providing new opportunities for clinical intervention. However, progress in sarcoidosis research has been hampered by the heterogeneity of clinical phenotypes and the lack of a consensus modeling system. Recently, reverse translational research, wherein observations made at the patient level catalyze hypothesis-driven research at the laboratory bench, has generated new discoveries regarding the immunopathogenic mechanisms of pulmonary granuloma formation, fibrogenesis, and disease model development. The purpose of this review is to highlight the promise and possibility of these novel investigative efforts.
Collapse
Affiliation(s)
- Angela Liu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Xiting Yan
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Charles S Dela Cruz
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Erica L Herzog
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| | - Changwan Ryu
- Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, Yale University, New Haven, Connecticut
| |
Collapse
|
16
|
Zhang H, Costabel U, Dai H. The Role of Diverse Immune Cells in Sarcoidosis. Front Immunol 2021; 12:788502. [PMID: 34868074 PMCID: PMC8640342 DOI: 10.3389/fimmu.2021.788502] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/04/2021] [Indexed: 12/19/2022] Open
Abstract
Sarcoidosis is a systemic inflammatory disorder of unknown etiology characterized by tissue infiltration with macrophages and lymphocytes and associated non-caseating granuloma formation. The disease primarily affects the lungs. Patients suffering from sarcoidosis show a wide range of clinical symptoms, natural history and disease outcomes. Originally described as a Th1-driven disease, sarcoidosis involves a complex interplay among diverse immune cells. This review highlights recent advances in the pathogenesis of sarcoidosis, with emphasis on the role of different immune cells. Accumulative evidence suggests Th17 cells, IFN-γ-producing Th17 cells or Th17.1 cells, and regulatory T (Treg) cells play a critical role. However, their specific actions, whether protective or pathogenic, remain to be clarified. Macrophages are also involved in granuloma formation, and M2 polarization may be predictive of fibrosis. Previously neglected cells including B cells, dendritic cells (DCs), natural killer (NK) cells and natural killer T (NKT) cells were studied more recently for their contribution to sarcoid granuloma formation. Despite these advances, the pathogenesis remains incompletely understood, indicating an urgent need for further research to reveal the distinct immunological events in this process, with hope to open up new therapeutic avenues and if possible, to develop preventive measures.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Peking Union Medical College, Beijing, China
| | - Ulrich Costabel
- Center for Interstitial and Rare Lung Diseases, Pneumology Department, Ruhrlandklinik, University Hospital, Essen, Germany
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China.,National Center for Respiratory Medicine, Beijing, China.,Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, China.,National Clinical Research Center for Respiratory Diseases, Beijing, China
| |
Collapse
|
17
|
Koth LL, Harmacek LD, White EK, Arger NK, Powers L, Werner BR, Magallon RE, Grewal P, Barkes BQ, Li L, Gillespie M, Collins SE, Cardenas J, Chen ES, Maier LA, Leach SM, O'Connor BP, Hamzeh NY. Defining CD4 T helper and T regulatory cell endotypes of progressive and remitting pulmonary sarcoidosis (BRITE): protocol for a US-based, multicentre, longitudinal observational bronchoscopy study. BMJ Open 2021; 11:e056841. [PMID: 34753769 PMCID: PMC8578977 DOI: 10.1136/bmjopen-2021-056841] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
INTRODUCTION Sarcoidosis is a multiorgan granulomatous disorder thought to be triggered and influenced by gene-environment interactions. Sarcoidosis affects 45-300/100 000 individuals in the USA and has an increasing mortality rate. The greatest gap in knowledge about sarcoidosis pathobiology is a lack of understanding about the underlying immunological mechanisms driving progressive pulmonary disease. The objective of this study is to define the lung-specific and blood-specific longitudinal changes in the adaptive immune response and their relationship to progressive and non-progressive pulmonary outcomes in patients with recently diagnosed sarcoidosis. METHODS AND ANALYSIS The BRonchoscopy at Initial sarcoidosis diagnosis Targeting longitudinal Endpoints study is a US-based, NIH-sponsored longitudinal blood and bronchoscopy study. Enrolment will occur over four centres with a target sample size of 80 eligible participants within 18 months of tissue diagnosis. Participants will undergo six study visits over 18 months. In addition to serial measurement of lung function, symptom surveys and chest X-rays, participants will undergo collection of blood and two bronchoscopies with bronchoalveolar lavage separated by 6 months. Freshly processed samples will be stained and flow-sorted for isolation of CD4 +T helper (Th1, Th17.0 and Th17.1) and T regulatory cell immune populations, followed by next-generation RNA sequencing. We will construct bioinformatic tools using this gene expression to define sarcoidosis endotypes that associate with progressive and non-progressive pulmonary disease outcomes and validate the tools using an independent cohort. ETHICS AND DISSEMINATION The study protocol has been approved by the Institutional Review Boards at National Jewish Hospital (IRB# HS-3118), University of Iowa (IRB# 201801750), Johns Hopkins University (IRB# 00149513) and University of California, San Francisco (IRB# 17-23432). All participants will be required to provide written informed consent. Findings will be disseminated via journal publications, scientific conferences, patient advocacy group online content and social media platforms.
Collapse
Affiliation(s)
- Laura L Koth
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Laura D Harmacek
- Center for Genes, Environment, & Health, National Jewish Health, Denver, Colorado, USA
| | - Elizabeth K White
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, USA
| | | | - Linda Powers
- Department of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Brenda R Werner
- Department of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Roman E Magallon
- Center for Genes, Environment, & Health, National Jewish Health, Denver, Colorado, USA
| | - Pineet Grewal
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Briana Q Barkes
- Division of Environmental and Occupational Health Sciences, National Jewish Health, Denver, Colorado, USA
| | - Li Li
- Division of Environmental and Occupational Health Sciences, National Jewish Health, Denver, Colorado, USA
| | - May Gillespie
- Division of Environmental and Occupational Health Sciences, National Jewish Health, Denver, Colorado, USA
| | - Sarah E Collins
- Division of Pulmonary and Critical Care Medicine, Baltimore, Maryland, USA
| | - Jessica Cardenas
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Edward S Chen
- Division of Pulmonary and Critical Care Medicine, Baltimore, Maryland, USA
| | - Lisa A Maier
- Division of Environmental and Occupational Health Sciences, National Jewish Health, Denver, Colorado, USA
| | - Sonia M Leach
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, USA
| | - Brian P O'Connor
- Center for Genes, Environment, & Health, National Jewish Health, Denver, Colorado, USA
| | - Nabeel Y Hamzeh
- Department of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
18
|
Abstract
Failure of regulatory T (Treg) cells to properly control immune responses leads invariably to autoimmunity and organ damage. Decreased numbers or impaired function of Treg cells, especially in the context of inflammation, has been documented in many human autoimmune diseases. Restoration of Treg cell fitness and/or expansion of their numbers using low-dose natural IL-2, the main cytokine driving Treg cell survival and function, has demonstrated clinical efficacy in early clinical trials. Genetically modified IL-2 with an extended half-life and increased selectivity for Treg cells is now in clinical development. Administration of IL-2 combined with therapies targeting other pathways involved in the expression of autoimmune diseases should further enhance its therapeutic potential. Ongoing clinical efforts that capitalize on the early clinical success of IL-2 treatment should bring the use of this cytokine to the forefront of biological treatments for autoimmune diseases.
Collapse
|
19
|
Dohrn MF, Ellrichmann G, Pjontek R, Lukas C, Panse J, Gold R, Schulz JB, Gess B, Tauber SC. Progressive multifocal leukoencephalopathy and immune reconstitution inflammatory syndrome in seven patients with sarcoidosis: a critical discussion of treatment and prognosis. Ther Adv Neurol Disord 2021; 14:17562864211035543. [PMID: 34377151 PMCID: PMC8326823 DOI: 10.1177/17562864211035543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/05/2021] [Indexed: 12/12/2022] Open
Abstract
Progressive multifocal leukoencephalopathy (PML) is a subacute brain infection by the opportunistic John Cunningham (JC) virus. Herein, we describe seven patients with PML, lymphopenia, and sarcoidosis, in three of whom PML was the first manifestation of sarcoidosis. At onset, the clinical picture comprised rapidly progressive spastic hemi- or limb pareses as well as disturbances of vision, speech, and orientation. Cerebral magnetic resonance imaging showed T2-hyperintense, confluent, mainly supratentorial lesions. Four patients developed punctate contrast enhancement as a radiological sign of an immune reconstitution inflammatory syndrome (IRIS), three of them having a fatal course. In the cerebrospinal fluid, the initial JC virus load (8–25,787 copies/ml) did not correlate with interindividual severity; however, virus load corresponded to clinical dynamics. Brain biopsies (n = 2), performed 2 months after symptom onset, showed spotted demyelination and microglial activation. All patients had lymphopenia in the range of 270–1150/µl. To control JC virus, three patients received a combination of mirtazapine and mefloquine, another two patients additionally took cidofovir. One patient was treated with cidofovir only, and one patient had a combined regimen with mirtazapine, mefloquine, cidofovir, intravenous interleukin 2, and JC capsid vaccination. To treat sarcoidosis, the four previously untreated patients received prednisolone. Three patients had taken immunosuppressants prior to PML onset, which were subsequently stopped as a potential accelerator of opportunistic infections. After 6–54 months of follow up, three patients reached an incomplete recovery, one patient progressed, but survived so far, and two patients died. One further patient was additionally diagnosed with lung cancer, which he died from after 24 months. We conclude that the combination of PML and sarcoidosis is a diagnostic and therapeutic challenge. PML can occur as the first sign of sarcoidosis without preceding immunosuppressive treatment. The development of IRIS might be an indicator of poor outcome.
Collapse
Affiliation(s)
- Maike F Dohrn
- Department of Neurology, Medical Faculty of the RWTH Aachen University, Pauwelsstr. 30, Aachen, 52074, Germany
| | - Gisa Ellrichmann
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Rastislav Pjontek
- Department of Diagnostic and Interventional Neuroradiology, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Carsten Lukas
- Department of Radiology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Jens Panse
- Department of Oncology, Hematology and Stem Cell Transplantation, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Jörg B Schulz
- Department of Neurology, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Burkhard Gess
- Department of Neurology, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| | - Simone C Tauber
- Department of Neurology, Medical Faculty of the RWTH Aachen University, Aachen, Germany
| |
Collapse
|
20
|
Cytokines and Leukocytes Subpopulations Profile in SARS-CoV-2 Patients Depending on the CT Score Severity. Viruses 2021; 13:v13050880. [PMID: 34064802 PMCID: PMC8151453 DOI: 10.3390/v13050880] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/28/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023] Open
Abstract
The role of the adaptive microenvironment components in severe acute respiratory syndrome coronavirus 2 (SARS-Cov-2) infection is widely researched, but remains unclear. Studying the common dynamics of adaptive immune response changes can help understand the pathogenesis of coronavirus disease 2019 (COVID-19), especially in critical patients. The aim of the present study was to determine the cytokines concentration and leukocyte subpopulations profiles in the severe COVID-19 (n = 23) and critical (n = 18) COVID-19 group distinguished by the computed tomography (CT) severity score. We observed lower percentage of lymphocyte subpopulation, higher neutrophils to lymphocytes ratio (NLR) and higher IL-6 concentration in critical COVID-19 group than in severe group. CT severity score was negative correlated with proportion of lymphocytes, lymphocytes T, CD4+ cells, Treg cells and NK cells and positive correlated with neutrophils, NLR, and IL-6. In critical group more correlations between cytokines and lymphocytes were observed, mainly between TNF-α, IL-1β and lymphocyte subpopulations. The collective assessment of the cytokine profile, leukocyte subpopulations and the CT severity score can help to characterize and differentiate patient in advanced COVID-19 than the study of single parameters. We have shown that the interconnection of elements of the adaptive microenvironment can play an important role in critical COVID-19 cases.
Collapse
|
21
|
Garman L, Pelikan RC, Rasmussen A, Lareau CA, Savoy KA, Deshmukh US, Bagavant H, Levin AM, Daouk S, Drake WP, Montgomery CG. Single Cell Transcriptomics Implicate Novel Monocyte and T Cell Immune Dysregulation in Sarcoidosis. Front Immunol 2020; 11:567342. [PMID: 33363531 PMCID: PMC7753017 DOI: 10.3389/fimmu.2020.567342] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/03/2020] [Indexed: 12/19/2022] Open
Abstract
Sarcoidosis is a systemic inflammatory disease characterized by infiltration of immune cells into granulomas. Previous gene expression studies using heterogeneous cell mixtures lack insight into cell-type-specific immune dysregulation. We performed the first single-cell RNA-sequencing study of sarcoidosis in peripheral immune cells in 48 patients and controls. Following unbiased clustering, differentially expressed genes were identified for 18 cell types and bioinformatically assessed for function and pathway enrichment. Our results reveal persistent activation of circulating classical monocytes with subsequent upregulation of trafficking molecules. Specifically, classical monocytes upregulated distinct markers of activation including adhesion molecules, pattern recognition receptors, and chemokine receptors, as well as enrichment of immunoregulatory pathways HMGB1, mTOR, and ephrin receptor signaling. Predictive modeling implicated TGFβ and mTOR signaling as drivers of persistent monocyte activation. Additionally, sarcoidosis T cell subsets displayed patterns of dysregulation. CD4 naïve T cells were enriched for markers of apoptosis and Th17/Treg differentiation, while effector T cells showed enrichment of anergy-related pathways. Differentially expressed genes in regulatory T cells suggested dysfunctional p53, cell death, and TNFR2 signaling. Using more sensitive technology and more precise units of measure, we identify cell-type specific, novel inflammatory and regulatory pathways. Based on our findings, we suggest a novel model involving four convergent arms of dysregulation: persistent hyperactivation of innate and adaptive immunity via classical monocytes and CD4 naïve T cells, regulatory T cell dysfunction, and effector T cell anergy. We further our understanding of the immunopathology of sarcoidosis and point to novel therapeutic targets.
Collapse
Affiliation(s)
- Lori Garman
- Oklahoma Medical Research Foundation, Genes and Human Disease, Oklahoma City, OK, United States
| | - Richard C Pelikan
- Oklahoma Medical Research Foundation, Genes and Human Disease, Oklahoma City, OK, United States
| | - Astrid Rasmussen
- Oklahoma Medical Research Foundation, Genes and Human Disease, Oklahoma City, OK, United States
| | - Caleb A Lareau
- Cell Circuits and Epigenomics Program, Broad Institute, Cambridge, MA, United States
| | - Kathryn A Savoy
- Oklahoma Medical Research Foundation, Genes and Human Disease, Oklahoma City, OK, United States
| | - Umesh S Deshmukh
- Oklahoma Medical Research Foundation, Arthritis and Clinical Immunology, Oklahoma City, OK, United States
| | - Harini Bagavant
- Oklahoma Medical Research Foundation, Arthritis and Clinical Immunology, Oklahoma City, OK, United States
| | - Albert M Levin
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, United States
| | - Salim Daouk
- Cardiovascular Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Wonder P Drake
- Vanderbilt University School of Medicine, Nashville, TN, United States
| | - Courtney G Montgomery
- Oklahoma Medical Research Foundation, Genes and Human Disease, Oklahoma City, OK, United States
| |
Collapse
|
22
|
Erlich-Malona N, Cahill J, Chaudhry S, Martin J, Rizvi S. Cardiac sarcoidosis requiring ICD placement and immune thrombocytopenia following alemtuzumab treatment for multiple sclerosis. Mult Scler Relat Disord 2020; 47:102599. [PMID: 33160137 DOI: 10.1016/j.msard.2020.102599] [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: 10/06/2020] [Revised: 10/19/2020] [Accepted: 10/21/2020] [Indexed: 10/23/2022]
Abstract
Alemtuzumab, an effective disease-modifying therapy for multiple sclerosis, carries a significant risk of secondary autoimmunity. We present a case of cardiac sarcoidosis and immune thrombocytopenia diagnosed in an MS patient two years after completing alemtuzumab treatment. We hypothesize that alemtuzumab-induced changes to the T regulatory cell population may be implicated in the development of sarcoidosis in MS patients.
Collapse
Affiliation(s)
- Natalie Erlich-Malona
- Brown University Department of Neurology, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA.
| | - Jonathan Cahill
- Brown University Department of Neurology, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
| | - Saima Chaudhry
- Brown University Department of Neurology, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
| | - Janice Martin
- Brown University Department of Neurology, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
| | - Syed Rizvi
- Brown University Department of Neurology, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
| |
Collapse
|
23
|
Key Players and Biomarkers of the Adaptive Immune System in the Pathogenesis of Sarcoidosis. Int J Mol Sci 2020; 21:ijms21197398. [PMID: 33036432 PMCID: PMC7582702 DOI: 10.3390/ijms21197398] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/03/2020] [Accepted: 10/05/2020] [Indexed: 12/16/2022] Open
Abstract
Sarcoidosis is a systemic inflammatory disease characterized by development of granulomas in the affected organs. Sarcoidosis is often a diagnosis of exclusion, and traditionally used tests for sarcoidosis demonstrate low sensitivity and specificity. We propose that accuracy of diagnosis can be improved if biomarkers of altered lymphocyte populations and levels of signaling molecules involved in disease pathogenesis are measured for patterns suggestive of sarcoidosis. These distinctive biomarkers can also be used to determine disease progression, predict prognosis, and make treatment decisions. Many subsets of T lymphocytes, including CD8+ T-cells and regulatory T-cells, have been shown to be dysfunctional in sarcoidosis, and the predominant CD4+ T helper cell subset in granulomas appears to be a strong indicator of disease phenotype and outcome. Studies of altered B cell populations, B cell signaling molecules, and immune complexes in sarcoidosis patients reveal promising biomarkers as well as possible explanations of disease etiology. Furthermore, examined biomarkers raise questions about new treatment methods and sarcoidosis antigens.
Collapse
|
24
|
Frye BC, Rump IC, Uhlmann A, Schubach F, Ihorst G, Grimbacher B, Zissel G, Quernheim JM. Safety and efficacy of abatacept in patients with treatment-resistant SARCoidosis (ABASARC) - protocol for a multi-center, single-arm phase IIa trial. Contemp Clin Trials Commun 2020; 19:100575. [PMID: 32551397 PMCID: PMC7292904 DOI: 10.1016/j.conctc.2020.100575] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 05/02/2020] [Accepted: 05/17/2020] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Sarcoidosis is a granulomatous systemic disease that becomes chronic in approximately one third of affected patients resulting in quality of life and functional impairment. Immunosuppressive drugs other than steroids represent alternative therapeutic options, but side effects like liver and bone marrow toxicity or increased susceptibility to infections limit their use. Pathophysiological studies in sarcoidosis patients demonstrate altered regulatory T-cell functions with a reduced expression of CTLA-4 (CD152) and prolonged inflammation. Therefore, interfering with CTLA-4 using abatacept might be a therapeutic option in sarcoidosis similar to rheumatoid arthritis therapy. METHODS/DESIGN This is a multicenter prospective open-labeled single arm phase II study addressing the safety of abatacept in sarcoidosis patients. 30 patients with chronic sarcoidosis requiring immunosuppressive therapy beyond 5 mg prednisolone equivalent will be treated with abatacept in combination with corticosteroids for one year in two centers.The primary endpoint is the number and characterization of severe infectious complications under treatment with abatacept.Secondary endpoints are the rate of all infections, patient-related outcomes (assessed by questionnaires), lung function and immunological parameters including alveolar inflammation assessed by bronchoaveolar lavage. DISCUSSION This is the first trial of abatacept in patients with sarcoidosis. It is hypothesized that administration of abatacept is safe in patients with chronic sarcoidosis and can limit ongoing inflammation. Patients' wellbeing is assessed by established questionnaires. Immunological work-up will highlight the effect of abatacept on inflammatory pathways in sarcoidosis. TRIAL REGISTRATION The trial has been registered at the German Clinical Trial Registry (Deutsches Register Klinischer Studien, DRKS) with the identity number DRKS00011660.
Collapse
Key Words
- 18FDG-PET-CT, 18Fluor-Desoxy-Glucose positron-emission tomography combined with computer tomography
- Abatacept
- BAL, bronchoalveolar lavage
- CMV, cytomegaly-virus
- Chronic sarcoidosis
- EBV, Epstein-Barr-Virus
- FVC, forced vital capacity
- GHS, general health score
- IFN-γ, Interferon-γ
- IL, interleukin
- KSQ, King's sarcoidosis questionnaire
- King's sarcoidosis questionnaire
- Patient-reported outcome
- Regulatory T-cells
- TLC, total lung capacity
- TNF, tumor-necrosis factor
- TReg, regulatory T-cells
- Therapy
Collapse
Affiliation(s)
- Björn C. Frye
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Ina Caroline Rump
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Annette Uhlmann
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- Clinical Trials Unit, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Fabian Schubach
- Clinical Trials Unit, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Gabriele Ihorst
- Clinical Trials Unit, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Bodo Grimbacher
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
- DZIF – German Center for Infection Research, Satellite Center Freiburg, Germany
- CIBSS – Centre for Integrative Biological Signalling Studies, University of Freiburg, Germany
- RESIST – Cluster of Excellence 2155 to Hanover Medical School, Satellite Center Freiburg, Germany
| | - Gernot Zissel
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Joachim Müller Quernheim
- Department of Pneumology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
25
|
Tarasidis A, Arce S. Immune response biomarkers as indicators of sarcoidosis presence, prognosis, and possible treatment: An Immunopathogenic perspective. Autoimmun Rev 2020; 19:102462. [PMID: 31917262 DOI: 10.1016/j.autrev.2020.102462] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 09/05/2019] [Indexed: 02/08/2023]
Abstract
Sarcoidosis has been a disease of puzzling occurrence and clinical course. Multiple immunological markers have been noted to be altered within sarcoidosis, however there is variable consistency among these reports. Previous studies have shown sarcoidosis to be a primary T cell-mediated disease, yet new data concerning B cell and mycobacterial involvement have been brought to light. The possibility of a uniform biomarker to characterize sarcoidosis presence, severity and prognosis greatly increases the movement towards directed and specialized treatment for this rare disease.
Collapse
Affiliation(s)
- Anna Tarasidis
- University of South Carolina School of Medicine, Greenville, SC, USA
| | - Sergio Arce
- Department of Biomedical Sciences, University of South Carolina School of Medicine Greenville, Greenville, SC 29605, USA.
| |
Collapse
|
26
|
Pediatric Sarcoidosis: A Review with Emphasis on Early Onset and High-Risk Sarcoidosis and Diagnostic Challenges. Diagnostics (Basel) 2019; 9:diagnostics9040160. [PMID: 31731423 PMCID: PMC6963233 DOI: 10.3390/diagnostics9040160] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/14/2022] Open
Abstract
Sarcoidosis is a non-necrotizing granulomatous inflammatory syndrome with multisystemic manifestations. We performed a systematic review of sarcoidosis in the pediatric population with particular emphases on early onset sarcoidosis, high-risk sarcoidosis, and newly reported or unusual sarcoid-related diseases. Blau Syndrome and early onset sarcoidosis/ BS-EOS are seen in children younger than five years old presenting with extra-thoracic manifestations but usually without lymphadenopathy and/or pulmonary involvement. The prevalence of high-risk sarcoidosis is very low in children and is further limited by the difficulty of diagnosis in symptomatic children and underdiagnosis in subclinical or asymptomatic patients. Reports of sarcoidal syndromes in users of E-cigarette/marijuana/other flavorings and their induction in cancer immunotherapies are of interests and may be challenging to differentiate from metastatic malignancy. The diagnostic considerations in pediatric sarcoidosis are to support a compatible clinicoradiographic presentation and the pathologic findings of non-necrotizing granulomas by ruling out granulomas of infective etiology. There is no absolutely reliable diagnostic test for sarcoidosis at present. The use of endoscopic bronchial ultrasound (EBUS) and transbronchial fine needle aspiration (TBNA) sampling of intrathoracic lymph nodes and lung, and for superficially accessible lesions, with cytopathological assessment and pathological confirmations provide fair diagnostic yield and excellent patient safety profile in children.
Collapse
|
27
|
Bennett D, Bargagli E, Refini RM, Rottoli P. New concepts in the pathogenesis of sarcoidosis. Expert Rev Respir Med 2019; 13:981-991. [DOI: 10.1080/17476348.2019.1655401] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- David Bennett
- Respiratory Diseases and Lung Transplantation Unit, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Elena Bargagli
- Respiratory Diseases and Lung Transplantation Unit, Azienda Ospedaliera Universitaria Senese, Siena, Italy
- Department of Medical and Surgical Sciences & Neurosciences, University of Siena, Siena, Italy
| | - Rosa Metella Refini
- Respiratory Diseases and Lung Transplantation Unit, Azienda Ospedaliera Universitaria Senese, Siena, Italy
- Department of Medical and Surgical Sciences & Neurosciences, University of Siena, Siena, Italy
| | - Paola Rottoli
- Department of Medical and Surgical Sciences & Neurosciences, University of Siena, Siena, Italy
- Regional Coordinator for Rare Respiratory Diseases for Tuscany, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| |
Collapse
|
28
|
Kaiser Y, Eklund A, Grunewald J. Moving target: shifting the focus to pulmonary sarcoidosis as an autoimmune spectrum disorder. Eur Respir J 2019; 54:13993003.021532018. [PMID: 31000677 DOI: 10.1183/13993003.021532018] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 04/01/2019] [Indexed: 12/27/2022]
Abstract
Despite more than a century of research, the causative agent(s) in sarcoidosis, a heterogeneous granulomatous disorder mainly affecting the lungs, remain(s) elusive. Following identification of genetic factors underlying different clinical phenotypes, increased understanding of CD4+ T-cell immunology, which is believed to be central to sarcoid pathogenesis, as well as the role of B-cells and other cells bridging innate and adaptive immunity, contributes to novel insights into the mechanistic pathways influencing disease resolution or chronicity. Hopefully, new perspectives and state-of-the-art technology will help to shed light on the still-elusive enigma of sarcoid aetiology. This perspective article highlights a number of recent advances in the search for antigenic targets in sarcoidosis, as well as the main arguments for sarcoidosis as a spectrum of autoimmune conditions, either as a result of an external (microbial) trigger and/or due to defective control mechanisms regulating the balance between T-cell activation and inhibition.
Collapse
Affiliation(s)
- Ylva Kaiser
- Respiratory Medicine Unit, Dept of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Anders Eklund
- Respiratory Medicine Unit, Dept of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Johan Grunewald
- Respiratory Medicine Unit, Dept of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
29
|
Inaoka PT, Shono M, Kamada M, Espinoza JL. Host-microbe interactions in the pathogenesis and clinical course of sarcoidosis. J Biomed Sci 2019; 26:45. [PMID: 31182092 PMCID: PMC6558716 DOI: 10.1186/s12929-019-0537-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 05/22/2019] [Indexed: 12/27/2022] Open
Abstract
Sarcoidosis is a rare inflammatory disease characterized by the development of granulomas in various organs, especially in the lungs and lymph nodes. Clinics of the disease largely depends on the organ involved and may range from mild symptoms to life threatening manifestations. Over the last two decades, significant advances in the diagnosis, clinical assessment and treatment of sarcoidosis have been achieved, however, the precise etiology of this disease remains unknown. Current evidence suggests that, in genetically predisposed individuals, an excessive immune response to unknown antigen/s is crucial for the development of sarcoidosis. Epidemiological and microbiological studies suggest that, at least in a fraction of patients, microbes or their products may trigger the immune response leading to sarcoid granuloma formation. In this article, we discuss the scientific evidence on the interaction of microbes with immune cells that may be implicated in the immunopathogenesis of sarcoidosis, and highlight recent studies exploring potential implications of human microbiota in the pathogenesis and the clinical course of sarcoidosis.
Collapse
Affiliation(s)
- Pleiades T Inaoka
- Department of Physical Therapy, School of Health Sciences, Kanazawa University, Kodatsuno, Kanazawa, 577-8502, Japan
| | - Masato Shono
- Faculty of Medicine, Kindai University, 377-2, Ohno-Higashi, Osaka-Sayama, Osaka, 577-8502, Japan
| | - Mishio Kamada
- Faculty of Medicine, Kindai University, 377-2, Ohno-Higashi, Osaka-Sayama, Osaka, 577-8502, Japan
| | - J Luis Espinoza
- Department of Hematology and Rheumatology, Kindai University Faculty of Medicine, 377-2, Ohno-Higashi, Osaka-Sayama, Osaka, 577-8502, Japan.
| |
Collapse
|
30
|
|
31
|
Interleukin 33 ameliorates disturbance of regulatory T cells in pulmonary sarcoidosis. Int Immunopharmacol 2018; 64:208-216. [DOI: 10.1016/j.intimp.2018.08.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 08/20/2018] [Accepted: 08/23/2018] [Indexed: 12/19/2022]
|
32
|
Molecular profiling of regulatory T cells in pulmonary sarcoidosis. J Autoimmun 2018; 94:56-69. [DOI: 10.1016/j.jaut.2018.07.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/11/2018] [Accepted: 07/15/2018] [Indexed: 11/23/2022]
|
33
|
Landi C, Carleo A, Cillis G, Rottoli P. Sarcoidosis: proteomics and new perspectives for improving personalized medicine. Expert Rev Proteomics 2018; 15:829-835. [DOI: 10.1080/14789450.2018.1528148] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Claudia Landi
- Laboratory of Functional Proteomics, Department of Life Sciences, University of Siena. Siena, Italy
- UOC Respiratory Diseases and Lung Transplantation, Department Internal and Specialist Medicine, University of Siena, Siena, Italy
| | - Alfonso Carleo
- UOC Respiratory Diseases and Lung Transplantation, Department Internal and Specialist Medicine, University of Siena, Siena, Italy
- Department of Pulmonology, Hannover Medical School, Hannover, Germany
| | - Giuseppe Cillis
- UOC Respiratory Diseases and Lung Transplantation, Department Internal and Specialist Medicine, University of Siena, Siena, Italy
| | - Paola Rottoli
- UOC Respiratory Diseases and Lung Transplantation, Department Internal and Specialist Medicine, University of Siena, Siena, Italy
| |
Collapse
|
34
|
Cornejo CM, Haun P, English J, Rosenbach M. Immune checkpoint inhibitors and the development of granulomatous reactions. J Am Acad Dermatol 2018; 81:1165-1175. [PMID: 30092327 DOI: 10.1016/j.jaad.2018.07.051] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 06/21/2018] [Accepted: 07/18/2018] [Indexed: 12/13/2022]
Abstract
Immune checkpoint inhibitors (ICPIs) have emerged as a frontline treatment for a growing list of malignancies. Disruption of the negative regulatory immune checkpoints by ICPIs has been associated with many immune-related adverse events. Granulomatous reactions, such as sarcoidosis-like reactions, granulomatous panniculitis, granuloma annulare, and granulomatous dermatitis, are uncommon but increasingly recognized immune-related adverse events seen in patients treated with ICPIs. The frequency and significance of these eruptions, including whether they portend responsiveness to treatment, remain unclear. Additionally, understanding the role of immune checkpoint blockade in these reactions may provide mechanistic insight into the relevant signaling pathways involved in sarcoidosis and other granulomatous disorders.
Collapse
Affiliation(s)
- Christine M Cornejo
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Paul Haun
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joseph English
- Department of Dermatology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Misha Rosenbach
- Department of Dermatology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
35
|
Th17-lineage cells in pulmonary sarcoidosis and Löfgren's syndrome: Friend or foe? J Autoimmun 2018; 87:82-96. [DOI: 10.1016/j.jaut.2017.12.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 12/12/2017] [Indexed: 01/17/2023]
|
36
|
Abstract
INTRODUCTION Sarcoidosis is a chronic granulomatous inflammatory disease that commonly causes lung disease, but can affect other vital organs and tissues. The cause of sarcoidosis is unknown, and current therapies are commonly limited by lack of efficacy, adverse side effects, and excessive cost. AREAS COVERED The manuscript will provide a review of current concepts relating to the pathogenesis of sarcoidosis, and how these disease mechanisms may be leveraged to develop more effective treatments for sarcoidosis. It provides only a brief summary of currently accepted therapy, while focusing more extensively on potential novel therapies. EXPERT OPINION Current sarcoidosis therapeutic agents primarily target the M1 or pro-inflammatory pathways. Agents that prevent M2 polarization, a regulatory phenotype favoring fibrosis, are attractive treatment alternatives that could potentially prevent fibrosis and associated life threatening complications. Effective treatment of sarcoidosis potentially requires simultaneous modulation both M1/M2 polarization instead of suppressing one pathway over the other to restore immune competent and inactive (M0) macrophages.
Collapse
Affiliation(s)
- Van Le
- a Department of Medicine , The Ohio State University Wexner Medical Center , Columbus , OH , USA
| | - Elliott D Crouser
- a Department of Medicine , The Ohio State University Wexner Medical Center , Columbus , OH , USA
| |
Collapse
|
37
|
The Pathogenesis of Pulmonary Sarcoidosis and Implications for Treatment. Chest 2017; 153:1432-1442. [PMID: 29224832 DOI: 10.1016/j.chest.2017.11.030] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/20/2017] [Accepted: 11/26/2017] [Indexed: 12/31/2022] Open
Abstract
Thoracic sarcoidosis is the most common form of sarcoidosis, encompassing a heterogeneous group of patients with a wide range of clinical features and associated outcomes. The distinction between isolated thoracic lymphadenopathy and pulmonary involvement matters. Morbidity is often higher, and long-term outcomes are worse for the latter. Although inflammatory infiltrates in pulmonary sarcoidosis may resolve, persistent disease activity is common and can result in lung fibrosis. Given the distinct clinical features and natural history of pulmonary sarcoidosis, its pathogenesis may differ in important ways from other sarcoidosis manifestations. This review highlights recent advances in the pathogenesis of pulmonary sarcoidosis, including the nature of the sarcoidosis antigen, the role of serum amyloid A and other host factors that contribute to alterations in innate immunity, factors that shape adaptive T-cell profiles in the lung, and how these mechanisms influence the maintenance of granulomatous inflammation in sarcoidosis. We discuss questions raised by recent findings, including the role of innate immunity in the pathogenesis, the meaning of immune cell exhaustion, and mechanisms that may contribute to lung fibrosis in sarcoidosis. We conclude with a reflection on when and how immunosuppressive therapies may be helpful for pulmonary sarcoidosis, a consideration of nonpharmacologic management strategies, and a survey of potential novel therapeutic targets for this vexing disease.
Collapse
|
38
|
T-cell immunology in sarcoidosis: Disruption of a delicate balance between helper and regulatory T-cells. Curr Opin Pulm Med 2017; 22:476-83. [PMID: 27379969 DOI: 10.1097/mcp.0000000000000303] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Although the aetiology of sarcoidosis is not yet completely understood, immunological changes within the T-cell compartment are characteristic for an exaggerated antigen-driven immune response. In this review, we describe the most recent findings on T-cell subset responses and regulation in sarcoidosis. We discuss how future immunological research can advance the field to unravel pathobiological mechanisms of this intriguingly complex disease. RECENT FINDINGS Research into the field of T-cell plasticity has recently challenged the long-held T helper type 1 (Th1) paradigm in sarcoidosis and striking parallels with autoimmune disorders and common variable immunodeficiency were recognized. For instance, it was demonstrated that Th17.1-cells rather than Th1-cells are responsible for the exaggerated IFN-γ production in pulmonary sarcoidosis. Furthermore, impaired regulatory T-cell function and alterations within the expression of co-inhibitory receptors that control T-cell responses, such as PD-1, CTLA-4 and BTNL2, raise new questions regarding T-cell regulation in pulmonary sarcoidosis. SUMMARY It becomes increasingly clear that Th17(.1)-cells and regulatory T-cells are key players in sarcoidosis T-cell immunology. New findings on plasticity and co-inhibitory receptor expression by these subsets help build a more comprehensive model for T-cell regulation in sarcoidosis and will finally shed light on the potential of new treatment modalities.
Collapse
|
39
|
|
40
|
Verwoerd A, Hijdra D, Vorselaars ADM, Crommelin HA, van Moorsel CHM, Grutters JC, Claessen AME. Infliximab therapy balances regulatory T cells, tumour necrosis factor receptor 2 (TNFR2) expression and soluble TNFR2 in sarcoidosis. Clin Exp Immunol 2016; 185:263-70. [PMID: 27158798 DOI: 10.1111/cei.12808] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2016] [Indexed: 12/19/2022] Open
Abstract
Sarcoidosis is a systemic granulomatous disease of unknown aetiology that most commonly affects the lungs. Although elevated levels of regulatory T cells (Tregs ) have been reported, the extent to which they play a role in sarcoidosis pathogenesis remains unclear. Tumour necrosis factor (TNF) is thought to be one of the driving forces behind granuloma formation, illustrated by the efficacy of infliximab in severe sarcoidosis. Tregs express TNF receptor 2 (TNFR2) highly. Here, we examined the influence of infliximab therapy on Tregs and (soluble) TNFR2 levels in sarcoidosis, and correlated these with response to therapy. We observed that relative frequencies of Tregs were significantly higher in patients (n = 54) compared to healthy controls (n = 26; median 6·73 versus 4·36%; P < 0·001) and decreased following therapy (4·95; P < 0·001). Baseline TNFR2 expression on Tregs was increased significantly in patients versus controls (99·4 versus 96·2%; P = 0·031), and also in responders to therapy versus non-responders (99·6 versus 97·3%; P = 0·012). Furthermore, baseline soluble TNFR2 (sTNFR2) was higher in responders than in non-responders (mean 174 versus 107 pg/ml; P = 0·015). After treatment, responders showed a significant reduction in sTNFR2 levels in peripheral blood (-44·7 pg/ml; P < 0·001), in contrast to non-responders (+3·59 pg/ml). Our results demonstrated that Treg frequencies and TNFR2 expression on Tregs are increased in sarcoidosis, followed by a decline during infliximab therapy, suggesting a pathophysiological role of this T cell subset. Interestingly, sTNFR2 levels at baseline differed significantly between responders and non-responders, making it a potential marker in predicting which patients might benefit from infliximab.
Collapse
Affiliation(s)
- A Verwoerd
- Department of Pulmonology, Interstitial Lung Diseases Centre of Excellence
| | - D Hijdra
- Department of Pulmonology, Interstitial Lung Diseases Centre of Excellence.,Department of Medical Microbiology and Immunology
| | - A D M Vorselaars
- Department of Pulmonology, Interstitial Lung Diseases Centre of Excellence
| | - H A Crommelin
- Department of Pulmonology, Interstitial Lung Diseases Centre of Excellence.,Department of Clinical Pharmacy, St Antonius Hospital, Nieuwegein
| | - C H M van Moorsel
- Department of Pulmonology, Interstitial Lung Diseases Centre of Excellence.,Division of Heart and Lungs, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - J C Grutters
- Department of Pulmonology, Interstitial Lung Diseases Centre of Excellence.,Division of Heart and Lungs, University Medical Centre Utrecht, Utrecht, the Netherlands
| | | |
Collapse
|
41
|
Liu Y, Qiu L, Wang Y, Aimurola H, Zhao Y, Li S, Xu Z. The Circulating Treg/Th17 Cell Ratio Is Correlated with Relapse and Treatment Response in Pulmonary Sarcoidosis Patients after Corticosteroid Withdrawal. PLoS One 2016; 11:e0148207. [PMID: 26845566 PMCID: PMC4742270 DOI: 10.1371/journal.pone.0148207] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 01/14/2016] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES Pulmonary sarcoidosis is an immune-mediated disease, and some patients can be effectively treated with corticosteroids. However, nearly half of all sarcoidosis patients relapse after corticosteroid withdrawal. Different subsets of CD4+ helper T cells participate in the immunopathogenesis of sarcoidosis. Thus, the aims of our study were to investigate whether the circulating subsets of CD4+ helper T cells were associated with sarcoidosis relapse and with its remission after retreatment. Additionally, we identified a useful biomarker for predicting the relapse and remission of sarcoidosis patients. METHODS Forty-two patients were enrolled in the present study who had previously been diagnosed with pulmonary sarcoidosis and treated with corticosteroids. The patients were allocated into either a stable group if they exhibited sustained remission (n = 22) or a relapse group if they experienced clinical or radiological recurrence after treatment withdrawal (n = 20). Peripheral blood cells were collected from these patients and analyzed to determine the frequencies of subsets of circulating CD4+ helper T cells by flow cytometry. The patients in the relapse group were retreated with corticosteroids and immunosuppressive agents and were then reevaluated to determine the frequencies of dynamic subsets of circulating CD4+ helper T cells after remission. RESULTS The frequencies of circulating Tregs were significantly increased concomitant with a decrease in the circulating Th17 cell frequency in the relapsed patients compared with the stable patients. The Treg/Th17 ratio was negatively correlated with sarcoidosis activity and was sensitive to retreatment. In addition, the percentage of isolated CD45RO+Ki67+ Tregs was higher in the patients who were stable and in those who recovered after retreatment than in those who relapsed. CONCLUSIONS An imbalance between Tregs and Th17 cells is associated with pulmonary sarcoidosis relapse after corticosteroid withdrawal. The circulating Treg/Th17 ratio could serve as an alternative marker for monitoring pulmonary sarcoidosis relapse after the end of corticosteroid treatment and for rapidly predicting the response to retreatment.
Collapse
Affiliation(s)
- Yongzhe Liu
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Lan Qiu
- Department of Internal Medicine, No. 2 Jikun Hospital, Urumchi, Xinjiang Uyghur Autonomous Region 830013, China
| | - Yanxun Wang
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Halimulati Aimurola
- Department of Respiratory Medicine, Chest Hospital of Xinjiang Uyghur Autonomous Region, Urumchi, Xinjiang Uyghur Autonomous Region 830049, China
| | - Yuyue Zhao
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Shan Li
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Zuojun Xu
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| |
Collapse
|