1
|
Belfeki N, Kammoun S, Ghriss N, Eldirani C, Mekinian A. Current concepts on pathogenesis, diagnosis and management of hepatic sarcoidosis. Rheumatol Int 2025; 45:130. [PMID: 40304755 DOI: 10.1007/s00296-025-05888-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Accepted: 04/10/2025] [Indexed: 05/02/2025]
Abstract
Sarcoidosis is an inflammatory multisystemic granulomatosis of unknown cause, with a wide range of clinical features, characterized by the presence of non-caseating granulomas. Liver is the third involved organ after lungs and lymph nodes, with a reported prevalence of hepatic involvement in 5 to 25% of systemic symptomatic sarcoidosis. The immunopathogenesis of sarcoidosis is still unknown but it seems to involve the innate and adaptive immune actors in response to a putative antigen in genetically predisposed individuals. Because of its paucisymptomatic presentation, hepatic sarcoidosis may be underdiagnosed. Unspecified impaired general condition, fever, abdominal pain, and jaundice are the main symptoms associated with liver sarcoidosis. Frequently, laboratory liver tests are abnormal. Imaging tools may reveal liver nodular enlargement but can be totally normal. Liver biopsy is often required to confirm the diagnosis. A meticulous workup is mandatory to rule out differential diagnosis of hepatic granuloma. Portal hypertension and liver cirrhosis are the most prevalent complications of hepatic sarcoidosis. Treatment is not necessary in all cases and first line treatment in symptomatic patients requires corticosteroids and/or ursodeoxycholic acid. Immunosuppressants and biologics could be used as second line agents. In severe cases, liver transplantation is indicated.
Collapse
Affiliation(s)
- Nabil Belfeki
- Department of Internal Medicine and Clinical Immunology, Groupe Hospitalier Sud-Ile de France, 270 Avenue Marc Jacquet, Melun, 77 000, France.
| | - Sonia Kammoun
- Department of Internal Medicine and Clinical Immunology, Groupe Hospitalier Sud-Ile de France, 270 Avenue Marc Jacquet, Melun, 77 000, France
| | - Nouha Ghriss
- Department of Internal Medicine and Clinical Immunology, Groupe Hospitalier Sud-Ile de France, 270 Avenue Marc Jacquet, Melun, 77 000, France
| | - Charif Eldirani
- Departement of Gastroenterology and Hepatology, Groupe Hospitalier Sud-Ile de France, Melun, France
| | - Arsene Mekinian
- Department of Internal Medicine, Inflammation-Immunopathology-Biotherapy, Assistance Publique - Hôpitaux de Paris, Hôpital Saint-Antoine Paris, Paris, France
- Sorbonne University, UMPC University Paris 06, INSERM U938, Centre de Recherche Saint- Antoine (CRSA), Paris, France
| |
Collapse
|
2
|
Xu D, Tao X, Fan Y, Teng Y. Sarcoidosis: molecular mechanisms and therapeutic strategies. MOLECULAR BIOMEDICINE 2025; 6:6. [PMID: 39904950 PMCID: PMC11794924 DOI: 10.1186/s43556-025-00244-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/06/2025] [Accepted: 01/13/2025] [Indexed: 02/06/2025] Open
Abstract
Sarcoidosis, a multisystemic granulomatous disease with unknown etiology, is characterized by formation of noncaseating granulomas, which can affect all organs. Recent studies have made outstanding achievement in understanding the pathology, etiology, genetics, and immune dysregulation involved in granuloma formation of sarcoidosis. Antigen stimulation in genetically predisposed individuals enhances the phagocytic activity of antigen-presenting cells, including macrophages and dendritic cells. CD4 + T cells initiate dysregulated immune responses and secrete significant quantities of inflammatory cytokines, including interleukin (IL)-2 and interferon-gamma (IFN-γ), which play a crucial role in modulating the aggregation and fusion of macrophages to form granulomas. The current therapeutic strategies focus on blocking the formation and spread of granulomas to protect organ function and alleviate symptoms. The efficacy of traditional treatments, such as glucocorticoids and immunosuppressants, has been confirmed in the management of sarcoidosis. Promising therapeutic agents encompass inhibitors of cytokines, like those targeting tumor necrosis factor (TNF)-α, as well as inhibitors of signaling pathways, such as Janus kinase (JAK) inhibitors, which exhibit favorable prospects for application. Although there has been progress in the identification of biomarkers for the diagnosis, prognosis, activity and severity of sarcoidosis, specific and sensitive biomarkers have yet to be identified. This review outlines recent advancements in the molecular mechanisms and therapeutic strategies for the sarcoidosis.
Collapse
Affiliation(s)
- Danfeng Xu
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaohua Tao
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yibin Fan
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yan Teng
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Levra S, Giannoccaro F, Chernovsky M, Carriero V, Arrigo E, Bertolini F, Balbi M, Pizzimenti S, Guida G, Ricciardolo FLM. Alveolar nitric oxide concentration as a potential biomarker of fibrosis and active disease in pulmonary sarcoidosis: a pilot study. J Breath Res 2025; 19:026003. [PMID: 39836984 DOI: 10.1088/1752-7163/adac82] [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: 11/13/2024] [Accepted: 01/21/2025] [Indexed: 01/23/2025]
Abstract
Sarcoidosis is considered a T-helper (Th) 1 related disease, but a transition from Th1 to Th2 pathway activation has been postulated in sarcoidosis-associated pulmonary fibrosis (SAPF). Fraction of exhaled nitric oxide (FENO) is a marker of Th2 airway inflammation, but alveolar concentration of nitric oxide (CANO) can be measured to assess Th2 inflammation in the periphery of the lung. The aim of this study is to assess whether CANO can be considered a biomarker of SAPF or active pulmonary sarcoidosis. In this single-center retrospective study, we compared exhaled NO levels of patients with pulmonary sarcoidosis without fibrosis (N= 11) with those obtained from patients with SAPF (N= 15). Clinical data, as well as respiratory function tests, were also analyzed. FENO (28.5 ± 16 ppb vs 30.9 ± 17.2 ppb,p= 0.72) and CANO (4.4 ± 3.5 ppb vs 3.2 ± 1.7 ppb,p= 0.73) levels did not differ significantly between patients with or without SAPF, even when dividing them according to treatment or disease activity. CANO appeared reduced in patients with active sarcoidosis (2.1 ± 0.8 ppb vs 4.1 ± 3 ppb,p< 0.05). In conclusion, CANO cannot be considered a biomarker of SAPF. Its lower level in patients with active disease confirms the prevalence of Th1 inflammation in granuloma formation and suggests its potential role as a biomarker of active pulmonary sarcoidosis, but further studies with larger samples are needed to confirm this hypothesis.
Collapse
Affiliation(s)
- Stefano Levra
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | | | - Maria Chernovsky
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Vitina Carriero
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Elisa Arrigo
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Francesca Bertolini
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - Maurizio Balbi
- Radiology Unit, Department of Oncology, San Luigi Gonzaga University Hospital, Orbassano, Turin, Italy
| | | | - Giuseppe Guida
- San Luigi Gonzaga University Hospital, Orbassano, Turin, Italy
| | - Fabio L M Ricciardolo
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
- San Luigi Gonzaga University Hospital, Orbassano, Turin, Italy
- Institute of Translational Pharmacology, National Research Council (IFT-CNR), section of Palermo, Italy
| |
Collapse
|
4
|
Ma C, Huang J, Zheng Y, Na Y, Wei J, Shan J, Meng K, Zhang X, Zhang S, Wen Y, Ding J. Anti-TL1A monoclonal antibody modulates the dysregulation of Th1/Th17 cells and attenuates granuloma formation in sarcoidosis by inhibiting the PI3K/AKT signaling pathway. Int Immunopharmacol 2024; 137:112360. [PMID: 38852524 DOI: 10.1016/j.intimp.2024.112360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/26/2024] [Accepted: 05/26/2024] [Indexed: 06/11/2024]
Abstract
Sarcoidosis is a systemic granulomatous disease characterized by non-caseating epithelioid cell granulomas. One of its immunological hallmarks is the differentiation of CD4 + naïve T cells into Th1/Th17 cells, accompanied by the release of numerous pro-inflammatory cytokines. The TL1A/DR3 signaling pathway plays a crucial role in activating effector lymphocytes, thereby triggering pro-inflammatory responses. The primary aim of this investigation was to scrutinize the impact of anti-TL1A monoclonal antibody on the dysregulation of Th1/Th17 cells and granuloma formation in sarcoidosis. Initially, the abnormal activation of the TL1A/DR3 signaling pathway in pulmonary tissues of sarcoidosis patients was confirmed using qPCR and immunohistochemistry techniques. Subsequently, employing a murine model of sarcoidosis, the inhibitory effects of anti-TL1A monoclonal antibody on the TL1A/DR3 signaling pathway in sarcoidosis were investigated through qPCR, immunohistochemistry, and Western blot experiments. The influence of anti-TL1A monoclonal antibody on granulomas was assessed through HE staining, while their effects on sarcoidosis Th1/Th17 cells and associated cytokine mRNA levels were evaluated using flow cytometry and qPCR, respectively. Immunofluorescence and Western blot experiments corroborated the inhibitory effects of anti-TL1A monoclonal antibody on the aberrant activation of the PI3K/AKT signaling pathway in sarcoidosis. The findings of this study indicate that the TL1A/DR3 signaling pathway is excessively activated in sarcoidosis. Anti-TL1A monoclonal antibody effectively inhibit this abnormal activation in sarcoidosis, thereby alleviating the dysregulation of Th1/Th17 cells and reducing the formation of pulmonary granulomas. This effect may be associated with the inhibition of the downstream PI3K/AKT signaling pathway. Anti-TL1A monoclonal antibody hold promise as a potential novel therapeutic intervention for sarcoidosis.
Collapse
Affiliation(s)
- Chengxing Ma
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiayi Huang
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yin Zheng
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yuqi Na
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Jia Wei
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jiajia Shan
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Kaifang Meng
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Xian Zhang
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Shiyun Zhang
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China
| | - Yanting Wen
- Department of Basic Medicine, Center of Translational Medicine, Jiangsu Key Laboratory of Molecular Medicine, Nanjing University Medical School, Nanjing, China.
| | - Jingjing Ding
- Department of Respiratory and Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
5
|
Heymans S, Van Linthout S, Kraus SM, Cooper LT, Ntusi NAB. Clinical Characteristics and Mechanisms of Acute Myocarditis. Circ Res 2024; 135:397-411. [PMID: 38963866 DOI: 10.1161/circresaha.124.324674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT05335928.
Collapse
Affiliation(s)
- Stephane Heymans
- Centre for Heart Failure Research, Department of Cardiology, Maastricht University, The Netherlands (S.H.)
- Department of Cardiovascular Sciences, University of Leuven, Belgium (S.H.)
| | - Sophie Van Linthout
- Berlin Institute of Health Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Germany (S.V.L.)
- German Centre for Cardiovascular Research, partner site Berlin, Germany (S.V.L.)
| | - Sarah Mignon Kraus
- Division of Cardiology, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur Hospital, South Africa (S.M.K., N.A.B.N.)
- South African Medical Research Council Extramural Unit on Intersection of Noncommunicable Diseases and Infectious Diseases, Cape Town, South Africa (S.M.K., N.A.B.N.)
| | - Leslie T Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL (L.T.C.)
| | - Ntobeko A B Ntusi
- Division of Cardiology, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur Hospital, South Africa (S.M.K., N.A.B.N.)
- South African Medical Research Council Extramural Unit on Intersection of Noncommunicable Diseases and Infectious Diseases, Cape Town, South Africa (S.M.K., N.A.B.N.)
- Cape Heart Institute, Faculty of Health Sciences, University of Cape Town, South Africa (N.A.B.N.)
- ARUA/Guild Cluster of Research Excellence on Noncommunicable Diseases and Associated Multiborbidity, South Africa (N.A.B.N.)
| |
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
|
Li X, Li Y, Lv S, Xu H, Ma R, Sun Z, Li Y, Guo C. Long-term respiratory exposure to amorphous silica nanoparticles promoted systemic inflammation and progression of fibrosis in a susceptible mouse model. CHEMOSPHERE 2022; 300:134633. [PMID: 35439488 DOI: 10.1016/j.chemosphere.2022.134633] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/26/2022] [Accepted: 04/13/2022] [Indexed: 06/14/2023]
Abstract
Exposure to amorphous silica nanoparticles (SiNPs) has increased dramatically, and concerns are growing about their potential health effects. However, their long-term systemic toxicity profile and underlying mechanisms following respiratory exposure still remains unexplored. It is well documented that the inhalation of ultrafine particles is firmly associated with adverse effects in humans. Environmental pollutants may contribute to diverse adverse effect or comorbidity in susceptible individuals. Thereby, we examined the long-term systemic effects of inhaled SiNPs using a sensitive mouse model (ApoE-/-) fed by a western diet. Male ApoE-/- mice were intratracheally instilled with SiNPs suspension at a dose of 1.5, 3.0 and 6.0 mg/kg·bw, respectively, once per week, 12 times in total. The histological analysis was conducted. The serum cytokine levels were quantified by RayBiotech antibody array. As a result, systemic histopathological alterations were noticed, mainly characterized by inflammation and fibrosis. More importantly, cytokine array analysis indicated the key role of mast cells accumulation in systemic inflammation and fibrosis progression induced by inhaled SiNPs. Collectively, our study firstly demonstrated that long-term exposure to inhaled SiNPs promoted the mast cell-dominated activation of inflammatory response, not only in the lung but also in heart, liver and kidney, etc., eventually leading to the progression of tissue fibrosis in ApoE-/- mice.
Collapse
Affiliation(s)
- Xueyan Li
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yan Li
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Songqing Lv
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Hailin Xu
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Ru Ma
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Zhiwei Sun
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Yanbo Li
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China; Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Caixia Guo
- Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
8
|
Zhang Y, Ye Y, Tang X, Wang H, Tanaka T, Tian R, Yang X, Wang L, Xiao Y, Hu X, Jin Y, Pang H, Du T, Liu H, Sun L, Xiao S, Dong R, Ferrucci L, Tian Z, Zhang S. CCL17 acts as a novel therapeutic target in pathological cardiac hypertrophy and heart failure. J Exp Med 2022; 219:213274. [PMID: 35687056 PMCID: PMC9194836 DOI: 10.1084/jem.20200418] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/22/2022] [Accepted: 06/01/2022] [Indexed: 11/18/2022] Open
Abstract
Circulating proteomic signatures of age are closely associated with aging and age-related diseases; however, the utility of changes in secreted proteins in identifying therapeutic targets for diseases remains unclear. Serum proteomic profiling of an age-stratified healthy population and further community-based cohort together with heart failure patients study demonstrated that circulating C-C motif chemokine ligand 17 (CCL17) level increased with age and correlated with cardiac dysfunction. Subsequent animal experiments further revealed that Ccll7-KO significantly repressed aging and angiotensin II (Ang II)–induced cardiac hypertrophy and fibrosis, accompanied by the plasticity and differentiation of T cell subsets. Furthermore, the therapeutic administration of an anti-CCL17 neutralizing antibody inhibited Ang II–induced pathological cardiac remodeling. Our findings reveal that chemokine CCL17 is identifiable as a novel therapeutic target in age-related and Ang II–induced pathological cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicong Ye
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hui Wang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Toshiko Tanaka
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Ran Tian
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xufei Yang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lun Wang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Xiao
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaomin Hu
- Department of Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ye Jin
- Department of Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haiyu Pang
- Department of Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tian Du
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Honghong Liu
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lihong Sun
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuo Xiao
- Thermo Fisher Scientific (China) Co., Ltd, Changning, Shanghai, China
| | - Ruijia Dong
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, Baltimore, MD
| | - Zhuang Tian
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuyang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Department of Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
9
|
Schrijver B, Kolijn PM, Berge JC, Nagtzaam NM, Rijswijk AL, Swagemakers SM, Spek PJ, Missotten TO, Velthoven ME, Hoog J, Hagen PM, Langerak AW, Dik WA. Vitreous proteomics, a gateway to improved understanding and stratification of diverse uveitis aetiologies. Acta Ophthalmol 2022; 100:403-413. [PMID: 34318583 PMCID: PMC9292680 DOI: 10.1111/aos.14993] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/06/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE The vitreous proteome might provide an attractive gateway to discriminate between various uveitis aetiologies and gain novel insights into the underlying pathophysiological processes. Here, we investigated 180 vitreous proteins to discover novel biomarkers and broaden disease insights by comparing (1). primary vitreoretinal lymphoma ((P)VRL) versus other aetiologies, (2). sarcoid uveitis versus tuberculosis (TB)-associated uveitis and (3). granulomatous (sarcoid and TB) uveitis versus other aetiologies. METHODS Vitreous protein levels were determined by proximity extension assay in 47 patients with intraocular inflammation and a prestudy diagnosis (cohort 1; training) and 22 patients with a blinded diagnosis (cohort 2; validation). Differentially expressed proteins identified by t-tests on cohort 1 were used to calculate Youden's indices. Pathway and network analysis was performed by ingenuity pathway analysis. A random forest classifier was trained to predict the diagnosis of blinded patients. RESULTS For (P)VRL stratification, the previously reported combined diagnostic value of IL-10 and IL-6 was confirmed. Additionally, CD70 was identified as potential novel marker for (P)VRL. However, the classifier trained on the entire cohort (cohort 1 and 2) relied primarily on the interleukin score for intraocular lymphoma diagnosis (ISOLD) or IL-10/IL-6 ratio and only showed a supportive role for CD70. Furthermore, sarcoid uveitis displayed increased levels of vitreous CCL17 as compared to TB-associated uveitis. CONCLUSION We underline the previously reported value of the ISOLD and the IL-10/IL-6 ratio for (P)VRL identification and present CD70 as a potentially valuable target for (P)VRL stratification. Finally, we also show that increased CCL17 levels might help to distinguish sarcoid uveitis from TB-associated uveitis.
Collapse
Affiliation(s)
- Benjamin Schrijver
- Department of Immunology Laboratory Medical Immunology Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - P. Martijn Kolijn
- Department of Immunology Laboratory Medical Immunology Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Josianne C.E.M. Berge
- Department of Ophthalmology Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Nicole M.A. Nagtzaam
- Department of Immunology Laboratory Medical Immunology Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Angelique L.C.T. Rijswijk
- Department of Immunology Laboratory Medical Immunology Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Sigrid M.A. Swagemakers
- Department of Bioinformatics Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Peter J. Spek
- Department of Bioinformatics Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | | | | | - Joeri Hoog
- Department of Ophthalmology Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - P. Martin Hagen
- Department of Immunology Laboratory Medical Immunology Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
- Department of Internal Medicine Section Clinical Immunology Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Anton W. Langerak
- Department of Immunology Laboratory Medical Immunology Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| | - Willem A. Dik
- Department of Immunology Laboratory Medical Immunology Erasmus MC University Medical Center Rotterdam Rotterdam the Netherlands
| |
Collapse
|
10
|
Byg KE, Illes Z, Sejbaek T, Lambertsen KL, Ellingsen T, Nielsen HH. Inflammatory profiles in plasma and cerebrospinal fluid of patients with neurosarcoidosis. J Neuroimmunol 2022; 367:577849. [DOI: 10.1016/j.jneuroim.2022.577849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 03/04/2022] [Accepted: 03/22/2022] [Indexed: 10/18/2022]
|
11
|
Schrijver B, La Distia Nora R, Nagtzaam NMA, Laar JAM, Hagen PM, Dik WA. Serum CCL17 distinguishes sarcoid uveitis from TB-uveitis and QFT-negative uveitis. Acta Ophthalmol 2022; 100:e1533-e1534. [PMID: 35080799 DOI: 10.1111/aos.15093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/21/2021] [Accepted: 12/29/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Benjamin Schrijver
- Laboratory Medical Immunology, Department of Immunology Erasmus MC, University Medical Center Rotterdam Rotterdam The Netherlands
| | - Rina La Distia Nora
- Department of Ophthalmology Faculty of Medicine University of Indonesia and Cipto Mangunkusumo Hospital Jakarta Indonesia
| | - Nicole M. A. Nagtzaam
- Laboratory Medical Immunology, Department of Immunology Erasmus MC, University Medical Center Rotterdam Rotterdam The Netherlands
| | - Jan A. M. Laar
- Laboratory Medical Immunology, Department of Immunology Erasmus MC, University Medical Center Rotterdam Rotterdam The Netherlands
- Department of Internal Medicine, Section Allergy & Clinical Immunology Erasmus MC, University Medical Center Rotterdam Rotterdam The Netherlands
| | - P. Martin Hagen
- Laboratory Medical Immunology, Department of Immunology Erasmus MC, University Medical Center Rotterdam Rotterdam The Netherlands
- Department of Internal Medicine, Section Allergy & Clinical Immunology Erasmus MC, University Medical Center Rotterdam Rotterdam The Netherlands
| | - Willem A. Dik
- Laboratory Medical Immunology, Department of Immunology Erasmus MC, University Medical Center Rotterdam Rotterdam The Netherlands
| |
Collapse
|
12
|
Effects of phenylethanol glycosides from Orobanche cernua Loefling on UVB-Induced skin photodamage: a comparative study. Photochem Photobiol Sci 2021; 20:599-614. [PMID: 33909279 DOI: 10.1007/s43630-021-00038-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/24/2021] [Indexed: 10/21/2022]
Abstract
Previous study has found that Orobanche cernua Loefling(OC) and its main ingredient, acteoside, possess excellently anti-photo-aging effect. In addition to acteoside, crenatoside, isoacteoside and 2'-acetylacteoside were also identified as the main phenylethanol glycosides (PhGs) in OC. To screen optimum effective substance and further clarify the photoprotective ingredients of OC, the effects of four major PhGs in OC were compared using UVB-irradiated HaCaT cells. Results indicated that acteoside, isoacteoside and 2'-acetylacteoside effectively decreased UVB-induced MMP-1 expression and stimulated type I procollagen synthesis through inhibition of MAPK/AP-1 and activation of TGF-β/Smad pathway. Moreover, acteoside and 2'-acetylacteoside significantly reduced UVB-induced ROS and TARC secretion, which is involved in the inhibition of NF-κβ/Iκβα and stimulation of Nrf2 antioxidant defense system. However, crenatoside did not show any effect on the regulation of signal cascades mentioned above. Together, our results suggested that 2'-acetylacteoside and isoacteoside also served as efficient agents against UV radiation-induced skin damage. Among them, acteoside and 2'-acetylacteoside showed a higher efficiency than that of isoacteoside, which possessed great potential in treating skin photo-damage.
Collapse
|
13
|
Cheikhi AM, Johnson ZI, Julian DR, Wheeler S, Feghali-Bostwick C, Conley YP, Lyons-Weiler J, Yates CC. Prediction of severity and subtype of fibrosing disease using model informed by inflammation and extracellular matrix gene index. PLoS One 2020; 15:e0240986. [PMID: 33095822 PMCID: PMC7584227 DOI: 10.1371/journal.pone.0240986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/06/2020] [Indexed: 11/19/2022] Open
Abstract
Fibrosis is a chronic disease with heterogeneous clinical presentation, rate of progression, and occurrence of comorbidities. Systemic sclerosis (scleroderma, SSc) is a rare rheumatic autoimmune disease that encompasses several aspects of fibrosis, including highly variable fibrotic manifestation and rate of progression. The development of effective treatments is limited by these variabilities. The fibrotic response is characterized by both chronic inflammation and extracellular remodeling. Therefore, there is a need for improved understanding of which inflammation-related genes contribute to the ongoing turnover of extracellular matrix that accompanies disease. We have developed a multi-tiered method using Naïve Bayes modeling that is capable of predicting level of disease and clinical assessment of patients based on expression of a curated 60-gene panel that profiles inflammation and extracellular matrix production in the fibrotic disease state. Our novel modeling design, incorporating global and parametric-based methods, was highly accurate in distinguishing between severity groups, highlighting the importance of these genes in disease. We refined this gene set to a 12-gene index that can accurately identify SSc patient disease state subsets and informs knowledge of the central regulatory pathways in disease progression.
Collapse
Affiliation(s)
- Amin M. Cheikhi
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States of America
| | - Zariel I. Johnson
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States of America
| | - Dana R. Julian
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States of America
- Department of Health Promotion and Development, University of Pittsburgh School of Nursing, Pittsburgh, PA, United States of America
| | - Sarah Wheeler
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
| | - Carol Feghali-Bostwick
- Department of Rheumatology & Immunology, Medical University of South Carolina, Charleston, SC, United States of America
| | - Yvette P. Conley
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States of America
| | - James Lyons-Weiler
- Genomic and Proteomic Core Laboratories, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Cecelia C. Yates
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States of America
- Department of Health Promotion and Development, University of Pittsburgh School of Nursing, Pittsburgh, PA, United States of America
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States of America
- * E-mail:
| |
Collapse
|
14
|
Kim YJ, Ahn HJ, Lee SH, Lee MH, Kang KS. Effects of conditioned media from human umbilical cord blood-derived mesenchymal stem cells in the skin immune response. Biomed Pharmacother 2020; 131:110789. [PMID: 33152947 DOI: 10.1016/j.biopha.2020.110789] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/12/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Atopic dermatitis (AD) is an inflammatory skin disease in which type 2 allergic inflammation plays a critical role. In this study, the anti-inflammatory effect of conditioned media from human umbilical cord blood-derived mesenchymal stem cells (USC-CM) was investigated in order to apply it as an effective treatment with a low risk of side effects that can overcome the limitations of AD treatment which is currently in use. We found that USC-CM has various growth factors and cytokines associated with anti-inflammatory effect. RT-PCR and ELISA analysis showed that USC-CM inhibited the levels of type 2 cytokine and chemokine Thymus and activation-regulated chemokine (TARC), TNF-α and IL-6 in TNF-α/IFN-γ-stimulated HaCaT cells. In addition, USC-CM inhibited IL-4 and IL-13 levels in Th2 cells. Therefore, the results of our study demonstrated that USC-CM has anti-inflammatory effect in TNF-α/IFN-γ-stimulated HaCaT cells which associated with the inhibition of the immunoglobulin (IgE) secretion by activating B cell line. Our In vivo results showed that when the USC-CM was applied to lesions of patients with the mild AD for 4 weeks, the skin barrier was strengthened by increasing the level of Corneometer and decreasing the value of transepidermal water loss (TEWL). In conclusion, the results suggest that USC-CM may have therapeutic effect for AD as cosmetics and drug materials.
Collapse
Affiliation(s)
- Yoon-Jin Kim
- Derma Science R&D Center, Primoris International CO., LTD., #1504, A Bldg., 60 Haahn-ro, Gwangmyeong-si, Gyeonggi-do, 14332, Republic of Korea
| | - Hee-Jin Ahn
- Cytotherapy R&D Center, PRIMORIS CO., LTD., #1504, A Bldg., 60 Haahn-ro, Gwangmyeong-si, Gyeonggi-do, 14332, Republic of Korea
| | - Seung-Hee Lee
- Stem Cells and Regenerative Bioengineering Institute, Kangstem Biotech Co., Ltd., 2nd Floor, Biotechnology Center, #81 Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Mi-Hye Lee
- GMP Center, Kangstem Biotech Co., Ltd., 6nd Floor, A Bldg., 60 Haahn-ro, Gwangmyeong-si, Gyeonggi-do, 14332, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea; Cytotherapy R&D Center, PRIMORIS CO., LTD., #1504, A Bldg., 60 Haahn-ro, Gwangmyeong-si, Gyeonggi-do, 14332, Republic of Korea.
| |
Collapse
|
15
|
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: 4.2] [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
|
16
|
Current perspectives on the immunopathogenesis of sarcoidosis. Respir Med 2020; 173:106161. [PMID: 32992264 DOI: 10.1016/j.rmed.2020.106161] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 08/28/2020] [Accepted: 09/16/2020] [Indexed: 12/25/2022]
Abstract
Sarcoidosis is an inflammatory systemic disease that commonly affects the lungs or lymph nodes but can manifest in other organs. Herein, we review the latest evidence establishing how innate and adaptive immune responses contribute to the pathogenesis and clinical course of sarcoidosis. We discuss the possible role of microbial organisms as etiologic agents in sarcoidosis and the evidence supporting sarcoidosis as an autoimmune disease. We also discuss how animal and in vitro human models have advanced our understanding of the immunopathogenesis of sarcoidosis. Finally, we discuss therapeutics for sarcoidosis and the effects on the immune system.
Collapse
|
17
|
Komatsu M, Yamamoto H, Yasuo M, Ushiki A, Nakajima T, Uehara T, Kawakami S, Hanaoka M. The utility of serum C-C chemokine ligand 1 in sarcoidosis: A comparison to IgG4-related disease. Cytokine 2020; 133:155123. [PMID: 32447247 DOI: 10.1016/j.cyto.2020.155123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023]
Abstract
We previously reported higher levels of C-C chemokine ligand (CCL) 1 in the bronchoalveolar lavage (BAL) fluid (BALF) of patients with sarcoidosis than in BALF of patients with immunoglobulin G4 (IgG4)-related disease (IgG4-RD), indicating that CCL1 might act as a marker of disease activity in sarcoidosis. Notably, less invasive sampling sources are desirable, as BAL cannot always be performed due to its inherent risk. In this study, we sought to decipher the correlation between serum levels of CCL1 and clinical characteristics of sarcoidosis. Serum samples were obtained from 44 patients with clinically confirmed sarcoidosis, 14 patients with IgG4-RD, and 14 healthy controls. The clinical and radiological findings were retrospectively evaluated. Serum levels of CCL1 were measured using a sandwich enzyme-linked immunosorbent assay. Serum levels of other 17 cytokines and chemokines were measured using a MILLIPLEX® MAP KIT and Luminex® magnetic beads. Serum levels of CCL1 were significantly higher in patients with sarcoidosis than in patients with IgG4-RD and healthy controls. Serum CCL1 was positively correlated with the degree of hilar lymph node swelling on chest computed tomography and serum levels of soluble interleukin 2 receptor. Positive correlations were also observed between serum CCL1 and total cell counts, lymphocyte counts in BALF, and serum T helper 1 mediators such as IP-10 and TNF-α in patients with sarcoidosis. Serum CCL1 levels were significantly elevated in sarcoidosis and correlated with clinical parameters of the disease. In addition, serum and BALF levels of CCL1 were positively correlated in a statistically significant manner. Although further research in this field is necessary, CCL1 might have the potential to be a reliable serological marker of disease activity in sarcoidosis.
Collapse
Affiliation(s)
- Masamichi Komatsu
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan.
| | - Hiroshi Yamamoto
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan.
| | - Masanori Yasuo
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan.
| | - Atsuhito Ushiki
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan.
| | - Tomoyuki Nakajima
- Department of Laboratory Medicine, Shinshu University School of Medicine, Matsumoto, Japan.
| | - Takeshi Uehara
- Department of Laboratory Medicine, Shinshu University School of Medicine, Matsumoto, Japan.
| | - Satoshi Kawakami
- Department of Radiology, Shinshu University School of Medicine, Matsumoto, Japan.
| | - Masayuki Hanaoka
- First Department of Internal Medicine, Shinshu University School of Medicine, Matsumoto, Japan.
| |
Collapse
|
18
|
Kishimoto I, Nguyen CTH, Kambe N, Ly NTM, Ueki Y, Ueda-Hayakawa I, Okamoto H. Circulating intermediate monocytes produce TARC in sarcoidosis. Allergol Int 2020; 69:310-312. [PMID: 31662224 DOI: 10.1016/j.alit.2019.09.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 09/18/2019] [Accepted: 09/28/2019] [Indexed: 12/22/2022] Open
|
19
|
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: 16] [Impact Index Per Article: 3.2] [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
|
20
|
Kalinauskaite-Zukauske V, Januskevicius A, Janulaityte I, Miliauskas S, Malakauskas K. Expression of eosinophil β chain-signaling cytokines receptors, outer-membrane integrins, and type 2 inflammation biomarkers in severe non-allergic eosinophilic asthma. BMC Pulm Med 2019; 19:158. [PMID: 31438916 PMCID: PMC6706886 DOI: 10.1186/s12890-019-0904-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 07/23/2019] [Indexed: 01/21/2023] Open
Abstract
Background Severe non-allergic eosinophilic asthma (SNEA) is a rare asthma phenotype associated with severe clinical course, frequent exacerbations, and resistance to therapy, including high steroid doses. The key feature is type 2 inflammation with predominant airway eosinophilia. Eosinophil maturation, activation, survivability, and recruitment are mainly induced by interleukin (IL)-3, IL-5 and granulocyte–macrophage colony-stimulating factor (GM-CSF) through their receptors on eosinophil surface and related with integrins activation states. The aim of the study was to estimate the expression of eosinophil β chain-signaling cytokines receptors, outer-membrane integrins, and serum-derived type 2 inflammation biomarkers in SNEA. Methods We examined 8 stable SNEA patients with high inhaled steroid doses, 12 steroid-free patients with non-severe allergic asthma (AA), 12 healthy subjects (HS). Blood eosinophils were isolated using Ficol gradient centrifugation and magnetic separation. Eosinophils were lysed, and mRNA was isolated. Gene expressions of IL-5Rα, IL-3Rα, GM-CSFRα, and α4β1, αMβ2 integrins were analyzed using quantitative real-time reverse transcription polymerase chain reaction. Type 2 inflammation activity was evaluated measuring exhaled nitric oxide concentration (FeNO) collected with the electrochemical sensing device. Serum IL-5, IL-3, GM-CSF, periostin, chemokine ligand (CCL) 17 and eotaxin concentrations were assessed by enzyme-linked immunosorbent assay. Results Eosinophils from SNEA patients demonstrated significantly increased gene expression of IL-3Rα, IL-5Rα and GM-CSFRα as well as α4, β1 and αM integrin subunits compared with the AA group. The highest IL-5 serum concentration was in the SNEA group; it significantly differed compared with AA and HS. GM-CSF serum levels were similar in the SNEA and AA groups and were significantly lower in the HS group. No differences in serum IL-3 concentration were found among all groups. Furthermore, serum levels of eotaxin, CCL17 and FeNO, but not periostin, differed in all groups, with the highest levels in SNEA patients. Conclusions Eosinophil demonstrated higher expression of IL-3, IL-5, GM-CSF α-chain receptors and α4, β1, αM integrins subunits in SNEA compared with the AA group. Additionally, SNEA patients had increased serum levels of IL-5, eotaxin and CCL-17. Trial registration ClinicalTrials.gov Identifier NCT03388359.
Collapse
Affiliation(s)
| | - Andrius Januskevicius
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Ieva Janulaityte
- Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Skaidrius Miliauskas
- Department of Pulmonology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Kestutis Malakauskas
- Department of Pulmonology, Lithuanian University of Health Sciences, Kaunas, Lithuania.,Laboratory of Pulmonology, Department of Pulmonology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
21
|
Ueda‐Hayakawa I, Nguyen CTH, Kishimoto I, Ly NTM, Okamoto H. Clinical characteristics of sarcoidosis patients with systemic sclerosis‐specific autoantibody: Possible involvement of thymus and activation‐regulated chemokine and a review of the published works. J Dermatol 2019; 46:577-583. [DOI: 10.1111/1346-8138.14932] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 04/24/2019] [Indexed: 11/29/2022]
Affiliation(s)
| | - Chuyen Thi Hong Nguyen
- Department of Dermatology Kansai Medical University Hirakata Osaka Japan
- Department of Dermatology and Venereology University of Medicine and Pharmacy Ho Chi Minh City Vietnam
| | - Izumi Kishimoto
- Department of Dermatology Kansai Medical University Hirakata Osaka Japan
| | - Nhung Thi My Ly
- Department of Dermatology Kansai Medical University Hirakata Osaka Japan
| | - Hiroyuki Okamoto
- Department of Dermatology Kansai Medical University Hirakata Osaka Japan
| |
Collapse
|
22
|
Bao L, Hao C, Liu S, Zhang L, Wang J, Wang D, Li Y, Yao W. Dendritic cells trigger imbalance of Th1/Th2 cells in silica dust exposure rat model via MHC-II, CD80, CD86 and IL-12. RSC Adv 2018; 8:26108-26115. [PMID: 35541981 PMCID: PMC9083086 DOI: 10.1039/c8ra03970d] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/07/2018] [Indexed: 12/24/2022] Open
Abstract
Silicosis is one of the most common occupational respiratory diseases caused by inhaling silica dust over a prolonged period of time, and the progression of silicosis is accompanied with chronic inflammation and progressive pulmonary fibrosis, in which dendritic cells (DCs), the most powerful antigen presentation cell (APC) in the immune response, play a crucial role. To investigate the role of DCs in the development of silicosis, we established an experimental silicosis rat model and examined the number of DCs and alveolar macrophages (AMs) in lung tissues using immunofluorescence over 84 days. Additionally, to obtain an overview of the immunological changes in rat lung tissues, a series of indicators including Th1/Th2 cells, IFN-γ, IL-4, MHC-II, CD80/86 and IL-12 were detected using flow cytometry and an enzyme-linked immunosorbent assay (ELISA) as well as a real-time polymerase chain reaction (PCR) assay. We observed that the number of DCs slightly increased at the inflammatory stage, and it increased significantly at the final stage of fibrosis. Polarization of Th1 cells and IFN-γ expressions were dominant during the inflammatory stage, whereas polarization of Th2 cells and IL-4 expressions were dominant during the fibrotic stage. The subsequent mechanistic study found that the expressions of MHC-II, CD80/86 and IL-12, which are the key molecules that connect DCs and Th cells, changed dynamically in the experimental silicosis rat model. The data obtained in this study indicated that the increase in DCs may contribute to polarization of Th1/Th2 cells via MHC-II, CD80/86, and IL-12 in silica dust-exposed rats.
Collapse
Affiliation(s)
- Lei Bao
- School of Public Health, Zhengzhou University No. 100 Science Avenue Zhengzhou Henan 450001 China +86-371-67781922 +86-371-67781922
| | - Changfu Hao
- School of Public Health, Zhengzhou University No. 100 Science Avenue Zhengzhou Henan 450001 China +86-371-67781922 +86-371-67781922
| | - Suna Liu
- The Third Affiliated Hospital of Zhengzhou University Zhengzhou Henan 450001 China
| | - Lin Zhang
- School of Public Health, Zhengzhou University No. 100 Science Avenue Zhengzhou Henan 450001 China +86-371-67781922 +86-371-67781922
| | - Juan Wang
- Hebei General Hospital Shijiazhuang Hebei 050000 China
| | - Di Wang
- School of Public Health, Zhengzhou University No. 100 Science Avenue Zhengzhou Henan 450001 China +86-371-67781922 +86-371-67781922
| | - Yiping Li
- School of Public Health, Zhengzhou University No. 100 Science Avenue Zhengzhou Henan 450001 China +86-371-67781922 +86-371-67781922
| | - Wu Yao
- School of Public Health, Zhengzhou University No. 100 Science Avenue Zhengzhou Henan 450001 China +86-371-67781922 +86-371-67781922
| |
Collapse
|