1
|
Liu M, Ma L, An W, Yang Y, Liu J, Jiang H, Yuan J, Sun X, Zhu J, Yan M, Wang L, Li Z, Liao Z, Sun C. Heterozygous Spink1 c.194+2T>C mutation promotes chronic pancreatitis after acute attack in mice. Pancreatology 2024; 24:677-689. [PMID: 38763786 DOI: 10.1016/j.pan.2024.05.514] [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] [Received: 01/15/2024] [Revised: 04/26/2024] [Accepted: 05/06/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND & AIMS Mutations in genes, including serine protease inhibitor Kazal-type 1 (SPINK1), influence disease progression following sentinel acute pancreatitis event (SAPE) attacks. SPINK1 c.194+2T > C intron mutation is one of the main mutants of SPINK1,which leads to the impairment of SPINK1 function by causing skipping of exon 3. Research on the pathogenesis of SAPE attacks would contribute to the understanding of the outcomes of acute pancreatitis. Therefore, the aim of the study was to clarify the role of SPINK1 c.194+2T > C mutation in the CP progression after an AP attack. METHODS SAPE attacks were induced in wildtype and SPINK mutant (Spink1 c.194+2T > C) mice by cerulein injection. The mice were sacrificed at 24 h, 14 d, 28 d, and 42 d post-SAPE. Data-independent acquisition (DIA) proteomic analysis was performed for the identification of differentially expressed protein in the pancreatic tissues. Functional analyses were performed using THP-1 and HPSCs. RESULTS Following SAPE attack, the Spink1 c.194+2T > C mutant mice exhibited a more severe acute pancreatitis phenotype within 24 h. In the chronic phase, the chronic pancreatitis phenotype was more severe in the Spink1 c.194+2T > C mutant mice after SAPE. Proteomic analysis revealed elevated IL-33 level in Spink1 c.194+2T > C mutant mice. Further in vitro analyses revealed that IL-33 induced M2 polarization of macrophages and activation of pancreatic stellate cells. CONCLUSION Spink1 c.194+2T > C mutation plays an important role in the prognosis of patients following SAPE. Heterozygous Spink1 c.194+2T > C mutation promotes the development of chronic pancreatitis after an acute attack in mice through elevated IL-33 level and the induction of M2 polarization in coordination with pancreatic stellate cell activation.
Collapse
Affiliation(s)
- Muyun Liu
- Department of Gastroenterology, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China; Department of Gastroenterology, NO. 905 Hospital of PLA Navy affiliated to Naval Medical University, Shanghai, 200050, China; Shanghai Institute of Pancreatic Diseases, Shanghai, 200433, China
| | - Lizhe Ma
- Department of Gastroenterology, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, 200433, China; Department of Gastroenterology, No 988 Hospital of PLA Joint Logistics Support Force, Zhengzhou, 450000, China
| | - Wei An
- Department of Gastroenterology, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, 200433, China
| | - Yaying Yang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, 400016, China
| | - Juncen Liu
- Department of Gastroenterology, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, 200433, China
| | - Hui Jiang
- Shanghai Institute of Pancreatic Diseases, Shanghai, 200433, China; Department of Pathology, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Jihang Yuan
- Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China
| | - Xiaoru Sun
- Department of Gastroenterology, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, 200433, China
| | - Jingyi Zhu
- Department of Gastroenterology, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, 200433, China
| | - Maoyun Yan
- Department of Gastroenterology, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, 200433, China
| | - Luowei Wang
- Department of Gastroenterology, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, 200433, China
| | - Zhaoshen Li
- Department of Gastroenterology, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, 200433, China
| | - Zhuan Liao
- Department of Gastroenterology, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, 200433, China.
| | - Chang Sun
- Department of Gastroenterology, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China; Shanghai Institute of Pancreatic Diseases, Shanghai, 200433, China.
| |
Collapse
|
2
|
Liu B, Jiang Q, Chen R, Zhang H, Xia Q, Shao C, Liu X, Wang M, Shi Y, Zhu J, Zhao R, Jiang H, Gao S, Li X, Zhou H, Yang C, Huang H. Tacrolimus alleviates pulmonary fibrosis progression through inhibiting the activation and interaction of ILC2 and monocytes. Int Immunopharmacol 2024; 132:111999. [PMID: 38581994 DOI: 10.1016/j.intimp.2024.111999] [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: 10/24/2023] [Revised: 03/27/2024] [Accepted: 04/01/2024] [Indexed: 04/08/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a heterogeneous group of lung diseases with different etiologies and characterized by progressive fibrosis. This disease usually causes pulmonary structural remodeling and decreased pulmonary function. The median survival of IPF patients is 2-5 years. Predominantly accumulation of type II innate immune cells accelerates fibrosis progression by secreting multiple pro-fibrotic cytokines. Group 2 innate lymphoid cells (ILC2) and monocytes/macrophages play key roles in innate immunity and aggravate the formation of pro-fibrotic environment. As a potent immunosuppressant, tacrolimus has shown efficacy in alleviating the progression of pulmonary fibrosis. In this study, we found that tacrolimus is capable of suppressing ILC2 activation, monocyte differentiation and the interaction of these two cells. This effect further reduced activation of monocyte-derived macrophages (Mo-M), thus resulting in a decline of myofibroblast activation and collagen deposition. The combination of tacrolimus and nintedanib was more effective than either drug alone. This study will reveal the specific process of tacrolimus alleviating pulmonary fibrosis by regulating type II immunity, and explore the potential feasibility of tacrolimus combined with nintedanib in the treatment of pulmonary fibrosis. This project will provide new ideas for clinical optimization of anti-pulmonary fibrosis drug strategies.
Collapse
Affiliation(s)
- Bowen Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China
| | - Qiuyan Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China
| | - Ruxuan Chen
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Huizhe Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, 300070 Tianjin, China
| | - Qin Xia
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, 300070 Tianjin, China
| | - Chi Shao
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Xiangning Liu
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Mengqi Wang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Yujie Shi
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Jingyan Zhu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, 300070 Tianjin, China
| | - Ruixi Zhao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China
| | - Haixia Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, 300070 Tianjin, China
| | - Shaoyan Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China
| | - Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, 300070 Tianjin, China
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, 300000 Tianjin, China; High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, 300070 Tianjin, China
| | - Hui Huang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
3
|
Dong Y, He L, Zhu Z, Yang F, Ma Q, Zhang Y, Zhang X, Liu X. The mechanism of gut-lung axis in pulmonary fibrosis. Front Cell Infect Microbiol 2024; 14:1258246. [PMID: 38362497 PMCID: PMC10867257 DOI: 10.3389/fcimb.2024.1258246] [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: 07/13/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024] Open
Abstract
Pulmonary fibrosis (PF) is a terminal change of a lung disease that is marked by damage to alveolar epithelial cells, abnormal proliferative transformation of fibroblasts, excessive deposition of extracellular matrix (ECM), and concomitant inflammatory damage. Its characteristics include short median survival, high mortality rate, and limited treatment effectiveness. More in-depth studies on the mechanisms of PF are needed to provide better treatment options. The idea of the gut-lung axis has emerged as a result of comprehensive investigations into the microbiome, metabolome, and immune system. This theory is based on the material basis of microorganisms and their metabolites, while the gut-lung circulatory system and the shared mucosal immune system act as the connectors that facilitate the interplay between the gastrointestinal and respiratory systems. The emergence of a new view of the gut-lung axis is complementary and cross-cutting to the study of the mechanisms involved in PF and provides new ideas for its treatment. This article reviews the mechanisms involved in PF, the gut-lung axis theory, and the correlation between the two. Exploring the gut-lung axis mechanism and treatments related to PF from the perspectives of microorganisms, microbial metabolites, and the immune system. The study of the gut-lung axis and PF is still in its early stages. This review systematically summarizes the mechanisms of PF related to the gut-lung axis, providing ideas for subsequent research and treatment of related mechanisms.
Collapse
Affiliation(s)
- Yawei Dong
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Lanlan He
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Zhongbo Zhu
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Fan Yang
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Quan Ma
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Respiratory Medicine, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yanmei Zhang
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xuhui Zhang
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Respiratory Medicine, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Xiping Liu
- Key Laboratory of Gansu Provincial Prescription Mining and Innovative Translational Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
- Gansu Provincial Traditional Chinese Medicine New Product Creation Engineering Laboratory, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| |
Collapse
|
4
|
Senoo S, Higo H, Taniguchi A, Kiura K, Maeda Y, Miyahara N. Pulmonary fibrosis and type-17 immunity. Respir Investig 2023; 61:553-562. [PMID: 37356133 DOI: 10.1016/j.resinv.2023.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/17/2023] [Accepted: 05/12/2023] [Indexed: 06/27/2023]
Abstract
Fibrosis of the lung can occur in idiopathic pulmonary fibrosis, collagen vascular diseases, and hypersensitivity pneumonitis, among other diseases. Transforming growth factor (TGF)-β, vascular epithelial growth factor, fibroblast growth factor, and platelet-derived growth factor contribute to the pathophysiology of fibrosis. TGF-β and other cytokines, including interleukin (IL)-1β, IL-6, and IL-23, activate type-17 immunity, which is involved in pulmonary fibrosis. The components of type-17 immunity include type-17 helper T cells, γδT cells, IL-17A-producing CD8-positive T cells, invariant NKT cells, and group 3 innate lymphoid cells. IL-17A, the main cytokine of type-17 immunity, is able to induce the epithelial-mesenchymal transition in epithelial cells via a production of TGF-β, directly stimulate fibroblasts and fibrocytes, and inhibit autophagy, which otherwise protects against pulmonary fibrosis. IL-23 induces type-17 immunity and plays an important role in the acute exacerbation of pulmonary fibrosis. Clinical studies have also linked type-17 immunity to the pathogenesis of pulmonary fibrosis. Consequently, targeting type-17 immunity may serve as a new therapeutic strategy to prevent the development or exacerbation of pulmonary fibrosis.
Collapse
Affiliation(s)
- Satoru Senoo
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Academic Field of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Hisao Higo
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan
| | - Akihiko Taniguchi
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Academic Field of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Katsuyuki Kiura
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan
| | - Yoshinobu Maeda
- Department of Hematology, Oncology, Allergy and Respiratory Medicine, Okayama University Academic Field of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Nobuaki Miyahara
- Department of Allergy and Respiratory Medicine, Okayama University Hospital, Okayama, Japan; Department of Medical Technology, Okayama University Academic Field of Health Sciences, Okayama, Japan.
| |
Collapse
|
5
|
Nedeva D, Kowal K, Mihaicuta S, Guidos Fogelbach G, Steiropoulos P, Jose Chong-Neto H, Tiotiu A. Epithelial alarmins: a new target to treat chronic respiratory diseases. Expert Rev Respir Med 2023; 17:773-786. [PMID: 37746733 DOI: 10.1080/17476348.2023.2262920] [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: 06/25/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
INTRODUCTION In response to injury, epithelial cells release alarmins including thymic stromal lymphopoietin (TSLP), high mobility group-box-1 (HMGB1), interleukin (IL)-33 and -25 that can initiate innate immune responses. These alarmins are recognized as activators of T2-immune responses characteristic for asthma, but recent evidence highlighted their role in non-T2 inflammation, airway remodeling, and pulmonary fibrosis making them an attractive therapeutic target for chronic respiratory diseases (CRD). AREAS COVERED In this review, firstly we discuss the role of TSLP, IL-33, IL-25, and HMGB1 in the pathogenesis of asthma, COPD, idiopathic pulmonary fibrosis, and cystic fibrosis according to the published data. In the second part, we summarize the current evidence concerning the efficacy of the antialarmin therapies in CRD. Recent clinical trials showed that anti-TSLP and IL-33/R antibodies can improve severe asthma outcomes. Blocking the IL-33-mediated pathway decreased the exacerbation rate in COPD patients with more important benefit for former-smokers. EXPERT OPINION Despite progress in the understanding of the alarmins' role in the pathogenesis of CRD, all their mechanisms of action are not yet identified. Blocking IL-33 and TSLP pathways offers an interesting option to treat severe asthma and COPD, but future investigations are needed to establish their place in the treatment strategies.
Collapse
Affiliation(s)
- Denislava Nedeva
- Clinic of Asthma and Allergology, UMBAL Alexandrovska, Medical University Sofia, Sofia, Bulgaria
| | - Krzysztof Kowal
- Department of Experimental Allergology and Immunology, Department of Internal Medicine and Allergology, Medical University of Bialystok, Bialystok, Poland
| | - Stefan Mihaicuta
- Center for Research and Innovation in Precision Medicine and Pharmacy, University of Medicine and Pharmacy, Timisoara, Romania
- Department of Pulmonology, "Victor Babes" University of Medicine and Pharmacy, Timisoara, Romania
| | | | - Paschalis Steiropoulos
- Department of Respiratory Medicine, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| | - Herberto Jose Chong-Neto
- Division of Allergy and Immunology, Complexo Hospital de Clinicas Federal University of Paraná, Curitiba, PR, Brazil
| | - Angelica Tiotiu
- Department of Pulmonology, University Hospital of Nancy, Vandœuvre-lès-Nancy, France
- Development, Adaptation and Disadvantage. Cardiorespiratory regulations and motor control (EA 3450 DevAH), University of Lorraine, Vandœuvre-lès-Nancy, France
| |
Collapse
|
6
|
Nakashima T, Omori K, Namba M, Yamaguchi K, Sakamoto S, Horimasu Y, Masuda T, Miyamoto S, Iwamoto H, Fujitaka K, Hamada H, Hattori N. Serum and bronchoalveolar lavage fluid levels of soluble B7H3 in patients with interstitial lung diseases. Respir Med 2023; 212:107224. [PMID: 37003499 DOI: 10.1016/j.rmed.2023.107224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 11/12/2022] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
BACKGROUND The B7 family member B7H3/CD276 was recently reported to be involved in the pathophysiology of idiopathic pulmonary fibrosis (IPF). However, the association of B7H3 with prognosis in patients with fibrosing interstitial lung diseases (ILDs), including IPF, remains unclear. This study was investigated to determine the potential of soluble B7H3 (sB7H3) as a biomarker to predict prognosis in patients with fibrosing ILDs. METHODS Patients with ILDs from various categories who underwent bronchoalveolar lavage (BAL) were included in the study. The relationship between sB7H3 levels in serum or BAL fluid (BALF) and clinical variables at the time of ILD diagnosis was studied retrospectively. All patients who met the fibrosing ILD criteria were followed for 5 years. RESULTS We found that coexisting malignancy affected the serum, but not the BALF, sB7H3 levels. There was no significant correlation between serum and BALF levels of sB7H3 in 49 ILD patients without malignancy (11 with sarcoidosis, 5 with drug-induced ILD, 22 with IPF, and 11 with ILD associated with systemic sclerosis). We also found that the BALF levels, but not serum levels, of sB7H3 at the time of ILD diagnosis had independent prognostic potential on 5-year survival in patients with fibrosing ILDs. Of note, patients with a higher level of BALF sB7H3 at diagnosis (≥0.100 ng/mL) showed significantly shorter survival than those with lower levels. CONCLUSIONS This study suggests that BALF sB7H3 could be a novel prognostic biomarker in a broad range of fibrosing ILD patients.
Collapse
Affiliation(s)
- Taku Nakashima
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan.
| | - Keitaro Omori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masashi Namba
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kakuhiro Yamaguchi
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shinjiro Sakamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yasushi Horimasu
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takeshi Masuda
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shintaro Miyamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroshi Iwamoto
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kazunori Fujitaka
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hironobu Hamada
- Department of Physical Analysis and Therapeutic Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
7
|
Stephenson KE, Porte J, Kelly A, Wallace WA, Huntington CE, Overed-Sayer CL, Cohen ES, Jenkins RG, John AE. The IL-33:ST2 axis is unlikely to play a central fibrogenic role in idiopathic pulmonary fibrosis. Respir Res 2023; 24:89. [PMID: 36949463 PMCID: PMC10035257 DOI: 10.1186/s12931-023-02334-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 01/18/2023] [Indexed: 03/24/2023] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a devastating interstitial lung disease (ILD) with limited treatment options. Interleukin-33 (IL-33) is proposed to play a role in the development of IPF however the exclusive use of prophylactic dosing regimens means that the therapeutic benefit of targeting this cytokine in IPF is unclear. METHODS IL-33 expression was assessed in ILD lung sections and human lung fibroblasts (HLFs) by immunohistochemistry and gene/protein expression and responses of HLFs to IL-33 stimulation measured by qPCR. In vivo, the fibrotic potential of IL-33:ST2 signalling was assessed using a murine model of bleomycin (BLM)-induced pulmonary fibrosis and therapeutic dosing with an ST2-Fc fusion protein. Lung and bronchoalveolar lavage fluid were collected for measurement of inflammatory and fibrotic endpoints. Human precision-cut lung slices (PCLS) were stimulated with transforming growth factor-β (TGFβ) or IL-33 and fibrotic readouts assessed. RESULTS IL-33 was expressed by fibrotic fibroblasts in situ and was increased by TGFβ treatment in vitro. IL-33 treatment of HLFs did not induce IL6, CXCL8, ACTA2 and COL1A1 mRNA expression with these cells found to lack the IL-33 receptor ST2. Similarly, IL-33 stimulation had no effect on ACTA2, COL1A1, FN1 and fibronectin expression by PCLS. Despite having effects on inflammation suggestive of target engagement, therapeutic dosing with the ST2-Fc fusion protein failed to reduce BLM-induced fibrosis measured by hydroxyproline content or Ashcroft score. CONCLUSIONS Together these findings suggest the IL-33:ST2 axis does not play a central fibrogenic role in the lungs with therapeutic blockade of this pathway unlikely to surpass the current standard of care for IPF.
Collapse
Affiliation(s)
- Katherine E Stephenson
- Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, UK.
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| | - Joanne Porte
- Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, UK
| | - Aoife Kelly
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | | | - Catherine L Overed-Sayer
- Bioscience COPD/IPF, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - E Suzanne Cohen
- Bioscience Asthma and Skin Immunity, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - R Gisli Jenkins
- National Heart and Lung Institute, Imperial College London, London, UK
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, Imperial College London, London, UK
- Interstitial lung disease unit, Royal Brompton Hospital, London, UK
| | - Alison E John
- Division of Respiratory Medicine, School of Medicine, University of Nottingham, Nottingham, UK
- National Heart and Lung Institute, Imperial College London, London, UK
- Margaret Turner Warwick Centre for Fibrosing Lung Disease, Imperial College London, London, UK
| |
Collapse
|
8
|
Löfdahl A, Nybom A, Wigén J, Dellgren G, Brunnström H, Wenglén C, Westergren-Thorsson G. Pulmonary 5-HT 2B receptor expression in fibrotic interstitial lung diseases. Acta Histochem 2023; 125:152024. [PMID: 36958084 DOI: 10.1016/j.acthis.2023.152024] [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: 11/01/2022] [Revised: 02/17/2023] [Accepted: 03/10/2023] [Indexed: 03/25/2023]
Abstract
Pulmonary fibrosis is a severe condition in interstitial lung diseases (ILD) such as idiopathic pulmonary fibrosis (IPF) and systemic sclerosis-ILD, where the underlying mechanism is not well defined and with no curative treatments available. Serotonin (5-HT) signaling via the 5-HT2B receptor has been recognized as a promising preclinical target for fibrosis. Despite this, the involvement of the 5-HT2B receptor in fibrotic ILD is widely unexplored. This work highlights the spatial pulmonary distribution of the 5-HT2B receptor in patients with IPF and systemic sclerosis-ILD. We show that the 5-HT2B receptor is located in typical pathological structures e.g. honeycomb cysts and weakly in fibroblast foci. Together with immunohistochemistry and immunofluorescence stainings of patient derived distal lung tissues, we identified cell targets for 5-HT2B receptor interference in type II alveolar epithelial cells, endothelial cells and M2 macrophages. Our results emphasize the role of 5-HT2B receptor as a target in lung fibrosis, warranting further consideration in targeting fibrotic ILDs.
Collapse
Affiliation(s)
- Anna Löfdahl
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Annika Nybom
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Jenny Wigén
- Lung Biology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Göran Dellgren
- Department of Cardiothoracic Surgery and Transplant Institute, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Hans Brunnström
- Lund University, Laboratory Medicine Region Skåne, Department of Clinical Sciences Lund, Pathology, Lund, Sweden
| | | | | |
Collapse
|
9
|
Assessment of sST2 Behaviors to Evaluate Severity/Clinical Impact of Acute Pulmonary Embolism. Int J Mol Sci 2023; 24:ijms24054591. [PMID: 36902022 PMCID: PMC10002919 DOI: 10.3390/ijms24054591] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 03/03/2023] Open
Abstract
Pulmonary embolism (PE) is a potentially life-threatening disorder. Beyond its usefulness in the prognostic stratification of heart failure, sST2 can represent a biomarker with high utility in several acute conditions. Our study was aimed to investigate whether sST2 can be used as a clinical marker of severity and prognostic outcome in acute PE. We enrolled 72 patients with documented PE and 38 healthy subjects; we measured the plasma concentrations of sST2 to evaluate the prognostic and severity performance of different levels of sST2 according to its association with the pulmonary embolism severity index (PESI) score and several parameters of respiratory function. PE patients had significantly higher levels of sST2 compared with healthy subjects (87.74 ± 17.1 vs. 17.1 ± 0.4 ng/mL, p < 0.001); we found higher PESI scores and serum lactate values in the group of patients with sST2 > 35 ng/mL compared with patients with sST2 < 35 ng/mL (138.7 ± 14.9 vs. 103.7 ± 15.1 and 2.43 ± 0.69 vs. 1.025 ± 0.05 mmol/L, respectively; p < 0.05). Patients with sST2 > 35 ng/mL showed higher radiological severity of PE compared with patients with sST2 < 35 ng/mL. Moreover, sST2 was the strongest parameter with a discriminative capacity for the development of acute respiratory failure and a PESI score >106 with respect to C reactive protein (CRP), creatinine, d-dimer, and serum lactate. We clearly demonstrated that sST2 significantly increased in PE and that its elevation was associated with disease severity. Therefore, sST2 may be used as a clinical marker in the evaluation of PE severity. However, further studies with larger patient populations are required to confirm these findings.
Collapse
|
10
|
Luzina IG, Lockatell V, Courneya JP, Mei Z, Fishelevich R, Kopach P, Pickering EM, Kang PH, Krupnick AS, Todd NW, Vogel SN, Atamas SP. Full-length IL-33 augments pulmonary fibrosis in an ST2- and Th2-independent, non-transcriptomic fashion. Cell Immunol 2023; 383:104657. [PMID: 36603504 PMCID: PMC9909894 DOI: 10.1016/j.cellimm.2022.104657] [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: 06/09/2022] [Revised: 11/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Mature IL-33 (MIL33) acting through its receptor, ST2, is known to regulate fibrosis. The precursor, full-length IL-33 (FLIL33), may function differently from MIL33 and independently of ST2. Here we report that genetic deletion of either IL-33 or ST2 attenuates pulmonary fibrosis in the bleomycin model, as does Cre-induced IL-33 deficiency in response to either acute or chronic bleomycin challenge. However, adenovirus-mediated gene delivery of FLIL33, but not MIL33, to the lungs of either wild-type or ST2-deficient mice potentiates the profibrotic effect of bleomycin without inducing a Th2 phenotype. In cultured mouse lung cells, FLIL33 overexpression induces moderate and distinct transcriptomic changes compared with a robust response induced by MIL33, whereas ST2 deletion abrogates the effects of both IL-33 forms. Thus, FLIL33 may contribute to fibrosis in an ST2-independent, Th2-independent, non-transcriptomic fashion, suggesting that pharmacological targeting of both FLIL33 and MIL33 may prove efficacious in patients with pulmonary fibrosis.
Collapse
Affiliation(s)
- Irina G Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States; Research Service, Baltimore VA Medical Center, Baltimore, MD, United States.
| | - Virginia Lockatell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jean-Paul Courneya
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Zhongcheng Mei
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rita Fishelevich
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Pavel Kopach
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Edward M Pickering
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Phillip H Kang
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Alexander S Krupnick
- Research Service, Baltimore VA Medical Center, Baltimore, MD, United States; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Nevins W Todd
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States; Research Service, Baltimore VA Medical Center, Baltimore, MD, United States
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Sergei P Atamas
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States; Research Service, Baltimore VA Medical Center, Baltimore, MD, United States; Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
11
|
Seyran M, Melanie S, Philip S, Amiq G, Fabian B. Allies or enemies? The effect of regulatory T cells and related T lymphocytes on the profibrotic environment in bleomycin-injured lung mouse models. Clin Exp Med 2022:10.1007/s10238-022-00945-7. [PMID: 36403186 PMCID: PMC10390389 DOI: 10.1007/s10238-022-00945-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/07/2022] [Indexed: 11/21/2022]
Abstract
AbstractIdiopathic pulmonary fibrosis (IPF) is characterized by permanent scarring of lung tissue and declining lung function, and is an incurable disease with increase in prevalence over the past decade. The current consensus is that aberrant wound healing following repeated injuries to the pulmonary epithelium is the most probable cause of IPF, with various immune inflammatory pathways having been reported to impact disease pathogenesis. While the role of immune cells, specifically T lymphocytes and regulatory T cells (Treg), in IPF pathogenesis has been reported and discussed recently, the pathogenic or beneficial roles of these cells in inducing or preventing lung fibrosis is still debated. This lack of understanding could be due in part to the difficulty in obtaining diseased human lung tissue for research purposes. For this reason, many animal models have been developed over the years to attempt to mimic the main clinical hallmarks of IPF: among these, inducing lung injury in rodents with the anti-cancer agent bleomycin has now become the most commonly studied animal model of IPF. Pulmonary fibrosis is the major side effect when bleomycin is administered for cancer treatment in human patients, and a similar effect can be observed after intra-tracheal administration of bleomycin to rodents. Despite many pathophysiological pathways of lung fibrosis having been investigated in bleomycin-injured animal models, one central facet still remains controversial, namely the involvement of specific T lymphocyte subsets, and in particular Treg, in disease pathogenesis. This review aims to summarize the major findings and conclusions regarding the involvement of immune cells and their receptors in the pathogenesis of IPF, and to elaborate on important parallels between animal models and the human disease. A more detailed understanding of the role of Treg and other immune cell subsets in lung injury and fibrosis derived from animal models is a critical basis for translating this knowledge to the development of new immune-based therapies for the treatment of human IPF.
Collapse
|
12
|
Xu X, Dai H, Zhang J. The potential role of interleukin (IL)-25/IL-33/thymic stromal lymphopoietin (TSLP) on the pathogenesis of idiopathic pulmonary fibrosis. THE CLINICAL RESPIRATORY JOURNAL 2022; 16:696-707. [PMID: 36082495 PMCID: PMC9629992 DOI: 10.1111/crj.13541] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/16/2022] [Indexed: 06/15/2023]
Abstract
OBJECTIVES Interleukin (IL)-25, IL-33, and thymic stromal lymphopoietin (TSLP) are the important drivers for excessive type-2 immunity. It has been well elucidated that IL-25/IL-33/TSLP plays an important role in allergic airway inflammation and remodeling, whereas their roles in idiopathic pulmonary fibrosis (IPF) still remained largely unclear. Herein, the aim of the review is to discuss the potential role and mechanism of IL-25/IL-33/TSLP on IPF by literature analysis and summary. DATA SOURCE We have done a literature search using the following terms: ("idiopathic pulmonary fibrosis" OR "IPF" OR "lung fibrosis") and (TSLP or "thymic stromal lymphopoietin" or IL-25 OR IL-17E OR IL-33) from the database of PubMed published in English up to July 2018. STUDY SELECTION We have totally found 58 articles by using the retrieval terms mentioned above. By careful title and abstract reading, 10 original research articles of high quality were enrolled for the full text reading and analysis. Two additional relevant studies were also included during the course of literature readings. RESULTS IL-25/IL-33/TSLP and their corresponding receptors, that is, IL-17BR/ST2L/TSLPR, are shown to be up-regulated both in IPF patients and bleomycin (BLM)-induced lung fibrosis mice model. IL-25 may promote lung fibrosis by activating IL-17BR+fibroblast and IL-17BR+ILC2 (type 2 innate lymphoid cell). Full length (fl)-IL-33, as a transcription factor mainly in the cell nucleus, mediated non-atopic lung inflammation and fibrosis by modulating expressions of several pro-fibrotic mediators, including transforming growth factor (TGF)-b1. By contrast, mature (m)-IL-33 potentiates lung fibrosis by recruiting ST2L+M2 macrophages and ST2L+ILC2 to enlarge type 2 immunity. TSLP was shown to directly promote CCL2 expression in primary human lung fibroblasts (pHLFs). CONCLUSION IL-25/IL-33/TSLP contributes to non-allergic lung fibrosis by mediating persistent abnormal epithelial-mesenchymal crosstalk. IL-25/IL-33/TSLP may serve the promising novel target for the treatment of IPF.
Collapse
Affiliation(s)
- Xuefeng Xu
- Department of Surgical Intensive Care Unit, Beijing An Zhen HospitalCapital Medical UniversityBeijingChina
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine, Center for Respiratory Diseases, China‐Japan Friendship HospitalNational Clinical Research Center for Respiratory DiseasesBeijingChina
| | - Jinglan Zhang
- Department of Surgical Intensive Care Unit, Beijing An Zhen HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
13
|
Guo Y, Mei Z, Li D, Banerjee A, Khalil MA, Burke A, Ritter J, Lau C, Kreisel D, Gelman AE, Jacobsen E, Luzina IG, Atamas SP, Krupnick AS. Ischemia reperfusion injury facilitates lung allograft acceptance through IL-33-mediated activation of donor-derived IL-5 producing group 2 innate lymphoid cells. Am J Transplant 2022; 22:1963-1975. [PMID: 35510760 PMCID: PMC9357103 DOI: 10.1111/ajt.17084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/11/2022] [Accepted: 04/30/2022] [Indexed: 01/25/2023]
Abstract
Pathways regulating lung alloimmune responses differ from most other solid organs and remain poorly explored. Based on our recent work identifying the unique role of eosinophils in downregulating lung alloimmunity, we sought to define pathways contributing to eosinophil migration and homeostasis. Using a murine lung transplant model, we have uncovered that immunosuppression increases eosinophil infiltration into the allograft in an IL-5-dependent manner. IL-5 production depends on immunosuppression-mediated preservation of donor-derived group 2 innate lymphoid cells (ILC2). We further describe that ischemia reperfusion injury upregulates the expression of IL-33, which functions as the dominant and nonredundant mediator of IL-5 production by graft-resident ILC2. Our work thus identifies unique cellular mechanisms that contribute to lung allograft acceptance. Notably, ischemia reperfusion injury, widely considered to be solely deleterious to allograft survival, can also downregulate alloimmune responses by initiating unique pathways that promote IL-33/IL-5/eosinophil-mediated tolerance.
Collapse
Affiliation(s)
- Yizhan Guo
- Department of Surgery, University of Maryland, Baltimore Maryland
| | - Zhongcheng Mei
- Department of Surgery, University of Maryland, Baltimore Maryland
| | - Dongge Li
- Department of Surgery, University of Maryland, Baltimore Maryland
| | - Anirban Banerjee
- Department of Surgery, University of Maryland, Baltimore Maryland
| | - May A. Khalil
- Department of Surgery, University of Maryland, Baltimore Maryland
| | - Allen Burke
- Department of Pathology, University of Maryland, Baltimore Maryland
| | - Jon Ritter
- Department of Pathology & Immunology, Washington University in St. Louis, St. Louis Missouri
| | - Christine Lau
- Department of Surgery, University of Maryland, Baltimore Maryland
| | - Daniel Kreisel
- Department of Pathology & Immunology, Washington University in St. Louis, St. Louis Missouri
- Department of Surgery, Washington University in St. Louis, St. Louis Missouri
| | - Andrew E. Gelman
- Department of Pathology & Immunology, Washington University in St. Louis, St. Louis Missouri
- Department of Surgery, Washington University in St. Louis, St. Louis Missouri
| | - Elizabeth Jacobsen
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic, Scottsdale, Arizona
| | - Irina G. Luzina
- Department of Medicine, University of Maryland, Baltimore Maryland
| | - Sergei P. Atamas
- Department of Surgery, University of Maryland, Baltimore Maryland
| | | |
Collapse
|
14
|
Nagashima R, Ishikawa H, Kuno Y, Kohda C, Iyoda M. IL-33 attenuates renal fibrosis via group2 innate lymphoid cells. Cytokine 2022; 157:155963. [PMID: 35868116 DOI: 10.1016/j.cyto.2022.155963] [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: 03/28/2022] [Revised: 06/20/2022] [Accepted: 07/04/2022] [Indexed: 11/03/2022]
Abstract
Renal fibrosis is a common pathway in the progression of various kidney diseases and injuries. Unilateral ureteral obstruction (UUO) induces renal fibrosis, and immune responses profoundly affect its pathogenesis. Group2 innate lymphoid cells (ILC2s) are strongly activated by interleukin (IL) -33, which is a member of IL-1 family and recognize as alarmin. ILC2s quickly produce large amounts of type 2 cytokines including IL-5 and IL-13, which are involved in inflammation, tissue homeostasis, and wound healing. However, the relationship between renal fibrosis and ILC2s has been unclear. In the present study, we investigated the roles of the ILC2/L-33 axis in renal fibrosis using a UUO model. We found that kidney ILC2s decreased in UUO-affected kidneys compared with their counterpart kidneys despite IL-33 upregulation. There was no effect of reactive oxygen species or TGF-β from reduced ILC2 caused by UUO. Pretreatment with IL-33 before UUO induced ILC2s and Tregs in kidneys and alleviated renal fibrosis. Furthermore, this protective effect was maintained even when CD4+T cells was depleted. These findings demonstrated that ILC2s play a predominant role in the suppressive function of renal fibrosis mediated by pretreatment with IL-33. In contrast, post-treatment with IL-33 after UUO increased ILC2s in kidneys but had no therapeutic effect on renal fibrosis. Our findings suggest that ILC2s have potential roles in the prevention of renal fibrosis and can serve as a therapeutic and diagnostic target.
Collapse
Affiliation(s)
- Ryuichi Nagashima
- Department of Microbiology and Immunology, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8555, Japan.
| | - Hiroki Ishikawa
- Department of Microbiology and Immunology, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Yoshihiro Kuno
- Department of Microbiology and Immunology, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8555, Japan; Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Chikara Kohda
- Department of Microbiology and Immunology, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8555, Japan
| | - Masayuki Iyoda
- Department of Microbiology and Immunology, Showa University School of Medicine, Shinagawa-ku, Tokyo 142-8555, Japan; Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
15
|
Zhang M, Duffen JL, Nocka KH, Kasaian MT. IL-13 Controls IL-33 Activity through Modulation of ST2. THE JOURNAL OF IMMUNOLOGY 2021; 207:3070-3080. [PMID: 34789557 DOI: 10.4049/jimmunol.2100655] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/13/2021] [Indexed: 12/12/2022]
Abstract
IL-33 is a multifunctional cytokine that mediates local inflammation upon tissue damage. IL-33 is known to act on multiple cell types including group 2 innate lymphoid cells (ILC2s), Th2 cells, and mast cells to drive production of Th2 cytokines including IL-5 and IL-13. IL-33 signaling activity through transmembrane ST2L can be inhibited by soluble ST2 (sST2), which acts as a decoy receptor. Previous findings suggested that modulation of IL-13 levels in mice lacking decoy IL-13Rα2, or mice lacking IL-13, impacted responsiveness to IL-33. In this study, we used Il13 -/- mice to investigate whether IL-13 regulates IL-33 activity by modulating the transmembrane and soluble forms of ST2. In Il13 -/- mice, the effects of IL-33 administration were exacerbated relative to wild type (WT). Il13 -/- mice administered IL-33 i.p. had heightened splenomegaly, more immune cells in the peritoneum including an expanded ST2L+ ILC2 population, increased eosinophilia in the spleen and peritoneum, and reduced sST2 in the circulation and peritoneum. In the spleen, lung, and liver of mice given IL-33, gene expression of both isoforms of ST2 was increased in Il13 -/- mice relative to WT. We confirmed fibroblasts to be an IL-13-responsive cell type that can regulate IL-33 activity through production of sST2. This study elucidates the important regulatory activity that IL-13 exerts on IL-33 through induction of IL-33 decoy receptor sST2 and through modulation of ST2L+ ILC2s.
Collapse
Affiliation(s)
- Melvin Zhang
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| | - Jennifer L Duffen
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| | - Karl H Nocka
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| | - Marion T Kasaian
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA
| |
Collapse
|
16
|
Burgoyne RA, Fisher AJ, Borthwick LA. The Role of Epithelial Damage in the Pulmonary Immune Response. Cells 2021; 10:cells10102763. [PMID: 34685744 PMCID: PMC8534416 DOI: 10.3390/cells10102763] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
Pulmonary epithelial cells are widely considered to be the first line of defence in the lung and are responsible for coordinating the innate immune response to injury and subsequent repair. Consequently, epithelial cells communicate with multiple cell types including immune cells and fibroblasts to promote acute inflammation and normal wound healing in response to damage. However, aberrant epithelial cell death and damage are hallmarks of pulmonary disease, with necrotic cell death and cellular senescence contributing to disease pathogenesis in numerous respiratory diseases such as idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and coronavirus disease (COVID)-19. In this review, we summarise the literature that demonstrates that epithelial damage plays a pivotal role in the dysregulation of the immune response leading to tissue destruction and abnormal remodelling in several chronic diseases. Specifically, we highlight the role of epithelial-derived damage-associated molecular patterns (DAMPs) and senescence in shaping the immune response and assess their contribution to inflammatory and fibrotic signalling pathways in the lung.
Collapse
Affiliation(s)
- Rachel Ann Burgoyne
- Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
| | - Andrew John Fisher
- Regenerative Medicine, Stem Cells and Transplantation Theme, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
- Institute of Transplantation, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| | - Lee Anthony Borthwick
- Fibrosis Research Group, Biosciences Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK;
- Fibrofind, Medical School, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- Correspondence: ; Tel.: +44-191-208-3112
| |
Collapse
|
17
|
Chen L, Alam A, Pac-Soo A, Chen Q, Shang Y, Zhao H, Yao S, Ma D. Pretreatment with valproic acid alleviates pulmonary fibrosis through epithelial-mesenchymal transition inhibition in vitro and in vivo. J Transl Med 2021; 101:1166-1175. [PMID: 34168289 PMCID: PMC8367813 DOI: 10.1038/s41374-021-00617-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) plays a crucial role in the development of pulmonary fibrosis. This study aims to investigate the effects of valproic acid (VPA) on EMT in vitro and in vivo. In vitro, EMT was induced by the administration of transforming growth factor-β1 (TGF-β1) in a human alveolar epithelial cell line (A549). The dose effects of VPA (0.1-3 mM) on EMT were subsequently evaluated at different timepoints. VPA (1 mM) was applied prior to the administration of TGF-β1 and the expression of E-cadherin, vimentin, p-Smad2/3 and p-Akt was assessed. In addition, the effects of a TGF-β type I receptor inhibitor (A8301) and PI3K-Akt inhibitor (LY294002) on EMT were evaluated. In vivo, the effects of VPA on bleomycin-induced lung fibrosis were evaluated by assessing variables such as survival rate, body weight and histopathological changes, whilst the expression of E-cadherin and vimentin in lung tissue was also evaluated. A8301 and LY294002 were used to ascertain the cellular signaling pathways involved in this model. The administration of VPA prior to TGF-β1 in A549 cells prevented EMT in both a time- and concentration-dependent manner. Pretreatment with VPA downregulated the expression of both p-Smad2/3 and p-Akt. A8301 administration increased the expression of E-cadherin and reduced the expression of vimentin. LY294002 inhibited Akt phosphorylation induced by TGF-β1 but failed to prevent EMT. Pretreatment with VPA both increased the survival rate and prevented the loss of body weight in mice with pulmonary fibrosis. Interestingly, both VPA and A8301 prevented EMT and facilitated an improvement in lung structure. Overall, pretreatment with VPA attenuated the development of pulmonary fibrosis by inhibiting EMT in mice, which was associated with Smad2/3 deactivation but without Akt cellular signal involvement.
Collapse
Affiliation(s)
- Lin Chen
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Anesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Azeem Alam
- Anesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Aurelie Pac-Soo
- Anesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - Qian Chen
- Anesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| | - You Shang
- Department of Critical Care Medicine, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hailin Zhao
- Anesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK.
| | - Shanglong Yao
- Department of Anesthesiology, Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Daqing Ma
- Anesthetics, Pain Medicine and Intensive Care, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Chelsea & Westminster Hospital, London, UK
| |
Collapse
|
18
|
Nagashima R, Iyoda M. The Roles of Kidney-Resident ILC2 in Renal Inflammation and Fibrosis. Front Immunol 2021; 12:688647. [PMID: 34381446 PMCID: PMC8350317 DOI: 10.3389/fimmu.2021.688647] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/13/2021] [Indexed: 12/24/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a recently discovered lymphocyte population with high cytokine productive capacity. Type-2 ILCs (ILC2s) are the most studied, and they exert a rapid type-2 immune response to eliminate helminth infections. Massive and sustainable ILC2 activation induces allergic tissue inflammation, so it is important to maintain correct ILC2 activity for immune homeostasis. The ILC2-activating cytokine IL-33 is released from epithelial cells upon tissue damage, and it is upregulated in various kidney disease mouse models and in kidney disease patients. Various kidney diseases eventually lead to renal fibrosis, which is a common pathway leading to end-stage renal disease and is a chronic kidney disease symptom. The progression of renal fibrosis is affected by the innate immune system, including renal-resident ILC2s; however, the roles of ILC2s in renal fibrosis are not well understood. In this review, we summarize renal ILC2 function and characterization in various kidney diseases and highlight the known and potential contributions of ILC2s to kidney fibrosis.
Collapse
Affiliation(s)
- Ryuichi Nagashima
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan
| | - Masayuki Iyoda
- Department of Microbiology and Immunology, Showa University School of Medicine, Tokyo, Japan
- Division of Nephrology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
19
|
Perkins TN, Oury TD. The perplexing role of RAGE in pulmonary fibrosis: causality or casualty? Ther Adv Respir Dis 2021; 15:17534666211016071. [PMID: 34275342 PMCID: PMC8293846 DOI: 10.1177/17534666211016071] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease in which most patients die within 3 years of diagnosis. With an unknown etiology, IPF results in progressive fibrosis of the lung parenchyma, diminishing normal lung function, which results in respiratory failure, and eventually, death. While few therapies are available to reduce disease progression, patients continue to advance toward respiratory failure, leaving lung transplantation the only viable option for survival. As incidence and mortality rates steadily increase, the need for novel therapeutics is imperative. The receptor for advanced glycation endproducts (RAGE) is most highly expressed in the lungs and plays a significant role in a number of chronic lung diseases. RAGE has long been linked to IPF; however, confounding data from both human and experimental studies have left an incomplete and perplexing story. This review examines the present understanding of the role of RAGE in human and experimental models of IPF, drawing parallels to recent advances in RAGE biology. Moreover, this review discusses the role of RAGE in lung injury response, type 2 immunity, and cellular senescence, and how such mechanisms may relate to RAGE as both a biomarker of disease progression and potential therapeutic target in IPF.The reviews of this paper are available via the supplemental material section.
Collapse
Affiliation(s)
- Timothy N Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, 3550 Terrace Street, S-784 Scaife Hall, Pittsburgh, PA 15261, USA
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
20
|
Planté-Bordeneuve T, Pilette C, Froidure A. The Epithelial-Immune Crosstalk in Pulmonary Fibrosis. Front Immunol 2021; 12:631235. [PMID: 34093523 PMCID: PMC8170303 DOI: 10.3389/fimmu.2021.631235] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 05/04/2021] [Indexed: 12/12/2022] Open
Abstract
Interactions between the lung epithelium and the immune system involve a tight regulation to prevent inappropriate reactions and have been connected to several pulmonary diseases. Although the distal lung epithelium and local immunity have been implicated in the pathogenesis and disease course of idiopathic pulmonary fibrosis (IPF), consequences of their abnormal interplay remain less well known. Recent data suggests a two-way process, as illustrated by the influence of epithelial-derived periplakin on the immune landscape or the effect of macrophage-derived IL-17B on epithelial cells. Additionally, damage associated molecular patterns (DAMPs), released by damaged or dying (epithelial) cells, are augmented in IPF. Next to “sterile inflammation”, pathogen-associated molecular patterns (PAMPs) are increased in IPF and have been linked with lung fibrosis, while outer membrane vesicles from bacteria are able to influence epithelial-macrophage crosstalk. Finally, the advent of high-throughput technologies such as microbiome-sequencing has allowed for the identification of a disease-specific microbial environment. In this review, we propose to discuss how the interplays between the altered distal airway and alveolar epithelium, the lung microbiome and immune cells may shape a pro-fibrotic environment. More specifically, it will highlight DAMPs-PAMPs pathways and the specificities of the IPF lung microbiome while discussing recent elements suggesting abnormal mucosal immunity in pulmonary fibrosis.
Collapse
Affiliation(s)
- Thomas Planté-Bordeneuve
- Pôle de pneumologie, O.R.L. et dermatologie, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Bruxelles, Belgium
| | - Charles Pilette
- Pôle de pneumologie, O.R.L. et dermatologie, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Bruxelles, Belgium.,Service de pneumologie, Cliniques universitaires Saint-Luc, Bruxelles, Belgium
| | - Antoine Froidure
- Pôle de pneumologie, O.R.L. et dermatologie, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Bruxelles, Belgium.,Service de pneumologie, Cliniques universitaires Saint-Luc, Bruxelles, Belgium
| |
Collapse
|
21
|
Mathä L, Martinez-Gonzalez I, Steer CA, Takei F. The Fate of Activated Group 2 Innate Lymphoid Cells. Front Immunol 2021; 12:671966. [PMID: 33968080 PMCID: PMC8100346 DOI: 10.3389/fimmu.2021.671966] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/07/2021] [Indexed: 12/20/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) reside in both mucosal and non-mucosal tissues and play critical roles in the first line of defense against parasites and irritants such as allergens. Upon activation by cytokines released from epithelial and stromal cells during tissue damage or stimulation, ILC2s produce copious amounts of IL-5 and IL-13, leading to type 2 inflammation. Over the past 10 years, ILC2 involvement in a variety of human diseases has been unveiled. However, questions remain as to the fate of ILC2s after activation and how that might impact their role in chronic inflammatory diseases such as asthma and fibrosis. Here, we review studies that have revealed novel properties of post-activation ILC2s including the generation of immunological memory, exhausted-like phenotype, transdifferentiation and activation-induced migration.
Collapse
Affiliation(s)
- Laura Mathä
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BC, Canada
| | | | - Catherine A Steer
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BC, Canada
| | - Fumio Takei
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
22
|
Mi LL, Zhu Y, Lu HY. A crosstalk between type 2 innate lymphoid cells and alternative macrophages in lung development and lung diseases (Review). Mol Med Rep 2021; 23:403. [PMID: 33786611 PMCID: PMC8025469 DOI: 10.3892/mmr.2021.12042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/08/2021] [Indexed: 12/14/2022] Open
Abstract
Type 2 innate lymphoid cells (ILC2s) are important innate immune cells that are involved in type 2 inflammation, in both mice and humans. ILC2s are stimulated by factors, including interleukin (IL)-33 and IL-25, and activated ILC2s secrete several cytokines that mediate type 2 immunity by inducing profound changes in physiology, including activation of alternative (M2) macrophages. M2 macrophages possess immune modulatory, phagocytic, tissue repair and remodeling properties, and can regulate ILC2s under infection. The present review summarizes the role of ILC2s as innate cells and M2 macrophages as anti-inflammatory cells, and discusses current literature on their important biological significance. The present review also highlights how the crosstalk between ILC2s and M2 macrophages contributes to lung development, induces pulmonary parasitic expulsion, exacerbates pulmonary viral and fungal infections and allergic airway diseases, and promotes the development of lung diseases, such as pulmonary fibrosis, chronic obstructive pulmonary disease and carcinoma of the lungs.
Collapse
Affiliation(s)
- Lan-Lan Mi
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Yue Zhu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Hong-Yan Lu
- Department of Pediatrics, The Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| |
Collapse
|
23
|
Full-length IL-33 regulates Smad3 phosphorylation and gene transcription in a distinctive AP2-dependent manner. Cell Immunol 2020; 357:104203. [DOI: 10.1016/j.cellimm.2020.104203] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/30/2020] [Accepted: 08/26/2020] [Indexed: 12/15/2022]
|
24
|
Tissue-Resident Type 2 Innate Lymphoid Cells Arrest Alveolarization in Bronchopulmonary Dysplasia. J Immunol Res 2020; 2020:8050186. [PMID: 33178840 PMCID: PMC7648679 DOI: 10.1155/2020/8050186] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/09/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a severe complication of the respiratory system associated with preterm birth. Type 2 innate lymphoid cells (ILC2s) play a major role in tissue homeostasis, inflammation, and wound healing. However, the role in BPD remains unclear. The present study showed that ILC2s, interleukin-4 (IL-4), IL-13, and anti-inflammatory (M2) macrophages increased significantly in BPD mice as compared to the control mice. Administration with recombinant mouse IL-33 amplified the above phenomena and aggravated the alveolar structural disorder and functional injury in mice subjected to BPD, and the opposite was true with anti-ST2 antibody. In addition, the depletion of ILC2s in BPD mice with anti-CD90.2 antibody substantially abolished the destructive effect on BPD. In the treatment of BPD with dexamethasone, the number of ILC2s and M2 macrophages and levels of IL-4 and IL-13 decreased with remission as compared to the control group. This study identified a major destructive role of the ILC2s in BPD that could be attenuated as a therapeutic strategy.
Collapse
|
25
|
Messing M, Jan-Abu SC, McNagny K. Group 2 Innate Lymphoid Cells: Central Players in a Recurring Theme of Repair and Regeneration. Int J Mol Sci 2020; 21:E1350. [PMID: 32079296 PMCID: PMC7072936 DOI: 10.3390/ijms21041350] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/13/2020] [Accepted: 02/13/2020] [Indexed: 12/25/2022] Open
Abstract
Innate lymphoid cells (ILCs) are recently discovered innate counterparts to the well-established T helper cell subsets and are most abundant at barrier surfaces, where they participate in tissue homeostasis and inflammatory responses against invading pathogens. Group 2 innate lymphoid cells (ILC2s) share cytokine and transcription factor expression profiles with type-2 helper T cells and are primarily associated with immune responses against allergens and helminth infections. Emerging data, however, suggests that ILC2s are also key regulators in other inflammatory settings; both in a beneficial context, such as the establishment of neonatal immunity, tissue repair, and homeostasis, and in the context of pathological tissue damage and disease, such as fibrosis development. This review focuses on the interactions of ILC2s with stromal cells, eosinophils, macrophages, and T regulatory cells that are common to the different settings in which type-2 immunity has been explored. We further discuss how an understanding of these interactions can reveal new avenues of therapeutic tissue regeneration, where the role of ILC2s is yet to be fully established.
Collapse
Affiliation(s)
- Melina Messing
- Division of Experimental Medicine, Faculty of Medicine, University of British Columbia, The Biomedical Research Centre, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada;
| | - Sia Cecilia Jan-Abu
- Department of Medical Genetics and School of Biomedical Engineering, Faculty of Applied Science and Faculty of Medicine, University of British Columbia, The Biomedical Research Centre, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada;
| | - Kelly McNagny
- Department of Medical Genetics and School of Biomedical Engineering, Faculty of Applied Science and Faculty of Medicine, University of British Columbia, The Biomedical Research Centre, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada;
| |
Collapse
|
26
|
Chang CP, Hu MH, Hsiao YP, Wang YC. ST2 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1240:83-93. [PMID: 32060890 DOI: 10.1007/978-3-030-38315-2_7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Suppression of tumorigenicity 2 (ST2), also known as interleukin-1 receptor-like 1 (IL1RL1), is one of the natural receptors of IL-33. Three major isoforms, ST2L (transmembrane form), sST2 (soluble form), and ST2V, are generated by alternative splicing. Damage to stromal cells induces necrosis and release of IL-33, which binds to heterodimeric ST2L/IL-1RAcP complex on the membrane of a variety of immune cells. This IL-33/ST2L signal induces transcription of the downstream inflammatory and anti-inflammatory genes by activating diverse intracellular kinases and factors to mount an adequate immune response, even in tumor microenvironment. For example, activation of IL-33/ST2L signal may trigger Th2-dependent M2 macrophage polarization to facilitate tumor progression. Notably, sST2 is a soluble form of ST2 that lacks a transmembrane domain but preserves an extracellular domain similar to ST2L, which acts as a "decoy" receptor for IL-33. sST2 has been shown to involve in the inflammatory tumor microenvironment and the progression of colorectal cancer, non-small cell lung cancer, and gastric cancer. Therefore, targeting the IL-33/ST2 axis becomes a promising new immunotherapy for treatment of many cancers. This chapter reviews the recent findings on IL-33/ST2L signaling in tumor microenvironment, the trafficking mode of sST2, and the pharmacological strategies to target IL-33/ST2 axis for cancer treatment.
Collapse
Affiliation(s)
- Chih-Peng Chang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Meng-Hsuan Hu
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Peng Hsiao
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ching Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
27
|
Nakashima T, Liu T, Hu B, Wu Z, Ullenbruch M, Omori K, Ding L, Hattori N, Phan SH. Role of B7H3/IL-33 Signaling in Pulmonary Fibrosis-induced Profibrogenic Alterations in Bone Marrow. Am J Respir Crit Care Med 2019; 200:1032-1044. [PMID: 31106564 PMCID: PMC6794107 DOI: 10.1164/rccm.201808-1560oc] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 05/14/2019] [Indexed: 01/12/2023] Open
Abstract
Rationale: The impact of lung insult on the bone marrow (BM) and subsequent disease is unknown.Objectives: To study alterations in the BM in response to lung injury/fibrosis and examine their impact on subsequent lung insult.Methods: BM cells from control or bleomycin-treated donor mice were transplanted into naive mice, which were subsequently evaluated for bleomycin-induced pulmonary fibrosis. In addition, the effect of prior bleomycin treatment on subsequent fibrosis was examined in wild-type and B7H3-knockout mice. Samples from patients with idiopathic pulmonary fibrosis were analyzed for potential clinical relevance of the findings.Measurements and Main Results: Recipient mice transplanted with BM from bleomycin-pretreated donors showed significant exacerbation of subsequent fibrosis with increased B7H3+ cell numbers and a T-helper cell type 2-skewed phenotype. Pretreatment with a minimally fibrogenic/nonfibrogenic dose of bleomycin also caused exacerbation, but not in B7H3-deficient mice. Exacerbation was not observed if the mice received naive BM cell transplant after the initial bleomycin pretreatment. Soluble B7H3 stimulated BM Ly6Chi monocytic cell expansion in vitro and caused similar expansion in the lung in vivo. Notably, soluble B7H3 was elevated in plasma of patients with idiopathic pulmonary fibrosis and in BAL fluid in those with acute exacerbation. Finally, ST2 deficiency diminished the bleomycin-induced B7H3 and IL-13 upregulation, suggesting a role for type 2 innate lymphoid cells.Conclusions: Pulmonary fibrosis caused significant alterations in BM with expansion and activation of monocytic cells, which enhanced fibrosis when transplanted to naive recipients with potential mediation by a novel role for B7H3 in the pathophysiology of pulmonary fibrosis in both mice and humans.
Collapse
Affiliation(s)
- Taku Nakashima
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; and
| | - Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Biao Hu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Zhe Wu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Matthew Ullenbruch
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Keitaro Omori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; and
| | - Lin Ding
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; and
| | - Sem H. Phan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
28
|
Epithelial Alarmins in Serum and Exhaled Breath in Patients with Idiopathic Pulmonary Fibrosis: A Prospective One-Year Follow-Up Cohort Study. J Clin Med 2019; 8:jcm8101590. [PMID: 31581688 PMCID: PMC6832270 DOI: 10.3390/jcm8101590] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/24/2019] [Accepted: 09/26/2019] [Indexed: 12/13/2022] Open
Abstract
Background: Recently, epithelial alarmins have been shown to play important roles in non-allergen driven respiratory diseases like idiopathic pulmonary fibrosis (IPF). Little is known about the expression of the epithelial alarmins in IPF. Methods: This study aimed to prospectively examine interleukin (IL)-25, IL-33, and thymic stromal lymphopoietin (TSLP) levels in the serum and exhaled breath condensate (EBC) in patients with IPF before and after one-year of antifibrotic treatment. A total of 82 volunteers, including 52 patients diagnosed with IPF that qualified for antifibrotic therapy as well as 30 controls, were examined. All study participants underwent baseline peripheral blood and EBC sampling. In 35 out of 52 IPF subjects, a follow-up sampling was performed after 12 months of antifibrotic treatment. Concentrations of alarmins in the serum and EBC were evaluated by means of ELISA. Results: Baseline TSLP concentrations were significantly elevated in patients with IPF compared to controls both in the serum (p < 0.05) and EBC (p < 0.0001). Baseline IL-25 and IL-33 serum and EBC levels did not differ significantly between IPF subjects and controls. Prospective analysis of changes in the epithelial alarmin levels showed significantly decreased IL-25 and TSLP EBC concentrations after 12 months of antifibrotic treatment (p < 0.05), which was observed in the subgroup of IPF patients treated with pirfenidone, but not in those treated with nintedanib. In stable patients with IPF over a study period (absolute forced vital capacity (FVC) % of predicted decline/year ≤ 5%, n = 25), a significant decrease in the EBC levels of both IL-25 and TSLP after 12 months of antifibrotic treatment was noted (p < 0.05), whereas, in progressor IPF patients (absolute FVC % of predicted decline/year > 5%, n = 10), a significant decrease was noted in the IL-25 EBC levels only (p < 0.05). Conclusions: Elevated TSLP levels in patients with IPF and their significant decrease in the lung compartment during antifibrotic therapy in stable patients with IPF, but not in progressors, support its significant contribution to pro-fibrotic type 2 immune responses in IPF. Noted changes in the epithelial alarmins concentration in the lung compartment during pirfenidone therapy may suggest its possible interaction with epithelial alarmins pathways in IPF.
Collapse
|
29
|
Majewski S, Tworek D, Szewczyk K, Kurmanowska Z, Antczak A, Górski P, Piotrowski WJ. Epithelial alarmin levels in exhaled breath condensate in patients with idiopathic pulmonary fibrosis: A pilot study. CLINICAL RESPIRATORY JOURNAL 2019; 13:652-656. [PMID: 31392802 DOI: 10.1111/crj.13075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/30/2019] [Accepted: 08/01/2019] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Interleukin (IL)-25, IL-33 and thymic stromal lymphopoietin (TSLP) are epithelial alarmins involved in innate immune responses and have been shown to play an important role in chronic lung diseases. No data are available regarding their levels in exhaled breath condensate (EBC) in idiopathic pulmonary fibrosis (IPF). OBJECTIVES To examine IL-25, IL-33 and TSLP levels in the EBC obtained from patients with IPF and compare them to those in healthy controls, patients with asthma and chronic obstructive pulmonary disease (COPD). METHODS Twenty-three patients with asthma, 25 patients with COPD, 15 patients with IPF and 16 healthy controls were studied. Concentrations of alarmins in the EBC were evaluated by means of ELISA. RESULTS IL-25 EBC levels were numerically lowest in IPF (25.33 ± 8.84 pg/ml). However, they did not differ significantly from healthy subjects (43.18 ± 5.53 pg/ml), but were significantly lower compared to asthma (72.07 ± 6.03 pg/ml; P < .001). IL-33 EBC levels were significantly increased in IPF (3.41 ± 0.55 pg/ml) compared to healthy controls (1.20 ± 0.60 pg/ml; P < .01) but did not differ from asthma (3.68 pg/ml) and COPD levels (2.47 ± 0.34 pg/ml). There were significant correlations between IL-33 EBC levels and lung diffusion capacity of carbon monoxide (DLco ) absolute (r = .63; P < .05) and % of predicted values (r = .67; P < .01) as well as with time since diagnosis (r = -.59; P < .05) in IPF subjects. TSLP was undetectable in examined samples. CONCLUSION IL-25 and IL-33 are detectable in the EBC obtained from IPF subjects. Increased levels of IL-33 compared to healthy controls indicate its possible role in the pathobiology of IPF.
Collapse
Affiliation(s)
- Sebastian Majewski
- Department of Pneumology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Damian Tworek
- Department of General and Oncological Pulmonology, Medical University of Lodz, Lodz, Poland
| | - Karolina Szewczyk
- Department of Pneumology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Zofia Kurmanowska
- Department of Pneumology and Allergy, Medical University of Lodz, Lodz, Poland
| | - Adam Antczak
- Department of General and Oncological Pulmonology, Medical University of Lodz, Lodz, Poland
| | - Paweł Górski
- Department of Pneumology and Allergy, Medical University of Lodz, Lodz, Poland
| | | |
Collapse
|
30
|
Kotsiou OS, Gourgoulianis KI, Zarogiannis SG. IL-33/ST2 Axis in Organ Fibrosis. Front Immunol 2018; 9:2432. [PMID: 30405626 PMCID: PMC6207585 DOI: 10.3389/fimmu.2018.02432] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/02/2018] [Indexed: 12/19/2022] Open
Abstract
Interleukin 33 (IL-33) is highly expressed in barrier sites, acting via the suppression of tumorigenicity 2 receptor (ST2). IL-33/ST2 axis has long been known to play a pivotal role in immunity and cell homeostasis by promoting wound healing and tissue repair. However, it is also involved in the loss of balance between extensive inflammation and tissue regeneration lead to remodeling, the hallmark of fibrosis. The aim of the current review is to critically evaluate the available evidence regarding the role of the IL-33/ST2 axis in organ fibrosis. The role of the axis in tissue remodeling is better understood considering its crucial role reported in organ development and regeneration. Generally, the IL-33/ST2 signaling pathway has mainly anti-inflammatory/anti-proliferative effects; however, chronic tissue injury is responsible for pro-fibrogenetic responses. Regarding pulmonary fibrosis mature IL-33 enhances pro-fibrogenic type 2 cytokine production in an ST2- and macrophage-dependent manner, while full-length IL-33 is also implicated in the pulmonary fibrotic process in an ST2-independent, Th2-independent fashion. In liver fibrosis, evidence indicate that when acute and massive liver damage occurs, the release of IL-33 might act as an activator of tissue-protective mechanisms, while in cases of chronic injury IL-33 plays the role of a hepatic fibrotic factor. IL-33 signaling has also been involved in the pathogenesis of acute and chronic pancreatitis. Moreover, IL-33 could be used as an early marker for ulcer-associated activated fibroblasts and myofibroblast trans-differentiation; thus one cannot rule out its potential role in inflammatory bowel disease-associated fibrosis. Similarly, the upregulation of the IL-33/ST2 axismay contribute to tubular cell injury and fibrosis via epithelial to mesenchymal transition (EMT) of various cell types in the kidneys. Of note, IL-33 exerts a cardioprotective role via ST2 signaling, while soluble ST2 has been demonstrated as a marker of myocardial fibrosis. Finally, IL-33 is a crucial cytokine in skin pathology responsible for abnormal fibroblast proliferation, leukocyte infiltration and morphologic differentiation of human endothelial cells. Overall, emerging data support a novel contribution of the IL-33/ST2 pathway in tissue fibrosis and highlight the significant role of the Th2 pattern of immune response in the pathophysiology of organ fibrosis.
Collapse
Affiliation(s)
- Ourania S. Kotsiou
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, Larissa, Greece
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, Larissa, Greece
| | - Konstantinos I. Gourgoulianis
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, Larissa, Greece
| | - Sotirios G. Zarogiannis
- Department of Respiratory Medicine, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, Larissa, Greece
- Department of Physiology, Faculty of Medicine, School of Health Sciences, University of Thessaly, BIOPOLIS, Larissa, Greece
| |
Collapse
|
31
|
Wang X, Zhao H, Ma C, Lv L, Feng J, Han S. Gallic acid attenuates allergic airway inflammation via suppressed interleukin-33 and group 2 innate lymphoid cells in ovalbumin-induced asthma in mice. Int Forum Allergy Rhinol 2018; 8:1284-1290. [PMID: 30191679 DOI: 10.1002/alr.22207] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 07/27/2018] [Accepted: 08/21/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Asthma is an inflammatory disease characterized by airway hyperresponsiveness. Gallic acid is a powerful anti-inflammatory agent. In this study we aimed to investigate the efficacy of gallic acid in asthma treatment and its mechanisms. METHODS An ovalbumin-induced asthma mouse model was generated. Pro-inflammatory cell infiltration and T helper (Th2)-associated cytokine release in the bronchoalveolar lavage fluid (BALF) were analyzed to reflect the severity of asthma in mice. An interleukin-33 (IL-33)-induced asthma mouse model was also generated to study the mechanism by which gallic acid could improve asthma. Group 2 lymphoid cells (ILC2s) were identified using flow cytometry. Proteins were detected using Western blotting. RESULTS Ovalbumin significantly increased the infiltration of pro-inflammatory cells, including eosinophils, macrophages, lymphocytes, and neutrophils, accompanied by enhanced airway hyperesponsiveness. Gallic acid reduced pro-inflammatory cell infiltration and improved airway hyperresponsiveness. Meanwhile, gallic acid reduced IL-5 and IL-13 levels in BALF and decreased expression of IL-33 in the lungs. Mechanistically, gallic acid inhibited MyD88 expression and downregulated nuclear factor (NF)-κB signaling to decrease IL-33 expression. CONCLUSIONS Gallic acid can mollify ovalbumin-induced asthma in mice, possibly by inhibiting IL-33-mediated ILC2 activation and subsequent Th2 cytokine release via downregulation of the MyD88/NF-κB signaling pathway. ©2018 ARSAAOA, LLC.
Collapse
Affiliation(s)
- Xinhua Wang
- Department of Respiratory Medicine, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Liangxi District, Wuxi, China
| | - Hongqing Zhao
- Department of Respiratory Medicine, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Liangxi District, Wuxi, China
| | - Chenhui Ma
- Department of Respiratory Medicine, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Liangxi District, Wuxi, China
| | - Lei Lv
- Department of Respiratory Medicine, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Liangxi District, Wuxi, China
| | - Jinping Feng
- Department of Respiratory Medicine, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Liangxi District, Wuxi, China
| | - Shuguang Han
- Department of Respiratory Medicine, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Liangxi District, Wuxi, China
| |
Collapse
|
32
|
Fanny M, Nascimento M, Baron L, Schricke C, Maillet I, Akbal M, Riteau N, Le Bert M, Quesniaux V, Ryffel B, Gombault A, Même S, Même W, Couillin I. The IL-33 Receptor ST2 Regulates Pulmonary Inflammation and Fibrosis to Bleomycin. Front Immunol 2018; 9:1476. [PMID: 29988569 PMCID: PMC6026799 DOI: 10.3389/fimmu.2018.01476] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 06/13/2018] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis is a progressive, devastating, and yet untreatable fibrotic disease of unknown origin. Interleukin-33 (IL-33), an IL-1 family member acts as an alarmin with pro-inflammatory properties when released after stress or cell death. Here, we investigated the role of IL-33 in the bleomycin (BLM)-induced inflammation and fibrosis model using mice IL-33 receptor [chain suppression of tumorigenicity 2 (ST2)] mice compared with C57BL/6 wild-type mice. Unexpectedly, 24 h post-BLM treatment ST2-deficient mice displayed augmented inflammatory cell recruitment, in particular by neutrophils, together with enhanced levels of chemokines and remodeling factors in the bronchoalveolar space and/or the lungs. At 11 days, lung remodeling and fibrosis were decreased with reduced M2 macrophages in the lung associated with M2-like cytokine profile in ST2-deficient mice, while lung cellular inflammation was decreased but with fluid retention (edema) increased. In vivo magnetic resonance imaging (MRI) analysis demonstrates a rapid development of edema detectable at day 7, which was increased in the absence of ST2. Our results demonstrate that acute neutrophilic pulmonary inflammation leads to the development of an IL-33/ST2-dependent lung fibrosis associated with the production of M2-like polarization. In addition, non-invasive MRI revealed enhanced inflammation with lung edema during the development of pulmonary inflammation and fibrosis in absence of ST2.
Collapse
Affiliation(s)
- Manoussa Fanny
- University of Orleans and CNRS, UMR7355, Orleans, France
| | | | - Ludivine Baron
- University of Orleans and CNRS, UMR7355, Orleans, France
| | | | | | - Myriam Akbal
- University of Orleans and CNRS, UMR7355, Orleans, France
| | - Nicolas Riteau
- University of Orleans and CNRS, UMR7355, Orleans, France
| | - Marc Le Bert
- University of Orleans and CNRS, UMR7355, Orleans, France
| | | | | | | | - Sandra Même
- University of Orleans and CNRS, UPR4301, Orleans, France
| | - William Même
- University of Orleans and CNRS, UPR4301, Orleans, France
| | | |
Collapse
|