1
|
Shi Z, Liu J, Qin J, Liang X, Ou X, Zhang T, Yan X, Hu Q, Huang W, Hu K. Astilbin Alleviates Radiation-Induced Pulmonary Fibrosis via circPRKCE Targeting the TGF-β/Smad7 Pathway to Inhibit Epithelial-Mesenchymal Transition. Biomedicines 2025; 13:689. [PMID: 40149664 PMCID: PMC11939908 DOI: 10.3390/biomedicines13030689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/29/2025] Open
Abstract
Purpose: This study aimed to clarify the protective effect of astilbin (AST) on radiation-induced pulmonary fibrosis (RIPF) and explore its underlying molecular mechanism, focusing on non-coding RNAs. Methods: Mouse lung epithelial cells (MLE-12 and TC-1) and C57BL/6J mice were used to establish in vitro radiation injury models and in vivo RIPF models, respectively. Cell viability, apoptosis, the epithelial-to-mesenchymal transition (EMT), and fibrosis-related markers were assessed using cell-counting kit-8 assays, Western blotting, immunohistochemistry, and histological staining. High-throughput sequencing identified differentially expressed circRNAs. The mechanistic studies included RNA-FISH, a dual-luciferase reporter assay, an RNA immunoprecipitation (RIP) assay, and loss-of-function experiments. Results: AST significantly alleviated radiation-induced apoptosis and EMT in vitro, as well as RIPF in vivo. AST treatment reduced collagen deposition, fibrosis-related protein expression, and EMT marker changes. High-throughput sequencing revealed that AST upregulated circPRKCE, a non-coding RNA that functions through a ceRNA mechanism by binding to miR-15b-5p, thereby promoting Smad7 expression and suppressing the TGF-β/Smad7 pathway. Knockdown of circPRKCE abolished AST's protective effects, confirming its pivotal role in mediating AST's anti-fibrotic activity. Conclusions: This study demonstrates that Astilbin alleviates radiation-induced pulmonary fibrosis via circPRKCE targeting the TGF-β/Smad7 pathway to inhibit EMT, suggesting AST as a potential therapeutic agent for managing this severe complication of radiotherapy.
Collapse
Affiliation(s)
- Zhiling Shi
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning 530021, China
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Jing Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning 530021, China
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Jing Qin
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
| | - Xian Liang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
| | - Xue Ou
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
- Department of Radiation Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, China
| | - Tingting Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
| | - Xueting Yan
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
| | - Qianxin Hu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
| | - Weimei Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning 530021, China
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| | - Kai Hu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China; (Z.S.); (J.L.); (J.Q.); (X.L.); (X.O.); (T.Z.); (X.Y.); (Q.H.)
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning 530021, China
- State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
2
|
Rajabinasab F, Hajimirzaei P, Ramezani F, Moayer F, Gorjipour F, Nikoofar A, Hassanzadeh L, Hamblin MR, Janzadeh A, Paydar R. Apigenin's Influence on Inflammatory and Epigenetic Responses in Rat Lungs After Radiotherapy. Curr Radiopharm 2025; 18:147-157. [PMID: 39444181 DOI: 10.2174/0118744710336823241011095632] [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: 06/11/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION The lung is a moderately radio-sensitive organ. When cells are damaged due to accidental radiation exposure or treatment, they release molecules that lead to the recruitment of immune cells, accumulating inflammatory cytokines at the site of damage. Apigenin (Api) is a natural flavonoid known for its anti-inflammatory properties. In this study, we investigated the radioprotective properties of Api in the lung. METHODS Thirty-six Wistar rats were randomly assigned to nine groups: control, radiation (Rad), CMC+Rad, Api10+Rad, and Api20+Rad. Api was administered with an intraperitoneal injection for 7 days, after which the rats were irradiated with 6 Gy whole-body X-ray. At 6 and 72 hours post-irradiation, the rats were euthanized, and their lung tissue was extracted. RESULTS Radiation led to increased alveolar wall thickness and the infiltration of macrophages and lymphocytes. Furthermore, the expression levels of inflammatory factors such as a nuclear factor of kappa light polypeptide gene enhancer in B-cells (NF-κB), Glycogen synthase kinase-3 beta (GSK-3β), transforming growth factor-beta1 (TGF-β1), and epigenetic factors including DNA methyltransferase 3a (DNMT3a) and Histone deacetylase 2 (HDAC2) were elevated in the lung tissue following radiation. Meanwhile, the expression level of IκB-α decreased. However, administration of Api (at both 10&20 mg/kg) reversed the adverse effects of radiation. CONCLUSION Api administration mitigated radiation-induced lung damage by reversing inflammatory and epigenetic changes.
Collapse
Affiliation(s)
- Fatemeh Rajabinasab
- Department of Radiation Sciences, Allied Medicine Faculty, Iran University of Medical Sciences, Tehran, Iran
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Pooya Hajimirzaei
- Department of Radiation Sciences, Allied Medicine Faculty, Iran University of Medical Sciences, Tehran, Iran
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Ramezani
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fariborz Moayer
- Department of Pathobiology, Faculty of Veterinary Medicine, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Fazel Gorjipour
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Nikoofar
- Department of Radiation Oncology, Iran University of Medical Sciences, Tehran, Iran
| | - Leila Hassanzadeh
- Department of Nuclear Medicine, School of Medicine, Rajaie Cardiovascular, Medical & Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Imaging, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, University of Johannesburg, Doornfontein, South Africa
| | - Atousa Janzadeh
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Paydar
- Department of Radiation Sciences, Allied Medicine Faculty, Iran University of Medical Sciences, Tehran, Iran
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Chen S, Song X, Lv C. Macrophages and Pulmonary Fibrosis. Curr Mol Med 2025; 25:416-430. [PMID: 39779550 DOI: 10.2174/0115665240286046240112112310] [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: 10/07/2023] [Revised: 12/27/2023] [Accepted: 01/04/2024] [Indexed: 01/11/2025]
Abstract
Most chronic respiratory diseases often lead to the clinical manifestation of pulmonary fibrosis. Inflammation and immune disorders are widely recognized as primary contributors to the onset of pulmonary fibrosis. Given that macrophages are predominantly responsible for inflammation and immune disorders, in this review, we first focused on the role of different subpopulations of macrophages in the lung and discussed the crosstalk between macrophages and other immune cells, such as neutrophils, regulatory T cells, NKT cells, and B lymphocytes during pulmonary fibrogenesis. Subsequently, we analyzed the interaction between macrophages and fibroblasts as a possible new research direction. Finally, we proposed that exosomes, which function as a means of communication between macrophages and target cells to maintain cellular homeostasis, are a strategy for targeting lung drugs in the future. By comprehending the mechanisms underlying the interplay between macrophages and other lung cells, we aim to enhance our understanding of pulmonary fibrosis, leading to improved diagnostics, preventative measures, and the potential development of macrophage-based therapeutics.
Collapse
Affiliation(s)
- Shengjun Chen
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, China
| | - Xiaodong Song
- Department of Cellular and Genetic Medicine, Binzhou Medical University, Yantai 264003, China
| | - Changjun Lv
- Department of Respiratory and Critical Care Medicine, Binzhou Medical University Hospital, Binzhou Medical University, Binzhou 256603, China
| |
Collapse
|
4
|
Wang Y, Zhang J, Shao C. Cytological changes in radiation-induced lung injury. Life Sci 2024; 358:123188. [PMID: 39481833 DOI: 10.1016/j.lfs.2024.123188] [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: 09/20/2024] [Revised: 10/20/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
Radiation-induced lung injury (RILI) is a prevalent complication associated with radiotherapy for thoracic tumors. Based on the pathological progression, it can be categorized into two stages: early radiation pneumonitis and late radiation pulmonary fibrosis. The occurrence of RILI not only constrains the therapeutic dose that can be administered to the tumor target area but also significantly impairs patients' health and quality of life, thereby limiting the efficacy and applicability of radiotherapy. To effectively prevent and mitigate the development of RILI, it is crucial to disclose its underlying mechanisms. This review aims to elucidate the specific mechanisms involved in RILI and to examine the roles of various cell types, including lung parenchymal cells and different immune cells. The functions and interactions of lung epithelial cells, pulmonary vascular endothelial cells, a variety of immune cells, and fibroblasts during different stages of inflammation, tissue repair, and fibrosis following radiation-induced lung injury are analyzed. A comprehensive understanding of the dynamic changes in these cellular components is anticipated to offer new strategies for the prevention of RILI.
Collapse
Affiliation(s)
- Yun Wang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China
| | - Jianghong Zhang
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, No. 2094 Xie-Tu Road, Shanghai 200032, China.
| |
Collapse
|
5
|
Chang S, Lv J, Wang X, Su J, Bian C, Zheng Z, Yu H, Bao J, Xin Y, Jiang X. Pathogenic mechanisms and latest therapeutic approaches for radiation-induced lung injury: A narrative review. Crit Rev Oncol Hematol 2024; 202:104461. [PMID: 39103129 DOI: 10.1016/j.critrevonc.2024.104461] [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: 10/13/2023] [Revised: 07/26/2024] [Accepted: 07/28/2024] [Indexed: 08/07/2024] Open
Abstract
The treatment of thoracic tumors with ionizing radiation can cause radiation-induced lung injury (RILI), which includes radiation pneumonitis and radiation-induced pulmonary fibrosis. Preventing RILI is crucial for controlling tumor growth and improving quality of life. However, the serious adverse effects of traditional RILI treatment methods remain a major obstacle, necessitating the development of novel treatment options that are both safe and effective. This review summarizes the molecular mechanisms of RILI and explores novel treatment options, including natural compounds, gene therapy, nanomaterials, and mesenchymal stem cells. These recent experimental approaches show potential as effective prevention and treatment options for RILI in clinical practice.
Collapse
Affiliation(s)
- Sitong Chang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China; Department of Radiation Oncology, the First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun 130021, China.
| | - Jincai Lv
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China; Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Xuanzhong Wang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China; Department of Radiation Oncology, the First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun 130021, China.
| | - Jing Su
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China; Department of Radiation Oncology, the First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun 130021, China.
| | - Chenbin Bian
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China; Department of Radiation Oncology, the First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun 130021, China.
| | - Zhuangzhuang Zheng
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China; Department of Radiation Oncology, the First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun 130021, China.
| | - Huiyuan Yu
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China; Department of Radiation Oncology, the First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun 130021, China.
| | - Jindian Bao
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China; Department of Radiation Oncology, the First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun 130021, China.
| | - Ying Xin
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China; Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China.
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University and College of Basic Medical Science, Jilin University, Changchun, China; Department of Radiation Oncology, the First Hospital of Jilin University, Changchun 130021, China; NHC Key Laboratory of Radiobiology, School of Public Health of Jilin University, Changchun 130021, China.
| |
Collapse
|
6
|
Zheng M, Liu Z, He Y. Radiation-induced fibrosis: Mechanisms and therapeutic strategies from an immune microenvironment perspective. Immunology 2024; 172:533-546. [PMID: 38561001 DOI: 10.1111/imm.13788] [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: 07/24/2023] [Accepted: 03/22/2024] [Indexed: 04/04/2024] Open
Abstract
Radiation-induced fibrosis (RIF) is a severe chronic complication of radiotherapy (RT) manifested by excessive extracellular matrix (ECM) components deposition within the irradiated area. The lung, heart, skin, jaw, pelvic organs and so on may be affected by RIF, which hampers body functions and quality of life. There is accumulating evidence suggesting that the immune microenvironment may play a key regulatory role in RIF. This article discussed the synergetic or antagonistic effects of immune cells and mediators in regulating RIF's development. Several potential preventative and therapeutic strategies for RIF were proposed based on the immunological mechanisms to provide clinicians with improved cognition and clinical treatment guidance.
Collapse
Affiliation(s)
- Mengting Zheng
- Department of Oral Maxillofacial & Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Zhonglong Liu
- Department of Oral Maxillofacial & Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yue He
- Department of Oral Maxillofacial & Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
7
|
Liu B, Wang Y, Ma L, Chen G, Yang Z, Zhu M. CCL22 Induces the Polarization of Immature Dendritic Cells into Tolerogenic Dendritic Cells in Radiation-Induced Lung Injury through the CCR4-Dectin2-PLC-γ2-NFATC2-Nr4a2-PD-L1 Signaling Pathway. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:268-282. [PMID: 38856585 DOI: 10.4049/jimmunol.2300718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 05/20/2024] [Indexed: 06/11/2024]
Abstract
Recruitment of immune cells to the injury site plays a pivotal role in the pathology of radiation-associated diseases. In this study, we investigated the impact of the chemokine CCL22 released from alveolar type II epithelial (AT2) cells after irradiation on the recruitment and functional changes of dendritic cells (DCs) in the development of radiation-induced lung injury (RILI). By examining changes in CCL22 protein levels in lung tissue of C57BL/6N mice with RILI, we discovered that ionizing radiation increased CCL22 expression in irradiated alveolar AT2 cells, as did MLE-12 cells after irradiation. A transwell migration assay revealed that CCL22 promoted the migration of CCR4-positive DCs to the injury site, which explained the migration of pulmonary CCR4-positive DCs in RILI mice in vivo. Coculture experiments demonstrated that, consistent with the response of regulatory T cells in the lung tissue of RILI mice, exogenous CCL22-induced DCs promoted regulatory T cell proliferation. Mechanistically, we demonstrated that Dectin2 and Nr4a2 are key targets in the CCL22 signaling pathway, which was confirmed in pulmonary DCs of RILI mice. As a result, CCL22 upregulated the expression of PD-L1, IL-6, and IL-10 in DCs. Consequently, we identified a mechanism in which CCL22 induced DC tolerance through the CCR4-Dectin2-PLC-γ2-NFATC2-Nr4a2-PD-L1 pathway. Collectively, these findings demonstrated that ionizing radiation stimulates the expression of CCL22 in AT2 cells to recruit DCs to the injury site and further polarizes them into a tolerant subgroup of CCL22 DCs to regulate lung immunity, ultimately providing potential therapeutic targets for DC-mediated RILI.
Collapse
Affiliation(s)
- Benbo Liu
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yilong Wang
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Liping Ma
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Guo Chen
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhihua Yang
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Maoxiang Zhu
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
8
|
Liang X, Huang X, Cai Z, Deng Y, Liu D, Hu J, Jin Z, Zhou X, Zhou H, Wang L. The S100 family is a prognostic biomarker and correlated with immune cell infiltration in pan-cancer. Discov Oncol 2024; 15:137. [PMID: 38684596 PMCID: PMC11058162 DOI: 10.1007/s12672-024-00945-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/20/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND The S100 protein family is a group of small molecular EF-hand calcium-binding proteins that play critical roles in various biological processes, including promotion of growth, metastasis and immune evasion of tumor. However, the potential roles of S100 protein family expression in tumor microenvironment (TME) cell infiltration in pan-cancer remain elusive. METHODS Herein, we conducted a comprehensive assessment of the expression patterns of the S100 protein family in pan-cancer, meticulously examining their correlation with characteristics of TME cell infiltration. The S100 score was constructed to quantify S100 family expression patterns of individual tumors. RESULTS The S100 family was a potent risk factor in many cancers. Clustering analysis based on the transcriptome patterns of S100 protein family identified two cancer clusters with distinct immunophenotypes and clinical characteristics. Cluster A, with lower S100 expression, exhibited lower immune infiltration, whereas, Cluster B, with higher S100 expression, featured higher immune infiltration. Interestingly, Cluster B had a poorer prognosis, likely due to an immune-excluded phenotype resulting from stromal activation. The analysis revealed robust enrichment of the TGFb and EMT pathways in the cohort exhibiting high S100 score, alongside a positive correlation between the S100 score and Treg levels, suggesting the manifestation of an immune-excluded phenotype in this group. Moreover, S100 families were associated with the prognosis of 22 different cancers and a noteworthy association was observed between high S100 score and an unfavorable response to anti-PD-1/L1 immunotherapy. Consistent findings across two independent immunotherapy cohorts substantiated the advantageous therapeutic outcomes and clinical benefits in patients displaying lower S100score. CONCLUSION Our analysis demonstrated the role of S100 family in formation of TME diversity and complexity, enabling deeper cognition of TME infiltration characterization and the development of personalized immunotherapy strategies targeting S100 family for unique tumor types.
Collapse
Affiliation(s)
- Xiaojie Liang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xiaoshan Huang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zihong Cai
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yeling Deng
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, 524000, China
| | - Dan Liu
- Department of Radiology, Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde), Southern Medical University, Foshan, China
| | - Jiayi Hu
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, 524000, China
| | - Zhihao Jin
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, 524000, China
| | - Xinyu Zhou
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, 524000, China
| | - Hongsheng Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
9
|
Wang M, Feng Y, Zhang P, Shen K, Su J, Zhong Y, Yang X, Lin S, Lu J. Jiawei Maxing Shigan Tang alleviates radiation-induced lung injury via TGF-β1/Smad signaling pathway mediated by regulatory T cells. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117389. [PMID: 37944875 DOI: 10.1016/j.jep.2023.117389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/26/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Radiation-induced lung injury (RILI) is a common complication during thoracic radiotherapy which impairs the quality of life in patients and limits radiation doses. Jiawei Maxing Shigan Tang (JMST), which is a modified decoction made of Ephedra, Apricot Kernel, Gypsum, and Licorice, can alleviate the symptoms of RILI in patients. Previous studies and preliminary findings suggested a potential molecular mechanism of JMST in the treatment of RILI. Further studies are needed. AIM OF THE STUDY To elucidate the mechanisms of how regulatory T cells (Tregs) promote RILI and the effect of JMST on Tregs, as well as the corresponding pathway. MATERIALS AND METHODS CD4+CD25+ Tregs were isolated from rats, and the supernatant's TGF-β1 level was examined by using enzyme-linked immunosorbent assay (ELISA). Type II alveolar epithelial cells (AECs) were co-cultured with the supernatant of Tregs, and the expression levels of epithelial-to-mesenchymal transition (EMT)-related and TGF-β1/Smad signaling pathway-related proteins were analyzed by using western blotting (WB). Afterward, the Tregs were incubated with different concentrations of JMST. The cell viability and TGF-β1 concentration were confirmed by cell counting kit-8 (CCK-8) assay and ELISA, respectively. The optimized concentration of JMST was applied in vitro and vivo experiments. The specific mechanism was investigated through the combination of using flow cytometry, lung histopathology analysis, ELISA, and WB. RESULTS Radiation could promote Tregs to secrete TGF-β1. After radiation, the expression levels of Smad2/3, phosphorylated Smad2/3 (p-Smad2/3), Smad4 and mesenchymal markers Vimentin and α-SMA were all increased, while the expression level of epithelial markers E-cadherin was decreased. The expression levels of these proteins were reversed after interventions involving Treg cell activation inhibition or TGF-β1 receptor inhibitor. JMST reduced the number of Tregs in lung tissue and alleviated the degree of pulmonary fibrosis. The expression of Smad2/3, p-Smad2/3, Smad4, TGF-β1, Vimentin, and α-SMA were significantly downregulated, while the E-cadherin was upregulated, through the intervention of JMST. CONCLUSION Tregs could mediate EMT through TGF-β1/Smad pathway. JMST inhibits EMT via TGF-β1/Smad pathway by regulating Tregs, therefore alleviating RILI.
Collapse
Affiliation(s)
- Menglei Wang
- The Third School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuqian Feng
- Hangzhou School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Pengcheng Zhang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Kezhan Shen
- Hangzhou School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingyang Su
- Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Yazhen Zhong
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Xuefei Yang
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China.
| | - Shengyou Lin
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Province Hospital of Chinese Medicine), Hangzhou, China.
| | - Jinhua Lu
- Department of Oncology, Hangzhou TCM Hospital of Zhejiang Chinese Medical University (Hangzhou Hospital of Traditional Chinese Medicine), Hangzhou, China.
| |
Collapse
|
10
|
Guo H, Yu R, Zhang H, Wang W. Cytokine, chemokine alterations and immune cell infiltration in Radiation-induced lung injury: Implications for prevention and management. Int Immunopharmacol 2024; 126:111263. [PMID: 38000232 DOI: 10.1016/j.intimp.2023.111263] [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: 10/22/2023] [Revised: 11/11/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023]
Abstract
Radiation therapy is one of the primary treatments for thoracic malignancies, with radiation-induced lung injury (RILI) emerging as its most prevalent complication. RILI encompasses early-stage radiation pneumonitis (RP) and the subsequent development of radiation pulmonary fibrosis (RPF). During radiation treatment, not only are tumor cells targeted, but normal tissue cells, including alveolar epithelial cells and vascular endothelial cells, also sustain damage. Within the lungs, ionizing radiation boosts the intracellular levels of reactive oxygen species across various cell types. This elevation precipitates the release of cytokines and chemokines, coupled with the infiltration of inflammatory cells, culminating in the onset of RP. This pulmonary inflammatory response can persist, spanning a duration from several months to years, ultimately progressing to RPF. This review aims to explore the alterations in cytokine and chemokine release and the influx of immune cells post-ionizing radiation exposure in the lungs, offering insights for the prevention and management of RILI.
Collapse
Affiliation(s)
- Haochun Guo
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China
| | - Ran Yu
- Department of Radiotherapy, Lianshui People's Hospital, Kangda College of Nanjing Medical University, Huai'an 223400, China; Jiangsu Nursing Vocational and Technical College, Huai'an 223400, China; School of Clinical Medicine, Medical College of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou 225009, China
| | - Haijun Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing 210009, China.
| | - Wanpeng Wang
- Department of Radiotherapy, Lianshui People's Hospital, Kangda College of Nanjing Medical University, Huai'an 223400, China; Jiangsu Nursing Vocational and Technical College, Huai'an 223400, China; School of Clinical Medicine, Medical College of Yangzhou University, Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou 225009, China.
| |
Collapse
|
11
|
Chen J, Wang C, Pan X, Zhan Y, Zhou W, Peng S, Chen C, Zhang M, Lan R, Wu J, Huang F, Hong J. Glycyrrhetinic Acid Mitigates Radiation-Induced Pulmonary Fibrosis via Inhibiting the Secretion of TGF-β1 by Treg Cells. Int J Radiat Oncol Biol Phys 2024; 118:218-230. [PMID: 37586613 DOI: 10.1016/j.ijrobp.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/18/2023]
Abstract
PURPOSE Radiation-induced pulmonary fibrosis (RIPF) is a common side effect of radiation therapy for thoracic tumors without effective prevention and treatment methods at present. The aim of this study was to explore whether glycyrrhetinic acid (GA) has a protective effect on RIPF and the underlying mechanism. METHODS AND MATERIALS A RIPF mouse model administered GA was used to determine the effect of GA on RIPF. The cocultivation of regulatory T (Treg) cells with mouse lung epithelial-12 cells or mouse embryonic fibroblasts and intervention with GA or transforming growth factor-β1 (TGF-β1) inhibitor to block TGF-β1 was conducted to study the mechanism by which GA alleviates RIPF. Furthermore, injection of Treg cells into GA-treated RIPF mice to upregulate TGF-β1 levels was performed to verify the roles of TGF-β1 and Treg cells. RESULTS GA intervention improved the damage to lung tissue structure and collagen deposition and inhibited Treg cell infiltration, TGF-β1 levels, epithelial mesenchymal transition (EMT), and myofibroblast (MFB) transformation in mice after irradiation. Treg cell-induced EMT and MFB transformation in vitro were prevented by GA, as well as a TGF-β1 inhibitor, by decreasing TGF-β1. Furthermore, reinfusion of Treg cells upregulated TGF-β1 levels and exacerbated RIPF in GA-treated RIPF mice. CONCLUSIONS GA can improve RIPF in mice, and the corresponding mechanisms may be related to the inhibition of TGF-β1 secreted by Treg cells to induce EMT and MFB transformation. Therefore, GA may be a promising therapeutic candidate for the clinical treatment of RIPF.
Collapse
Affiliation(s)
- Jinmei Chen
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Caihong Wang
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaoxian Pan
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yuping Zhan
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Weitong Zhou
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Shaoli Peng
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Chun Chen
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Mingwei Zhang
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ruilong Lan
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Central Lab, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jiandong Wu
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Fei Huang
- Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Central Lab, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
| | - Jinsheng Hong
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
12
|
Yu Z, Xu C, Song B, Zhang S, Chen C, Li C, Zhang S. Tissue fibrosis induced by radiotherapy: current understanding of the molecular mechanisms, diagnosis and therapeutic advances. J Transl Med 2023; 21:708. [PMID: 37814303 PMCID: PMC10563272 DOI: 10.1186/s12967-023-04554-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 09/22/2023] [Indexed: 10/11/2023] Open
Abstract
Cancer remains the leading cause of death around the world. In cancer treatment, over 50% of cancer patients receive radiotherapy alone or in multimodal combinations with other therapies. One of the adverse consequences after radiation exposure is the occurrence of radiation-induced tissue fibrosis (RIF), which is characterized by the abnormal activation of myofibroblasts and the excessive accumulation of extracellular matrix. This phenotype can manifest in multiple organs, such as lung, skin, liver and kidney. In-depth studies on the mechanisms of radiation-induced fibrosis have shown that a variety of extracellular signals such as immune cells and abnormal release of cytokines, and intracellular signals such as cGAS/STING, oxidative stress response, metabolic reprogramming and proteasome pathway activation are involved in the activation of myofibroblasts. Tissue fibrosis is extremely harmful to patients' health and requires early diagnosis. In addition to traditional serum markers, histologic and imaging tests, the diagnostic potential of nuclear medicine techniques is emerging. Anti-inflammatory and antioxidant therapies are the traditional treatments for radiation-induced fibrosis. Recently, some promising therapeutic strategies have emerged, such as stem cell therapy and targeted therapies. However, incomplete knowledge of the mechanisms hinders the treatment of this disease. Here, we also highlight the potential mechanistic, diagnostic and therapeutic directions of radiation-induced fibrosis.
Collapse
Affiliation(s)
- Zuxiang Yu
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Chaoyu Xu
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Bin Song
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang, 621099, China
| | - Shihao Zhang
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Chong Chen
- Department of Gastroenterology, The First People's Hospital of Xuzhou, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221200, China
| | - Changlong Li
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China.
- Department of Molecular Biology and Biochemistry, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China.
| | - Shuyu Zhang
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, China.
- The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, 610051, China.
- NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang, 621099, China.
| |
Collapse
|
13
|
Pan X, Wang C, Zhan Y, Chen J, Wang Z, Lan R, Chen J, Zhang W, Chen C, Zhang M, Huang F, Hong J. A Subset of Breg Cells, B10, Contributes to the Development of Radiation-Induced Pulmonary Fibrosis. Int J Radiat Oncol Biol Phys 2023; 117:237-251. [PMID: 37054996 DOI: 10.1016/j.ijrobp.2023.03.077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 03/07/2023] [Accepted: 03/30/2023] [Indexed: 04/15/2023]
Abstract
PURPOSE Radiation-induced pulmonary fibrosis (RIPF) is a serious side effect of radiation therapy, but the underlying mechanisms are unknown. B10 cells, as negative B regulatory cells, play important roles in regulating inflammation and autoimmunity. However, the role of B10 cells in RIPF progression is unclear. The aim of this study was to determine the role of B10 cells in aggravating RIPF and the underlying mechanism. METHODS AND MATERIALS The role of B10 cells in RIPF was studied by constructing mouse models of RIPF and depleting B10 cells with an anti-CD22 antibody. The mechanism of B10 cells in RIPF was further explored through cocultivation of B10 cells and MLE-12 or NIH3T3 cells and administration of an interleukin (IL)-10 antibody to block IL-10. RESULTS B10 cell numbers increased significantly during the early stage in the RIPF mouse models compared with the controls. In addition, depleting B10 cells with the anti-CD22 antibody attenuated the development of lung fibrosis in mice. Subsequently, we confirmed that B10 cells induced epithelial-mesenchymal transition and the transformation of myofibroblasts via activation of STAT3 signaling in vitro. After blockade of IL-10, it was verified that IL-10 secreted by B10 cells mediates the epithelial-mesenchymal transition of myofibroblasts, thereby promoting RIPF. CONCLUSIONS Our study uncovers a novel role for IL-10-secreting B10 cells that could be a new target of research for relieving RIPF.
Collapse
Affiliation(s)
- Xiaoxian Pan
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical Universisty, Fuzhou 350212, China
| | - Caihong Wang
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical Universisty, Fuzhou 350212, China
| | - Yuping Zhan
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical Universisty, Fuzhou 350212, China
| | - Jinmei Chen
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical Universisty, Fuzhou 350212, China; Key Laboratory of Radiation Biology of Fujian higher education institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Zeng Wang
- Key Laboratory of Radiation Biology of Fujian higher education institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Central Lab, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Ruilong Lan
- Provincial Key Laboratory of Precision Medicine for Cancer, Fuzhou 350005, China; Central Lab, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Junying Chen
- Key Laboratory of Radiation Biology of Fujian higher education institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Central Lab, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Weijian Zhang
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical Universisty, Fuzhou 350212, China; Key Laboratory of Radiation Biology of Fujian higher education institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Chun Chen
- School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Mingwei Zhang
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical Universisty, Fuzhou 350212, China; Key Laboratory of Radiation Biology of Fujian higher education institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Fei Huang
- Provincial Key Laboratory of Precision Medicine for Cancer, Fuzhou 350005, China; Central Lab, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China.
| | - Jinsheng Hong
- Department of Radiotherapy, Cancer Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical Universisty, Fuzhou 350212, China; Key Laboratory of Radiation Biology of Fujian higher education institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China.
| |
Collapse
|
14
|
Zhang YC, Zhang YT, Wang Y, Zhao Y, He LJ. What role does PDL1 play in EMT changes in tumors and fibrosis? Front Immunol 2023; 14:1226038. [PMID: 37649487 PMCID: PMC10463740 DOI: 10.3389/fimmu.2023.1226038] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 07/28/2023] [Indexed: 09/01/2023] Open
Abstract
Epithelial-mesenchymal transformation (EMT) plays a pivotal role in embryonic development, tissue fibrosis, repair, and tumor invasiveness. Emerging studies have highlighted the close association between EMT and immune checkpoint molecules, particularly programmed cell death ligand 1 (PDL1). PDL1 exerts its influence on EMT through bidirectional regulation. EMT-associated factors, such as YB1, enhance PDL1 expression by directly binding to its promoter. Conversely, PDL1 signaling triggers downstream pathways like PI3K/AKT and MAPK, promoting EMT and facilitating cancer cell migration and invasion. Targeting PDL1 holds promise as a therapeutic strategy for EMT-related diseases, including cancer and fibrosis. Indeed, PDL1 inhibitors, such as pembrolizumab and nivolumab, have shown promising results in clinical trials for various cancers. Recent research has also indicated their potential benefit in fibrosis treatment in reducing fibroblast activation and extracellular matrix deposition, thereby addressing fibrosis. In this review, we examine the multifaceted role of PDL1 in immunomodulation, growth, and fibrosis promotion. We discuss the challenges, mechanisms, and clinical observations related to PDL1, including the limitations of the PD1/PDL1 axis in treatment and PD1-independent intrinsic PDL1 signaling. Our study highlights the dynamic changes in PDL1 expression during the EMT process across various tumor types. Through interplay between PDL1 and EMT, we uncover co-directional alterations, regulatory pathways, and diverse changes resulting from PDL1 intervention in oncology. Additionally, our findings emphasize the dual role of PDL1 in promoting fibrosis and modulating immune responses across multiple diseases, with potential implications for therapeutic approaches. We particularly investigate the therapeutic potential of targeting PDL1 in type II EMT fibrosis: strike balance between fibrosis modulation and immune response regulation. This analysis provides valuable insights into the multifaceted functions of PDL1 and contributes to our understanding of its complex mechanisms and therapeutic implications.
Collapse
Affiliation(s)
- Yun-Chao Zhang
- Department of Nephrology, Xi Jing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yu-Ting Zhang
- Department of Nephrology, Xi Jing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yi Wang
- Department of Nephrology, Xi Jing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ya Zhao
- Department of Medical Microbiology and Parasitology, Fourth Military Medical University, Xi'an, China
| | - Li-Jie He
- Department of Nephrology, Xi Jing Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
15
|
Galluzzi L, Aryankalayil MJ, Coleman CN, Formenti SC. Emerging evidence for adapting radiotherapy to immunotherapy. Nat Rev Clin Oncol 2023:10.1038/s41571-023-00782-x. [PMID: 37280366 DOI: 10.1038/s41571-023-00782-x] [Citation(s) in RCA: 147] [Impact Index Per Article: 73.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2023] [Indexed: 06/08/2023]
Abstract
Immunotherapy has revolutionized the clinical management of many malignancies but is infrequently associated with durable objective responses when used as a standalone treatment approach, calling for the development of combinatorial regimens with superior efficacy and acceptable toxicity. Radiotherapy, the most commonly used oncological treatment, has attracted considerable attention as a combination partner for immunotherapy owing to its well-known and predictable safety profile, widespread clinical availability, and potential for immunostimulatory effects. However, numerous randomized clinical trials investigating radiotherapy-immunotherapy combinations have failed to demonstrate a therapeutic benefit compared with either modality alone. Such a lack of interaction might reflect suboptimal study design, choice of end points and/or administration of radiotherapy according to standard schedules and target volumes. Indeed, radiotherapy has empirically evolved towards radiation doses and fields that enable maximal cancer cell killing with manageable toxicity to healthy tissues, without much consideration of potential radiation-induced immunostimulatory effects. Herein, we propose the concept that successful radiotherapy-immunotherapy combinations might require modifications of standard radiotherapy regimens and target volumes to optimally sustain immune fitness and enhance the antitumour immune response in support of meaningful clinical benefits.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Molykutty J Aryankalayil
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - C Norman Coleman
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
16
|
zhao T, Wang L, Zhang Y, Ye W, Liu J, Wu H, Wang F, Tang T, Li Z. Qi-Dong-Huo-Xue-Yin balances the immune microenvironment to protect against LPS induced acute lung injury. Front Pharmacol 2023; 14:1200058. [PMID: 37292149 PMCID: PMC10244563 DOI: 10.3389/fphar.2023.1200058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/09/2023] [Indexed: 06/10/2023] Open
Abstract
COVID-19 induces acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) and leads to severe immunological changes that threatens the lives of COVID-19 victims. Studies have shown that both the regulatory T cells and macrophages were deranged in COVID-19-induced ALI. Herbal drugs have long been utilized to adjust the immune microenvironment in ALI. However, the underlying mechanisms of herbal drug mediated ALI protection are largely unknown. This study aims to understand the cellular mechanism of a traditional Chinese medicine, Qi-Dong-Huo-Xue-Yin (QD), in protecting against LPS induced acute lung injury in mouse models. Our data showed that QD intrinsically promotes Foxp3 transcription via promoting acetylation of the Foxp3 promoter in CD4+ T cells and consequently facilitates CD4+CD25+Foxp3+ Tregs development. Extrinsically, QD stabilized β-catenin in macrophages to expedite CD4+CD25+Foxp3+ Tregs development and modulated peripheral blood cytokines. Taken together, our results illustrate that QD promotes CD4+CD25+Foxp3+ Tregs development via intrinsic and extrinsic pathways and balanced cytokines within the lungs to protect against LPS induced ALI. This study suggests a potential application of QD in ALI related diseases.
Collapse
Affiliation(s)
- Tian zhao
- Department of Respiratory Medicine, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Le Wang
- The Second Clinical Medical College Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, China
| | - Yongjun Zhang
- The Cancer Hospital of the University of Chinese Academy of Sciences Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
| | - Wu Ye
- Department of Respiratory Medicine, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Juan Liu
- Department of Respiratory Medicine, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Haiyan Wu
- Department of Respiratory Medicine, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Fei Wang
- Department of Respiratory Medicine, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Tingyu Tang
- Department of Respiratory Medicine, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Zhijun Li
- Department of Respiratory Medicine, Zhejiang Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
17
|
Zhang J, Hu Z, Horta CA, Yang J. Regulation of epithelial-mesenchymal transition by tumor microenvironmental signals and its implication in cancer therapeutics. Semin Cancer Biol 2023; 88:46-66. [PMID: 36521737 DOI: 10.1016/j.semcancer.2022.12.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 12/06/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022]
Abstract
Epithelial-mesenchymal transition (EMT) has been implicated in various aspects of tumor development, including tumor invasion and metastasis, cancer stemness, and therapy resistance. Diverse stroma cell types along with biochemical and biophysical factors in the tumor microenvironment impinge on the EMT program to impact tumor progression. Here we provide an in-depth review of various tumor microenvironmental signals that regulate EMT in cancer. We discuss the molecular mechanisms underlying the role of EMT in therapy resistance and highlight new therapeutic approaches targeting the tumor microenvironment to impact EMT and tumor progression.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Zhimin Hu
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Calista A Horta
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Jing Yang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA; Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA.
| |
Collapse
|
18
|
Guo X, Du L, Ma N, Zhang P, Wang Y, Han Y, Huang X, Zhang Q, Tan X, Lei X, Qu B. Monophosphoryl lipid A ameliorates radiation-induced lung injury by promoting the polarization of macrophages to the M1 phenotype. J Transl Med 2022; 20:597. [DOI: 10.1186/s12967-022-03804-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
Abstract
Background
Radiation-induced lung injury (RILI) often occurs during clinical chest radiotherapy and acute irradiation from accidental nuclear leakage. This study explored the role of monophosphoryl lipid A (MPLA) in RILI.
Materials and Methods
The entire thoracic cavity of C57BL/6N mice was irradiated at 20 Gy with or without pre-intragastric administration of MPLA. HE staining, Masson trichrome staining, and TUNEL assay were used to assess lung tissue injury after treatment. The effect of irradiation on the proliferation of MLE-12 cells was analyzed using the Clonogenic assay. The effect of MPLA on the apoptosis of MLE-12 cells was analyzed using flow cytometry. Expression of γ-H2AX and epithelial-mesenchymal transition (EMT) markers in MLE-12 cells was detected by immunofluorescence and Western blot, respectively.
Results
MPLA attenuated early pneumonitis and late pulmonary fibrosis after thoracic irradiation and reversed radiation-induced EMT in C57 mice. MPLA further promoted proliferation and inhibited apoptosis of irradiated MLE-12 cells in vitro. Mechanistically, the radioprotective effect of MPLA was mediated by exosomes secreted by stimulated macrophages. Macrophage-derived exosomes modulated DNA damage in MLE-12 cells after irradiation. MPLA promoted the polarization of RAW 264.7 cells to the M1 phenotype. The exosomes secreted by M1 macrophages suppressed EMT in MLE-12 cells after irradiation.
Conclusion
MPLA is a novel treatment strategy for RILI. Exosomes derived from macrophages are key to the radioprotective role of MPLA in RILI.
Collapse
|
19
|
Mesenchymal Stem Cell-Derived Extracellular Vesicles as Idiopathic Pulmonary Fibrosis Microenvironment Targeted Delivery. Cells 2022; 11:cells11152322. [PMID: 35954166 PMCID: PMC9367455 DOI: 10.3390/cells11152322] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 02/05/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) affects an increasing number of people globally, yet treatment options remain limited. At present, conventional treatments depending on drug therapy do not show an ideal effect in reversing the lung damage or extending the lives of IPF patients. In recent years, more and more attention has focused on extracellular vesicles (EVs) which show extraordinary therapeutic effects in inflammation, fibrosis disease, and tissue damage repair in many kinds of disease therapy. More importantly, EVs can be modified or used as a drug or cytokine delivery tool, targeting injury sites to enhance treatment efficiency. In light of this, the treatment strategy of mesenchymal stem cell-extracellular vesicles (MSC-EVs) targeting the pulmonary microenvironment for IPF provides a new idea for the treatment of IPF. In this review, we summarized the inflammation, immune dysregulation, and extracellular matrix microenvironment (ECM) disorders in the IPF microenvironment in order to reveal the treatment strategy of MSC-EVs targeting the pulmonary microenvironment for IPF.
Collapse
|
20
|
Wang Y, Huang B, Jin T, Ocansey DKW, Jiang J, Mao F. Intestinal Fibrosis in Inflammatory Bowel Disease and the Prospects of Mesenchymal Stem Cell Therapy. Front Immunol 2022; 13:835005. [PMID: 35370998 PMCID: PMC8971815 DOI: 10.3389/fimmu.2022.835005] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
Intestinal fibrosis is an important complication of inflammatory bowel disease (IBD). In the course of the development of fibrosis, certain parts of the intestine become narrowed, significantly destroying the structure and function of the intestine and affecting the quality of life of patients. Chronic inflammation is an important initiating factor of fibrosis. Unfortunately, the existing anti-inflammatory drugs cannot effectively prevent and alleviate fibrosis, and there is no effective anti-fibrotic drug, which makes surgical treatment the mainstream treatment for intestinal fibrosis and stenosis. Mesenchymal stem cells (MSCs) are capable of tissue regeneration and repair through their self-differentiation, secretion of cytokines, and secretion of extracellular vesicles. MSCs have been shown to play an important therapeutic role in the fibrosis of many organs. However, the role of MSC in intestinal fibrosis largely remained unexplored. This review summarizes the mechanism of intestinal fibrosis, including the role of immune cells, TGF-β, and the gut microbiome and metabolites. Available treatment options for fibrosis, particularly, MSCs are also discussed.
Collapse
Affiliation(s)
- Yifei Wang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Bin Huang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- General Surgery Department, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
| | - Tao Jin
- Department of Gastrointestinal and Endoscopy, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
- Directorate of University Health Services, University of Cape Coast, Cape Coast, Ghana
| | - Jiajia Jiang
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
- *Correspondence: Jiajia Jiang, ; Fei Mao,
| | - Fei Mao
- Aoyang Institute of Cancer, Affiliated Aoyang Hospital of Jiangsu University, Suzhou, China
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
- *Correspondence: Jiajia Jiang, ; Fei Mao,
| |
Collapse
|
21
|
Jost T, Schultz AK, Frey B, Vu J, Fietkau R, Distel LV, Hecht M. Influence of alectinib and crizotinib on ionizing radiation - in vitro analysis of ALK/ROS1-wildtype lung tissue cells. Neoplasia 2022; 27:100780. [PMID: 35278911 PMCID: PMC8914391 DOI: 10.1016/j.neo.2022.100780] [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: 10/15/2021] [Revised: 02/06/2022] [Accepted: 02/18/2022] [Indexed: 10/31/2022]
Abstract
(1) Background: Just little is known about the interaction of ALK/ROS1-targeting kinase inhibitors with ionizing radiation (IR), particularly regarding side effects. We investigated the toxicity in two different lung cell lines both ALK/ROS1 wildtype (healthy and tumor origin) as representatives for normal lung tissue; (2) Methods: Human lung cell line BEAS-2B and malignant A549 lung cancer cells (ALK/ROS1 wt) were treated with alectinib or crizotinib, 2 Gy irradiation or a combination of KI and IR. Cell toxicity was analyzed by cell death (Annexin, 7AAD), colony forming, migration assay and live-cell imaging (TMRM, DRAQ7, Caspase3/7). Cell cycle (Hoechst) were analyzed by flow cytometry; (3) Results: Crizotinib led to higher cell death rates than alectinib, when cells were treated with 10 µM KI. Alectinib induced a more intense growth inhibition of colonies. Both inhibitors showed additive effects in combination with irradiation. Combination treatment (IR + KI) does not lead to synergistic effect on neither cell death nor colony forming; (4) Conclusions: The influence of simultaneous KI and IR was studied in non-mutated ALK/ROS1 cell lines. Both KIs seems to be well tolerated in combination with thoracic radiotherapy and lacked synergistic reinforcement in cellular toxicity. This supports the feasibility of ALK/ROS1 inhibition in combination with thoracic irradiation in future clinical trials.
Collapse
Affiliation(s)
- Tina Jost
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen 91054, Germany.
| | - Ann-Kristin Schultz
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany.
| | - Benjamin Frey
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen 91054, Germany; Deutsches Zentrum Immuntherapie, Erlangen, Germany.
| | - Jennifer Vu
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany.
| | - Rainer Fietkau
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen 91054, Germany.
| | - Luitpold V Distel
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen 91054, Germany.
| | - Markus Hecht
- Department of Radiation Oncology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen 91054, Germany.
| |
Collapse
|
22
|
Dai S, Wen Y, Luo P, Ma L, Liu Y, Ai J, Shi C. Therapeutic implications of exosomes in the treatment of radiation injury. BURNS & TRAUMA 2022; 10:tkab043. [PMID: 35071650 PMCID: PMC8778593 DOI: 10.1093/burnst/tkab043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/11/2021] [Indexed: 12/28/2022]
Abstract
Radiotherapy is one of the main cancer treatments, but it may damage normal tissue and cause various side effects. At present, radioprotective agents used in clinics have side effects such as nausea, vomiting, diarrhea and hypotension, which limit their clinical application. It has been found that exosomes play an indispensable role in radiation injury. Exosomes are lipid bilayer vesicles that carry various bioactive substances, such as proteins, lipids and microRNA (miRNA), that play a key role in cell-to-cell communication and affect tissue injury and repair. In addition, studies have shown that radiation can increase the uptake of exosomes in cells and affect the composition and secretion of exosomes. Here, we review the existing studies and discuss the effects of radiation on exosomes and the role of exosomes in radiation injury, aiming to provide new insights for the treatment of radiation injury.
Collapse
Affiliation(s)
| | | | | | | | | | - Junhua Ai
- Correspondence. Junhua Ai, ; Chunmeng Shi,
| | | |
Collapse
|
23
|
Kobayashi S, Nagafuchi Y, Okubo M, Sugimori Y, Hatano H, Yamada S, Nakano M, Yoshida R, Takeshima Y, Ota M, Tsuchida Y, Iwasaki Y, Setoguchi K, Kubo K, Okamura T, Yamamoto K, Shoda H, Fujio K. Dysregulation of the gene signature of effector regulatory T cells in the early phase of systemic sclerosis. Rheumatology (Oxford) 2022; 61:4163-4174. [PMID: 35040949 DOI: 10.1093/rheumatology/keac031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 01/11/2022] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES We evaluated flow-cytometric and transcriptome features of peripheral blood immune cells from early-phase (disease duration < 5 years) systemic sclerosis (SSc) in comparison to late-phase SSc. METHODS Fifty Japanese patients with SSc (12 early SSc cases and 38 late SSc cases) and 50 age- and sex-matched healthy controls were enrolled. A comparison of flow-cytometric subset proportions and RNA-sequencing of 24 peripheral blood immune cell subsets was performed. We evaluated differentially expressed genes (DEGs), characterized the co-expressed gene modules, and estimated the composition of subpopulations by deconvolution based on single-cell RNA-sequencing data. As a disease control, idiopathic inflammatory myositis (IIM) patients were also evaluated. RESULTS Analyzing the data from early and late SSc, Fraction II effector regulatory T cell (Fr. II eTreg) genes showed a remarkable differential gene expression, which was enriched for genes related to oxidative phosphorylation. Although the flow-cytometric proportion of Fr. II eTregs was not changed in early SSc, deconvolution indicated expansion of the activated subpopulation. Co-expressed gene modules of Fr. II eTregs demonstrated enrichment of the DEGs of early SSc and correlation with the proportion of the activated subpopulation. These results suggested that DEGs in Fr. II eTregs from patients with early SSc were closely associated with the increased proportion of the activated subpopulation. Similar dysregulation of Fr. II eTregs was also observed in data from patients with early IIM. CONCLUSIONS RNA-seq of immune cells indicated the dysregulation of Fr. II eTregs in early SSc with increased proportion of the activated subpopulation.
Collapse
Affiliation(s)
- Satomi Kobayashi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo.,Department of Medicine and Rheumatology, Tokyo Metropolitan Geriatric Hospital, Japan. 35-2 Sakaechou, Itabashi-ku, 173-0015, Japan, Tokyo, Tokyo
| | - Yasuo Nagafuchi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo.,Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Mai Okubo
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo
| | - Yusuke Sugimori
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo
| | - Hiroaki Hatano
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo
| | - Saeko Yamada
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo
| | - Masahiro Nakano
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo
| | - Ryochi Yoshida
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo
| | - Yusuke Takeshima
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo
| | - Mineto Ota
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo.,Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Yumi Tsuchida
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo
| | - Yukiko Iwasaki
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo
| | - Keigo Setoguchi
- Department of Rheumatology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Japan. 3-18-22 Honkomagome, Bunkyo-ku, 113-8677, Japan, Tokyo, Tokyo
| | - Kanae Kubo
- Department of Medicine and Rheumatology, Tokyo Metropolitan Geriatric Hospital, Japan. 35-2 Sakaechou, Itabashi-ku, 173-0015, Japan, Tokyo, Tokyo
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo.,Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kazuhiko Yamamoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo.,Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Japan. 1-7-22 Suehiro-cho, Tsurumi-ku, Kanagawa, 230-0045, Japan, Yokohama, Yokohama
| | - Hirofumi Shoda
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Japan. 7-3-1 Hongo, Bunkyo-ku, 113-8655, Japan, Tokyo, Tokyo
| |
Collapse
|
24
|
Bertho A, Dos Santos M, Braga-Cohen S, Buard V, Paget V, Guipaud O, Tarlet G, Milliat F, François A. Preclinical Model of Stereotactic Ablative Lung Irradiation Using Arc Delivery in the Mouse: Is Fractionation Worthwhile? Front Med (Lausanne) 2022; 8:794324. [PMID: 35004768 PMCID: PMC8739220 DOI: 10.3389/fmed.2021.794324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/07/2021] [Indexed: 11/23/2022] Open
Abstract
Lung stereotactic body radiation therapy is characterized by a reduction in target volumes and the use of severely hypofractionated schedules. Preclinical modeling became possible thanks to rodent-dedicated irradiation devices allowing accurate beam collimation and focal lung exposure. Given that a great majority of publications use single dose exposures, the question we asked in this study was as follows: in incremented preclinical models, is it worth using fractionated protocols or should we continue focusing solely on volume limitation? The left lungs of C57BL/6JRj mice were exposed to ionizing radiation using arc therapy and 3 × 3 mm beam collimation. Three-fraction schedules delivered over a period of 1 week were used with 20, 28, 40, and 50 Gy doses per fraction. Lung tissue opacification, global histological damage and the numbers of type II pneumocytes and club cells were assessed 6 months post-exposure, together with the gene expression of several lung cells and inflammation markers. Only the administration of 3 × 40 Gy or 3 × 50 Gy generated focal lung fibrosis after 6 months, with tissue opacification visible by cone beam computed tomography, tissue scarring and consolidation, decreased club cell numbers and a reactive increase in the number of type II pneumocytes. A fractionation schedule using an arc-therapy-delivered three fractions/1 week regimen with 3 × 3 mm beam requires 40 Gy per fraction for lung fibrosis to develop within 6 months, a reasonable time lapse given the mouse lifespan. A comparison with previously published laboratory data suggests that, in this focal lung irradiation configuration, administering a Biological Effective Dose ≥ 1000 Gy should be recommended to obtain lung fibrosis within 6 months. The need for such a high dose per fraction challenges the appropriateness of using preclinical highly focused fractionation schedules in mice.
Collapse
Affiliation(s)
- Annaïg Bertho
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Morgane Dos Santos
- Laboratory of Radiobiology of Accidental Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Sarah Braga-Cohen
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Valérie Buard
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Vincent Paget
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Olivier Guipaud
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Georges Tarlet
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Fabien Milliat
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| | - Agnès François
- Laboratory of Radiobiology of Medical Exposures, Institute for Radioprotection and Nuclear Safety (IRSN), Research Department in Radiobiology and Regenerative Medicine, Fontenay-aux-Roses, France
| |
Collapse
|
25
|
Scott EN, Gocher AM, Workman CJ, Vignali DAA. Regulatory T Cells: Barriers of Immune Infiltration Into the Tumor Microenvironment. Front Immunol 2021; 12:702726. [PMID: 34177968 PMCID: PMC8222776 DOI: 10.3389/fimmu.2021.702726] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
Regulatory T cells (Tregs) are key immunosuppressive cells that promote tumor growth by hindering the effector immune response. Tregs utilize multiple suppressive mechanisms to inhibit pro-inflammatory responses within the tumor microenvironment (TME) by inhibition of effector function and immune cell migration, secretion of inhibitory cytokines, metabolic disruption and promotion of metastasis. In turn, Tregs are being targeted in the clinic either alone or in combination with other immunotherapies, in efforts to overcome the immunosuppressive TME and increase anti-tumor effects. However, it is now appreciated that Tregs not only suppress cells intratumorally via direct engagement, but also serve as key interactors in the peritumor, stroma, vasculature and lymphatics to limit anti-tumor immune responses prior to tumor infiltration. We will review the suppressive mechanisms that Tregs utilize to alter immune and non-immune cells outside and within the TME and discuss how these mechanisms collectively allow Tregs to create and promote a physical and biological barrier, resulting in an immune-excluded or limited tumor microenvironment.
Collapse
Affiliation(s)
- Ellen N. Scott
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Angela M. Gocher
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
| | - Creg J. Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
| | - Dario A. A. Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, PA, United States
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| |
Collapse
|
26
|
Wang D, Liu Z, Yan Z, Liang X, Liu X, Liu Y, Wang P, Bai C, Gu Y, Zhou PK. MiRNA-155-5p inhibits epithelium-to-mesenchymal transition (EMT) by targeting GSK-3β during radiation-induced pulmonary fibrosis. Arch Biochem Biophys 2020; 697:108699. [PMID: 33259794 DOI: 10.1016/j.abb.2020.108699] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023]
Abstract
Radiation-induced pulmonary fibrosis (RIPF) is a major lung complication in using radiotherapy to treat thoracic diseases. MicroRNAs (miRNAs) are reported to be the therapeutic targets for many diseases. However, the miRNAs involved in the pathogenesis of RIPF are rarely studied as potential therapeutic targets. Alveolar epithelial cells participate in RIPF formation by undergoing epithelial-mesenchymal transition (EMT). Here we demonstrated the critical role of miR-155-5p in radiation-induced EMT and RIPF. Using the previously established EMT cell model, we found that miR-155-5p was significantly down-regulated through high-throughput sequencing. Irradiation could decrease the expression of miR-155-5p in intro and in vivo, and it was inversely correlated to RIPF formation. Ectopic miR-155-5p expression inhibited radiation-induced-EMT in vitro and in vivo. Knockdown of glycogen synthase kinase-3β (GSK-3β), the functional target of miR-155-5p, reversed the induction of EMT and enhanced the phosphorylation of p65, a subunit of NF-κB, which were mediated by the down-regulation of miR-155-5p. Moreover, our finding demonstrated that ectopic miR-155-5p expression alleviated RIPF in mice by the GSK-3β/NF-κB pathway. Thus, radiation downregulates miR-155-5p in alveolar epithelial cells that induces EMT, which contributes to RIPF using GSK-3β/NF-κB pathway. Our observation provides further understanding on the regulation of RIPF and identifies potential therapeutic targets.
Collapse
Affiliation(s)
- Duo Wang
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Zheng Liu
- School of Public Health, University of South China, Hengyang, Hunan Province, 421001, PR China
| | - Ziyan Yan
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Xinxin Liang
- School of Public Health, University of South China, Hengyang, Hunan Province, 421001, PR China
| | - Xiaochang Liu
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China; School of Public Health, University of South China, Hengyang, Hunan Province, 421001, PR China
| | - Yuhao Liu
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Ping Wang
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Chenjun Bai
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Yongqing Gu
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China; School of Public Health, University of South China, Hengyang, Hunan Province, 421001, PR China.
| | - Ping-Kun Zhou
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, PR China.
| |
Collapse
|
27
|
Li L, Nie X, Yi M, Qin W, Li F, Wu B, Yuan X. Aerosolized Thyroid Hormone Prevents Radiation Induced Lung Fibrosis. Front Oncol 2020; 10:528686. [PMID: 33042829 PMCID: PMC7523090 DOI: 10.3389/fonc.2020.528686] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 08/20/2020] [Indexed: 12/09/2022] Open
|
28
|
Guo T, Zou L, Ni J, Zhou Y, Ye L, Yang X, Zhu Z. Regulatory T Cells: An Emerging Player in Radiation-Induced Lung Injury. Front Immunol 2020; 11:1769. [PMID: 32849634 PMCID: PMC7417370 DOI: 10.3389/fimmu.2020.01769] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/01/2020] [Indexed: 12/25/2022] Open
Abstract
Regulatory T cells (Tregs), which have long been recognized as essential regulators of both inflammation and autoimmunity, also impede effective antitumor immune response due to their immunosuppressive properties. Combined radiotherapy and immunotherapeutic interventions focusing on the removal of Tregs have recently garnered interest as a promising strategy to reverse immunosuppression. Meanwhile, Tregs are emerging as a key player in the pathogenesis of radiation-induced lung injury (RILI), a frequent and potentially life-threatening complication of thoracic radiotherapy. Recognition of the critical role of Tregs in RILI raises the important question of whether radiotherapy combined with Treg-targeting immunotherapy offers any beneficial effects in the protection of normal lung tissue. This present review focuses on the contributions of Tregs to RILI, with particular emphasis on the suspected differential role of Tregs in the pneumonitic phase and fibrotic phase of RILI. We also introduce recent progress on the potential mechanisms by which Tregs modulate RILI and the crosstalk among Tregs, other infiltrating T cells, fibrocytes, and resident epithelial cells driving disease pathogenesis. Finally, we discuss whether Tregs also hold promise as a potential target for immunotherapeutic interventions for RILI.
Collapse
Affiliation(s)
- Tiantian Guo
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liqing Zou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jianjiao Ni
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yue Zhou
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Luxi Ye
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xi Yang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhengfei Zhu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
29
|
Regulatory T cells are a double-edged sword in pulmonary fibrosis. Int Immunopharmacol 2020; 84:106443. [PMID: 32334385 DOI: 10.1016/j.intimp.2020.106443] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/23/2020] [Accepted: 03/23/2020] [Indexed: 02/06/2023]
Abstract
Pulmonary fibrosis (PF) is a chronic progressive interstitial lung disease. The pathogenesis of PF has not been clearly elucidated, and there is no obvious effective treatment to arrest the progression of PF to date. A long-term chronic inflammatory response and inappropriate repair process after lung injury are important causes and pathological processes of PF. As an influential type of the body's immune cells, regulatory T cells (Tregs) play an irreplaceable role in inhibiting the inflammatory response and promoting the repair of lung tissue. However, the exact roles of Tregs in the process of PF have not been clearly established, and the available literature concerning the roles of Tregs in PF are contradictory. First, Tregs can advance the progression of pulmonary fibrosis by secreting platelet-derived growth factor (PDGF), transforming growth factor-β (TGF-β) and other related factors, promoting epithelial-mesenchymal transition (EMT) and affecting the Th1 and Th2 balance, etc. Second, Tregs can inhibit PF by promoting the repair of epithelial cell damage, inhibiting the accumulation of fibroblasts, and strongly inhibiting the production and function of other related pro-inflammatory factors and pro-inflammatory cells. Accordingly, in this review, we focus on the multiple roles of Tregs in different models and different pulmonary fibrosis phases, thereby providing theoretical support for a better understanding of the multiple roles of these cells in PF and a theoretical basis for identifying targets for PF therapy.
Collapse
|
30
|
Expression Analysis of the Mediators of Epithelial to Mesenchymal Transition and Early Risk Assessment of Therapeutic Failure in Laryngeal Carcinoma. JOURNAL OF ONCOLOGY 2019; 2019:5649846. [PMID: 31885577 PMCID: PMC6926423 DOI: 10.1155/2019/5649846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 07/26/2019] [Accepted: 08/10/2019] [Indexed: 02/08/2023]
Abstract
Laryngeal squamous cell carcinoma (LSCC) is an aggressive malignancy which lacks early predictors of prognosis. Here, we hypothesized that expression and prognostic characterization of the critical mediators of epithelial to mesenchymal transition (EMT) may provide key information in this regard. Linear regression and multiple correspondence analyses were performed on immunohistochemical data obtained from 20 invasive tumors. Principal component and unsupervised hierarchical clustering were used to analyze the dataset patterns associating with LSCC metastatic profile. Survival and death risk assessments were performed using Kaplan–Meier and hazard ratio tests. Data mining analysis using CHAID decision tree and logistic regression analysis was applied to define the predictive value of the risk factors of tumor aggressiveness. Our analyses showed, that in invasive LSCC tumors, cells associating with a mesenchymal profile were likely to exhibit enhanced NOS2, TGF-β, and IL-17A expression levels, concomitantly to NF-κB nuclear translocation. IHC data deciphering determined that EMT induction was also linked to the enrichment of the tumors with CD68+ populations and IL-10 signal. Strikingly, dataset cluster analysis showed that these signatures could define distinct patterns of invasive tumors, where NOS2 associated with IL-10 expression, and TGF-β and IL-17A signals associated with MMP-9 activation. Decision tree analysis identified IL-17A as a possible predictor of LSCC aggressiveness. Altogether, our results show that distinct immunological patterns would support the acquisition of EMT features in invasive LSCC and suggest that IL-17A may be useful in the early identification of patients “at-risk” of therapeutic failure.
Collapse
|
31
|
Paluskievicz CM, Cao X, Abdi R, Zheng P, Liu Y, Bromberg JS. T Regulatory Cells and Priming the Suppressive Tumor Microenvironment. Front Immunol 2019; 10:2453. [PMID: 31681327 PMCID: PMC6803384 DOI: 10.3389/fimmu.2019.02453] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 10/01/2019] [Indexed: 12/20/2022] Open
Abstract
Treg play a central role in maintenance of self tolerance and homeostasis through suppression of self-reactive T cell populations. In addition to that role, Treg also survey cancers and suppress anti-tumor immune responses. Thus, understanding the unique attributes of Treg-tumor interactions may permit control of this pathologic suppression without interfering with homeostatic self-tolerance. This review will define the unique role of Treg in cancer growth, and the ways by which Treg inhibit a robust anti-tumor immune response. There will be specific focus placed on Treg homing to the tumor microenvironment (TME), TME formation of induced Treg (iTreg), mechanisms of suppression that underpin cancer immune escape, and trophic nonimmunologic effects of Treg on tumor cells.
Collapse
Affiliation(s)
| | - Xuefang Cao
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Reza Abdi
- Division of Renal Medicine, Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Pan Zheng
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Yang Liu
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jonathan S. Bromberg
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
32
|
Gong L, Wu X, Li X, Ni X, Gu W, Wang X, Ji H, Hu L, Zhu L. S1PR3 deficiency alleviates radiation-induced pulmonary fibrosis through the regulation of epithelial-mesenchymal transition by targeting miR-495-3p. J Cell Physiol 2019; 235:2310-2324. [PMID: 31489649 DOI: 10.1002/jcp.29138] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 08/23/2019] [Indexed: 12/22/2022]
Abstract
Radiation-induced pulmonary fibrosis (RIPF) is a life-threatening complication of thoracic radiotherapy, which contributes to continued deterioration in pulmonary function. Sphingosine-1 phosphate receptor 3 (S1PR3) has been identified as a crucial molecule in fibrosis. Accumulating evidence indicated that the inhibition of the S1PRs ameliorates fibrogenesis. Thus, this study aims to explore whether S1PR3 participates in RIPF and elucidates the molecular mechanisms underlying S1PR3-modulated epithelial-mesenchymal transition (EMT) in transforming growth factor-β1-induced pulmonary epithelia. A recombinant adeno-associated viral-mediated S1PR3 (AAV-S1PR3) gene therapy analyzed the effect of S1PR3 gene deficiency on the altered histology structure and molecular mechanisms in the lung of mice with whole-lung irradiation. Compared with the AAV-negative control mice, AAV-mediated S1PR3 knockdown in the lung of mice attenuated pulmonary fibrosis induced by the radiation, as indicated by the alleviation of collagen accumulation, lessened histopathological alterations, and the suppression of inflammatory cells infiltration. S1PR3 deficiency reversed the RIPF concomitantly with abrogated EMT-related protein (α-smooth muscle actin). Consistently, S1PR3-deficient pulmonary epithelia inhibited the EMT process changes and fibrosis formation. Furthermore, S1PR3 was designated as one of the target genes for microRNA-495-3p (miR-495-3p). The inhibition of miR-495-3p promoted the expression of S1PR3 in pulmonary epithelia, whereas the overexpression of miR-495-3p inhibited the S1PR3/SMAD2/3 pathway and suppressed the EMT process. Collectively, miR-495-3p might be a negative regulator of the EMT process in fibrosis formation by inhibiting the targeted S1PR3 gene. These results established a link between the S1PR3 gene, the EMT process, and the fibrosis, suggesting the pharmacological blockage of S1PR3 as a potential therapeutic strategy for RIPF.
Collapse
Affiliation(s)
- Linjing Gong
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xu Wu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinyi Li
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoying Ni
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenyu Gu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xinyuan Wang
- Department of Orthopaedics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haiying Ji
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lijuan Hu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lei Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
33
|
Landskron G, De la Fuente López M, Dubois-Camacho K, Díaz-Jiménez D, Orellana-Serradell O, Romero D, Sepúlveda SA, Salazar C, Parada-Venegas D, Quera R, Simian D, González MJ, López-Köstner F, Kronberg U, Abedrapo M, Gallegos I, Contreras HR, Peña C, Díaz-Araya G, Roa JC, Hermoso MA. Interleukin 33/ST2 Axis Components Are Associated to Desmoplasia, a Metastasis-Related Factor in Colorectal Cancer. Front Immunol 2019; 10:1394. [PMID: 31281317 PMCID: PMC6598075 DOI: 10.3389/fimmu.2019.01394] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 06/03/2019] [Indexed: 12/24/2022] Open
Abstract
In colorectal cancer (CRC), cancer-associated fibroblasts (CAFs) are the most abundant component from the tumor microenvironment (TM). CAFs facilitate tumor progression by inducing angiogenesis, immune suppression and invasion, thus altering the organization/composition of the extracellular matrix (i.e., desmoplasia) and/or activating epithelial-mesenchymal transition (EMT). Soluble factors from the TM can also contribute to cell invasion through secretion of cytokines and recently, IL-33/ST2 pathway has gained huge interest as a protumor alarmin, promoting progression to metastasis by inducing changes in TM. Hence, we analyzed IL-33 and ST2 content in tumor and healthy tissue lysates and plasma from CRC patients. Tissue localization and distribution of these molecules was evaluated by immunohistochemistry (using localization reference markers α-smooth muscle actin or α-SMA and E-cadherin), and clinical/histopathological information was obtained from CRC patients. In vitro experiments were conducted in primary cultures of CAFs and normal fibroblasts (NFs) isolated from tumor and healthy tissue taken from CRC patients. Additionally, migration and proliferation analysis were performed in HT29 and HCT116 cell lines. It was found that IL-33 content increases in left-sided CRC patients with lymphatic metastasis, with localization in tumor epithelia associated with abundant desmoplasia. Although ST2 content showed similarities between tumor and healthy tissue, a decreased immunoreactivity was observed in left-sided tumor stroma, associated to metastasis related factors (advanced stages, abundant desmoplasia, and presence of tumor budding). A principal component analysis (including stromal and epithelial IL-33/ST2 and α-SMA immunoreactivity with extent of desmoplasia) allowed us to distinguish clusters of low, intermediate and abundant desmoplasia, with potential to develop a diagnostic signature with benefits for further therapeutic targets. IL-33 transcript levels from CAFs directly correlated with CRC cell line migration induced by CAFs conditioned media, with rhIL-33 inducing a mesenchymal phenotype in HT29 cells. These results indicate a role of IL-33/ST2 in tumor microenvironment, specifically in the interaction between CAFs and epithelial tumor cells, thus contributing to invasion and metastasis in left-sided CRC, most likely by activating desmoplasia.
Collapse
Affiliation(s)
- Glauben Landskron
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| | - Marjorie De la Fuente López
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile.,Research Sub-direction, Academic Direction, Clinica Las Condes, Santiago, Chile
| | - Karen Dubois-Camacho
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| | - David Díaz-Jiménez
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| | - Octavio Orellana-Serradell
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| | - Diego Romero
- Pathology Department, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Santiago A Sepúlveda
- Pathology Department, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Christian Salazar
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| | - Daniela Parada-Venegas
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| | - Rodrigo Quera
- Inflammatory Bowel Disease Program, Gastroenterology Department, Clinica Las Condes, Santiago, Chile
| | - Daniela Simian
- Research Sub-direction, Academic Direction, Clinica Las Condes, Santiago, Chile
| | - María-Julieta González
- Cell and Molecular Biology Program, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | | | - Udo Kronberg
- Coloproctology Department, Clinica Las Condes, Santiago, Chile
| | - Mario Abedrapo
- Coloproctology Department, Clinica Las Condes, Santiago, Chile.,Coloproctology Surgery Department, Hospital Clinico Universidad de Chile, Santiago, Chile
| | - Iván Gallegos
- Pathology Department, Hospital Clinico Universidad de Chile, Santiago, Chile
| | - Héctor R Contreras
- Department of Basic and Clinic Oncology, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Cristina Peña
- Medical Oncology Department, Ramon y Cajal University Hospital, IRYCIS, CIBERONC, Madrid, Spain
| | - Guillermo Díaz-Araya
- Molecular Pharmacology Laboratory, Faculty of Chemical Pharmaceutical Sciences, Universidad de Chile, Santiago, Chile
| | - Juan Carlos Roa
- Pathology Department, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Marcela A Hermoso
- Immunology Program, Innate Immunity Laboratory, Faculty of Medicine, Biomedical Sciences Institute, Universidad de Chile, Santiago, Chile
| |
Collapse
|
34
|
Singh S, Chakrabarti R. Consequences of EMT-Driven Changes in the Immune Microenvironment of Breast Cancer and Therapeutic Response of Cancer Cells. J Clin Med 2019; 8:jcm8050642. [PMID: 31075939 PMCID: PMC6572359 DOI: 10.3390/jcm8050642] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/30/2019] [Accepted: 05/04/2019] [Indexed: 02/06/2023] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is a process through which epithelial cells lose their epithelial characteristics and cell–cell contact, thus increasing their invasive potential. In addition to its well-known roles in embryonic development, wound healing, and regeneration, EMT plays an important role in tumor progression and metastatic invasion. In breast cancer, EMT both increases the migratory capacity and invasive potential of tumor cells, and initiates protumorigenic alterations in the tumor microenvironment (TME). In particular, recent evidence has linked increased expression of EMT markers such as TWIST1 and MMPs in breast tumors with increased immune infiltration in the TME. These immune cells then provide cues that promote immune evasion by tumor cells, which is associated with enhanced tumor progression and metastasis. In the current review, we will summarize the current knowledge of the role of EMT in the biology of different subtypes of breast cancer. We will further explore the correlation between genetic switches leading to EMT and EMT-induced alterations within the TME that drive tumor growth and metastasis, as well as their possible effect on therapeutic response in breast cancer.
Collapse
Affiliation(s)
- Snahlata Singh
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Rumela Chakrabarti
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
35
|
Qu H, Liu L, Liu Z, Qin H, Liao Z, Xia P, Yang Y, Li B, Gao F, Cai J. Blocking TBK1 alleviated radiation-induced pulmonary fibrosis and epithelial-mesenchymal transition through Akt-Erk inactivation. Exp Mol Med 2019; 51:1-17. [PMID: 30988282 PMCID: PMC6465273 DOI: 10.1038/s12276-019-0240-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/25/2018] [Accepted: 12/11/2018] [Indexed: 12/23/2022] Open
Abstract
As a common serious complication of thoracic radiotherapy, radiation-induced pulmonary fibrosis (RIPF) severely limits radiation therapy approaches. Epithelial–mesenchymal transition (EMT) is a direct contributor to the fibroblast pool during fibrogenesis, and prevention of EMT is considered an effective strategy to inhibit tissue fibrosis. Our previous study revealed that TANK-binding kinase 1 (TBK1) regulates EMT in lung cancer cells. In the present study, we aimed to investigate the therapeutic potential of targeting TBK1 to prevent RIPF and EMT progression. We found radiation-induced EMT and pulmonary fibrosis in normal alveolar epithelial cells and lung tissues. TBK1 knockdown or inhibition significantly reversed EMT in vivo and in vitro and attenuated pulmonary fibrosis and collagen deposition. Moreover, we observed that TBK1 was elevated in a time- and dose-dependent manner by radiation. Meanwhile, radiation also induced time- and dose-dependent activation of AKT and ERK, each of whose inhibitors suppressed radiation-induced EMT. Intriguingly, silencing of TBK1 with shRNA also blocked the radiation-induced activation of AKT and ERK signaling. The ERK inhibitor did not obviously affect the expression of TBK1 or phosphorylated AKT, while AKT inhibition suppressed activation of ERK without changing the expression of TBK1. Finally, we found that a TBK1 inhibitor inhibited inflammatory cytokine expression in a RIPF model and Amlexanox protected normal cells and mice from ionizing radiation. In conclusion, our results indicate that the TBK1–AKT–ERK signaling pathway regulates radiation-induced EMT in normal alveolar epithelial cells, suggesting that TBK1 is a potential target for pulmonary fibrosis prevention during cancer radiotherapy. The risk of scarred tissues and respiratory distress during radiation treatment of lung cancer could be reduced by targeting an enzyme that alters healthy cells. Lung cancer radiotherapy often causes pulmonary fibrosis, excessive growth of fibrous tissues in the lungs, involving the transition of normal epithelial cells into an invasive form of multipotent stem cells. The development of pulmonary fibrosis limits the clinical application of radiotherapy. Hongjin Qu and co-workers at the Second Military University in Shanghai, China, previously demonstrated that the TANK-binding kinase 1 (TBK1) enzyme regulates this transition. Now, the team have shown that levels of TBK1 itself increased during radiation treatment, together with two proteins that would normally suppress alterations in healthy cells. Inhibiting TBK1, both in cell cultures and mouse models, reversed the cell transitions and prevented pulmonary fibrosis.
Collapse
Affiliation(s)
- Hongjin Qu
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, 800, Xiangyin Road, 200433, Shanghai, P. R. China
| | - Lei Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, 800, Xiangyin Road, 200433, Shanghai, P. R. China
| | - Zhe Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, 800, Xiangyin Road, 200433, Shanghai, P. R. China
| | - Hongran Qin
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, 800, Xiangyin Road, 200433, Shanghai, P. R. China
| | - Zebin Liao
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, 800, Xiangyin Road, 200433, Shanghai, P. R. China
| | - Penglin Xia
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, 800, Xiangyin Road, 200433, Shanghai, P. R. China
| | - Yanyong Yang
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, 800, Xiangyin Road, 200433, Shanghai, P. R. China
| | - Bailong Li
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, 800, Xiangyin Road, 200433, Shanghai, P. R. China.
| | - Fu Gao
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, 800, Xiangyin Road, 200433, Shanghai, P. R. China.
| | - Jianming Cai
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, 800, Xiangyin Road, 200433, Shanghai, P. R. China.
| |
Collapse
|
36
|
Liu Z, Liang X, Li X, Liu X, Zhu M, Gu Y, Zhou P. MiRNA-21 functions in ionizing radiation-induced epithelium-to-mesenchymal transition (EMT) by downregulating PTEN. Toxicol Res (Camb) 2019; 8:328-340. [PMID: 31160967 DOI: 10.1039/c9tx00019d] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 02/26/2019] [Indexed: 12/19/2022] Open
Abstract
Radiation-induced pulmonary fibrosis (RIPF) results from thoracic radiotherapy and severely limits the use of radiotherapy. Recent studies suggest that epithelium-to-mesenchymal transition (EMT) contributes to pulmonary fibrosis. Although miRNA dysregulation participates in a variety of pathophysiologic processes, their roles in fibrotic lung diseases and EMT are unclear. In this study, we aimed to identify key miRNAs involved in this process using a mouse model of RIPF previously established by irradiation with a single dose (20 Gy) of 60Co γ-rays. At 2-weeks post-irradiation, a set of significantly upregulated miRNAs was identified in lung tissue by miRNA array analysis. This included miR-21, which has been reported to contribute to the pulmonary fibrotic response induced by stereotactic body radiotherapy. Here, we showed that miR-21 expression increased in parallel with EMT progression in the lungs of irradiated mice. Ectopic miR-21 expression promoted EMT progression in lung epithelial cells. Furthermore, downregulation of miR-21 expression by transfection of its inhibitor inhibited ionizing radiation (IR)-induced EMT. Knockdown of PTEN, which is the functional target of miR-21, reversed the attenuation of IR-induced EMT mediated by miR-21 downregulation. Radiation treatment decreased PTEN expression and increased Akt phosphorylation; these effects were abolished by the miR-21 inhibitor. MiR-21 overexpression in lung epithelial cell also downregulated PTEN expression and upregulated Akt phosphorylation. In conclusion, we have demonstrated that miR-21 functions as a key regulator of IR-induced EMT in lung epithelial cells via the PTEN/Akt pathway. Targeting miR-21 is implicated as a novel therapeutic strategy for the prevention of RIPF.
Collapse
Affiliation(s)
- Zheng Liu
- School of Public Health , University of South China , Hengyang , Hunan Province 421001 , P. R. China . ; .,Beijing Key Laboratory for Radiobiology , Beijing Institute of Radiation Medicine , Beijing 100850 , P. R. China
| | - Xin Liang
- Graduate School , Anhui Medical University , Hefei , Anhui province 230032 , P. R. China
| | - Xueping Li
- School of Life Science , Shihezi University , Shihezi , Xinjiang Province 832003 , P. R. China
| | - Xiaodan Liu
- Beijing Key Laboratory for Radiobiology , Beijing Institute of Radiation Medicine , Beijing 100850 , P. R. China
| | - Maoxiang Zhu
- Beijing Key Laboratory for Radiobiology , Beijing Institute of Radiation Medicine , Beijing 100850 , P. R. China
| | - Yongqing Gu
- School of Public Health , University of South China , Hengyang , Hunan Province 421001 , P. R. China . ; .,Beijing Key Laboratory for Radiobiology , Beijing Institute of Radiation Medicine , Beijing 100850 , P. R. China
| | - Pingkun Zhou
- School of Public Health , University of South China , Hengyang , Hunan Province 421001 , P. R. China . ; .,Beijing Key Laboratory for Radiobiology , Beijing Institute of Radiation Medicine , Beijing 100850 , P. R. China
| |
Collapse
|
37
|
Chen Z, Wu Z, Ning W. Advances in Molecular Mechanisms and Treatment of Radiation-Induced Pulmonary Fibrosis. Transl Oncol 2019; 12:162-169. [PMID: 30342294 PMCID: PMC6197541 DOI: 10.1016/j.tranon.2018.09.009] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 09/11/2018] [Accepted: 09/11/2018] [Indexed: 02/06/2023] Open
Abstract
Radiation-induced pulmonary fibrosis (RIPF) is a common complication in patients with lung cancer and breast cancer after receiving thoracic radiotherapy. The average incidence of RIPF is 16%-28% after radiotherapy. RIPF includes a heterogeneous group of lung disorders characterized by progressive and irreversible destruction of lung architecture and disruption of gas exchange. The clinical signs of RIPF include increasing dyspnea, deteriorating lung function, and accumulation of interstitial fluid, eventually leading to respiratory failure. No medical therapy for RIPF has been approved for routine clinical use despite the apparent need for an effective treatment. Numerous signaling pathways are involved in the initiation and progression of RIPF. Also, various approaches for RIPF treatments have focused on several aspects of the current understanding of the molecular pathology of RIPF. This review used the mechanistic categories of associated cell signaling pathways, epithelial cell dysfunction and senescence, abnormal lung remodeling, and aberrant innate and adaptive immunity to review the published literature on RIPF to date and then to identify potential areas for the effective treatment of RIPF.
Collapse
Affiliation(s)
- Zhongjie Chen
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| | - Zhiqiang Wu
- Department of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wen Ning
- State Key Laboratory of Medical Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China.
| |
Collapse
|
38
|
Jin H, Kang GY, Jeon S, Kim JM, Park YN, Cho J, Lee YS. Identification of molecular signatures involved in radiation-induced lung fibrosis. J Mol Med (Berl) 2018; 97:37-47. [PMID: 30406363 PMCID: PMC6326977 DOI: 10.1007/s00109-018-1715-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 12/28/2022]
Abstract
In radiotherapy, radiation (IR)-induced lung fibrosis has severe and dose-limiting side effects. To elucidate the molecular effects of IR fibrosis, we examined the fibrosis process in irradiated mouse lung tissues. High focal IR (90 Gy) was exposed to a 3-mm volume of the left lung in C57BL6 mice. In the diffused irradiation, 20 Gy dose delivered with a 7-mm collimator almost covered the entire left lung. Histological examination for lung tissues of both irradiated and neighboring regions was done for 4 weeks after irradiation. Long-term effects (12 months) of 20Gy IR were compared on a diffuse region of the left lung and non-irradiated right lung. Fibrosis was initiated as early as 2 weeks after IR in the irradiated lung region and neighboring region. Upregulation of gtse1 in both 90Gy-irradiated and neighboring regions was observed. Upregulation of fgl1 in both 20Gy diffused irradiated and non-irradiated lungs was identified. When gtse1 or flg1 was knock-downed, TGFβ or IR-induced epithelial-mesenchymal transition was inhibited, accompanied with the inhibition of cellular migration, suggesting fibrosis responsible genes. Immunofluorescence analysis using mouse fibrotic lung tissues suggested that fibrotic regions showed increased expressions of Gtse1 and Fgl1, indicating novel molecular signatures of gtse1and fgl1 for IR-induced lung fibrosis. Even though their molecular mechanisms and IR doses or irradiated volumes for lung fibrosis may be different, these genes may be novel targets for understanding IR-induced lung fibrosis and in treatment strategies. KEY MESSAGES: Upregulation of gtse1 by 90Gy focal irradiation and upregulation of fgl1 by 20Gy diffused irradiation are identified in mouse lung fibrosis model. Gtse1 and Fgl1 are involved in radiation or TGFβ-induced epithelial-mesenchymal transition. Radiation-induced fibrotic regions of mouse lungs showed increased expressions of Gtse1 and Fgl1. Gtse1 and Fgl1 are suggested to be novel targets for radiation-induced lung fibrosis.
Collapse
Affiliation(s)
- Hee Jin
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-750, South Korea
| | - Ga-Young Kang
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-750, South Korea
| | - Seulgi Jeon
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-750, South Korea
| | - Jin-Mo Kim
- Department of Radiation Oncology, Yonsei University Health System, Seoul, 120-749, South Korea
| | - You Na Park
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-750, South Korea
| | - Jaeho Cho
- Department of Radiation Oncology, Yonsei University Health System, Seoul, 120-749, South Korea
| | - Yun-Sil Lee
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 120-750, South Korea.
| |
Collapse
|
39
|
Lierova A, Jelicova M, Nemcova M, Proksova M, Pejchal J, Zarybnicka L, Sinkorova Z. Cytokines and radiation-induced pulmonary injuries. JOURNAL OF RADIATION RESEARCH 2018; 59:709-753. [PMID: 30169853 PMCID: PMC6251431 DOI: 10.1093/jrr/rry067] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/11/2018] [Indexed: 05/20/2023]
Abstract
Radiation therapy is one of the most common treatment strategies for thorax malignancies. One of the considerable limitations of this therapy is its toxicity to normal tissue. The lung is the major dose-limiting organ for radiotherapy. That is because ionizing radiation produces reactive oxygen species that induce lesions, and not only is tumor tissue damaged, but overwhelming inflammatory lung damage can occur in the alveolar epithelium and capillary endothelium. This damage may result in radiation-induced pneumonitis and/or fibrosis. While describing the lung response to irradiation generally, the main focus of this review is on cytokines and their roles and functions within the individual stages. We discuss the relationship between radiation and cytokines and their direct and indirect effects on the formation and development of radiation injuries. Although this topic has been intensively studied and discussed for years, we still do not completely understand the roles of cytokines. Experimental data on cytokine involvement are fragmented across a large number of experimental studies; hence, the need for this review of the current knowledge. Cytokines are considered not only as molecular factors involved in the signaling network in pathological processes, but also for their diagnostic potential. A concentrated effort has been made to identify the significant immune system proteins showing positive correlation between serum levels and tissue damages. Elucidating the correlations between the extent and nature of radiation-induced pulmonary injuries and the levels of one or more key cytokines that initiate and control those damages may improve the efficacy of radiotherapy in cancer treatment and ultimately the well-being of patients.
Collapse
Affiliation(s)
- Anna Lierova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Marcela Jelicova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Marketa Nemcova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Magdalena Proksova
- Department of Molecular Pathology and Biology, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Jaroslav Pejchal
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Lenka Zarybnicka
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
| | - Zuzana Sinkorova
- Department of Radiobiology, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic
- Corresponding author. Department of Radiobiology, Faculty of Military Health Sciences, University of Defence in Brno, Trebesska 1575, 500 01 Hradec Kralove, Czech Republic. Tel.: +420 973 253 219.
| |
Collapse
|
40
|
Nguyen HQ, To NH, Zadigue P, Kerbrat S, De La Taille A, Le Gouvello S, Belkacemi Y. Ionizing radiation-induced cellular senescence promotes tissue fibrosis after radiotherapy. A review. Crit Rev Oncol Hematol 2018; 129:13-26. [PMID: 30097231 DOI: 10.1016/j.critrevonc.2018.06.012] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 06/08/2018] [Accepted: 06/15/2018] [Indexed: 02/06/2023] Open
Abstract
Ionizing radiation-exposure induces a variety of cellular reactions, such as senescence and apoptosis. Senescence is a permanent arrest state of the cell division, which can be beneficial or detrimental for normal tissue via an inflammatory response and senescence-associated secretion phenotype. Damage to healthy cells and their microenvironment is considered as an important source of early and late complications with an increased risk of morbidity in patients after radiotherapy (RT). In addition, the benefit/risk ratio may depend on the radiation technique/dose used for cancer eradication and the irradiated volume of healthy tissues. For radiation-induced fibrosis risk, the knowledge of mechanisms and potential prevention has become a crucial point to determining radiation parameters and patients' intrinsic radiosensitivity. This review summarizes our understanding of ionizing radiation-induced senescent cell in fibrogenesis. This mechanism may provide new insights for therapeutic modalities for better risk/benefit ratios after RT in the new era of personalized treatments.
Collapse
Affiliation(s)
- Hoang Quy Nguyen
- University of Paris Saclay, University of Paris Est Créteil (UPEC), France, University of Medicine and Pharmacy, Ho Chi Minh City, Viet Nam; INSERM U955 Team 07, Créteil, France
| | - Nhu Hanh To
- INSERM U955 Team 07, Créteil, France; APHP, Department of Radiation Oncology and Henri Mondor Breast Cancer and, Henri Mondor University Hospital, University of Paris Est Créteil (UPEC), France
| | | | - Stéphane Kerbrat
- INSERM U955 Team 04, University of Paris Est Créteil (UPEC), France
| | - Alexandre De La Taille
- INSERM U955 Team 07, Créteil, France; APHP, Department of Urology, Henri Mondor University Hospital, University of Paris Est Créteil (UPEC), Créteil, France
| | - Sabine Le Gouvello
- INSERM U955 Team 04, University of Paris Est Créteil (UPEC), France; APHP, Department of Biology & Pathology, Henri Mondor University Hospital, University of Paris Est Créteil (UPEC), Créteil, France
| | - Yazid Belkacemi
- INSERM U955 Team 07, Créteil, France; APHP, Department of Radiation Oncology and Henri Mondor Breast Cancer and, Henri Mondor University Hospital, University of Paris Est Créteil (UPEC), France.
| |
Collapse
|
41
|
Abstract
Normal tissue injury from irradiation is an unfortunate consequence of radiotherapy. Technologic improvements have reduced the risk of normal tissue injury; however, toxicity causing treatment breaks or long-term side effects continues to occur in a subset of patients. The molecular events that lead to normal tissue injury are complex and span a variety of biologic processes, including oxidative stress, inflammation, depletion of injured cells, senescence, and elaboration of proinflammatory and profibrogenic cytokines. This article describes selected recent advances in normal tissue radiobiology.
Collapse
Affiliation(s)
- Deborah E Citrin
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| | - James B Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
42
|
Fan K, Yang C, Fan Z, Huang Q, Zhang Y, Cheng H, Jin K, Lu Y, Wang Z, Luo G, Yu X, Liu C. MUC16 C terminal-induced secretion of tumor-derived IL-6 contributes to tumor-associated Treg enrichment in pancreatic cancer. Cancer Lett 2018; 418:167-175. [PMID: 29337110 DOI: 10.1016/j.canlet.2018.01.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/28/2017] [Accepted: 01/08/2018] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is the most lethal tumor. CA125 (gene symbol MUC16) is an important serum marker for pancreatic cancer diagnosis and treatment. High serum CA125 is related to metabolic tumor burden and poor prognosis. The circulating Treg subset is another independent prognostic factor for pancreatic cancer. Our unpublished data indicated that the circulating Treg proportion might be related to the serum CA125 level. However, the potential relationship and underlying mechanism of MUC16 and Treg in pancreatic cancer tissues remain unclear. In this study, we found that pancreatic cancer tissues were positive for both MUC16 C terminal (MUC16c) and Foxp3 expression and that their expression was correlated. MUC16c released into the cytoplasm via EGF induction significantly increased IL-6 expression and secretion. The PI3K/AKT pathway may participate in the regulation of IL-6 expression and secretion. By treating CD4+ T cells with IL-6 or co-culturing the cells with pancreatic cancer cells, tumor-derived IL-6 was identified to promote Foxp3 expression and Treg differentiation, which was significantly inhibited by the JAK2 inhibitor AG-490. In sum, our study demonstrated that the relationship between the MUC16c level and Foxp3 expression in the local tumor environment was consistent with that of the serum CA125 level and circulating Treg proportion in the systemic environment. MUC16c promoted Foxp3 expression and tumor-associated Treg enrichment in tumor tissues through tumor-secreted IL-6 activation of the JAK2/STAT3 pathway. These findings may provide deeper insight into potential pancreatic cancer therapy approaches.
Collapse
Affiliation(s)
- Kun Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, PR China; Department of Oncology, Shanghai Medical College, Fudan University, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China
| | - Chao Yang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, PR China; Department of Oncology, Shanghai Medical College, Fudan University, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China
| | - Zhiyao Fan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, PR China; Department of Oncology, Shanghai Medical College, Fudan University, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China
| | - Qiuyi Huang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, PR China; Department of Oncology, Shanghai Medical College, Fudan University, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China
| | - Yiyin Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, PR China; Department of Oncology, Shanghai Medical College, Fudan University, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China
| | - He Cheng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, PR China; Department of Oncology, Shanghai Medical College, Fudan University, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China
| | - Kaizhou Jin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, PR China; Department of Oncology, Shanghai Medical College, Fudan University, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China
| | - Yu Lu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, PR China; Department of Oncology, Shanghai Medical College, Fudan University, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China
| | - Zhengshi Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, PR China; Department of Oncology, Shanghai Medical College, Fudan University, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China
| | - Guopei Luo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, PR China; Department of Oncology, Shanghai Medical College, Fudan University, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, PR China; Department of Oncology, Shanghai Medical College, Fudan University, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China.
| | - Chen Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, PR China; Department of Oncology, Shanghai Medical College, Fudan University, PR China; Shanghai Pancreatic Cancer Institute, Shanghai, 200032, PR China; Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, PR China.
| |
Collapse
|
43
|
Posttreatment Immune Parameters Predict Cancer Control and Pneumonitis in Stage I Non-Small-Cell Lung Cancer Patients Treated With Stereotactic Ablative Radiotherapy. Clin Lung Cancer 2018. [PMID: 29519614 DOI: 10.1016/j.cllc.2017.12.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Stereotactic ablative body radiotherapy (SABR) represents an exciting, tolerable, and highly effective form of radiotherapy. Ongoing investigations into the interactions between radiotherapy and the immune system have uncovered new mechanisms that can be exploited to improve efficacy. We determined whether baseline or posttreatment immune parameters could predict disease control and toxicity in stage I non-small-cell lung cancer (NSCLC) patients treated with SABR. PATIENTS AND METHODS Peripheral blood samples were collected from 62 patients 24 hours before treatment and within 4 weeks after treatment for lymphocyte subset count analysis. All peripheral blood samples were analyzed by flow cytometry. Associated parameters were evaluated to determine their association with progression-free survival (PFS) and symptomatic radiation pneumonitis (grade 2 or higher). The survival rates were estimated with Kaplan-Meier and multivariable analyses using binary logistic regression analysis or a Cox proportional hazards model. RESULTS At a median follow-up time of 36.0 months, the PFS rates for years 1, 2, and 3 were 91.0%, 82.5%, and 48.9%, respectively. The multivariable logistic regression analysis showed that only proportion of lung receiving 20 Gy of radiotherapy (odds ratio = 1.41; 95% confidence interval, 1.05-1.87; P = .023) and mean lung dose (odds ratio = 2.02; 95% confidence interval, 1.16-3.53; P = .016) were associated with symptomatic radiation pneumonitis (grade 2 or higher). Moreover, the immune parameters had no predictive value. In the multivariable Cox regression analysis, an elevated posttreatment cytotoxic CD8+ T-cell level was an independent prognostic factor for longer PFS in stage I NSCLC (hazard ratio, 1.16; 95% confidence interval, 1.01-1.28; P = .01). CONCLUSION A higher posttreatment cytotoxic CD8+ T-cell level was predictive of better PFS in stage I NSCLC patients receiving SABR. Thus, enhancing tumor antigen-specific cellular immunity by combining radiotherapy and immunotherapy might be a crucial strategy for improving survival in these patients.
Collapse
|
44
|
β-catenin, Twist and Snail: Transcriptional regulation of EMT in smokers and COPD, and relation to airflow obstruction. Sci Rep 2017; 7:10832. [PMID: 28883453 PMCID: PMC5589881 DOI: 10.1038/s41598-017-11375-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 08/23/2017] [Indexed: 12/20/2022] Open
Abstract
COPD is characterised by poorly reversible airflow obstruction usually due to cigarette smoking. The transcription factor clusters of β-catenin/Snail1/Twist has been implicated in the process of epithelial mesenchymal transition (EMT), an intermediate between smoking and airway fibrosis, and indeed lung cancer. We have investigated expression of these transcription factors and their "cellular localization" in bronchoscopic airway biopsies from patients with COPD, and in smoking and non-smoking controls. An immune-histochemical study compared cellular protein expression of β-catenin, Snail1 and Twist, in these subject groups in 3 large airways compartment: epithelium (basal region), reticular basement membrane (Rbm) and underlying lamina propria (LP). β-catenin and Snail1 expression was generally high in all subjects throughout the airway wall with marked cytoplasmic to nuclear shift in COPD (P < 0.01). Twist expression was generalised in the epithelium in normal but become more basal and nuclear with smoking (P < 0.05). In addition, β-catenin and Snail1 expression, and to lesser extent of Twist, was related to airflow obstruction and to expression of a canonical EMT biomarker (S100A4). The β-catenin-Snail1-Twist transcription factor cluster is up-regulated and nuclear translocated in smokers and COPD, and their expression is closely related to both EMT activity and airway obstruction.
Collapse
|
45
|
Distinct Roles of Wnt/ β-Catenin Signaling in the Pathogenesis of Chronic Obstructive Pulmonary Disease and Idiopathic Pulmonary Fibrosis. Mediators Inflamm 2017; 2017:3520581. [PMID: 28588349 PMCID: PMC5447271 DOI: 10.1155/2017/3520581] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/29/2017] [Accepted: 04/12/2017] [Indexed: 02/07/2023] Open
Abstract
Wnt signaling pathways are tightly controlled under a physiological condition, under which they play key roles in many biological functions, including cell fate specification and tissue regeneration. Increasing lines of evidence recently demonstrated that a dysregulated activation of Wnt signaling, particularly the Wnt/β-catenin signaling, was involved in the pathogenesis of chronic pulmonary diseases, such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). In this respect, Wnt signaling interacts with other cellular signaling pathways to regulate the initiation and pathogenic procedures of airway inflammation and remodeling, pulmonary myofibroblast proliferation, epithelial-to-mesenchymal transition (EMT), and development of emphysema. Intriguingly, Wnt/β-catenin signaling is activated in IPF; an inhibition of this signaling leads to an alleviation of pulmonary inflammation and fibrosis in experimental models. Conversely, Wnt/β-catenin signaling is inactivated in COPD tissues, and its reactivation results in an amelioration of airspace enlargement with a restored alveolar epithelial structure and function in emphysema models. These studies thus imply distinct mechanisms of Wnt/β-catenin signaling in the pathogenesis of these two chronic pulmonary diseases, indicating potential targets for COPD and IPF treatments. This review article aims to summarize the involvement and pathogenic roles of Wnt signaling pathways in the COPD and IPF, with a focus on the implication of Wnt/β-catenin signaling as underlying mechanisms and therapeutic targets in these two incurable diseases.
Collapse
|
46
|
Wirsdörfer F, Jendrossek V. The Role of Lymphocytes in Radiotherapy-Induced Adverse Late Effects in the Lung. Front Immunol 2016; 7:591. [PMID: 28018357 PMCID: PMC5155013 DOI: 10.3389/fimmu.2016.00591] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/29/2016] [Indexed: 12/31/2022] Open
Abstract
Radiation-induced pneumonitis and fibrosis are dose-limiting side effects of thoracic irradiation. Thoracic irradiation triggers acute and chronic environmental lung changes that are shaped by the damage response of resident cells, by the resulting reaction of the immune system, and by repair processes. Although considerable progress has been made during the last decade in defining involved effector cells and soluble mediators, the network of pathophysiological events and the cellular cross talk linking acute tissue damage to chronic inflammation and fibrosis still require further definition. Infiltration of cells from the innate and adaptive immune systems is a common response of normal tissues to ionizing radiation. Herein, lymphocytes represent a versatile and wide-ranged group of cells of the immune system that can react under specific conditions in various ways and participate in modulating the lung environment by adopting pro-inflammatory, anti-inflammatory, or even pro- or anti-fibrotic phenotypes. The present review provides an overview on published data about the role of lymphocytes in radiation-induced lung disease and related damage-associated pulmonary diseases with a focus on T lymphocytes and B lymphocytes. We also discuss the suspected dual role of specific lymphocyte subsets during the pneumonitic phase and fibrotic phase that is shaped by the environmental conditions as well as the interaction and the intercellular cross talk between cells from the innate and adaptive immune systems and (damaged) resident epithelial cells and stromal cells (e.g., endothelial cells, mesenchymal stem cells, and fibroblasts). Finally, we highlight potential therapeutic targets suited to counteract pathological lymphocyte responses to prevent or treat radiation-induced lung disease.
Collapse
Affiliation(s)
- Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen , Essen , Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen , Essen , Germany
| |
Collapse
|
47
|
Mont S, Davies SS, Roberts second LJ, Mernaugh RL, McDonald WH, Segal BH, Zackert W, Kropski JA, Blackwell TS, Sekhar KR, Galligan JJ, Massion PP, Marnett LJ, Travis EL, Freeman ML. Accumulation of isolevuglandin-modified protein in normal and fibrotic lung. Sci Rep 2016; 6:24919. [PMID: 27118599 PMCID: PMC4847119 DOI: 10.1038/srep24919] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 04/07/2016] [Indexed: 12/27/2022] Open
Abstract
Protein lysine modification by γ-ketoaldehyde isomers derived from arachidonic acid, termed isolevuglandins (IsoLGs), is emerging as a mechanistic link between pathogenic reactive oxygen species and disease progression. However, the questions of whether covalent modification of proteins by IsoLGs are subject to genetic regulation and the identity of IsoLG-modified proteins remain unclear. Herein we show that Nrf2 and Nox2 are key regulators of IsoLG modification in pulmonary tissue and report on the identity of proteins analyzed by LC-MS following immunoaffinity purification of IsoLG-modified proteins. Gene ontology analysis revealed that proteins in numerous cellular pathways are susceptible to IsoLG modification. Although cells tolerate basal levels of modification, exceeding them induces apoptosis. We found prominent modification in a murine model of radiation-induced pulmonary fibrosis and in idiopathic pulmonary fibrosis, two diseases considered to be promoted by gene-regulated oxidant stress. Based on these results we hypothesize that IsoLG modification is a hitherto unrecognized sequelae that contributes to radiation-induced pulmonary injury and IPF.
Collapse
Affiliation(s)
- Stacey Mont
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Sean S. Davies
- Division of Clinical Pharmacology, Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - L. Jackson Roberts second
- Division of Clinical Pharmacology, Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Raymond L. Mernaugh
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - W. Hayes McDonald
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37240, USA
- Proteomics Laboratory and Mass Spectrometry Research Center, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Brahm H. Segal
- Department of Medicine, Department of Immunology, Roswell Park Cancer Institute, and University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, 14263, USA
| | - William Zackert
- Division of Clinical Pharmacology, Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Jonathan A. Kropski
- Division of Pulmonary & Critical Care, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Timothy S. Blackwell
- Division of Pulmonary & Critical Care, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Konjeti R. Sekhar
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - James J. Galligan
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Pierre P. Massion
- Division of Pulmonary & Critical Care, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Lawrence J. Marnett
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN 37240, USA
- A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Vanderbilt Institute of Chemical Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Elizabeth L. Travis
- Department of Experimental Radiation Oncology, Division of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Michael L. Freeman
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| |
Collapse
|