1
|
Zhang J, Ma L, He L, Xu Q, Ding Y, Wang L. MicroRNA-541-3p/Rac2 signaling bridges radiation-induced lung injury and repair. Noncoding RNA Res 2025; 12:10-19. [PMID: 40026446 PMCID: PMC11869541 DOI: 10.1016/j.ncrna.2025.01.010] [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: 10/20/2024] [Revised: 01/18/2025] [Accepted: 01/26/2025] [Indexed: 03/05/2025] Open
Abstract
Background While radiation-induced lung injury decreases quality of life and suppresses efficacy of radiotherapy, to date, the relationship between radiation-induced lung injury and repair remains unclear. Our previous studies revealed that TNFRSF10B-RIPK1/RIPK3-MLKL signaling induces necroptosis of alveolar epithelial cells and potentiates radiation-induced lung injury. We also found that microRNA-541-3p is differentially expressed in radiation-damaged lungs. The connection between microRNA-541-3p, TNFRSF10B signaling, and TGFβ1 signaling is also unclear. Objective This study was performed to explore the regulatory effects of microRNA-541-3p on TNFRSF10B and TGFβ1 signaling. Methods Mouse alveolar epithelial cells were transfected with a vector expressing microRNA-541-3p to regulate expression of target genes. Flow cytometry, polymerase chain reaction, and western blotting were used to analyze cell necroptosis, target gene expression, and target protein expression, respectively. Results Overexpression of microRNA-541-3p positively regulated TNFRSF10B-RIPK1/RIPK3-MLKL signaling through Rac2 to induce cell necroptosis. MicroRNA-541-3p negatively regulates Rac2. MicroRNA-541-3p and Rac2 regulate the expression of Tgf-beta1 and its encoded proteins. Conclusions The Rac2 gene synchronously regulates TNFRSF10B-RIPK1/RIPK3-MLKL and TGFβ1 signaling. MicroRNA-541-3P/Rac2 act as mediators of radiation damage and repair signaling.
Collapse
Affiliation(s)
- Jiandong Zhang
- Clinical School of Medicine, Henan University of Science and Technology, Luoyang City, Henan Province, 471023, China
- The First Affiliated Hospital of Henan University of Science and Technology, Luoyang City, Henan Province, 471023, China
| | - Lei Ma
- Department of Radiation Oncology, Nanyang First People's Hospital Affiliated to Henan University, Nanyang City, Henan Province, 473000, China
| | - Limin He
- Department of Radiation Oncology, Nanyang First People's Hospital Affiliated to Henan University, Nanyang City, Henan Province, 473000, China
| | - Quanxiao Xu
- Department of Oncology, the Second Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, 215000, China
| | - Yan Ding
- Department of Radiation Oncology, Nanyang First People's Hospital Affiliated to Henan University, Nanyang City, Henan Province, 473000, China
| | - Lidong Wang
- Clinical School of Medicine, Henan University of Science and Technology, Luoyang City, Henan Province, 471023, China
- The First Affiliated Hospital of Henan University of Science and Technology, Luoyang City, Henan Province, 471023, China
| |
Collapse
|
2
|
Dong Z, Wang R, Yi J, Wei W, Wang M, Wei X, Shen Y, Wang Z, Jin S, Liu Z. A novel role for the regulatory NRP1 in immune and inflammatory reactions during radiation-induced lung injury. Int J Biol Macromol 2025; 308:142307. [PMID: 40118426 DOI: 10.1016/j.ijbiomac.2025.142307] [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: 12/09/2024] [Revised: 03/02/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Neuropilin-1 (NRP1) is a transmembrane protein with diverse functions in tumor biology and immune system regulation. Despite extensive research, its specific function in radiation-induced lung injury (RILI) remains unclear. In this study, we aimed to explore the influence of NRP1 on immune and inflammatory responses mediated by regulatory T cells (Tregs) in RILI. Initial findings revealed that radiation increases the number of NRP1-expressing Tregs, which correlated with RILI severity. Inhibiting Tregs in mice effectively suppressed radiation-induced Treg differentiation and promoted helper T cell 1 (Th1) and Th2 cytokine expression, thereby shifting T cell polarization toward a Th1 phenotype and slowing RILI progression. However, Treg inhibition alone did not entirely prevent RILI, as skin damage and inflammatory factor expression persisted. Subsequent specific NRP1 knockdown in alveolar epithelial cell-II altered the inflammatory gene network, suppressed TGF-β signaling, reduced Treg levels, decreased IL-17 A and INF-γ expression, and shifted Th cell polarization toward Th2, alleviating RILI. In conclusion, this study reveals a novel mechanism by which NRP1 regulates immune and inflammatory responses in RILI, offering a promising avenue for developing innovative therapeutic strategies.
Collapse
Affiliation(s)
- Zhuo Dong
- Cancer Center, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Guangzhou 510280, China; Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510280, China
| | - Rui Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130000, China
| | - Junxuan Yi
- Cancer Center, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Guangzhou 510280, China; Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510280, China
| | - Wei Wei
- Department of Radiotherapy, Chinese PLA General Hospital, Beijing 100000, China
| | - Mingwei Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130000, China
| | - Xinfeng Wei
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130000, China
| | - Yannan Shen
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130000, China
| | - Zhicheng Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130000, China
| | - Shunzi Jin
- Cancer Center, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Guangzhou 510280, China; NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130000, China.
| | - Zhigang Liu
- Cancer Center, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Guangzhou 510280, China; Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
3
|
Zheng C, Zhang L, Sun Y, Ma Y, Zhang Y. Alveolar epithelial cell dysfunction and epithelial-mesenchymal transition in pulmonary fibrosis pathogenesis. Front Mol Biosci 2025; 12:1564176. [PMID: 40343260 PMCID: PMC12058482 DOI: 10.3389/fmolb.2025.1564176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/11/2025] [Indexed: 05/11/2025] Open
Abstract
Pulmonary fibrosis (PF) is a progressive and lethal interstitial lung disease characterized by aberrant scar formation and destruction of alveolar architecture. Dysfunctional alveolar epithelial cells (AECs) play a central role in initiating PF, where chronic injury triggers apoptosis and disrupts epithelial homeostasis, leading to epithelial-mesenchymal transition (EMT). This dynamic reprogramming process causes AECs to shed epithelial markers and adopt a mesenchymal phenotype, fueling fibroblast activation and pathological extracellular matrix (ECM) deposition. This review systematically explores the multi-layered mechanisms driving AECs dysfunction and EMT, focusing on core signaling axes such as transforming growth factor-β (TGF-β)/Smad, WNT/β-catenin, NF-κB-BRD4, and nuclear factor erythroid 2-related factor 2 (Nrf2), which regulate EMT and fibroblast-ECM interactions. It also highlights emerging regulators, including metabolic reprogramming, exosomal miRNA trafficking, and immune-epithelial interactions. Furthermore, understanding these mechanisms is essential for developing targeted therapeutic strategies to modulate these pathways and halt or reverse fibrosis progression, offering critical insights into potential clinical treatments for PF.
Collapse
Affiliation(s)
- Caopei Zheng
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Ling Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yuqing Sun
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Yingmin Ma
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yulin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Beijing Research Center for Respiratory Infectious Diseases, Beijing, China
| |
Collapse
|
4
|
Us SB, Bayrak G, Ballı E, Büyükakıllı B. Histological, immunohistochemical and electron-microscopic evaluation of different radiotherapy doses effects on rat's lung. Tissue Cell 2025; 95:102860. [PMID: 40139078 DOI: 10.1016/j.tice.2025.102860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/22/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Radiation-related complications occur in the lungs during radiotherapy for intrathoracic tumors. Lung damage caused by radiation in the long term varies depending on the radiation dose received. The incidence of pulmonary toxicity has decreased with the advancement of radiotherapy techniques such as intensity-modulated RT (IMRT) and image-guided RT (IGRT). This study aimed to examine the damage caused by different radiation doses applied with the IMRT technique, both histochemically and histopathologically, and to emphasize the effect of a low dose (5 Gy). METHODS AND MATERIALS A total of 24 rats were divided into 4 groups: control (Group 1), 5 Gy (Group 2), 10 Gy (Group 3), and 20 Gy (Group 4). Helical IMRT plans were made using tomotherapy to ensure that the thorax received the entire dose designated for each group. The rats in groups 2, 3, and 4 were exposed to radiation doses of 5 Gy, 10 Gy, and 20 Gy, respectively. After 180 days, the morphological and immunohistochemical features and the number of apoptotic cells in the lung tissues were examined using electron microscopy and light microscopy. Morphological, inflammatory (IL-1β, IL-10, and TNF-α), and apoptotic index values were compared statistically. RESULTS This study observed that morphological, inflammatory, and apoptotic cell damage in the lungs gradually increased in a dose-dependent manner compared to the control group. However, at the low radiation dose of 5 Gy, the severity of lung damage was relatively less than at the higher doses (10 Gy and 20 Gy). CONCLUSION In conclusion, this study found that the severity of lung damage was less at a low radiation dose (5 Gy) compared to higher doses (10 Gy and 20 Gy). This emphasizes the need to limit the maximum dose and the irradiated volume during thoracic radiotherapy.
Collapse
Affiliation(s)
- Songül Barlaz Us
- Department of Radiation Oncology, Mersin University, Mersin, Türkiye
| | - Gülsen Bayrak
- Department of Histology-Embryology, Usak University, Usak, Türkiye.
| | - Ebru Ballı
- Department of Histology-Embryology, Mersin University, Mersin, Türkiye
| | | |
Collapse
|
5
|
Zhang P, Wang J, Miao J, Zhu P. The dual role of tissue regulatory T cells in tissue repair: return to homeostasis or fibrosis. Front Immunol 2025; 16:1560578. [PMID: 40114929 PMCID: PMC11922884 DOI: 10.3389/fimmu.2025.1560578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/18/2025] [Indexed: 03/22/2025] Open
Abstract
Tissue resident regulatory T cells (tissue Tregs) are vital for maintaining immune homeostasis and controlling inflammation. They aid in repairing damaged tissues and influencing the progression of fibrosis. However, despite extensive research on how tissue Tregs interact with immune and non-immune cells during tissue repair, their pro- and anti-fibrotic effects in chronic tissue injury remain unclear. Understanding how tissue Tregs interact with various cell types, as well as their roles in chronic injury and fibrosis, is crucial for uncovering the mechanisms behind these conditions. In this review, we describe the roles of tissue Tregs in repair and fibrosis across different tissues and explore potential strategies for regulating tissue homeostasis. These insights hold promise for providing new perspectives and approaches for the treatment of irreversible fibrotic diseases.
Collapse
Affiliation(s)
- Peiyan Zhang
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jiawei Wang
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jinlin Miao
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ping Zhu
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
6
|
Zhang Y, Shen WX, Li P, Chen MB, Zhou LN. Serum Interleukin Levels Predict Occurrence of Acute Radiation Pneumonitis and Overall Survival in Thoracic Tumours. CLIN INVEST MED 2025; 48:29-38. [PMID: 40131214 DOI: 10.3138/cim-2024-0262] [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] [Indexed: 03/26/2025]
Abstract
BACKGROUND Radiation-induced lung injury (RILI) is a significant adverse effect of thoracic radiotherapy, potentially impacting patient prognosis. The risk factors for acute radiation pneumonitis (RP) have not been fully clarified. The present study evaluated the predictive value of serum interleukins (ILs) in the occurrence of RP and overall survival in patients with thoracic cancers. METHODS This single-centre retrospective observational study enrolled 435 thoracic cancer patients who underwent chest radiation therapy. Serum levels of IL-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12p70, IL-17, TNF-α, IFN-γ, IFN-α were measured by cytometric bead array before radiotherapy. The relationship between clinical characteristics, serum IL levels and the occurrence of RP were analyzed. Cox regression and Kaplan-Meier methods were also performed to investigate the prognostic role of serum IL levels in these patients. RESULTS The incidence of RP in these patients was 17.01%. Elevated serum levels of IL-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12p70, TNF-α, IFN-α were all associated with the occurrence of RP. High levels of IL-1β, IL-4, and IL-12p70 were correlated with more severe pneumonitis. Univariate and multivariate logistic regression analysis identified serum IL-6 level as an independent prognostic factor in patients receiving thoracic radiotherapy. CONCLUSIONS Serum interleukin levels are linked to the development of acute RP in patients receiving thoracic radiotherapy. Serum IL-6 could serve as a valuable biomarker in identifying patients at high risk for RP, potentially guiding individualized therapeutic strategies and improving patient management in radiotherapy. Future research should focus on validating IL-6's role in larger cohorts and exploring its integration into clinical practice for the early prediction of RILI.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Oncology & Radiotherapy, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, China
| | - Wen-Xiang Shen
- Department of Oncology & Radiotherapy, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, China
| | - Ping Li
- Department of Oncology & Radiotherapy, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, China
| | - Min-Bin Chen
- Department of Oncology & Radiotherapy, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, China
| | - Li-Na Zhou
- Department of Oncology & Radiotherapy, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, China
| |
Collapse
|
7
|
Gockeln L, Wirsdörfer F, Jendrossek V. CD73/adenosine dynamics in treatment-induced pneumonitis: balancing efficacy with risks of adverse events in combined radio-immunotherapies. Front Cell Dev Biol 2025; 12:1471072. [PMID: 39872847 PMCID: PMC11769960 DOI: 10.3389/fcell.2024.1471072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/23/2024] [Indexed: 01/30/2025] Open
Abstract
Consolidation with PD-1/PD-L1-based immune checkpoint blockade after concurrent platinum-based chemo-radiotherapy has become the new standard of care for advanced stage III unresectable non-small cell lung cancer (NSCLC) patients. In order to further improve therapy outcomes, innovative combinatorial treatment strategies aim to target additional immunosuppressive barriers in the tumor microenvironment such as the CD73/adenosine pathway. CD73 and adenosine are known as crucial endogenous regulators of lung homeostasis and inflammation, but also contribute to an immunosuppressive tumor microenvironment. Furthermore, the CD73/adenosine pathway can also limit the immune-activating effects of cytotoxic therapies by degrading the pro-inflammatory danger molecule ATP, which is released into the tumor microenvironment and normal lung tissue upon therapy-induced cell damage. Thus, while targeting CD73 may enhance the efficacy of radio-immunotherapies in cancer treatment by mitigating tumor immune escape and improving immune-mediated tumor killing, it also raises concerns about increased immune-related adverse events (irAEs) in the normal tissue. In fact, combined radio-immunotherapies bear an increased risk of irAEs in the lungs, and additional pharmacologic inhibition of CD73 may further enhance the risk of overwhelming or overlapping pulmonary toxicity and thereby limit therapy outcome. This review explores how therapeutic interventions targeting CD73/adenosine dynamics could enhance radiation-induced immune activation in combined radio-immunotherapies, whilst potentially driving irAEs in the lung. We specifically investigate the interactions between radiotherapy and the CD73/adenosine pathway in radiation pneumonitis. Additionally, we compare the incidence of (radiation) pneumonitis reported in relevant trials to determine if there is an increased risk of irAEs in the clinical setting. By understanding these dynamics, we aim to inform future strategies for optimizing radio-immunotherapy regimens, ensuring effective cancer control while preserving pulmonary integrity and patient quality of life.
Collapse
Affiliation(s)
| | | | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
8
|
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
|
9
|
Ma L, Jin Y, Aili A, Xu L, Wang X, Xiao L, Zhao W, Yin S, Liu B, Yuan X. High-dose vitamin C attenuates radiation-induced pulmonary fibrosis by targeting S100A8 and S100A9. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167358. [PMID: 39025374 DOI: 10.1016/j.bbadis.2024.167358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/19/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
Radiation-induced pulmonary fibrosis (RIPF) is a frequently encountered late complication in patients undergoing radiation therapy, presenting a substantial risk to patient mortality and quality of life. The pathogenesis of RIPF remains unclear, and current treatment options are limited in efficacy. High-dose vitamin C has demonstrated potential when used in conjunction with other adjuvant therapies due to potent anticancer properties. However, the potential relationship between high-dose vitamin C and RIPF has not yet been explored in existing literature. In our study, the RIPF model and the LLC tumor model were used as two animal models to explore how high-dose vitamin C can improve RIPF without hampering the antitumour efficacy of radiotherapy. The impact of high-dose vitamin C on RIPF was assessed through various assays, including micro-CT, HE staining, Masson staining, and immunohistochemistry. Our results indicated that administering high-dose vitamin C 2 days before radiation and continuing for a duration of 6 weeks significantly inhibited the progression of RIPF. In order to explore the mechanism by which high-dose vitamin C attenuates RIPF, we utilized RNA-seq analysis of mouse lung tissue in conjunction with publicly available databases. Our findings indicated that high-dose vitamin C inhibits the differentiation of fibroblasts into myofibroblasts by targeting S100A8 and S100A9 derived from neutrophils. Additionally, the combination of high-dose vitamin C and radiation demonstrated enhanced inhibition of tumor growth in a murine LLC tumor model. These results revealed that the combination of radiotherapy and high-dose vitamin C may offer a promising therapeutic approach for the clinical management of thoracic tumors and the prevention of RIPF.
Collapse
Affiliation(s)
- Li Ma
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Jin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aifeina Aili
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Xu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingyan Xiao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiyu Yin
- Department of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
10
|
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
|
11
|
Wang X, Zhang H, XinZhang, Liu Y. Abscopal effect: from a rare phenomenon to a new frontier in cancer therapy. Biomark Res 2024; 12:98. [PMID: 39228005 PMCID: PMC11373306 DOI: 10.1186/s40364-024-00628-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 07/30/2024] [Indexed: 09/05/2024] Open
Abstract
Radiotherapy (RT) controls local lesions, meantime it has the capability to induce systemic response to inhibit distant, metastatic, non-radiated tumors, which is referred to as the "abscopal effect". It is widely recognized that radiotherapy can stimulate systemic immune response. This provides a compelling theoretical basis for the combination of immune therapy combined with radiotherapy(iRT). Indeed, this phenomenon has also been observed in clinical treatment, bringing significant clinical benefits to patients, and a series of basic studies are underway to amplify this effect. However, the molecular mechanisms of immune response induced by RT, determination of the optimal treatment regimen for iRT, and how to amplify the abscopal effect. In order to amplify and utilize this effect in clinical management, these key issues require to be well addressed; In this review, we comprehensively summarize the growing consensus and emphasize the emerging limitations of enhancing the abscopal effect with radiotherapy or immunotherapy. Finally, we discuss the prospects and barriers to the current clinical translational applications.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - Haoyu Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China
| | - XinZhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China.
| | - Yong Liu
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Otolaryngology Major Disease Research Key Laboratory of Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- Clinical Research Center for Laryngopharyngeal and Voice Disorders in Hunan Province, 87 Xiangya Road, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders (Xiangya Hospital), Changsha, 410008, Hunan, China.
| |
Collapse
|
12
|
Taghizadeh-Hesary F. "Reinforcement" by Tumor Microenvironment: The Seventh "R" of Radiobiology. Int J Radiat Oncol Biol Phys 2024; 119:727-733. [PMID: 38032584 DOI: 10.1016/j.ijrobp.2023.09.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/11/2023] [Accepted: 09/16/2023] [Indexed: 12/01/2023]
Affiliation(s)
- Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Clinical Oncology Department, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Liang L, Huang Y, Chen L, Shi Z, Wang H, Zhang T, Li Z, Mi J, Fan T, Lu Y, Chen F, Huang W, Hu K. Radioprotective efficacy of Astilbin in mitigating radiation-induced lung injury through inhibition of p53 acetylation. ENVIRONMENTAL TOXICOLOGY 2023; 38:2967-2980. [PMID: 37598414 DOI: 10.1002/tox.23931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/19/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023]
Abstract
Radiation-induced lung injury (RILI) is a common side effect in thoracic tumor patients undergoing radiotherapy. At present, there is no ideal radio-protective agent which is widely used in RILI treatment. Astilbin (AST), a bioactive flavonoid, exhibits various biological effects, including anti-inflammatory, antioxidant, and anti-fibrotic activities, which partly result from reducing oxidative stress and inflammation in various pathogenic conditions. However, the protective efficacy of AST to ameliorate RILI has not been reported. In this study, we employed network pharmacology, RNA sequencing, and experimental evaluation to reveal the effects and pharmacological mechanism of AST to treat RILI in vivo and in vitro. We observed that AST reduced radiation-induced apoptosis, DNA damage, inflammatory reactions, and the reactive oxygen species (ROS) level in human normal lung epithelial cells BEAS-2B. Further study showed that AST treatment significantly ameliorated RILI by reducing the radiation-induced pathology changes and inflammatory reaction of lung tissue in C57BL/6J mice. Mechanistically, the expression of epithelial-mesenchymal transition (EMT) markers and radiation-triggered acetylation of the p53 protein were alleviated by AST treatment. Furthermore, AST alleviated the acetylation of p53 after intervention of Trichostatin A (TSA). Our data indicate that AST can alleviate RILI by inhibiting inflammatory reactions and the EMT process through decreasing the expression of p53 acetylation. In conclusion, our study suggests that AST has great potential to be a new protective and therapeutic compound for RILI.
Collapse
Affiliation(s)
- Lixing Liang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Yaqin Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Liuyin Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Zhiling Shi
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Housheng Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Tingting Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Zhixun Li
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Jinglin Mi
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Ting Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Yushuang Lu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Fuli Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Weimei Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| | - Kai Hu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, China
| |
Collapse
|
16
|
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: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
17
|
Dąbrowska A, Grubba M, Balihodzic A, Szot O, Sobocki BK, Perdyan A. The Role of Regulatory T Cells in Cancer Treatment Resistance. Int J Mol Sci 2023; 24:14114. [PMID: 37762416 PMCID: PMC10531820 DOI: 10.3390/ijms241814114] [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: 08/16/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Despite tremendous progress in cancer treatment in recent years, treatment resistance is still a major challenge for a great number of patients. One of the main causes is regulatory T lymphocytes (Tregs), which suppress excessive inflammatory responses via the secretion of immunosuppressive cytokines and upregulate the immune checkpoints. Their abundance causes an immunosuppressive reprogramming of the tumor environment, which is ideal for tumor growth and drug inefficiency. Hence, regiments that can regain tumor immunogenicity are a promising strategy to overcome Tregs-mediated drug resistance. However, to develop effective therapeutic regimens, it is essential to understand the molecular mechanisms of Treg-mediated resistance. In this article, we gathered a comprehensive summary of the current knowledge on molecular mechanisms and the role of Tregs in cancer treatment resistance, including cancer immunotherapy, targeted therapy, chemotherapy, and radiotherapy.
Collapse
Affiliation(s)
- Anna Dąbrowska
- Student Scientific Circle of Oncology and Radiotherapy, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Magdalena Grubba
- Student Scientific Circle of Oncology and Radiotherapy, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Amar Balihodzic
- Division of Oncology, Department of Internal Medicine, Comprehensive Cancer Center Graz, Medical University of Graz, 8036 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Olga Szot
- Student Scientific Circle of Oncology and Radiotherapy, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Bartosz Kamil Sobocki
- Student Scientific Circle of Oncology and Radiotherapy, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Adrian Perdyan
- 3P-Medicine Laboratory, Medical University of Gdansk, 80-210 Gdansk, Poland
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
18
|
Groves AM, Misra R, Clair G, Hernady E, Olson H, Orton D, Finkelstein J, Marples B, Johnston CJ. Influence of the irradiated pulmonary microenvironment on macrophage and T cell dynamics. Radiother Oncol 2023; 183:109543. [PMID: 36813173 PMCID: PMC10238652 DOI: 10.1016/j.radonc.2023.109543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/29/2022] [Accepted: 02/04/2023] [Indexed: 02/22/2023]
Abstract
BACKGROUND The lung is sensitive to radiation, increasing normal tissue toxicity risks following radiation therapy. Adverse outcomes include pneumonitis and pulmonary fibrosis, which result from dysregulated intercellular communication within the pulmonary microenvironment. Although macrophages are implicated in these pathogenic outcomes, the impact of their microenvironment is not well understood. MATERIALS AND METHODS C57BL/6J mice received 6Gyx5 irradiation to the right lung. Macrophage and T cell dynamics were investigated in ipsilateral right lungs, contralateral left lungs and non-irradiated control lungs 4-26wk post exposure. Lungs were evaluated by flow cytometry, histology and proteomics. RESULTS Following uni-lung irradiation, focal regions of macrophage accumulation were noted in both lungs by 8wk, however by 26wk fibrotic lesions were observed only in ipsilateral lungs. Infiltrating and alveolar macrophages populations expanded in both lungs, however transitional CD11b + alveolar macrophages persisted only in ipsilateral lungs and expressed lower CD206. Concurrently, arginase-1 + macrophages accumulated in ipsilateral but not contralateral lungs at 8 and 26wk post exposure, while CD206 + macrophages were absent from these accumulations. While radiation expanded CD8 + T cells in both lungs, T regulatory cells only increased in ipsilateral lungs. Unbiased proteomics analysis of immune cells revealed a substantial number of differentially expressed proteins in ipsilateral lungs when compared to contralateral lungs and both differed from non-irradiated controls. CONCLUSIONS Pulmonary macrophage and T cell dynamics are impacted by the microenvironmental conditions that develop following radiation exposure, both locally and systemically. While macrophages and T cells infiltrate and expand in both lungs, they diverge phenotypically depending on their environment.
Collapse
Affiliation(s)
- Angela M Groves
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.
| | - Ravi Misra
- Department of Pediatrics, Division of Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Geremy Clair
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Eric Hernady
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Heather Olson
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Danny Orton
- Biological Science Division, Pacific Northwest National Laboratory, Richland, WA 99354, USA
| | - Jacob Finkelstein
- Department of Pediatrics, Division of Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Brian Marples
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Carl J Johnston
- Department of Pediatrics, Division of Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| |
Collapse
|
19
|
Sakagami M, Inokuchi H, Mukumoto N, Itoyama H, Hamaura N, Yamagishi M, Mukumoto N, Matsuda S, Kabata D, Shibuya K. Clinical features and risk factors for interstitial lung disease spreading in low-dose irradiated areas after definitive radiotherapy with or without durvalumab consolidation therapy for patients with non-small cell lung cancer. Radiat Oncol 2023; 18:87. [PMID: 37217919 DOI: 10.1186/s13014-023-02276-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 05/04/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND The current standard of care for patients with unresectable locally advanced non-small cell lung cancer (NSCLC) is chemoradiotherapy (CRT) combined with durvalumab consolidation therapy. However, radiotherapy (RT) always carries the risk of radiation pneumonitis (RP), which can preclude durvalumab continuation. In particular, the spread of interstitial lung disease (ILD) in low-dose areas or extending beyond the RT field often makes it difficult to determine the safety of continuation or rechallenging of durvalumab. Thus, we retrospectively analyzed ILD/RP after definitive RT with and without durvalumab, with assessment of radiologic features and dose distribution in RT. METHODS We retrospectively evaluated the clinical records, CT imaging, and radiotherapy planning data of 74 patients with NSCLC who underwent definitive RT at our institution between July 2016 and July 2020. We assessed the risk factors for recurrence within one year and occurrence of ILD/RP. RESULTS Kaplan-Meier method showed that ≥ 7 cycles of durvalumab significantly improved 1-year progression free survival (PFS) (p < 0.001). Nineteen patients (26%) were diagnosed with ≥ Grade 2 and 7 (9.5%) with ≥ Grade 3 ILD/RP after completing RT. There was no significant correlation between durvalumab administration and ≥ Grade 2 ILD/RP. Twelve patients (16%) developed ILD/RP that spread outside the high-dose (> 40 Gy) area, of whom 8 (67%) had ≥ Grade 2 and 3 (25%) had Grade 3 symptoms. In unadjusted and multivariate Cox proportional-hazards models adjusted for V20 (proportion of the lung volume receiving ≥ 20 Gy), high HbA1c level was significantly correlated with ILD/RP pattern spreading outside the high-dose area (hazard ratio, 1.842; 95% confidence interval, 1.35-2.51). CONCLUSIONS Durvalumab improved 1-year PFS without increasing the risk of ILD/RP. Diabetic factors were associated with ILD/RP distribution pattern spreading in the lower dose area or outside RT fields, with a high rate of symptoms. Further study of the clinical background of patients including diabetes is needed to safely increase the number of durvalumab doses after CRT.
Collapse
Affiliation(s)
- Mai Sakagami
- Department of Radiation Oncology, Graduate School of Medicine, Osaka City University, Osaka, Japan.
| | - Haruo Inokuchi
- Department of Radiation Oncology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Nobutaka Mukumoto
- Department of Radiation Oncology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Hiroshige Itoyama
- Department of Radiation Oncology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Nobunari Hamaura
- Department of Radiation Oncology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Mutsumi Yamagishi
- Department of Radiation Oncology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Naoki Mukumoto
- Department of Radiation Oncology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Shogo Matsuda
- Department of Radiation Oncology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Daijiro Kabata
- Department of Medical Statistics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| | - Keiko Shibuya
- Department of Radiation Oncology, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan
| |
Collapse
|
20
|
Identification of S100A9 as a Potential Inflammation-Related Biomarker for Radiation-Induced Lung Injury. J Clin Med 2023; 12:jcm12030733. [PMID: 36769382 PMCID: PMC9917937 DOI: 10.3390/jcm12030733] [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: 11/23/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Radiation-induced lung injury (RILI), a potentially fatal and dose-limiting complication of radiotherapy for thoracic tumors, is divided into early reversible pneumonitis and irreversible advanced-stage fibrosis. Early detection and intervention contribute to improving clinical outcomes of patients. However, there is still a lack of reliable biomarkers for early prediction and clinical diagnosis of RILI. Given the central role of inflammation in the initiation and progression of RILI, we explored specific inflammation-related biomarkers during the development of RILI in this study. Two expression profiles from the Gene Expression Omnibus (GEO) database were downloaded, in which 75 differentially expressed genes (DEGs) were screened out. Combining Gene Oncology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis and protein-protein interaction (PPI) network analysis, we identified four inflammation-related hub genes in the progression of RILI-MMP9, IL-1β, CCR1 and S100A9. The expression levels of the hub genes were verified in RILI mouse models, with S100A9 showing the highest level of overexpression. The level of S100A9 in bronchoalveolar lavage fluid (BALF) and the expression of S100A9 in lung tissues were positively correlated with the degree of inflammation in RILI. The results above indicate that S100A9 is a potential biomarker for the early prediction and diagnosis of the development of RILI.
Collapse
|
21
|
Hou G, Li J, Liu W, Wei J, Xin Y, Jiang X. Mesenchymal stem cells in radiation-induced lung injury: From mechanisms to therapeutic potential. Front Cell Dev Biol 2022; 10:1100305. [PMID: 36578783 PMCID: PMC9790971 DOI: 10.3389/fcell.2022.1100305] [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: 11/16/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
Radiotherapy (RT) is an effective treatment option for multiple thoracic malignant tumors, including lung cancers, thymic cancers, and tracheal cancers. Radiation-induced lung injury (RILI) is a serious complication of radiotherapy. Radiation causes damage to the pulmonary cells and tissues. Multiple factors contribute to the progression of Radiation-induced lung injury, including genetic alterations, oxidative stress, and inflammatory responses. Especially, radiation sources contribute to oxidative stress occurrence by direct excitation and ionization of water molecules, which leads to the decomposition of water molecules and the generation of reactive oxygen species (ROS), reactive nitrogen species (RNS). Subsequently, reactive oxygen species and reactive nitrogen species overproduction can induce oxidative DNA damage. Immune cells and multiple signaling molecules play a major role in the entire process. Mesenchymal stem cells (MSCs) are pluripotent stem cells with multiple differentiation potentials, which are under investigation to treat radiation-induced lung injury. Mesenchymal stem cells can protect normal pulmonary cells from injury by targeting multiple signaling molecules to regulate immune cells and to control balance between antioxidants and prooxidants, thereby inhibiting inflammation and fibrosis. Genetically modified mesenchymal stem cells can improve the natural function of mesenchymal stem cells, including cellular survival, tissue regeneration, and homing. These reprogrammed mesenchymal stem cells can produce the desired products, including cytokines, receptors, and enzymes, which can contribute to further advances in the therapeutic application of mesenchymal stem cells. Here, we review the molecular mechanisms of radiation-induced lung injury and discuss the potential of Mesenchymal stem cells for the prevention and treatment of radiation-induced lung injury. Clarification of these key issues will make mesenchymal stem cells a more fantastic novel therapeutic strategy for radiation-induced lung injury in clinics, and the readers can have a comprehensive understanding in this fields.
Collapse
Affiliation(s)
- Guowen Hou
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, and Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China,Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Jinjie Li
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Wenyun Liu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Jinlong Wei
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, and Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China,Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China,*Correspondence: Ying Xin, ; Xin Jiang,
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, and Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China,Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China,*Correspondence: Ying Xin, ; Xin Jiang,
| |
Collapse
|
22
|
Pazzaglia S, Eidemüller M, Lumniczky K, Mancuso M, Ramadan R, Stolarczyk L, Moertl S. Out-of-field effects: lessons learned from partial body exposure. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2022; 61:485-504. [PMID: 36001144 PMCID: PMC9722818 DOI: 10.1007/s00411-022-00988-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/03/2022] [Indexed: 05/27/2023]
Abstract
Partial body exposure and inhomogeneous dose delivery are features of the majority of medical and occupational exposure situations. However, mounting evidence indicates that the effects of partial body exposure are not limited to the irradiated area but also have systemic effects that are propagated outside the irradiated field. It was the aim of the "Partial body exposure" session within the MELODI workshop 2020 to discuss recent developments and insights into this field by covering clinical, epidemiological, dosimetric as well as mechanistic aspects. Especially the impact of out-of-field effects on dysfunctions of immune cells, cardiovascular diseases and effects on the brain were debated. The presentations at the workshop acknowledged the relevance of out-of-field effects as components of the cellular and organismal radiation response. Furthermore, their importance for the understanding of radiation-induced pathologies, for the discovery of early disease biomarkers and for the identification of high-risk organs after inhomogeneous exposure was emphasized. With the rapid advancement of clinical treatment modalities, including new dose rates and distributions a better understanding of individual health risk is urgently needed. To achieve this, a deeper mechanistic understanding of out-of-field effects in close connection to improved modelling was suggested as priorities for future research. This will support the amelioration of risk models and the personalization of risk assessments for cancer and non-cancer effects after partial body irradiation.
Collapse
Affiliation(s)
- S. Pazzaglia
- Laboratory of Biomedical Technologies, ENEA CR-Casaccia, Via Anguillarese 301, 00123 Rome, Italy
| | - M. Eidemüller
- Institute of Radiation Medicine, Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - K. Lumniczky
- Department of Radiobiology and Radiohygiene, Unit of Radiation Medicine, National Public Health Centre, Albert Florian u. 2-6, 1097 Budapest, Hungary
| | - M. Mancuso
- Laboratory of Biomedical Technologies, ENEA CR-Casaccia, Via Anguillarese 301, 00123 Rome, Italy
| | - R. Ramadan
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - L. Stolarczyk
- Danish Centre for Particle Therapy, Palle Juul-Jensens Boulevard 25, 8200 Aarhus N, Denmark
| | - S. Moertl
- Federal Office for Radiation Protection, Ingolstädter Landstr. 1, 85764 Oberschleißheim, Germany
| |
Collapse
|
23
|
Zhang Y, Qiu L, Ren Y, Cheng Z, Li L, Yao S, Zhang C, Luo Z, Lu H. A meta-learning approach to improving radiation response prediction in cancers. Comput Biol Med 2022; 150:106163. [PMID: 37070625 DOI: 10.1016/j.compbiomed.2022.106163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/18/2022] [Accepted: 10/01/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE Predicting the efficacy of radiotherapy in individual patients has drawn widespread attention, but the limited sample size remains a bottleneck for utilizing high-dimensional multi-omics data to guide personalized radiotherapy. We hypothesize the recently developed meta-learning framework could address this limitation. METHODS AND MATERIALS By combining gene expression, DNA methylation, and clinical data of 806 patients who had received radiotherapy from The Cancer Genome Atlas (TCGA), we applied the Model-Agnostic Meta-Learning (MAML) framework to tasks consisting of pan-cancer data, to obtain the best initial parameters of a neural network for a specific cancer with smaller number of samples. The performance of meta-learning framework was compared with four traditional machine learning methods based on two training schemes, and tested on Cancer Cell Line Encyclopedia (CCLE) and Chinese Glioma Genome Atlas (CGGA) datasets. Moreover, biological significance of the models was investigated by survival analysis and feature interpretation. RESULTS The mean AUC (Area under the ROC Curve) [95% confidence interval] of our models across nine cancer types was 0.702 [0.691-0.713], which improved by 0.166 on average over other the four machine learning methods on two training schemes. Our models performed significantly better (p < 0.05) in seven cancer types and performed comparable to the other predictors in the rest of two cancer types. The more pan-cancer samples were used to transfer meta-knowledge, the greater the performance improved (p < 0.05). The predicted response scores that our models generated were negatively correlated with cell radiosensitivity index in four cancer types (p < 0.05), while not statistically significant in the other three cancer types. Moreover, the predicted response scores were shown to be prognostic factors in seven cancer types and eight potential radiosensitivity-related genes were identified. CONCLUSIONS For the first time, we established the meta-learning approach to improving individual radiation response prediction by transferring common knowledge from pan-cancer data with MAML framework. The results demonstrated the superiority, generalizability, and biological significance of our approach.
Collapse
|
24
|
Ma L, Ye Y, Lu H, Xing Y, Zhao Z, Quan C, Jia Z, Lu Y, Li Y, Zhou G. A Study on the Radiosensitivity of Radiation-Induced Lung Injury at the Acute Phase Based on Single-Cell Transcriptomics. Front Immunol 2022; 13:941976. [PMID: 35967301 PMCID: PMC9364823 DOI: 10.3389/fimmu.2022.941976] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/22/2022] [Indexed: 11/25/2022] Open
Abstract
Background and Aims Radiation-induced lung injury (RILI) is the most common complication associated with chest tumors, such as lung and breast cancers, after radiotherapy; however, the pathogenic mechanisms are unclear. Single-cell RNA sequencing has laid the foundation for studying RILI at the cellular microenvironmental level. This study focused on changes during the acute pneumonitis stage of RILI at the cellular microenvironmental level and investigated the interactions between different cell types. Methods An acute RILI model in mice and a single-cell transcriptional library were established. Intercellular communication networks were constructed to study the heterogeneity and intercellular interactions among different cell types. Results A single-cell transcriptome map was established in a mouse model of acute lung injury. In total, 18,500 single-cell transcripts were generated, and 10 major cell types were identified. The heterogeneity and radiosensitivity of each cell type or subtype in the lung tissues during the acute stage were revealed. It was found that immune cells had higher radiosensitivity than stromal cells. Immune cells were highly heterogeneous in terms of radiosensitivity, while some immune cells had the characteristics of radiation resistance. Two groups of radiation-induced Cd8+Mki67+ T cells and Cd4+Cxcr6+ helper T cells were identified. The presence of these cells was verified using immunofluorescence. The ligand-receptor interactions were analyzed by constructing intercellular communication networks. These explained the origins of the cells and revealed that they had been recruited from endothelial cells to the inflammatory site. Conclusions This study revealed the heterogeneity of in vivo radiosensitivity of different cell types in the lung at the initial stage post irradiation
Collapse
Affiliation(s)
- Luyu Ma
- Beijing Institute of Radiation Medicine, Beijing, China
- Department of Rehabilitation Medicine, Eighth Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yumeng Ye
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Hao Lu
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Yuan Xing
- The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Zhen Zhao
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Cheng Quan
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhaoqian Jia
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Yiming Lu
- Beijing Institute of Radiation Medicine, Beijing, China
- *Correspondence: Gangqiao Zhou, ; Yang Li, ; Yiming Lu,
| | - Yang Li
- Beijing Institute of Radiation Medicine, Beijing, China
- Department of Pharmacy, Academy of Life Sciences, Anhui Medical University, Hefei, China
- *Correspondence: Gangqiao Zhou, ; Yang Li, ; Yiming Lu,
| | - Gangqiao Zhou
- Beijing Institute of Radiation Medicine, Beijing, China
- Collaborative Innovation Center for Personalized Cancer Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- *Correspondence: Gangqiao Zhou, ; Yang Li, ; Yiming Lu,
| |
Collapse
|
25
|
Single-Cell Transcriptome Analysis of Radiation Pneumonitis Mice. Antioxidants (Basel) 2022; 11:antiox11081457. [PMID: 35892659 PMCID: PMC9331247 DOI: 10.3390/antiox11081457] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/14/2022] [Accepted: 07/22/2022] [Indexed: 12/10/2022] Open
Abstract
Radiation-induced lung injury (RILI), especially radiation pneumonitis (RP), is a common clinical complication associated with thoracic radiotherapy for malignant tumors. However, the specific contributions of each cell subtype to this process are unknown. Here, we provide the single-cell pathology landscape of the RP in a mouse model by unbiased single-cell RNA-seq (scRNA-seq). We found a decline of type 2 alveolar cells in the RP lung tissue, with an expansion of macrophages, especially the Fabp4low and Spp1high subgroup, while Fabp4high macrophages were almost depleted. We observed an elevated expression of multiple mitochondrial genes in the RP group, indicating a type 2 alveolar cell (AT2) response to oxidative stress. We also calculated the enrichment of a cGAS-STING signaling pathway, which may be involved in regulating inflammatory responses and cancer progression in AT2 cells of PR mice. We delineate markers and transcriptional states, identify a type 2 alveolar cell, and uncover fundamental determinants of lung fibrosis and inflammatory response in RP lung tissue of mice.
Collapse
|
26
|
Zhang XZ, Chen MJ, Fan PM, Su TS, Liang SX, Jiang W. Prediction of the Mechanism of Sodium Butyrate against Radiation-Induced Lung Injury in Non-Small Cell Lung Cancer Based on Network Pharmacology and Molecular Dynamic Simulations and Molecular Dynamic Simulations. Front Oncol 2022; 12:809772. [PMID: 35837112 PMCID: PMC9275827 DOI: 10.3389/fonc.2022.809772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundRadiation-induced lung injury (RILI) is a severe side effect of radiotherapy for non-small cell lung cancer (NSCLC) ,and one of the major hindrances to improve the efficacy of radiotherapy. Previous studies have confirmed that sodium butyrate (NaB) has potential of anti-radiation toxicity. However, the mechanism of the protective effect of NaB against RILI has not yet been clarified. This study aimed to explore the underlying protective mechanisms of NaB against RILI in NSCLC through network pharmacology, molecular docking, molecular dynamic simulations and in vivo experiments.MethodsThe predictive target genes of NaB were obtained from the PharmMapper database and the literature review. The involved genes of RILI and NSCLC were predicted using OMIM and GeneCards database. The intersectional genes of drug and disease were identified using the Venny tool and uploaded to the Cytoscape software to identify 5 core target genes of NaB associated with RILI. The correlations between the 5 core target genes and EGFR, PD-L1, immune infiltrates, chemokines and chemokine receptors were analyzed using TIMER 2.0, TIMER and TISIDB databases. We constructed the mechanism maps of the 3 key signaling pathways using the KEGG database based on the results of GO and KEGG analyses from Metascape database. The 5 core target genes and drug were docked using the AutoDock Vina tool and visualized using PyMOL software. GROMACS software was used to perform 100 ns molecular dynamics simulation. Irradiation-induced lung injury model in mice were established to assess the therapeutic effects of NaB.ResultsA total of 51 intersectional genes involved in NaB against RILI in NSCLC were identified. The 5 core target genes were AKT1, TP53, NOTCH1, SIRT1, and PTEN. The expressions of the 5 core target genes were significantly associated with EGFR, PD-L1, immune infiltrates, chemokines and chemokine receptors, respectively. The results from GO analysis of the 51 intersectional genes revealed that the biological processes were focused on the regulation of smooth muscle cell proliferation, oxidative stress and cell death, while the three key KEGG pathways were enriched in PI3K-Akt signal pathway, p53 signal pathway, and FOXO signal pathway. The docking of NaB with the 5 core target genes showed affinity and stability, especially AKT1. In vivo experiments showed that NaB treatment significantly protected mice from RILI, with reduced lung histological damage. In addition, NaB treatment significantly inhibited the PI3K/Akt signaling pathway.ConclusionsNaB may protect patients from RILI in NSCLC through multiple target genes including AKT1, TP53, NOTCH1, SIRT1 and PTEN, with multiple signaling pathways involving, including PI3K-Akt pathway, p53 pathway, and FOXO pathways. Our findings effectively provide a feasible theoretical basis to further elucidate the mechanism of NaB in the treatment of RILI.
Collapse
Affiliation(s)
- Xiao-zhen Zhang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Mao-jian Chen
- Department of Respiratory Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ping-ming Fan
- Department of Breast-Thoracic Tumor Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Ting-shi Su
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Shi-xiong Liang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
- *Correspondence: Wei Jiang, ; Shi-xiong Liang,
| | - Wei Jiang
- Department of Respiratory Oncology, Guangxi Medical University Cancer Hospital, Nanning, China
- *Correspondence: Wei Jiang, ; Shi-xiong Liang,
| |
Collapse
|
27
|
Lapierre A, Bourillon L, Larroque M, Gouveia T, Bourgier C, Ozsahin M, Pèlegrin A, Azria D, Brengues M. Improving Patients' Life Quality after Radiotherapy Treatment by Predicting Late Toxicities. Cancers (Basel) 2022; 14:2097. [PMID: 35565227 PMCID: PMC9099838 DOI: 10.3390/cancers14092097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/14/2022] [Accepted: 04/16/2022] [Indexed: 11/26/2022] Open
Abstract
Personalized treatment and precision medicine have become the new standard of care in oncology and radiotherapy. Because treatment outcomes have considerably improved over the last few years, permanent side-effects are becoming an increasingly significant issue for cancer survivors. Five to ten percent of patients will develop severe late toxicity after radiotherapy. Identifying these patients before treatment start would allow for treatment adaptation to minimize definitive side effects that could impair their long-term quality of life. Over the last decades, several tests and biomarkers have been developed to identify these patients. However, out of these, only the Radiation-Induced Lymphocyte Apoptosis (RILA) assay has been prospectively validated in multi-center cohorts. This test, based on a simple blood draught, has been shown to be correlated with late radiation-induced toxicity in breast, prostate, cervical and head and neck cancer. It could therefore greatly improve decision making in precision radiation oncology. This literature review summarizes the development and bases of this assay, as well as its clinical results and compares its results to the other available assays.
Collapse
Affiliation(s)
- Ariane Lapierre
- IRCM, INSERM, University Montpellier, ICM, 34298 Montpellier, France; (A.L.); (L.B.); (M.L.); (T.G.); (C.B.); (A.P.); (D.A.)
- Department of Radiotherapy-Oncology, Lyon-Sud Hospital Center, 69310 Pierre-Bénite, France
| | - Laura Bourillon
- IRCM, INSERM, University Montpellier, ICM, 34298 Montpellier, France; (A.L.); (L.B.); (M.L.); (T.G.); (C.B.); (A.P.); (D.A.)
| | - Marion Larroque
- IRCM, INSERM, University Montpellier, ICM, 34298 Montpellier, France; (A.L.); (L.B.); (M.L.); (T.G.); (C.B.); (A.P.); (D.A.)
| | - Tiphany Gouveia
- IRCM, INSERM, University Montpellier, ICM, 34298 Montpellier, France; (A.L.); (L.B.); (M.L.); (T.G.); (C.B.); (A.P.); (D.A.)
| | - Céline Bourgier
- IRCM, INSERM, University Montpellier, ICM, 34298 Montpellier, France; (A.L.); (L.B.); (M.L.); (T.G.); (C.B.); (A.P.); (D.A.)
| | | | - André Pèlegrin
- IRCM, INSERM, University Montpellier, ICM, 34298 Montpellier, France; (A.L.); (L.B.); (M.L.); (T.G.); (C.B.); (A.P.); (D.A.)
| | - David Azria
- IRCM, INSERM, University Montpellier, ICM, 34298 Montpellier, France; (A.L.); (L.B.); (M.L.); (T.G.); (C.B.); (A.P.); (D.A.)
| | - Muriel Brengues
- IRCM, INSERM, University Montpellier, ICM, 34298 Montpellier, France; (A.L.); (L.B.); (M.L.); (T.G.); (C.B.); (A.P.); (D.A.)
| |
Collapse
|
28
|
DeDreu J, Pal-Ghosh S, Mattapallil MJ, Caspi RR, Stepp MA, Menko AS. Uveitis-mediated immune cell invasion through the extracellular matrix of the lens capsule. FASEB J 2021; 36:e21995. [PMID: 34874579 PMCID: PMC9300120 DOI: 10.1096/fj.202101098r] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/20/2021] [Accepted: 10/04/2021] [Indexed: 12/05/2022]
Abstract
While the eye is considered an immune privileged site, its privilege is abrogated when immune cells are recruited from the surrounding vasculature in response to trauma, infection, aging, and autoimmune diseases like uveitis. Here, we investigate whether in uveitis immune cells become associated with the lens capsule and compromise its privilege in studies of C57BL/6J mice with experimental autoimmune uveitis. These studies show that at D14, the peak of uveitis in these mice, T cells, macrophages, and Ly6G/Ly6C+ immune cells associate with the lens basement membrane capsule, burrow into the capsule matrix, and remain integrated with the capsule as immune resolution is occurring at D26. 3D surface rendering image analytics of confocal z‐stacks and scanning electron microscopy imaging of the lens surface show the degradation of the lens capsule as these lens‐associated immune cells integrate with and invade the lens capsule, with a subset infiltrating both epithelial and fiber cell regions of lens tissue, abrogating its immune privilege. Those immune cells that remain on the surface often become entwined with a fibrillar net‐like structure. Immune cell invasion of the lens capsule in uveitis has not been described previously and may play a role in induction of lens and other eye pathologies associated with autoimmunity.
Collapse
Affiliation(s)
- JodiRae DeDreu
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Sonali Pal-Ghosh
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Mary J Mattapallil
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Rachel R Caspi
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mary Ann Stepp
- Department of Anatomy and Cell Biology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA.,Department of Ophthalmology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - A Sue Menko
- Department of Pathology, Anatomy and Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Department of Ophthalmology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
29
|
Liu X, Shao C, Fu J. Promising Biomarkers of Radiation-Induced Lung Injury: A Review. Biomedicines 2021; 9:1181. [PMID: 34572367 PMCID: PMC8470495 DOI: 10.3390/biomedicines9091181] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/05/2021] [Accepted: 09/06/2021] [Indexed: 12/15/2022] Open
Abstract
Radiation-induced lung injury (RILI) is one of the main dose-limiting side effects in patients with thoracic cancer during radiotherapy. No reliable predictors or accurate risk models are currently available in clinical practice. Severe radiation pneumonitis (RP) or pulmonary fibrosis (PF) will reduce the quality of life, even when the anti-tumor treatment is effective for patients. Thus, precise prediction and early diagnosis of lung toxicity are critical to overcome this longstanding problem. This review summarizes the primary mechanisms and preclinical animal models of RILI reported in recent decades, and analyzes the most promising biomarkers for the early detection of lung complications. In general, ideal integrated models considering individual genetic susceptibility, clinical background parameters, and biological variations are encouraged to be built up, and more prospective investigations are still required to disclose the molecular mechanisms of RILI as well as to discover valuable intervention strategies.
Collapse
Affiliation(s)
- Xinglong Liu
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China;
| | - Chunlin Shao
- Institute of Radiation Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China;
| | - Jiamei Fu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| |
Collapse
|
30
|
Corso CR, Mulinari Turin de Oliveira N, Maria-Ferreira D. Susceptibility to SARS-CoV-2 infection in patients undergoing chemotherapy and radiation therapy. J Infect Public Health 2021; 14:766-771. [PMID: 34022735 PMCID: PMC7980522 DOI: 10.1016/j.jiph.2021.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 02/22/2021] [Accepted: 03/14/2021] [Indexed: 01/08/2023] Open
Abstract
The outbreak of the new coronavirus disease (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has rapidly become a public health emergency of international concern, especially affecting the elderly people and patients with chronic disease, such as hypertension and respiratory syndromes. Patients undergoing chemotherapy treatment (e.g., bleomycin, cyclophosphamide, methotrexate, monoclonal antibodies, and paclitaxel therapy) are vulnerable to the development of respiratory syndromes induced by chemotherapeutic agents and are also more susceptible to viral infections as they are immunosuppressed. Neutropenia is an important risk factor for increased vulnerability to infections, as a respiratory syndrome involves an array of immune cells maintaining the balance between pathogen clearance and immunopathology. However, the differential diagnosis of pulmonary symptoms in cancer patients is broad, with complications being related to the malignancy itself, treatment toxicity, and infections. The risk factors depend on the specific type of cancer, chemotherapy, patient characteristics, and comorbidities. Thus, this review discusses the main events implicated in immunosuppression caused by chemotherapy and radiation therapy and the association of immunosuppression and other factors with SARS-CoV-2 infection susceptibility in cancer patients; and, importantly, how to deal with this situation in face of the current pandemic scenario.
Collapse
Affiliation(s)
- Claudia Rita Corso
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Paraná, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, Paraná, Brazil
| | - Natalia Mulinari Turin de Oliveira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Paraná, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, Paraná, Brazil
| | - Daniele Maria-Ferreira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Paraná, Brazil; Programa de Pós-graduação em Biotecnologia Aplicada à Saúde da Criança e do Adolescente, Faculdades Pequeno Príncipe, Curitiba, Paraná, Brazil.
| |
Collapse
|
31
|
Wang HT, Han JT, Hu DH. [Research advances on the role and mechanism of inflammatory response in the formation of hypertrophic scars and keloids]. ZHONGHUA SHAO SHANG ZA ZHI = ZHONGHUA SHAOSHANG ZAZHI = CHINESE JOURNAL OF BURNS 2021; 37:490-494. [PMID: 34044530 PMCID: PMC11917354 DOI: 10.3760/cma.j.cn501120-20200310-00143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Hypertrophic scars and keloids are common sequelae after wound healing, with a high incidence, which seriously affect the patient's quality of life. However, there is still a lack of effective prevention and treatment methods, mainly because the pathogenesis of scars is not clear. Current research believe that inflammatory response plays a critical role in the process of scar formation, and through the researches on the mechanisms it is hopeful to find new potential therapeutic drug targets for the prevention and treatment of hypertrophic scars and keloids. This article mainly reviews the research progresses on the role and mechanism of inflammatory cytokines and inflammatory cells in the formation of hypertrophic scars and keloids, as well as drugs, microRNAs, and exosomes, etc., for the treatment of hypertrophic scars and keloids by inhibiting inflammatory response.
Collapse
Affiliation(s)
- H T Wang
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - J T Han
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| | - D H Hu
- Department of Burns and Cutaneous Surgery, Burn Center of PLA, the First Affiliated Hospital of Air Force Medical University, Xi'an 710032, China
| |
Collapse
|