1
|
Dunne VL, Ghita-Pettigrew M, Redmond KM, Small DM, Weldon S, Taggart CC, Prise KM, Hanna GG, Butterworth KT. PTEN Depletion Increases Radiosensitivity in Response to Ataxia Telangiectasia-Related-3 (ATR) Inhibition in Non-Small Cell Lung Cancer (NSCLC). Int J Mol Sci 2024; 25:7817. [PMID: 39063060 PMCID: PMC11277409 DOI: 10.3390/ijms25147817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Radiotherapy (RT) treatment is an important strategy for the management of non-small cell lung cancer (NSCLC). Local recurrence amongst patients with late-stage NSCLC remains a challenge. The loss of PTEN has been associated with radio-resistance. This study aimed to examine the efficacy of RT combined with ataxia telangiectasia-mutated Rad3-related (ATR) inhibition using Ceralasertib in phosphatase and tensin homolog (PTEN)-depleted NSCLC cells and to assess early inflammatory responses indicative of radiation pneumonitis (RP) after combined-modality treatment. Small hairpin RNA (shRNA) transfections were used to generate H460 and A549 PTEN-depleted models. Ceralasertib was evaluated as a single agent and in combination with RT in vitro and in vivo. Histological staining was used to assess immune cell infiltration in pneumonitis-prone C3H/NeJ mice. Here, we report that the inhibition of ATR in combination with RT caused a significant reduction in PTEN-depleted NSCLC cells, with delayed DNA repair and reduced cell viability, as shown by an increase in cells in Sub G1. Combination treatment in vivo significantly inhibited H460 PTEN-depleted tumour growth in comparison to H460 non-targeting PTEN-expressing (NT) cell-line-derived xenografts (CDXs). Additionally, there was no significant increase in infiltrating macrophages or neutrophils except at 4 weeks, whereby combination treatment significantly increased macrophage levels relative to RT alone. Overall, our study demonstrates that ceralasertib and RT combined preferentially sensitises PTEN-depleted NSCLC models in vitro and in vivo, with no impact on early inflammatory response indicative of RP. These findings provide a rationale for evaluating ATR inhibition in combination with RT in NSCLC patients with PTEN mutations.
Collapse
Affiliation(s)
- Victoria L. Dunne
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (M.G.-P.); (K.M.R.); (D.M.S.); (K.M.P.); (K.T.B.)
| | - Mihaela Ghita-Pettigrew
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (M.G.-P.); (K.M.R.); (D.M.S.); (K.M.P.); (K.T.B.)
| | - Kelly M. Redmond
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (M.G.-P.); (K.M.R.); (D.M.S.); (K.M.P.); (K.T.B.)
| | - Donna M. Small
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (M.G.-P.); (K.M.R.); (D.M.S.); (K.M.P.); (K.T.B.)
| | - Sinéad Weldon
- Airway Innate Immunity Research Group (AiiR), Wellcome Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7AE, UK; (S.W.); (C.C.T.)
| | - Clifford C. Taggart
- Airway Innate Immunity Research Group (AiiR), Wellcome Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast BT9 7AE, UK; (S.W.); (C.C.T.)
| | - Kevin M. Prise
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (M.G.-P.); (K.M.R.); (D.M.S.); (K.M.P.); (K.T.B.)
| | - Gerard G. Hanna
- Northern Ireland Cancer Centre, Belfast Health and Social Care Trust, Belfast BT9 7AB, UK;
| | - Karl T. Butterworth
- Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, Belfast BT9 7AE, UK; (M.G.-P.); (K.M.R.); (D.M.S.); (K.M.P.); (K.T.B.)
| |
Collapse
|
2
|
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: 1.0] [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
|
3
|
Wang H, Gao Y, Wang L, Yu Y, Zhang J, Liu C, Song Y, Xu H, Wang J, Lou H, Dong T. Lung specific homing of diphenyleneiodonium chloride improves pulmonary fibrosis by inhibiting macrophage M2 metabolic program. J Adv Res 2023; 44:213-225. [PMID: 36725191 PMCID: PMC9936526 DOI: 10.1016/j.jare.2022.04.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Pulmonary fibrosis (PF) is a fatal disease with a variable and unpredictable course. Effective clinical treatment for PF remains a challenge due to low drug accumulation in lungs and imbalanced polarization of pro/anti-fibrotic macrophages. OBJECTIVES To identify the alteration of immunometabolism in the pulmonary macrophages and investigate the feasibility of specific inhibition of M2 activation of macrophages as an effective anti-PF strategy in vivo. METHODS The high-content screening system was used to select lung-specific homing compounds that can modulate macrophage polarization. Imaging mass spectrometry (IMS) conjugated with chemical proteomics approach was conducted to explore the cells and proteins targeted by diphenyleneiodonium chloride (DPI). A bleomycin-induced fibrotic mouse model was established to examine the in vivo effect of DPI. RESULTS Pulmonary macrophages of PF at late stage exhibited predominantly the M2 phenotype with decreased glycolysis metabolism. DPI was demonstrated to inhibit profibrotic activation of macrophages in the preliminary screening. Notably, IMS conjugated with chemical proteomics approach revealed DPI specifically targeted pulmonary macrophages, leading to the efficient protection from bleomycin-induced pulmonary fibrosis in mice. Mechanistically, DPI upregulated glycolysis and suppressed M2 programming in fibrosis mice, thus resulting in pro-fibrotic cytokine inhibition, hydroxyproline biosynthesis, and collagen deposition, with a concomitant increase in alveolar airspaces. CONCLUSIONS DPI mediated glycolysis in lung and accordingly suppressed M2 programming, resulting in improved lung fibrosis.
Collapse
Affiliation(s)
- Huirui Wang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yinghui Gao
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Li Wang
- Respiratory Medicines, The Affiliated Hospital of Yanan University, Yan'an, Shaanxi, China
| | - Yang Yu
- Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiaozhen Zhang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunyu Liu
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yaxin Song
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haochuan Xu
- Peking University Health Science Center, Beijing, China
| | - Jingcheng Wang
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hongxiang Lou
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Ting Dong
- Department of Natural Product Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
4
|
Guo X, Du L, Ma N, Zhang P, Wang Y, Han Y, Huang X, Zhang Q, Tan X, Lei X, Qu B. Monophosphoryl lipid A ameliorates radiation-induced lung injury by promoting the polarization of macrophages to the M1 phenotype. J Transl Med 2022; 20:597. [DOI: 10.1186/s12967-022-03804-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
Abstract
Background
Radiation-induced lung injury (RILI) often occurs during clinical chest radiotherapy and acute irradiation from accidental nuclear leakage. This study explored the role of monophosphoryl lipid A (MPLA) in RILI.
Materials and Methods
The entire thoracic cavity of C57BL/6N mice was irradiated at 20 Gy with or without pre-intragastric administration of MPLA. HE staining, Masson trichrome staining, and TUNEL assay were used to assess lung tissue injury after treatment. The effect of irradiation on the proliferation of MLE-12 cells was analyzed using the Clonogenic assay. The effect of MPLA on the apoptosis of MLE-12 cells was analyzed using flow cytometry. Expression of γ-H2AX and epithelial-mesenchymal transition (EMT) markers in MLE-12 cells was detected by immunofluorescence and Western blot, respectively.
Results
MPLA attenuated early pneumonitis and late pulmonary fibrosis after thoracic irradiation and reversed radiation-induced EMT in C57 mice. MPLA further promoted proliferation and inhibited apoptosis of irradiated MLE-12 cells in vitro. Mechanistically, the radioprotective effect of MPLA was mediated by exosomes secreted by stimulated macrophages. Macrophage-derived exosomes modulated DNA damage in MLE-12 cells after irradiation. MPLA promoted the polarization of RAW 264.7 cells to the M1 phenotype. The exosomes secreted by M1 macrophages suppressed EMT in MLE-12 cells after irradiation.
Conclusion
MPLA is a novel treatment strategy for RILI. Exosomes derived from macrophages are key to the radioprotective role of MPLA in RILI.
Collapse
|
5
|
Abstract
We investigated the dynamics of the gene expression of M1 and M2 macrophage markers during skin wound healing in mice. Expression of M1-macrophage markers, such as Il12a, Tnf, Il6, Il1b, and Nos2 was upregulated after wounding and peaked at 1 or 3 days after injury, and that of M2-macrophage markers such as Mrc1, Cd163, Ccl17, Arg, and Tgfb1, peaked at 6 days after injury. Consistent with these findings, using triple-color immunofluorescence analysis revealed that F4/80+CD80+ M1 macrophages were more abundant than F4/80+CD206+ M2 macrophages on day 3 in mouse wound specimens, and that M2 macrophages were prominently detected in day 6 wounds. For application in forensic practice, we examined macrophage polarization using human wound specimens. The average ratios of CD68+iNOS+ M1 macrophages to CD68+CD163+ M2 macrophages (M1/M2 ratios) were greater than 2.5 for the wounds aged 2-5 days. Out of 11 wounds aged 1-5 days, five samples had the M1/M2 ratios of > 3.0. These observations propose that the M1/M2 ratios of 3.0 would indicate a wound age of 1-5 days as the forensic opinion. This study showed that M1 and M2 macrophages in human skin wound might be a promising marker for wound age determination.
Collapse
|
6
|
Kim SY, Kim JM, Lee SR, Kim HJ, Lee JH, Choi HL, Lee YJ, Lee YS, Cho J. Efferocytosis and enhanced FPR2 expression following apoptotic cell instillation attenuate radiation-induced lung inflammation and fibrosis. Biochem Biophys Res Commun 2022; 601:38-44. [DOI: 10.1016/j.bbrc.2022.02.075] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/15/2022] [Accepted: 02/19/2022] [Indexed: 12/13/2022]
|
7
|
Cartwright JK, Moreno FG. Presentation of Eagle syndrome following radiation therapy to carcinoma of the larynx. Clin Case Rep 2022; 10:e05325. [PMID: 35127092 PMCID: PMC8795837 DOI: 10.1002/ccr3.5325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/05/2021] [Accepted: 01/14/2022] [Indexed: 11/16/2022] Open
Abstract
Eagle syndrome is a rare clinical condition that is characterized by either an elongated styloid process or a calcified stylohyoid ligament. This report describes the case of a 35-year-old woman who presented with Eagle syndrome following the treatment of recurrent laryngeal carcinoma with ionizing radiation.
Collapse
Affiliation(s)
- Jake K. Cartwright
- Quillen College of MedicineEast Tennessee State UniversityMountain HomeTennesseeUSA
| | - Francisco G. Moreno
- Otolaryngology—Head and Neck SurgeryFacial Plastic SurgeryKnoxvilleTennesseeUSA
| |
Collapse
|
8
|
Nepon H, Safran T, Reece EM, Murphy AM, Vorstenbosch J, Davison PG. Radiation-Induced Tissue Damage: Clinical Consequences and Current Treatment Options. Semin Plast Surg 2021; 35:181-188. [PMID: 34526866 PMCID: PMC8432995 DOI: 10.1055/s-0041-1731464] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Radiation therapy is a valuable tool in the treatment of numerous malignancies but, in certain cases, can also causes significant acute and chronic damage to noncancerous neighboring tissues. This review focuses on the pathophysiology of radiation-induced damage and the clinical implications it has for plastic surgeons across breast reconstruction, osteoradionecrosis, radiation-induced skin cancers, and wound healing. The current understanding of treatment modalities presented here include hyperbaric oxygen therapy, autologous fat grafting and stem cells, and pharmaceutical agents.
Collapse
Affiliation(s)
- Hillary Nepon
- Division of Experimental Surgery, McGill University, Montreal, Quebec, Canada
| | - Tyler Safran
- Division of Plastic Surgery, McGill University, Montreal, Quebec, Canada
| | - Edward M. Reece
- Michael E. DeBakey Department of Surgery, Division of Plastic Surgery, Baylor College of Medicine, Houston, Texas
| | - Amanda M. Murphy
- Division of Plastic Surgery, McGill University, Montreal, Quebec, Canada
| | | | - Peter G. Davison
- Division of Plastic Surgery, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Ying H, Fang M, Hang QQ, Chen Y, Qian X, Chen M. Pirfenidone modulates macrophage polarization and ameliorates radiation-induced lung fibrosis by inhibiting the TGF-β1/Smad3 pathway. J Cell Mol Med 2021; 25:8662-8675. [PMID: 34327818 PMCID: PMC8435416 DOI: 10.1111/jcmm.16821] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 07/05/2021] [Accepted: 07/13/2021] [Indexed: 01/10/2023] Open
Abstract
Radiation-induced lung injury (RILI) mainly contributes to the complications of thoracic radiotherapy. RILI can be divided into radiation pneumonia (RP) and radiation-induced lung fibrosis (RILF). Once RILF occurs, patients will eventually develop irreversible respiratory failure; thus, a new treatment strategy to prevent RILI is urgently needed. This study explored the therapeutic effect of pirfenidone (PFD), a Food and Drug Administration (FDA)-approved drug for (IPF) treatment, and its mechanism in the treatment of RILF. In vivo, C57BL/6 mice received a 50 Gy dose of X-ray radiation to the whole thorax with or without the administration of PFD. Collagen deposition and fibrosis in the lung were reversed by PFD treatment, which was associated with reduced M2 macrophage infiltration and inhibition of the transforming growth factor-β1 (TGF-β1)/Drosophila mothers against the decapentaplegic 3 (Smad3) signalling pathway. Moreover, PFD treatment decreased the radiation-induced expression of TGF-β1 and phosphorylation of Smad3 in alveolar epithelial cells (AECs) and vascular endothelial cells (VECs). Furthermore, IL-4-induced M2 macrophage polarization and IL-13-induced M2 macrophage polarization were suppressed by PFD treatment in vitro, resulting in reductions in the release of arginase-1 (ARG-1), chitinase 3-like 3 (YM-1) and TGF-β1. Notably, the PFD-induced inhibitory effects on M2 macrophage polarization were associated with downregulation of nuclear factor kappa-B (NF-κB) p50 activity. Additionally, PFD could significantly inhibit ionizing radiation-induced chemokine secretion in MLE-12 cells and consequently impair the migration of RAW264.7 cells. PFD could also eliminate TGF-β1 from M2 macrophages by attenuating the activation of TGF-β1/Smad3. In conclusion, PFD is a potential therapeutic agent to ameliorate fibrosis in RILF by reducing M2 macrophage infiltration and inhibiting the activation of TGF-β1/Smad3.
Collapse
Affiliation(s)
- Hangjie Ying
- Institute of Basic Medicine and Cancer (IBMC), The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, China.,Zhejiang Key Laboratory of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Min Fang
- Institute of Basic Medicine and Cancer (IBMC), The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, China.,Zhejiang Key Laboratory of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China.,The Department of Thoracic Radiotherapy, Zhejiang Cancer Hospital, Hangzhou, China
| | - Qing Qing Hang
- The Second Clinical Medical College of Zhejiang, Chinese Medical University, Hangzhou, China
| | - Yamei Chen
- Institute of Basic Medicine and Cancer (IBMC), The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, China.,Zhejiang Key Laboratory of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xu Qian
- Institute of Basic Medicine and Cancer (IBMC), The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, China.,The Department of Clinical Laboratory, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ming Chen
- Institute of Basic Medicine and Cancer (IBMC), The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital, Chinese Academy of Sciences, Hangzhou, China.,Zhejiang Key Laboratory of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou, China.,The Department of Thoracic Radiotherapy, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
10
|
Rana T, Jiang C, Liu G, Miyata T, Antony V, Thannickal VJ, Liu RM. PAI-1 Regulation of TGF-β1-induced Alveolar Type II Cell Senescence, SASP Secretion, and SASP-mediated Activation of Alveolar Macrophages. Am J Respir Cell Mol Biol 2020; 62:319-330. [PMID: 31513752 PMCID: PMC7055702 DOI: 10.1165/rcmb.2019-0071oc] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 09/11/2019] [Indexed: 12/16/2022] Open
Abstract
Senescence of alveolar type II (ATII) cells, progenitors of the alveolar epithelium, is a pathological feature and contributes importantly to the pathogenesis of idiopathic pulmonary fibrosis. Despite recognition of the importance of ATII cell senescence in idiopathic pulmonary fibrosis pathogenesis, how ATII cell senescence is regulated and how senescent ATII cells contribute to lung fibrogenesis remain unclear. In this study, we show that TGF-β1 (transforming growth factor-β1), a most ubiquitous and potent profibrotic cytokine, induces plasminogen activator inhibitor-1 (PAI-1), a cell senescence and fibrosis mediator, and p16 as well as senescence, but not apoptosis, in primary mouse ATII cells. We also found that senescent ATII cells secrete various cytokines and chemokines, including IL-4 and IL-13, which stimulate the expression of genes associated with a profibrotic phenotype in alveolar macrophages. Similar responses were also observed in TGF-β1-treated rat ATII (L2) and rat macrophage NR8383 cells. Deletion of PAI-1 or inhibition of PAI-1 activity with a small molecule PAI-1 inhibitor, however, blocks TGF-β1-induced senescence as well as a senescence-associated secretory phenotype in ATII and L2 cells and, consequently, the stimulatory effects of the conditioned medium from senescent ATII/L2 cells on macrophages. Moreover, we show that silencing p16 ameliorates PAI-1 protein-induced ATII cell senescence and secretion of profibrotic mediators. Our data suggest that PAI-1 mediates TGF-β1-induced ATII cell senescence and secretion of profibrotic mediators through inducing p16, and they also suggest that senescent ATII cells contribute to lung fibrogenesis in part by activating alveolar macrophages through secreting profibrotic and proinflammatory mediators.
Collapse
Affiliation(s)
- Tapasi Rana
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Chunsun Jiang
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Gang Liu
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Toshio Miyata
- United Centers for Advanced Research and Translational Medicine, Tohoku University, Tohoku, Japan
| | - Veena Antony
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Victor J. Thannickal
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Rui-Ming Liu
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| |
Collapse
|
11
|
Beach TA, Groves AM, Williams JP, Finkelstein JN. Modeling radiation-induced lung injury: lessons learned from whole thorax irradiation. Int J Radiat Biol 2020; 96:129-144. [PMID: 30359147 PMCID: PMC6483900 DOI: 10.1080/09553002.2018.1532619] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/12/2018] [Accepted: 09/13/2018] [Indexed: 12/25/2022]
Abstract
Models of thoracic irradiation have been developed as clinicians and scientists have attempted to decipher the events that led up to the pulmonary toxicity seen in human subjects following radiation treatment. The most common model is that of whole thorax irradiation (WTI), applied in a single dose. Mice, particularly the C57BL/6J strain, has been frequently used in these investigations, and has greatly informed our current understanding of the initiation and progression of radiation-induced lung injury (RILI). In this review, we highlight the sequential progression and dynamic nature of RILI, focusing primarily on the vast array of information that has been gleaned from the murine model. Ample evidence indicates a wide array of biological responses that can be seen following irradiation, including DNA damage, oxidative stress, cellular senescence and inflammation, all triggered by the initial exposure to ionizing radiation (IR) and heterogeneously maintained throughout the temporal progression of injury, which manifests as acute pneumonitis and later fibrosis. It appears that the early responses of specific cell types may promote further injury, disrupting the microenvironment and preventing a return to homeostasis, although the exact mechanisms driving these responses remains somewhat unclear. Attempts to either prevent or treat RILI in preclinical models have shown some success by targeting these disparate radiobiological processes. As our understanding of the dynamic cellular responses to radiation improves through the use of such models, so does the likelihood of preventing or treating RILI.
Collapse
Affiliation(s)
- Tyler A. Beach
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642
- These authors contributed equally to this publication
| | - Angela M. Groves
- Department of Pediatrics and Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
- These authors contributed equally to this publication
| | - Jacqueline P. Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY 14642
| | - Jacob N. Finkelstein
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642
- Department of Pediatrics and Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642
| |
Collapse
|
12
|
de Leve S, Wirsdörfer F, Jendrossek V. Targeting the Immunomodulatory CD73/Adenosine System to Improve the Therapeutic Gain of Radiotherapy. Front Immunol 2019; 10:698. [PMID: 31024543 PMCID: PMC6460721 DOI: 10.3389/fimmu.2019.00698] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 03/14/2019] [Indexed: 12/23/2022] Open
Abstract
Extracellular adenosine is a potent endogenous immunosuppressive mediator critical to the maintenance of homeostasis in various normal tissues including the lung. Adenosine is either released from stressed or injured cells or generated from extracellular adenine nucleotides by the concerted action of the ectoenzymes ectoapyrase (CD39) and 5′ ectonucleotidase (CD73) that catabolize ATP to adenosine. An acute CD73-dependent increase of adenosine in normal tissues mostly exerts tissue protective functions whereas chronically increased adenosine-levels in tissues exposed to DNA damaging chemotherapy or radiotherapy promote pathologic remodeling processes and fibrosis for example in the skin and the lung. Importantly, cancer cells also express CD73 and high CD73 expression in the tumor tissue has been linked to poor overall survival and recurrence free survival in patients suffering from breast and ovarian cancer. CD73 and adenosine support growth-promoting neovascularization, metastasis, and survival in cancer cells. In addition, adenosine can promote tumor intrinsic or therapy-induced immune escape by various mechanisms that dampen the immune system. Consequently, modulating CD73 or cancer-derived adenosine in the tumor microenvironment emerges as an attractive novel therapeutic strategy to limit tumor progression, improve antitumor immune responses, avoid therapy-induced immune deviation, and potentially limit normal tissue toxicity. However, the role of CD73/adenosine signaling in the tumor and normal tissue responses to radiotherapy and its use as therapeutic target to improve the outcome of radiotherapy approaches is less understood. The present review will highlight the dual role of CD73 and adenosine in tumor and tissue responses to radiotherapy with a special focus to the lung. It will also discuss the potential benefits and risks of pharmacologic modulation of the CD73/adenosine system to increase the therapeutic gain of radiotherapy or combined radioimmunotherapy in cancer treatment.
Collapse
Affiliation(s)
- Simone de Leve
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
13
|
Zhou C, Moustafa MR, Cao L, Kriegsmann M, Winter M, Schwager C, Jones B, Wang S, Bäuerle T, Zhou PK, Schnölzer M, Weichert W, Debus J, Abdollahi A. Modeling and multiscale characterization of the quantitative imaging based fibrosis index reveals pathophysiological, transcriptome and proteomic correlates of lung fibrosis induced by fractionated irradiation. Int J Cancer 2019; 144:3160-3173. [PMID: 30536712 PMCID: PMC6590477 DOI: 10.1002/ijc.32059] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 11/15/2018] [Accepted: 11/21/2018] [Indexed: 12/24/2022]
Abstract
Pulmonary fibrosis represents a leading cause of morbidity and mortality worldwide. Therapy induced lung fibrosis constitutes a pivotal dose‐limiting side effect of radiotherapy and other anticancer agents. We aimed to develop objective criteria for assessment of fibrosis and discover pathophysiological and molecular correlates of lung fibrosis as a function of fractionated whole thoracic irradiation. Dose–response series of fractionated irradiation was utilized to develop a non‐invasive and quantitative measure for the degree of fibrosis – the fibrosis index (FI). The correlation of FI with histopathology, blood‐gas, transcriptome and proteome responses of the lung tissue was analyzed. Macrophages infiltration and polarization was assessed by immunohistochemistry. Fibrosis development followed a slow kinetic with maximum lung fibrosis levels detected at 24‐week post radiation insult. FI favorably correlated with radiation dose and surrogates of lung fibrosis i.e., enhanced pro‐inflammatory response, tissue remodeling and extracellular matrix deposition. The loss of lung architecture correlated with decreased epithelial marker, loss of microvascular integrity with decreased endothelial and elevated mesenchymal markers. Lung fibrosis was further attributed to a switch of the inflammatory state toward a macrophage/T‐helper cell type 2‐like (M2/Th2) polarized phenotype. Together, the multiscale characterization of FI in radiation‐induced lung fibrosis (RILF) model identified pathophysiological, transcriptional and proteomic correlates of fibrosis. Pathological immune response and endothelial/epithelial to mesenchymal transition were discovered as critical events governing lung tissue remodeling. FI will be instrumental for deciphering the molecular mechanisms governing lung fibrosis and discovery of novel targets for treatment of this devastating disease with an unmet medical need. What's new? The development of fibrosis scar tissue in the lungs is a dose‐limiting effect of radiotherapy for thoracic malignancies. Molecular mechanisms driving radiation‐induced lung fibrosis (RILF), however, remain unclear. In this study, a fibrosis index (FI) was devised to quantitatively detect spatial and temporal kinetics of lung fibrosis development. Multi‐scale characterization of FI uncovered mechanisms governing lung fibrosis, including perturbation of immune balance and microvascular integrity. Radiation dose and FI were correlated with an inflammatory switch toward a macrophage/T‐helper cell type 2‐like polarized phenotype. The findings open the way for further mechanistic study and the discovery of therapeutic targets for RILF.
Collapse
Affiliation(s)
- Cheng Zhou
- Translational Radiation Oncology, German Cancer Consortium (DKTK), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Centre (HIT), Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Heidelberg, Germany.,Division of Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation research in Oncology (NCRO), Heidelberg, Germany.,Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mahmoud R Moustafa
- Translational Radiation Oncology, German Cancer Consortium (DKTK), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Centre (HIT), Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Heidelberg, Germany.,Division of Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation research in Oncology (NCRO), Heidelberg, Germany.,Department of Clinical Pathology, Suez Canal University, Ismailia, Egypt
| | - Liji Cao
- Division of Medical Physics in Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mark Kriegsmann
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Winter
- Division of Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation research in Oncology (NCRO), Heidelberg, Germany.,Department of Functional Proteome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christian Schwager
- Translational Radiation Oncology, German Cancer Consortium (DKTK), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Centre (HIT), Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Heidelberg, Germany.,Division of Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation research in Oncology (NCRO), Heidelberg, Germany
| | - Bleddyn Jones
- Gray Laboratory, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Shijun Wang
- Department of Pediatric Nephrology, Gastroenterology & Metabolic Diseases, Hannover Medical School, Hannover, Germany
| | - Tobias Bäuerle
- Institute of Radiology, University Hospital Erlangen, Erlangen, Germany
| | - Ping-Kun Zhou
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Martina Schnölzer
- Division of Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation research in Oncology (NCRO), Heidelberg, Germany.,Department of Functional Proteome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University of Munich (TUM), Munich, Germany
| | - Juergen Debus
- Translational Radiation Oncology, German Cancer Consortium (DKTK), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Centre (HIT), Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Heidelberg, Germany.,Division of Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation research in Oncology (NCRO), Heidelberg, Germany
| | - Amir Abdollahi
- Translational Radiation Oncology, German Cancer Consortium (DKTK), National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Ion-Beam Therapy Centre (HIT), Department of Radiation Oncology, Heidelberg University Hospital (UKHD), Heidelberg, Germany.,Division of Molecular and Translational Radiation Oncology, Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation research in Oncology (NCRO), Heidelberg, Germany
| |
Collapse
|
14
|
Abstract
Soy isoflavones have demonstrated chemopreventive and anticancer properties in epidemiology and biological studies, in addition to their function as antioxidants in prevention of cardiovascular disease. We have explored the potential of soy isoflavones, as a safe biological approach, to enhance the efficacy of radiotherapy for local tumor control and limit normal tissue damage in solid tumors. This review presents studies investigating the interaction between soy isoflavones and radiation in different malignancies, including prostate cancer, renal cell carcinoma, and nonsmall cell lung cancer. Soy isoflavones were found to be potent sensitizers of cancer cells to radiation causing increased cell killing in vitro in human tumor cell lines and greater tumor inhibition in vivo in preclinical orthotopic murine tumor models. In the course of these studies, radioprotection of normal tissues and organs in the field of radiation was observed both in a clinical trial for prostate cancer and in preclinical models. The mechanisms of radiosensitization and radioprotection mediated by soy isoflavones are discussed and emphasize the role of soy isoflavones in increasing radiation effect on tumor and mitigating inflammatory responses induced by radiation in normal tissues. Soy isoflavones could be used as a safe, nontoxic complementary strategy that simultaneously increases radiation effectiveness on the malignancy while reducing damage in normal tissues in the field of radiation.
Collapse
Affiliation(s)
- Gilda G Hillman
- Department of Oncology, Radiation Oncology Division, and Department of Biochemistry, Microbiology and Immunology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI.
| |
Collapse
|
15
|
Du Y, Ren P, Wang Q, Jiang SK, Zhang M, Li JY, Wang LL, Guan DW. Cannabinoid 2 receptor attenuates inflammation during skin wound healing by inhibiting M1 macrophages rather than activating M2 macrophages. JOURNAL OF INFLAMMATION-LONDON 2018; 15:25. [PMID: 30534003 PMCID: PMC6278147 DOI: 10.1186/s12950-018-0201-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/19/2018] [Indexed: 12/17/2022]
Abstract
Background The anti-inflammatory properties of the cannabinoid 2 receptor (CB2R) in injury and inflammatory diseases have been widely substantiated. Specifically, the anti-inflammatory effect of CB2R may be achieved by regulating macrophage polarisation. Several research findings suggested that the activation of CB2R could attenuate inflammation by reducing pro-inflammatory M1 macrophage polarisation and promoting anti-inflammatory M2 polarisation. However, considering CB2R inhibits fibrosis and M2 promotes fibrosis, that the activation of CB2R may lead to an increase in M2 macrophages seems contradictory. Therefore, we hypothesised that the activation of CB2R to attenuate inflammation is not achieved by up-regulating M2 macrophages. Methods We established an incised wound model using mouse skin and used this to evaluate the effect of CB2R agonists (JWH133 or GP1a) and an antagonist (AM630) on wound healing. At various post-injury intervals, we used western blot analysis, immunofluorescence staining, enzyme-linked immunosorbent assay and quantitative reverse transcription polymerase chain reaction assays to determine CB2R protein expression, M1/M2 macrophage infiltration, and the protein and gene expression of M1/M2-associated markers and cytokines in skin lesions. Results Activation of CB2R significantly reduced M1 macrophage infiltration and slightly increased M2 macrophage infiltration. Similarly, gene expression and protein levels of M1-associated markers and cytokines (interleukin [IL]-6, IL-12, CD86 and inducible nitric oxide synthase) were significantly down-regulated after CB2R agonist administration; in contrast, markers and cytokines were increased in the CB2R antagonist–treated group. Conversely, the administration of agonists slightly increased gene expression and protein levels of M2-associated markers and cytokines (IL-4, IL-10, CD206 and arginase-1 [Arg-1]); however, a statistical significance at most time points post-injury was not noted. Conclusion In summary, our findings suggested that during incised skin wound healing in mice, increased levels of CB2R may affect inflammation by regulating M1 rather than M2 macrophage subtype polarisation. These results offer a novel understanding of the molecular mechanisms involved in the inhibition of inflammation by CBR2 that may lead to new treatments for cutaneous inflammation.
Collapse
Affiliation(s)
- Yu Du
- 1Department of Forensic Pathology, China Medical University School of Forensic Medicine, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Department of Forensic Medicine, Criminal Investigation Police University of China, Shenyang, 110854 China.,Collaborative Laboratory of Intelligentized Forensic Science, Shenyang, 110033 China
| | - Peng Ren
- 1Department of Forensic Pathology, China Medical University School of Forensic Medicine, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Department of Forensic Medicine, Criminal Investigation Police University of China, Shenyang, 110854 China.,Collaborative Laboratory of Intelligentized Forensic Science, Shenyang, 110033 China
| | - Qi Wang
- 4Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, Guangzhou, 510515 China
| | - Shu-Kun Jiang
- 1Department of Forensic Pathology, China Medical University School of Forensic Medicine, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Collaborative Laboratory of Intelligentized Forensic Science, Shenyang, 110033 China
| | - Miao Zhang
- 1Department of Forensic Pathology, China Medical University School of Forensic Medicine, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Collaborative Laboratory of Intelligentized Forensic Science, Shenyang, 110033 China
| | - Jiao-Yong Li
- 1Department of Forensic Pathology, China Medical University School of Forensic Medicine, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Collaborative Laboratory of Intelligentized Forensic Science, Shenyang, 110033 China
| | - Lin-Lin Wang
- 1Department of Forensic Pathology, China Medical University School of Forensic Medicine, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Collaborative Laboratory of Intelligentized Forensic Science, Shenyang, 110033 China
| | - Da-Wei Guan
- 1Department of Forensic Pathology, China Medical University School of Forensic Medicine, No.77, Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning Province People's Republic of China.,Collaborative Laboratory of Intelligentized Forensic Science, Shenyang, 110033 China
| |
Collapse
|
16
|
Zhang L, Wang Y, Wu G, Xiong W, Gu W, Wang CY. Macrophages: friend or foe in idiopathic pulmonary fibrosis? Respir Res 2018; 19:170. [PMID: 30189872 PMCID: PMC6127991 DOI: 10.1186/s12931-018-0864-2] [Citation(s) in RCA: 224] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 08/15/2018] [Indexed: 12/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a prototype of lethal, chronic, progressive interstitial lung disease of unknown etiology. Over the past decade, macrophage has been recognized to play a significant role in IPF pathogenesis. Depending on the local microenvironments, macrophages can be polarized to either classically activated (M1) or alternatively activated (M2) phenotypes. In general, M1 macrophages are responsible for wound healing after alveolar epithelial injury, while M2 macrophages are designated to resolve wound healing processes or terminate inflammatory responses in the lung. IPF is a pathological consequence resulted from altered wound healing in response to persistent lung injury. In this review, we intend to summarize the current state of knowledge regarding the process of macrophage polarization and its mediators in the pathogenesis of pulmonary fibrosis. Our goal is to update the understanding of the mechanisms underlying the initiation and progression of IPF, and by which, we expect to provide help for developing effective therapeutic strategies in clinical settings.
Collapse
Affiliation(s)
- Lei Zhang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Yi Wang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Guorao Wu
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Weining Xiong
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China
| | - Weikuan Gu
- Departments of Orthopaedic Surgery-Campbell Clinic, and Pathology, University of Tennessee Health Science Center (UTHSC), Memphis, TN, 38163, USA
| | - Cong-Yi Wang
- The Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Health, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.
| |
Collapse
|
17
|
Chen C, Yang S, Zhang M, Zhang Z, Zhang SB, Wu B, Hong J, Zhang W, Lin J, Okunieff P, Zhang L. Triptolide mitigates radiation-induced pneumonitis via inhibition of alveolar macrophages and related inflammatory molecules. Oncotarget 2018; 8:45133-45142. [PMID: 28415830 PMCID: PMC5542172 DOI: 10.18632/oncotarget.16456] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 03/14/2017] [Indexed: 11/29/2022] Open
Abstract
Ionizing radiation-induced pulmonary injury is a major limitation of radiotherapy for thoracic tumors. We have demonstrated that triptolide (TPL) could alleviate IR-induced pneumonia and pulmonary fibrosis. In this study, we explored the underlying mechanism by which TPL mitigates the effects of radiotoxicity. The results showed that: (1) Alveolar macrophages (AMs) were the primary inflammatory cells infiltrating irradiated lung tissues and were maintained at a high level for at least 17 days, which TPL could reduce by inhibiting of the production of macrophage inflammatory protein-2 (MIP-2) and its receptor CXCR2. (2) Stimulated by the co-cultured irradiated lung epithelium, AMs produced a panel of inflammative molecules (IMs), such as cytokines (TNF-α, IL-6, IL-1α, IL-1β) and chemokines (MIP-2, MCP-1, LIX). TPL-treated AMs could reduce the production of these IMs. Meanwhile, AMs isolated from irradiated lung tissue secreted significantly high levels of IMs, which could be dramatically reduced by TPL. (3) TPL suppressed the phagocytosis of AMs as well as ROS production. Our results indicate that TPL mitigates radiation-induced pulmonary inflammation through the inhibition of the infiltration, IM secretion, and phagocytosis of AMs.
Collapse
Affiliation(s)
- Chun Chen
- Department of Pharmacology, School of Pharmacy, Fujian Medical University, Fuzhou, China 350122
| | - Shanmin Yang
- Department of Radiation Oncology, University of Florida, Gainesville, Florida 32610, USA
| | - Mei Zhang
- Department of Radiation Oncology, University of Florida, Gainesville, Florida 32610, USA
| | - Zhenhuan Zhang
- Department of Radiation Oncology, University of Florida, Gainesville, Florida 32610, USA
| | - Steven B Zhang
- Department of Radiation Oncology, University of Florida, Gainesville, Florida 32610, USA
| | - Bing Wu
- Fujian Platform for Medical Research at First Affiliated Hospital, Fujian Key Lab of Individualized Active Immunotherapy and Key Laboratory of Radiation Biology of Fujian Province Universities, Fuzhou, China 350005
| | - Jinsheng Hong
- Fujian Platform for Medical Research at First Affiliated Hospital, Fujian Key Lab of Individualized Active Immunotherapy and Key Laboratory of Radiation Biology of Fujian Province Universities, Fuzhou, China 350005
| | - Weijian Zhang
- Fujian Platform for Medical Research at First Affiliated Hospital, Fujian Key Lab of Individualized Active Immunotherapy and Key Laboratory of Radiation Biology of Fujian Province Universities, Fuzhou, China 350005
| | - Jianhua Lin
- Fujian Platform for Medical Research at First Affiliated Hospital, Fujian Key Lab of Individualized Active Immunotherapy and Key Laboratory of Radiation Biology of Fujian Province Universities, Fuzhou, China 350005
| | - Paul Okunieff
- Department of Radiation Oncology, University of Florida, Gainesville, Florida 32610, USA
| | - Lurong Zhang
- Department of Radiation Oncology, University of Florida, Gainesville, Florida 32610, USA.,Fujian Platform for Medical Research at First Affiliated Hospital, Fujian Key Lab of Individualized Active Immunotherapy and Key Laboratory of Radiation Biology of Fujian Province Universities, Fuzhou, China 350005
| |
Collapse
|
18
|
Wu Q, Allouch A, Martins I, Modjtahedi N, Deutsch E, Perfettini JL. Macrophage biology plays a central role during ionizing radiation-elicited tumor response. Biomed J 2017; 40:200-211. [PMID: 28918908 PMCID: PMC6136289 DOI: 10.1016/j.bj.2017.06.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 06/01/2017] [Accepted: 06/11/2017] [Indexed: 12/13/2022] Open
Abstract
Radiation therapy is one of the major therapeutic modalities for most solid tumors. The anti-tumor effect of radiation therapy consists of the direct tumor cell killing, as well as the modulation of tumor microenvironment and the activation of immune response against tumors. Radiation therapy has been shown to promote immunogenic cells death, activate dendritic cells and enhance tumor antigen presentation and anti-tumor T cell activation. Radiation therapy also programs innate immune cells such as macrophages that leads to either radiosensitization or radioresistance, according to different tumors and different radiation regimen studied. The mechanisms underlying radiation-induced macrophage activation remain largely elusive. Various molecular players such as NF-κB, MAPKs, p53, reactive oxygen species, inflammasomes have been involved in these processes. The skewing to a pro-inflammatory phenotype thus results in the activation of anti-tumor immune response and enhanced radiotherapy effect. Therefore, a comprehensive understanding of the mechanism of radiation-induced macrophage activation and its role in tumor response to radiation therapy is crucial for the development of new therapeutic strategies to enhance radiation therapy efficacy.
Collapse
Affiliation(s)
- Qiuji Wu
- Cell Death and Aging Team, Gystave Roussy Cancer Campus, Villejuif, France; Laboratory of Molecular Radiotherapy, INSERM U1030, Gystave Roussy Cancer Campus, Villejuif, France; Gystave Roussy Cancer Campus, Villejuif, France; Université Paris Sud - Paris Saclay, Villejuif, France; Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, Hubei, China; Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, Hubei, China
| | - Awatef Allouch
- Cell Death and Aging Team, Gystave Roussy Cancer Campus, Villejuif, France; Laboratory of Molecular Radiotherapy, INSERM U1030, Gystave Roussy Cancer Campus, Villejuif, France; Gystave Roussy Cancer Campus, Villejuif, France; Université Paris Sud - Paris Saclay, Villejuif, France
| | - Isabelle Martins
- Cell Death and Aging Team, Gystave Roussy Cancer Campus, Villejuif, France; Laboratory of Molecular Radiotherapy, INSERM U1030, Gystave Roussy Cancer Campus, Villejuif, France; Gystave Roussy Cancer Campus, Villejuif, France; Université Paris Sud - Paris Saclay, Villejuif, France
| | - Nazanine Modjtahedi
- Cell Death and Aging Team, Gystave Roussy Cancer Campus, Villejuif, France; Laboratory of Molecular Radiotherapy, INSERM U1030, Gystave Roussy Cancer Campus, Villejuif, France; Gystave Roussy Cancer Campus, Villejuif, France; Université Paris Sud - Paris Saclay, Villejuif, France
| | - Eric Deutsch
- Laboratory of Molecular Radiotherapy, INSERM U1030, Gystave Roussy Cancer Campus, Villejuif, France; Gystave Roussy Cancer Campus, Villejuif, France; Université Paris Sud - Paris Saclay, Villejuif, France
| | - Jean-Luc Perfettini
- Cell Death and Aging Team, Gystave Roussy Cancer Campus, Villejuif, France; Laboratory of Molecular Radiotherapy, INSERM U1030, Gystave Roussy Cancer Campus, Villejuif, France; Gystave Roussy Cancer Campus, Villejuif, France; Université Paris Sud - Paris Saclay, Villejuif, France.
| |
Collapse
|
19
|
de Leve S, Wirsdörfer F, Cappuccini F, Schütze A, Meyer AV, Röck K, Thompson LF, Fischer JW, Stuschke M, Jendrossek V. Loss of CD73 prevents accumulation of alternatively activated macrophages and the formation of prefibrotic macrophage clusters in irradiated lungs. FASEB J 2017; 31:2869-2880. [PMID: 28325757 DOI: 10.1096/fj.201601228r] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 03/06/2017] [Indexed: 12/17/2022]
Abstract
While radiotherapy is a mainstay for cancer therapy, pneumonitis and fibrosis constitute dose-limiting side effects of thorax and whole body irradiation. So far, the contribution of immune cells to disease progression is largely unknown. Here we studied the role of ecto-5'-nucelotidase (CD73)/adenosine-induced changes in the myeloid compartment in radiation-induced lung fibrosis. C57BL/6 wild-type or CD73-/- mice received a single dose of whole thorax irradiation (WTI, 15 Gy). Myeloid cells were characterized in flow cytometric, histologic, and immunohistochemical analyses as well as RNA analyses. WTI induced a pronounced reduction of alveolar macrophages in both strains that recovered within 6 wk. Fibrosis development in wild-type mice was associated with a time-dependent deposition of hyaluronic acid (HA) and increased expression of markers for alternative activation on alveolar macrophages. These include the antiinflammatory macrophage mannose receptor and arginase-1. Further, macrophages accumulated in organized clusters and expressed profibrotic mediators at ≥25 wk after irradiation (fibrotic phase). Irradiated CD73-/- mice showed an altered regulation of components of the HA system and no clusters of alternatively activated macrophages. We speculate that accumulation of alternatively activated macrophages in organized clusters represents the origins of fibrotic foci after WTI and is promoted by a cross-talk between HA, CD73/adenosine signaling, and other profibrotic mediators.-De Leve, S., Wirsdörfer, F., Cappuccini, F., Schütze, A., Meyer, A. V., Röck, K., Thompson, L. F., Fischer, J. W., Stuschke, M., Jendrossek, V. Loss of CD73 prevents accumulation of alternatively activated macrophages and the formation of prefibrotic macrophage clusters in irradiated lungs.
Collapse
Affiliation(s)
- Simone de Leve
- Institute of Cell Biology (Cancer Research), University Hospital Essen, Essen, Germany
| | - Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, Essen, Germany
| | - Federica Cappuccini
- Institute of Cell Biology (Cancer Research), University Hospital Essen, Essen, Germany
| | - Alexandra Schütze
- Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Alina V Meyer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, Essen, Germany
| | - Katharina Röck
- Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Linda F Thompson
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Jens W Fischer
- Pharmacology and Clinical Pharmacology, University Hospital, Heinrich-Heine-University, Düsseldorf, Germany
| | - Martin Stuschke
- Department of Radiation Oncology, University Hospital Essen, Essen, Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, Essen, Germany;
| |
Collapse
|
20
|
Standardized Herbal Formula PM014 Inhibits Radiation-Induced Pulmonary Inflammation in Mice. Sci Rep 2017; 7:45001. [PMID: 28322297 PMCID: PMC5359558 DOI: 10.1038/srep45001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 02/17/2017] [Indexed: 12/27/2022] Open
Abstract
Radiation therapy is widely used for thoracic cancers. However, it occasionally causes radiation-induced lung injuries, including pneumonitis and fibrosis. Chung-Sang-Bo-Ha-Tang (CSBHT) has been traditionally used to treat chronic pulmonary disease in Korea. PM014, a modified herbal formula derived from CSBHT, contains medicinal herbs of seven species. In our previous studies, PM014 exhibited anti-inflammatory effects in a chronic obstructive pulmonary disease model. In this study, we have evaluated the effects of PM014 on radiation-induced lung inflammation. Mice in the treatment group were orally administered PM014 six times for 2 weeks. Effects of PM014 on radiation pneumonitis were evaluated based on histological findings and differential cell count in bronchoalveolar lavage fluid. PM014 treatment significantly inhibited immune cell recruitment and collagen deposition in lung tissue. Normal lung volume, evaluated by radiological analysis, in PM014-treated mice was higher compared to that in irradiated control mice. PM014-treated mice exhibited significant changes in inspiratory capacity, compliance and tissue damping and elastance. Additionally, PM014 treatment resulted in the downregulation of inflammatory cytokines, chemokines, and fibrosis-related genes and a reduction in the transforming growth factor-β1-positive cell population in lung tissue. Thus, PM014 is a potent therapeutic agent for radiation-induced lung fibrosis and inflammation.
Collapse
|
21
|
Kalita B, Ranjan R, Singh A, Yashavarddhan MH, Bajaj S, Gupta ML. A Combination of Podophyllotoxin and Rutin Attenuates Radiation Induced Gastrointestinal Injury by Negatively Regulating NF-κB/p53 Signaling in Lethally Irradiated Mice. PLoS One 2016; 11:e0168525. [PMID: 28036347 PMCID: PMC5201299 DOI: 10.1371/journal.pone.0168525] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 12/03/2016] [Indexed: 12/15/2022] Open
Abstract
Development of an effective radio protector to minimise radiation-inflicted damages have largely failed owing to inherent toxicity of most of the agents examined so far. This study is centred towards delivering protection to lethally irradiated mice by pre-administration of a safe formulation G-003M (combination of podophyllotoxin and rutin) majorly through regulation of inflammatory and cell death pathways in mice. Single intramuscular dose of G-003M injected 60 min prior to 9 Gy exposure rescued 89% of whole body lethally irradiated C57BL/6J mice. Studies have revealed reduction in radiation induced reactive oxygen species (ROS), nitric oxide (NO) generation, prostaglandin E2 (PGE2) levels and intestinal apoptosis in G-003M pre-treated mice intestine. Restricted nuclear translocation of redox-sensitive Nuclear factor-κB (NF-κB) and subsequent downregulation of cyclo-oxygenase 2 (COX-2), inducible nitric oxide synthase (iNOS; EC 1.14.13.39) and tumor necrosis factor (TNF-α) levels demonstrated the anti-inflammatory effect that G-003M exerts. Support to early hematopoietic recovery was exhibited through G-003M mediated induction of granulocyte colony stimulating factor (G-CSF) and interleukin (IL-6) levels in lethally irradiated mice. Considerable attenuation in radiation induced morphological damage to the intestinal villi, crypts and mucosal layers was observed in G-003M pre-treated mice. Additionally, our formulation did not reduce the sensitivity of tumor tissue to radiation. Altogether, these results suggest that G-003M ameliorates the deleterious effects of radiation exposure by minimising ROS and NO generation and effectively regulating inflammatory and cell death pathways. Mechanism of protection elucidated in the current study demonstrates that G-003M can be used as a safe and effective radio protective agent in radiotherapy for human application.
Collapse
Affiliation(s)
- Bhargab Kalita
- Division of Radioprotective Drug Development and Research, Institute of Nuclear Medicine and Allied Sciences, Brig.S.K Mazumdar Marg, Delhi, INDIA
| | - Rajiv Ranjan
- Division of Radioprotective Drug Development and Research, Institute of Nuclear Medicine and Allied Sciences, Brig.S.K Mazumdar Marg, Delhi, INDIA
| | - Abhinav Singh
- Division of Radioprotective Drug Development and Research, Institute of Nuclear Medicine and Allied Sciences, Brig.S.K Mazumdar Marg, Delhi, INDIA
| | - M. H. Yashavarddhan
- Division of Radioprotective Drug Development and Research, Institute of Nuclear Medicine and Allied Sciences, Brig.S.K Mazumdar Marg, Delhi, INDIA
| | - Sania Bajaj
- Division of Radioprotective Drug Development and Research, Institute of Nuclear Medicine and Allied Sciences, Brig.S.K Mazumdar Marg, Delhi, INDIA
| | - Manju Lata Gupta
- Division of Radioprotective Drug Development and Research, Institute of Nuclear Medicine and Allied Sciences, Brig.S.K Mazumdar Marg, Delhi, INDIA
- * E-mail:
| |
Collapse
|
22
|
Duru N, Zhang Y, Gernapudi R, Wolfson B, Lo PK, Yao Y, Zhou Q. Loss of miR-140 is a key risk factor for radiation-induced lung fibrosis through reprogramming fibroblasts and macrophages. Sci Rep 2016; 6:39572. [PMID: 27996039 PMCID: PMC5172237 DOI: 10.1038/srep39572] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 11/23/2016] [Indexed: 12/14/2022] Open
Abstract
Radiation-induced lung fibrosis (RILF) is a common side effect for patients with thoracic cancer receiving radiation therapy. RILF is characterized by excessive collagen deposition mediated by TGF-β1 and its downstream factor SMAD3, but the exact molecular mechanism leading to fibrosis is yet to be determined. The present study investigated the impact of miR-140 on RILF development. Herein, we first found that loss of miR-140 is a marker of fibrotic lung tissue in vivo one-year post-radiation treatment. We showed that miR-140 knockout primary lung fibroblasts have a higher percentage of myofibroblasts compared to wild type primary lung fibroblasts, and that loss of miR-140 expression leads to increased activation of TGF-β1 signaling as well as increased myofibroblast differentiation. We also identified fibronectin as a novel miR-140 target gene in lung fibroblasts. Finally, we have shown that miR-140 deficiency promotes accumulation of M2 macrophages in irradiated lung tissues. These data suggest that miR-140 is a key protective molecule against RILF through inhibiting myofibroblast differentiation and inflammation.
Collapse
Affiliation(s)
- Nadire Duru
- Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yongshu Zhang
- Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ramkishore Gernapudi
- Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Benjamin Wolfson
- Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Pang-Kuo Lo
- Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yuan Yao
- Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Qun Zhou
- Department of Biochemistry and Molecular Biology, Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
23
|
Adenovirus-mediated Foxp3 expression in lung epithelial cells ameliorates acute radiation-induced pneumonitis in mice. Gene Ther 2016; 24:104-112. [PMID: 27996966 DOI: 10.1038/gt.2016.86] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 11/11/2016] [Accepted: 12/05/2016] [Indexed: 12/11/2022]
Abstract
Forkhead transcription factor 3 (Foxp3) has a critical role in regulatory T cells (Treg). There are an increasing number of researches concerning the functions of Foxp3 in other cells, including lung epithelial cells besides Treg. However, the roles of Foxp3 in lung epithelial cells remain poorly understood. To examine the potential therapeutic benefits of Foxp3 for lung inflammation, this study investigates the effect of adenovirus-mediated Foxp3 overexpression in a radiation-induced lung damage model. Foxp3-EGFP expressing adenovirus was administered by intratracheal injection three times over 14 days after focal X-ray irradiation. To evaluate effects of Foxp3 overexpression in radiation-induced lung inflammation, immune cell profiles of bronchoalveolar lavage (BAL) fluid were analyzed. Foxp3 gene-delivered mice showed significant inhibition of immune cell infiltration, such as eosinophils, lymphocytes, macrophages and neutrophils in BAL fluid. Histopathological analysis also showed that Foxp3 overexpression inhibits inflammatory cell recruitment and collagen deposition in lung tissues. In addition, expression of inflammatory and fibrosis-related genes was decreased in the Foxp3 expression adenovirus-infected group. These results suggest that Foxp3 expression in lungs holds considerable therapeutic potential for attenuating inflammation and fibrosis in radiation-induced lung injury.
Collapse
|
24
|
Duru N, Wolfson B, Zhou Q. Mechanisms of the alternative activation of macrophages and non-coding RNAs in the development of radiation-induced lung fibrosis. World J Biol Chem 2016; 7:231-239. [PMID: 27957248 PMCID: PMC5124699 DOI: 10.4331/wjbc.v7.i4.231] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/17/2016] [Accepted: 10/25/2016] [Indexed: 02/05/2023] Open
Abstract
Radiation-induced lung fibrosis (RILF) is a common side effect of thoracic irradiation therapy and leads to high mortality rates after cancer treatment. Radiation injury induces inflammatory M1 macrophage polarization leading to radiation pneumonitis, the first stage of RILF progression. Fibrosis occurs due to the transition of M1 macrophages to the anti-inflammatory pro-fibrotic M2 phenotype, and the resulting imbalance of macrophage regulated inflammatory signaling. Non-coding RNA signaling has been shown to play a large role in the regulation of the M2 mediated signaling pathways that are associated with the development and progression of fibrosis. While many studies show the link between M2 macrophages and fibrosis, there are only a few that explore their distinct role and the regulation of their signaling by non-coding RNA in RILF. In this review we summarize the current body of knowledge describing the roles of M2 macrophages in RILF, with an emphasis on the expression and functions of non-coding RNAs.
Collapse
|
25
|
Groves AM, Johnston CJ, Misra RS, Williams JP, Finkelstein JN. Effects of IL-4 on pulmonary fibrosis and the accumulation and phenotype of macrophage subpopulations following thoracic irradiation. Int J Radiat Biol 2016; 92:754-765. [PMID: 27539247 DOI: 10.1080/09553002.2016.1222094] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE Thoracic irradiation injures lung parenchyma, triggering inflammation and immune cell activation, leading to pneumonitis and fibrosis. Macrophage polarization contributes to these processes. Since IL-4 promotes pro-fibrotic macrophage activation, its role in radiation-induced lung injury was investigated. MATERIALS AND METHODS Lung macrophage subpopulations were characterized from 3-26 weeks following exposure of WT and IL-4-/- mice to 0 or 12.5 Gray single dose thoracic irradiation. RESULTS Loss of IL-4 did not prevent fibrosis, but blunted macrophage accumulation within the parenchyma. At 3 weeks following exposure, cell numbers and expression of F4/80 and CD206, an alternative activation marker, decreased in alveolar macrophages but increased in infiltrating macrophages in WT mice. Loss of IL-4 impaired recovery of these markers in alveolar macrophages and blunted expansion of these populations in infiltrating macrophages. CD206+ cells were evident in fibrotic regions of WT mice only, however Arg-1+ cells increased in fibrotic regions in IL-4-/- mice only. Radiation-induced proinflammatory Ly6C expression was more apparent in alveolar and interstitial macrophages from IL-4-/- mice. CONCLUSIONS IL-4 loss did not prevent alternative macrophage activation and fibrosis in irradiated mice. Instead, a role is indicated for IL-4 in maintenance of macrophage populations in the lung following high single dose thoracic irradiation.
Collapse
Affiliation(s)
- Angela M Groves
- a Department of Pediatrics M&D Neonatology, University of Rochester Medical Center , Rochester , NY , USA
| | - Carl J Johnston
- a Department of Pediatrics M&D Neonatology, University of Rochester Medical Center , Rochester , NY , USA.,b Department of Environmental Medicine , University of Rochester Medical Center , Rochester , NY , USA
| | - Ravi S Misra
- a Department of Pediatrics M&D Neonatology, University of Rochester Medical Center , Rochester , NY , USA
| | - Jacqueline P Williams
- b Department of Environmental Medicine , University of Rochester Medical Center , Rochester , NY , USA
| | - Jacob N Finkelstein
- a Department of Pediatrics M&D Neonatology, University of Rochester Medical Center , Rochester , NY , USA.,b Department of Environmental Medicine , University of Rochester Medical Center , Rochester , NY , USA
| |
Collapse
|
26
|
Zhang X, Wu J, Ye B, Wang Q, Xie X, Shen H. Protective effect of curcumin on TNBS-induced intestinal inflammation is mediated through the JAK/STAT pathway. Altern Ther Health Med 2016; 16:299. [PMID: 27544348 PMCID: PMC4992287 DOI: 10.1186/s12906-016-1273-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 08/09/2016] [Indexed: 11/12/2022]
Abstract
Background Curcumin displays a protective role in rat models of intestinal inflammation. However, the mechanism of how curcumin affects on intestinal inflammation is less known. The purpose of the current study is to explore the signal pathway in which the curcumin protecting rat from intestinal inflammation. Methods The intestinal inflammation rat models were made by TNBS treatment. Curcumin was added to their diet 5 days before the TNBS instillation. After that, body weight change, score of macroscopic assessment of disease activity and microscopic scoring were utilized to analyse the severity of the induced inflammation. In addition, the level of pro-inflammatory cytokines and anti-inflammatory were detected to determine the effect of curcumin on intestinal inflammation. The JAK/STAT pathway of pro-inflammation response was also evaluated. Finally, the impact of curcumin on apoptosis in intestinal inflammation was assessed by TUNEL staining. Results Rats pretreated with curcumin significantly reversed the decrease of body weight and increase of colon weight derived from TNBS-induced colitis. Histological improvement was observed in response to curcumin. In addition, curcumin attenuated TNBS-induced secretion of pro-inflammatory cytokines and M1/M2 ratio, while stimulated the secretion of anti-inflammatory cytokines. The inhibition of pro-inflammation response was mediated by SOCS-1, which could efficiently suppress JAK/STAT pathways. Furthermore, curcumin efficiently suppressed the TNBS-induced apoptosis, and reduced the accumulation of cytochrome C in cytosol. Conclusion The anti-inflammatory effect of curcumin is realized by enhancing SOCS-1 expression and inhibiting JAK/STAT pathways. Curcumin also plays an anti-apoptotic role in TNBS-induced intestinal inflammation. We propose that curcumin may have therapeutic implications for human intestinal inflammation.
Collapse
|
27
|
Williams JP, Calvi L, Chakkalakal JV, Finkelstein JN, O’Banion MK, Puzas E. Addressing the Symptoms or Fixing the Problem? Developing Countermeasures against Normal Tissue Radiation Injury. Radiat Res 2016; 186:1-16. [PMID: 27332954 PMCID: PMC4991354 DOI: 10.1667/rr14473.1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jacqueline P. Williams
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Laura Calvi
- Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Joe V. Chakkalakal
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| | - Jacob N. Finkelstein
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
- Department of Pediatrics and Neonatology, University of Rochester Medical Center, Rochester, New York
| | - M. Kerry O’Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, New York
| | - Edward Puzas
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
28
|
Chen J, Wang Y, Mei Z, Zhang S, Yang J, Li X, Yao Y, Xie C. Radiation-induced lung fibrosis in a tumor-bearing mouse model is associated with enhanced Type-2 immunity. JOURNAL OF RADIATION RESEARCH 2016; 57:133-41. [PMID: 26703457 PMCID: PMC4795947 DOI: 10.1093/jrr/rrv077] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 10/09/2015] [Indexed: 05/09/2023]
Abstract
Lung fibrosis may be associated with Type-2 polarized inflammation. Herein, we aim to investigate whether radiation can initiate a Type-2 immune response and contribute to the progression of pulmonary fibrosis in tumor-bearing animals. We developed a tumor-bearing mouse model with Lewis lung cancer to receive either radiation therapy alone or radiation combined with Th1 immunomodulator unmethylated cytosine-phosphorothioate-guanine containing oligodeoxynucleotide (CpG-ODN). The Type-2 immune phenotype in tumors and the histological grade of lung fibrosis were evaluated in mice sacrificed three weeks after irradiation. Mouse lung tissues were analyzed for hydroxyproline and the expression of Type-1/Type-2 key transcription factors (T-bet/GATA-3). The concentration of Type-1/Type-2 cytokines in serum was measured by cytometric bead array. Lung fibrosis was observed to be more serious in tumor-bearing mice than in normal mice post-irradiation. The fibrosis score in irradiated tumor-bearing mice on Day 21 was 4.33 ± 0.82, which was higher than that of normal mice (2.00 ± 0.63; P < 0.05). Hydroxyproline and GATA-3 expression were increased in the lung tissues of tumor-bearing mice following irradiation. CpG-ODN attenuated fibrosis by markedly decreasing GATA-3 expression. Serum IL-13 and IL-5 were elevated, whereas INF-γ and IL-12 expression were decreased in irradiated tumor-bearing mice. These changes were reversed after CpG-ODN treatment. Thus, Type-2 immunity in tumors appeared to affect the outcome of radiation damage and might be of interest for future studies on developing approaches in which Type-1-related immunotherapy and radiotherapy are used in combination.
Collapse
Affiliation(s)
- Jing Chen
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China
| | - Yacheng Wang
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China
| | - Zijie Mei
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China
| | - Shimin Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China
| | - Jie Yang
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China
| | - Xin Li
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China
| | - Ye Yao
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital, Wuhan University, 169 Dong Hu Road, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
29
|
Groves AM, Johnston CJ, Misra RS, Williams JP, Finkelstein JN. Whole-Lung Irradiation Results in Pulmonary Macrophage Alterations that are Subpopulation and Strain Specific. Radiat Res 2015; 184:639-49. [PMID: 26632857 DOI: 10.1667/rr14178.1] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Exposure of the lung to radiation produces injury and inflammatory responses that result in microenvironmental alterations, which can promote the development of pneumonitis and/or pulmonary fibrosis. It has been shown that after other toxic insults, macrophages become phenotypically polarized in response to microenvironmental signals, orchestrating the downstream inflammatory responses. However, their contribution to the development of the late consequences of pulmonary radiation exposure remains unclear. To address this issue, fibrosis-prone C57BL/6J mice or pneumonitis-prone C3H/HeJ mice were whole-lung irradiated with 0 or 12.5 Gy and lung digests were collected between 3 and 26 weeks after radiation exposure. CD45(+) leukocytes were isolated and characterized by flow cytometry, and alveolar, interstitial and infiltrating macrophages were also detected. Ly6C, expressed by pro-inflammatory monocytes and macrophages, and mannose receptor (CD206), a marker of alternative activation, were assessed in each subpopulation. While the total number of pulmonary macrophages was depleted at 3 weeks after lung irradiation relative to age-matched controls in both C57 and C3H mice, identification of discrete subpopulations showed that this loss in cell number occurred in the alveolar, but not the interstitial or infiltrating, subsets. In the alveolar macrophages of both C57 and C3H mice, this correlated with a loss in the proportion of cells that expressed CD206 and F4/80. In contrast, in interstitial and infiltrating macrophages, the proportion of cells expressing these markers was increased at several time points after irradiation, with this response generally more pronounced in C3H mice. Radiation exposure was also associated with elevations in the proportion of alveolar and interstitial macrophage subpopulations expressing Ly6C and F4/80, with this response occurring at earlier time points in C57 mice. Although the radiation dose used in this study was not isoeffective for the inflammatory response in the two strains, the differences observed in the responses of these discrete macrophage populations between the fibrosis-prone versus pneumonitis-prone mice nonetheless suggest a possible role for these cells in the development of long-term consequences of pulmonary radiation exposure.
Collapse
Affiliation(s)
- Angela M Groves
- a Department of Pediatrics and Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Carl J Johnston
- a Department of Pediatrics and Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, New York; and.,b Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Ravi S Misra
- a Department of Pediatrics and Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, New York; and
| | - Jacqueline P Williams
- b Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| | - Jacob N Finkelstein
- a Department of Pediatrics and Neonatology, University of Rochester School of Medicine and Dentistry, Rochester, New York; and.,b Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
30
|
Abernathy LM, Fountain MD, Rothstein SE, David JM, Yunker CK, Rakowski J, Lonardo F, Joiner MC, Hillman GG. Soy Isoflavones Promote Radioprotection of Normal Lung Tissue by Inhibition of Radiation-Induced Activation of Macrophages and Neutrophils. J Thorac Oncol 2015; 10:1703-12. [PMID: 26709479 PMCID: PMC6876621 DOI: 10.1097/jto.0000000000000677] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Radiation therapy for lung cancer is limited by toxicity to normal lung tissue that results from an inflammatory process, leading to pneumonitis and fibrosis. Soy isoflavones mitigate inflammatory infiltrates and radiation-induced lung injury, but the cellular immune mediators involved in the radioprotective effect are unknown. METHODS Mice received a single dose of 10 Gy radiation delivered to the lungs and daily oral treatment of soy isoflavones. At different time points, mice were either processed to harvest bronchoalveolar lavage fluid for differential cell counting and lungs for flow cytometry or immunohistochemistry studies. RESULTS Combined soy and radiation led to a reduction in infiltration and activation of alveolar macrophages and neutrophils in both the bronchoalveolar and lung parenchyma compartments. Soy treatment protected F4/80CD11c interstitial macrophages, which are known to play an immunoregulatory role and are decreased by radiation. Furthermore, soy isoflavones reduced the levels of nitric oxide synthase 2 expression while increasing arginase-1 expression after radiation, suggesting a switch from proinflammatory M1 macrophage to an anti-inflammatory M2 macrophage phenotype. Soy also prevented the influx of activated neutrophils in lung caused by radiation. CONCLUSIONS Soy isoflavones inhibit the infiltration and activation of macrophages and neutrophils induced by radiation in lungs. Soy isoflavones-mediated modulation of macrophage and neutrophil responses to radiation may contribute to a mechanism of resolution of radiation-induced chronic inflammation leading to radioprotection of lung tissue.
Collapse
Affiliation(s)
- Lisa M. Abernathy
- Department of Oncology, Division of Radiation Oncology, Wayne State University School of Medicine, Detroit, MI 48201, U.S.A
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI 48201, U.S.A
| | - Matthew D. Fountain
- Department of Oncology, Division of Radiation Oncology, Wayne State University School of Medicine, Detroit, MI 48201, U.S.A
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI 48201, U.S.A
| | - Shoshana E. Rothstein
- Department of Oncology, Division of Radiation Oncology, Wayne State University School of Medicine, Detroit, MI 48201, U.S.A
| | - John M. David
- Department of Oncology, Division of Radiation Oncology, Wayne State University School of Medicine, Detroit, MI 48201, U.S.A
| | - Christopher K. Yunker
- Department of Oncology, Division of Radiation Oncology, Wayne State University School of Medicine, Detroit, MI 48201, U.S.A
| | - Joseph Rakowski
- Department of Oncology, Division of Radiation Oncology, Wayne State University School of Medicine, Detroit, MI 48201, U.S.A
| | - Fulvio Lonardo
- Department of Pathology, Wayne State University School of Medicine, Detroit, MI 48201, U.S.A
| | - Michael C. Joiner
- Department of Oncology, Division of Radiation Oncology, Wayne State University School of Medicine, Detroit, MI 48201, U.S.A
| | - Gilda G. Hillman
- Department of Oncology, Division of Radiation Oncology, Wayne State University School of Medicine, Detroit, MI 48201, U.S.A
- Department of Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI 48201, U.S.A
| |
Collapse
|
31
|
Jiao Y, Chen H, Gu T, Wang L, Postlethwaite A, Gu W. Molecular network of important genes for systemic sclerosis-related progressive lung fibrosis. BMC Res Notes 2015; 8:544. [PMID: 26444860 PMCID: PMC4596290 DOI: 10.1186/s13104-015-1510-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 09/21/2015] [Indexed: 01/06/2023] Open
Abstract
Background Considerable progress has been made in illuminating the pathological events for systemic sclerosis (SSc)-related progressive lung fibrosis. The molecular events that lead to SSc-related progressive lung fibrosis need to be defined. Some important genes have been identified from a recent study in humans. We aim to construct and compare the similarities and differences of molecular pathways between SSc-related progressive lung fibrosis and normal lungs of humans and mice. Methods We used the analytical approach of association of key genes in SSc-related progressive lung fibrosis. We first identified the probes for genes of SSc-related progressive lung fibrosis and analyzed the pathways in human lung using data generated by microarray. We then analyzed the gene pathways in mouse lung for similar sets of probes. Gene expression data from livers were used to compare with that in lung in both humans and mice. Results Our analysis indicated that, in humans, the expression levels of genes for macrophage activation are more strongly associated with each other than that in mice. In both humans and mice, the associations of these genes are much greater in the lung than that in the liver. The association in gene expression between humans and mice are similar for IFN-regulated genes and profibrotic/Tgfβ-regulated genes. Conclusion Our analysis reveals the differences and similarities of the network of key genes between humans and mice during the molecular processes that eventually lead to fibrosis in the lung. Electronic supplementary material The online version of this article (doi:10.1186/s13104-015-1510-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yan Jiao
- Departments of Orthopedic Surgery and BME, Campbell Clinic, University of Tennessee Health Science Center, 956 Court Avenue Rm A302, Memphis, TN, 38163-0001, USA. .,Mudanjiang Medical College, Mudanjiang, 157001, Heilongjiang, People's Republic of China.
| | - Hong Chen
- The First Hospital of Qiqihaer City, 30 Gongyuan Road, Longsha District, Qiqihaer, 161005, Heilongjiang, People's Republic of China.
| | - Tianshu Gu
- Departments of Orthopedic Surgery and BME, Campbell Clinic, University of Tennessee Health Science Center, 956 Court Avenue Rm A302, Memphis, TN, 38163-0001, USA. .,Hebei Medical University, Shijiazhuang, 050011, Hebei, People's Republic of China.
| | - Lishi Wang
- Departments of Orthopedic Surgery and BME, Campbell Clinic, University of Tennessee Health Science Center, 956 Court Avenue Rm A302, Memphis, TN, 38163-0001, USA.
| | - Arnold Postlethwaite
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163-0001, USA. .,Department of Veterans Affairs Medical Center, Memphis, TN, 38104, USA.
| | - Weikuan Gu
- Departments of Orthopedic Surgery and BME, Campbell Clinic, University of Tennessee Health Science Center, 956 Court Avenue Rm A302, Memphis, TN, 38163-0001, USA. .,Department of Veterans Affairs Medical Center, Memphis, TN, 38104, USA.
| |
Collapse
|
32
|
Straub JM, New J, Hamilton CD, Lominska C, Shnayder Y, Thomas SM. Radiation-induced fibrosis: mechanisms and implications for therapy. J Cancer Res Clin Oncol 2015; 141:1985-94. [PMID: 25910988 DOI: 10.1007/s00432-015-1974-6] [Citation(s) in RCA: 380] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/15/2015] [Indexed: 01/01/2023]
Abstract
PURPOSE Radiation-induced fibrosis (RIF) is a long-term side effect of external beam radiation therapy for the treatment of cancer. It results in a multitude of symptoms that significantly impact quality of life. Understanding the mechanisms of RIF-induced changes is essential to developing effective strategies to prevent long-term disability and discomfort following radiation therapy. In this review, we describe the current understanding of the etiology, clinical presentation, pathogenesis, treatment, and directions of future therapy for this condition. METHODS A literature review of publications describing mechanisms or treatments of RIF was performed. Specific databases utilized included PubMed and clinicaltrials.gov, using keywords "Radiation-Induced Fibrosis," "Radiotherapy Complications," "Fibrosis Therapy," and other closely related terms. RESULTS RIF is the result of a misguided wound healing response. In addition to causing direct DNA damage, ionizing radiation generates reactive oxygen and nitrogen species that lead to localized inflammation. This inflammatory process ultimately evolves into a fibrotic one characterized by increased collagen deposition, poor vascularity, and scarring. Tumor growth factor beta serves as the primary mediator in this response along with a host of other cytokines and growth factors. Current therapies have largely been directed toward these molecular targets and their associated signaling pathways. CONCLUSION Although RIF is widely prevalent among patients undergoing radiation therapy and significantly impacts quality of life, there is still much to learn about its pathogenesis and mechanisms. Current treatments have stemmed from this understanding, and it is anticipated that further elucidation will be essential for the development of more effective therapies.
Collapse
Affiliation(s)
- Jeffrey M Straub
- Department of Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, 3901 Rainbow Boulevard, 3020A Wahl Hall East, Kansas City, KS, 66160, USA
| | - Jacob New
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Chase D Hamilton
- Department of Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, 3901 Rainbow Boulevard, 3020A Wahl Hall East, Kansas City, KS, 66160, USA
| | - Chris Lominska
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Yelizaveta Shnayder
- Department of Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, 3901 Rainbow Boulevard, 3020A Wahl Hall East, Kansas City, KS, 66160, USA
| | - Sufi M Thomas
- Department of Otolaryngology-Head and Neck Surgery, University of Kansas Medical Center, 3901 Rainbow Boulevard, 3020A Wahl Hall East, Kansas City, KS, 66160, USA. .,Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA. .,Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
33
|
Linard C, Tissedre F, Busson E, Holler V, Leclerc T, Strup-Perrot C, Couty L, L'homme B, Benderitter M, Lafont A, Lataillade JJ, Coulomb B. Therapeutic potential of gingival fibroblasts for cutaneous radiation syndrome: comparison to bone marrow-mesenchymal stem cell grafts. Stem Cells Dev 2015; 24:1182-93. [PMID: 25584741 DOI: 10.1089/scd.2014.0486] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mesenchymal stem cell (MSC) therapy has recently been investigated as a potential treatment for cutaneous radiation burns. We tested the hypothesis that injection of local gingival fibroblasts (GFs) would promote healing of radiation burn lesions and compared results with those for MSC transplantation. Human clinical- grade GFs or bone marrow-derived MSCs were intradermally injected into mice 21 days after local leg irradiation. Immunostaining and real-time PCR analysis were used to assess the effects of each treatment on extracellular matrix remodeling and inflammation in skin on days 28 and 50 postirradiation. GFs induced the early development of thick, fully regenerated epidermis, skin appendages, and hair follicles, earlier than MSCs did. The acceleration of wound healing by GFs involved rearrangement of the deposited collagen, modification of the Col/MMP/TIMP balance, and modulation of the expression and localization of tenascin-C and of the expression of growth factors (VEGF, EGF, and FGF7). As MSC treatment did, GF injection decreased the irradiation-induced inflammatory response and switched the differentiation of macrophages toward an M2-like phenotype, characterized by CD163(+) macrophage infiltration and strong expression of arginase-1. These findings indicate that GFs are an attractive target for regenerative medicine, for easier to collect, can grow in culture, and promote cutaneous wound healing in irradiation burn lesions.
Collapse
Affiliation(s)
- Christine Linard
- 1 Institut de Radioprotection et de Sûreté Nucléaire (IRSN) , Fontenay-aux-Roses, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ballinger MN, Newstead MW, Zeng X, Bhan U, Mo XM, Kunkel SL, Moore BB, Flavell R, Christman JW, Standiford TJ. IRAK-M promotes alternative macrophage activation and fibroproliferation in bleomycin-induced lung injury. THE JOURNAL OF IMMUNOLOGY 2015; 194:1894-904. [PMID: 25595781 DOI: 10.4049/jimmunol.1402377] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Idiopathic pulmonary fibrosis is a devastating lung disease characterized by inflammation and the development of excessive extracellular matrix deposition. Currently, there are only limited therapeutic intervenes to offer patients diagnosed with pulmonary fibrosis. Although previous studies focused on structural cells in promoting fibrosis, our study assessed the contribution of macrophages. Recently, TLR signaling has been identified as a regulator of pulmonary fibrosis. IL-1R-associated kinase-M (IRAK-M), a MyD88-dependent inhibitor of TLR signaling, suppresses deleterious inflammation, but may paradoxically promote fibrogenesis. Mice deficient in IRAK-M (IRAK-M(-/-)) were protected against bleomycin-induced fibrosis and displayed diminished collagen deposition in association with reduced production of IL-13 compared with wild-type (WT) control mice. Bone marrow chimera experiments indicated that IRAK-M expression by bone marrow-derived cells, rather than structural cells, promoted fibrosis. After bleomycin, WT macrophages displayed an alternatively activated phenotype, whereas IRAK-M(-/-) macrophages displayed higher expression of classically activated macrophage markers. Using an in vitro coculture system, macrophages isolated from in vivo bleomycin-challenged WT, but not IRAK-M(-/-), mice promoted increased collagen and α-smooth muscle actin expression from lung fibroblasts in an IL-13-dependent fashion. Finally, IRAK-M expression is upregulated in peripheral blood cells from idiopathic pulmonary fibrosis patients and correlated with markers of alternative macrophage activation. These data indicate expression of IRAK-M skews lung macrophages toward an alternatively activated profibrotic phenotype, which promotes collagen production, leading to the progression of experimental pulmonary fibrosis.
Collapse
Affiliation(s)
- Megan N Ballinger
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210;
| | - Michael W Newstead
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109
| | - Xianying Zeng
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109
| | - Urvashi Bhan
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109
| | - Xiaokui M Mo
- Department of Biomedical Informatics, Center for Biostatistics, The Ohio State University, Columbus, OH 43221
| | - Steven L Kunkel
- Department of Pathology, University of Michigan Medical Center, Ann Arbor, MI 48109; and
| | - Bethany B Moore
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109
| | - Richard Flavell
- Department of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520
| | - John W Christman
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210
| | - Theodore J Standiford
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI 48109
| |
Collapse
|
35
|
Van der Meeren A, Moureau A, Griffiths NM. Macrophages as key elements of Mixed-oxide [U-Pu(O2)] distribution and pulmonary damage after inhalation? Int J Radiat Biol 2014; 90:1095-103. [PMID: 25029673 DOI: 10.3109/09553002.2014.943848] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
UNLABELLED Abstract Purpose: To investigate the consequences of alveolar macrophage (AM) depletion on Mixed OXide fuel (MOX: U, Pu oxide) distribution and clearance, as well as lung damage following MOX inhalation. MATERIALS AND METHODS Rats were exposed to MOX by nose only inhalation. AM were depleted with intratracheal administration of liposomal clodronate at 6 weeks. Lung changes, macrophage activation, as well as local and systemic actinide distribution were studied up to 3 months post-inhalation. RESULTS Clodronate administration modified excretion/retention patterns of α activity. At 3 months post-inhalation lung retention was higher in clodronate-treated rats compared to Phosphate Buffered Saline (PBS)-treated rats, and AM-associated α activity was also increased. Retention in liver was higher in clodronate-treated rats and fecal and urinary excretions were lower. Three months after inhalation, rats exhibited lung fibrotic lesions and alveolitis, with no marked differences between the two groups. Foamy macrophages of M2 subtype [inducible Nitric Oxide Synthase (iNOS) negative but galectin-3 positive] were frequently observed, in correlation with the accumulation of MOX particles. AM from all MOX-exposed rats showed increased chemokine levels as compared to sham controls. CONCLUSION Despite the transient reduced AM numbers in clodronate-treated animals no major differences on lung damage were observed as compared to non-treated rats after MOX inhalation. The higher lung activity retention in rats receiving clodronate seems to be part of a general inflammatory response and needs further investigation.
Collapse
Affiliation(s)
- Anne Van der Meeren
- Laboratoire de RadioToxicologie, CEA/DSV/iRCM, Bruyères le Châtel , Arpajon , France
| | | | | |
Collapse
|
36
|
Horton JA, Hudak KE, Chung EJ, White AO, Scroggins BT, Burkeen JF, Citrin DE. Mesenchymal stem cells inhibit cutaneous radiation-induced fibrosis by suppressing chronic inflammation. Stem Cells 2014; 31:2231-41. [PMID: 23897677 DOI: 10.1002/stem.1483] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Revised: 06/05/2013] [Accepted: 06/18/2013] [Indexed: 12/16/2022]
Abstract
Exposure to ionizing radiation (IR) can result in the development of cutaneous fibrosis, for which few therapeutic options exist. We tested the hypothesis that bone marrow-derived mesenchymal stem cells (BMSC) would favorably alter the progression of IR-induced fibrosis. We found that a systemic infusion of BMSC from syngeneic or allogeneic donors reduced skin contracture, thickening, and collagen deposition in a murine model. Transcriptional profiling with a fibrosis-targeted assay demonstrated increased expression of interleukin-10 (IL-10) and decreased expression of IL-1β in the irradiated skin of mice 14 days after receiving BMSC. Similarly, immunoassay studies demonstrated durable alteration of these and several additional inflammatory mediators. Immunohistochemical studies revealed a reduction in infiltration of proinflammatory classically activated CD80(+) macrophages and increased numbers of anti-inflammatory regulatory CD163(+) macrophages in irradiated skin of BMSC-treated mice. In vitro coculture experiments confirmed that BMSC induce expression of IL-10 by activated macrophages, suggesting polarization toward a regulatory phenotype. Furthermore, we demonstrated that tumor necrosis factor-receptor 2 (TNF-R2) mediates IL-10 production and transition toward a regulatory phenotype during coculture with BMSC. Taken together, these data demonstrate that systemic infusion of BMSC can durably alter the progression of radiation-induced fibrosis by altering macrophage phenotype and suppressing local inflammation in a TNF-R2-dependent fashion.
Collapse
Affiliation(s)
- Jason A Horton
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Zhang J, Li B, Ding X, Sun M, Li H, Yang M, Zhou C, Yu H, Liu H, Yu G. Genetic variants in inducible nitric oxide synthase gene are associated with the risk of radiation-induced lung injury in lung cancer patients receiving definitive thoracic radiation. Radiother Oncol 2014; 111:194-8. [PMID: 24746566 DOI: 10.1016/j.radonc.2014.03.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 02/02/2014] [Accepted: 03/09/2014] [Indexed: 01/20/2023]
Abstract
BACKGROUND AND PURPOSE Nitric oxide (NO), mainly synthesized by inducible nitric oxide synthase (NOS2) in pathological conditions, plays an important role in cytotoxicity, inflammation and fibrosis. Elevations in exhaled NO after thoracic radiation have been reported to predict radiation-induced lung injury (RILI). This study examined whether genetic variations in NOS2 gene is associated with the risk of RILI. MATERIAL AND METHODS A cohort of 301 patients between 2009 and 2011 were genotyped for 21 single nucleotide polymorphisms (SNPs) in the NOS2 gene by the Sequenom MassArray system. Kaplan-Meier cumulative probability was used to assess RILI risk and Cox proportional hazards analyses were performed to evaluate the effect of NOS2 genotypes on RILI. RESULTS Multivariate analysis found that three SNPs (rs2297518, rs1137933 and rs16949) in NOS2 were significantly associated with risk of RILI⩾2 (P value=0.001, 0.000092, 0.001, respectively) after adjusting for other covariates. Their associations were independent of radiation dose and mean lung dose. Further haplotype analysis indicated that the ATC haplotype of three SNPs is associated with reducing the risk of developing RILI. CONCLUSION Our results demonstrate that genetic variants of NOS2 may serve as a reliable predictor of RILI in lung cancer patients treated with thoracic radiation.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital, Shandong Academy of Medical Sciences, Jinan, PR China; Shandong's Key Laboratory of Radiation Oncology, Jinan, PR China; Department of Radiation Oncology, Cancer Hospital, Tianjin Medical University, PR China
| | - Baosheng Li
- Department of Radiation Oncology, Shandong Cancer Hospital, Shandong Academy of Medical Sciences, Jinan, PR China; Shandong's Key Laboratory of Radiation Oncology, Jinan, PR China.
| | - Xiuping Ding
- Department of Radiation Oncology, Shandong Cancer Hospital, Shandong Academy of Medical Sciences, Jinan, PR China; Shandong's Key Laboratory of Radiation Oncology, Jinan, PR China
| | - Mingping Sun
- Department of Radiation Oncology, Shandong Cancer Hospital, Shandong Academy of Medical Sciences, Jinan, PR China; Shandong's Key Laboratory of Radiation Oncology, Jinan, PR China
| | - Hongsheng Li
- Department of Radiation Oncology, Shandong Cancer Hospital, Shandong Academy of Medical Sciences, Jinan, PR China; Shandong's Key Laboratory of Radiation Oncology, Jinan, PR China
| | - Ming Yang
- College of Life Science and Technology, Beijing University of Chemical Technology, PR China
| | - Changchun Zhou
- Department of Radiation Oncology, Shandong Cancer Hospital, Shandong Academy of Medical Sciences, Jinan, PR China; Shandong's Key Laboratory of Radiation Oncology, Jinan, PR China
| | - Haiying Yu
- Department of Radiology, Shandong Cancer Hospital, Jinan, PR China
| | - Hong Liu
- Shandong Provincial Institute of Dermatology and Venereology, Jinan, PR China
| | - Gongqi Yu
- Shandong Provincial Institute of Dermatology and Venereology, Jinan, PR China
| |
Collapse
|
38
|
Redente EF, Keith RC, Janssen W, Henson PM, Ortiz LA, Downey GP, Bratton DL, Riches DWH. Tumor necrosis factor-α accelerates the resolution of established pulmonary fibrosis in mice by targeting profibrotic lung macrophages. Am J Respir Cell Mol Biol 2014; 50:825-37. [PMID: 24325577 PMCID: PMC4068926 DOI: 10.1165/rcmb.2013-0386oc] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 12/04/2013] [Indexed: 01/13/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a relentless, fibrotic parenchymal lung disease in which alternatively programmed macrophages produce profibrotic molecules that promote myofibroblast survival and collagen synthesis. Effective therapies to treat patients with IPF are lacking, and conventional therapy may be harmful. We tested the hypothesis that therapeutic lung delivery of the proinflammatory cytokine tumor necrosis factor (TNF)-α into wild-type fibrotic mice would reduce the profibrotic milieu and accelerate the resolution of established pulmonary fibrosis. Fibrosis was assessed in bleomycin-instilled wild-type and TNF-α(-/-) mice by measuring hydroxyproline levels, static compliance, and Masson's trichrome staining. Macrophage infiltration and programming status was assessed by flow cytometry of enzymatically digested lung and in situ immunostaining. Pulmonary delivery of TNF-α to wild-type mice with established pulmonary fibrosis was found to reduce their fibrotic burden, to improve lung function and architecture, and to reduce the number and programming status of profibrotic alternatively programmed macrophages. In contrast, fibrosis and alternative macrophage programming were prolonged in bleomycin-instilled TNF-α(-/-) mice. To address the role of the reduced numbers of alternatively programmed macrophages in the TNF-α-induced resolution of established pulmonary fibrosis, we conditionally depleted macrophages in MAFIA (MAcrophage Fas-Induced Apoptosis) mice. Conditional macrophage depletion phenocopied the resolution of established pulmonary fibrosis observed after therapeutic TNF-α delivery. Taken together, our results show for the first time that TNF-α is involved in the resolution of established pulmonary fibrosis via a mechanism involving reduced numbers and programming status of profibrotic macrophages. We speculate that pulmonary delivery of TNF-α or augmenting its signaling pathway represent a novel therapeutic strategy to resolve established pulmonary fibrosis.
Collapse
Affiliation(s)
| | - Rebecca C. Keith
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine
| | - William Janssen
- Department of Medicine, National Jewish Health, Denver, Colorado
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine
| | - Peter M. Henson
- Program in Cell Biology, Department of Pediatrics, and
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine
- Integrated Department of Immunology, and
| | - Luis A. Ortiz
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh Pennsylvania
| | - Gregory P. Downey
- Department of Medicine, National Jewish Health, Denver, Colorado
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine
- Integrated Department of Immunology, and
| | | | - David W. H. Riches
- Program in Cell Biology, Department of Pediatrics, and
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine
- Integrated Department of Immunology, and
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, Colorado; and
| |
Collapse
|
39
|
Zhao X, Yang H, Jiang G, Ni M, Deng Y, Cai J, Li Z, Shen F, Tao X. Simvastatin attenuates radiation-induced tissue damage in mice. JOURNAL OF RADIATION RESEARCH 2014; 55:257-64. [PMID: 24105712 PMCID: PMC3951077 DOI: 10.1093/jrr/rrt115] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 08/17/2013] [Accepted: 08/21/2013] [Indexed: 05/18/2023]
Abstract
The aim of this study was to investigate the protective effect of simvastatin against radiation-induced tissue injury in mice. Mice were radiated with 4 Gy or 8 Gy after 20 mg/kg/d simvastatin treatment over 2 weeks. Morphological changes were observed in the jejunum and bone marrow, and apoptotic cells were determined in both tissues. Peripheral blood cells were counted, and the superoxide dismutase (SOD) activity and the malondialdehyde (MDA) level in tissues of both thymus and spleen were measured. Compared with the radiation-only group, 20 mg/kg/d simvastatin administration significantly increased the mean villi height and decreased apoptotic cells in jejunum tissue, and stimulated regeneration and reduced apoptotic cells in bone marrow. Peripheral blood cell analysis revealed that simvastatin treatment induced a larger number of red blood cells and increased the hemoglobin level present after 4 Gy of radiation. Interestingly, it was also found that the number of peripheral endothelial progenitor cells was markedly increased following simvastatin administration. Antioxidant determination for tissues displayed that simvastatin therapy increased the SOD activity after both 4 and 8 Gy of radiation, but only decreased the MDA level after 4 Gy. Simvastatin ameliorated radiation-induced tissue damage in mice. The radioprotective effect of simvastatin was possibly related to inhibition of apoptosis and improvement of oxygen-carrying and antioxidant activities.
Collapse
Affiliation(s)
- Xinbin Zhao
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, 415 Feng-Yang Road, Shanghai 200003, China
| | - Hong Yang
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, 415 Feng-Yang Road, Shanghai 200003, China
| | - Guojun Jiang
- Department of Pharmacy, Xiaoshan Hospital, 728 Yu-Cai-Bei Road, Hangzhou, Zhejiang 311202, China
| | - Min Ni
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, 415 Feng-Yang Road, Shanghai 200003, China
| | - Yaping Deng
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, 325 Guo-He Road, Shanghai 200433, China
| | - Jian Cai
- Department of Pharmacy, Xiaoshan Hospital, 728 Yu-Cai-Bei Road, Hangzhou, Zhejiang 311202, China
| | - Zhangpeng Li
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, 325 Guo-He Road, Shanghai 200433, China
| | - Fuming Shen
- Department of Pharmacology, School of Pharmacy, Second Military Medical University, 325 Guo-He Road, Shanghai 200433, China
- Department of Pharmacy, Shanghai Tenth People's Hospital, Tongji University, 301 Yan-Chang-Zhong Road, Shanghai 200072, China
| | - Xia Tao
- Department of Pharmacy, Changzheng Hospital, Second Military Medical University, 415 Feng-Yang Road, Shanghai 200003, China
- Corresponding author. Tel: +86-21-8188-6182; Fax: +86-21-6549-3951;
| |
Collapse
|
40
|
Ding NH, Li JJ, Sun LQ. Molecular mechanisms and treatment of radiation-induced lung fibrosis. Curr Drug Targets 2013; 14:1347-56. [PMID: 23909719 PMCID: PMC4156316 DOI: 10.2174/13894501113149990198] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 07/11/2013] [Accepted: 07/11/2013] [Indexed: 02/06/2023]
Abstract
Radiation-induced lung fibrosis (RILF) is a severe side effect of radiotherapy in lung cancer patients that presents as a progressive pulmonary injury combined with chronic inflammation and exaggerated organ repair. RILF is a major barrier to improving the cure rate and well-being of lung cancer patients because it limits the radiation dose that is required to effectively kill tumor cells and diminishes normal lung function. Although the exact mechanism is unclear, accumulating evidence suggests that various cells, cytokines and regulatory molecules are involved in the tissue reorganization and immune response modulation that occur in RILF. In this review, we will summarize the general symptoms, diagnostics, and current understanding of the cells and molecular factors that are linked to the signaling networks implicated in RILF. Potential approaches for the treatment of RILF will also be discussed. Elucidating the key molecular mediators that initiate and control the extent of RILF in response to therapeutic radiation may reveal additional targets for RILF treatment to significantly improve the efficacy of radiotherapy for lung cancer patients.
Collapse
Affiliation(s)
- Nian-Hua Ding
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China
| | - Jian Jian Li
- Department of Radiation Oncology, NCI-Designated Comprehensive Cancer Center, University of California at Davis, Sacramento, CA 95817, USA
| | - Lun-Quan Sun
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410078, China
| |
Collapse
|
41
|
Lacavé-Lapalun JV, Benderitter M, Linard C. Flagellin or lipopolysaccharide treatment modified macrophage populations after colorectal radiation of rats. J Pharmacol Exp Ther 2013; 346:75-85. [PMID: 23596059 DOI: 10.1124/jpet.113.204040] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Radiation-induced acute intestinal toxicity remains a major limitation to the delivery of tumoricidal doses of colorectal irradiation. Recent reports indicate that Toll-like receptor (TLR) agonists TLR4 and TLR5 protect against toxicity due to intestinal irradiation. The phenotype (M1 or M2) of macrophages expressing TLRs may play a role in tissue repair. The aim was to investigate whether administration of TLR4 agonist lipopolysaccharide (LPS) or TLR5 agonist flagellin after irradiation modified the recruitment and phenotype of colonic macrophages and improved tissue damage. Rats were exposed to single 20- or 27-Gy doses of colorectal irradiation. TLR4 agonist LPS or TLR5 agonist flagellin (at 50 or 200 µg/rat) was administered i.p. 3 days after irradiation. Flow cytometric analysis, immunostaining, and real-time polymerase chain reaction analysis were used to assess the M1/M2 phenotype and crypt cell proliferation 7 days after irradiation. Irradiation (20 and 27 Gy) increased TLR4⁺ and TLR5⁺ macrophage frequency in the mucosa. LPS or flagellin administration maintained this elevated frequency after the 27-Gy irradiation. LPS and flagellin drove macrophages toward the anti-inflammatory M2 phenotype by increasing Arg1 and CD163 expression and microenvironmental effector molecules (C-C motif chemokine 22, transforming growth factor-β1, and interleukin-10). Proliferating cell nuclear antigen immunostaining, Ki67 expression, and antimicrobial factor Reg3γ showed that the M2 shift correlated with epithelial regeneration. In conclusion, administration of either LPS or flagellin after colorectal irradiation may provide effective protection against epithelial remodeling. This tissue repair was associated with an M2 macrophage shift. Using TLR agonists to moderately activate innate immunity should be considered as a strategy for protecting healthy tissue from irradiation.
Collapse
Affiliation(s)
- Jean-Victor Lacavé-Lapalun
- Laboratory of Radiopathology and Experimental Therapies, Institute for Radiological Protection and Nuclear Safety, Fontenay-aux-Roses, France
| | | | | |
Collapse
|
42
|
Radiation-induced lung injury is mitigated by blockade of gastrin-releasing peptide. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1248-54. [PMID: 23395092 DOI: 10.1016/j.ajpath.2012.12.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 10/27/2012] [Accepted: 12/12/2012] [Indexed: 02/06/2023]
Abstract
Gastrin-releasing peptide (GRP), secreted by pulmonary neuroendocrine cells, mediates oxidant-induced lung injury in animal models. Considering that GRP blockade abrogates pulmonary inflammation and fibrosis in hyperoxic baboons, we hypothesized that ionizing radiation triggers GRP secretion, contributing to inflammatory and fibrotic phases of radiation-induced lung injury (RiLI). Using C57BL/6 mouse model of pulmonary fibrosis developing ≥20 weeks after high-dose thoracic radiation (15 Gy), we injected small molecule 77427 i.p. approximately 1 hour after radiation then twice weekly for up to 20 weeks. Sham controls were anesthetized and placed in the irradiator without radiation. Lung paraffin sections were immunostained and quantitative image analyses performed. Mice exposed to radiation plus PBS had increased interstitial CD68(+) macrophages 4 weeks after radiation and pulmonary neuroendocrine cells hyperplasia 6 weeks after radiation. Ten weeks later radiation plus PBS controls had significantly increased pSmad2/3(+) nuclei/cm(2). GRP blockade with 77427 treatment diminished CD68(+), GRP(+), and pSmad2/3(+) cells. Finally, interstitial fibrosis was evident 20 weeks after radiation by immunostaining for α-smooth muscle actin and collagen deposition. Treatment with 77427 abrogated interstitial α-smooth muscle actin and collagen. Sham mice given 77427 did not differ significantly from PBS controls. Our data are the first to show that GRP blockade decreases inflammatory and fibrotic responses to radiation in mice. GRP blockade is a novel radiation fibrosis mitigating agent that could be clinically useful in humans exposed to radiation therapeutically or unintentionally.
Collapse
|
43
|
Du ZZ, Ren H, Song JF, Zhang LF, Lin F, Wang HY. Rabbit model of radiation-induced lung injury. ASIAN PAC J TROP MED 2013; 6:237-41. [PMID: 23375041 DOI: 10.1016/s1995-7645(13)60031-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2012] [Revised: 12/15/2012] [Accepted: 02/15/2013] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To explore the feasibility of establishing an animal model of chronic radiation-induced lung injury. METHODS Twenty-eight New Zealand white rabbits were randomly divided into 3 groups (the right lung irradiation group, the whole lung irradiation group and the control group). Animal model of radiation-induced lung injury was established by high-does radiotherapy in the irradiation groups, then all rabbits underwent CT and pathological examinations at 1, 2, 4, 8, 12, 16 weeks, respectively after radiation. RESULTS Within 4 weeks of irradiation, some rabbits in the right lung irradiation group and whole lung irradiation group died. CT and pathological examinations all showed acute radiation pneumonitis. At 8-12 weeks after irradiation, CT scanning showed ground glass samples signs, patchy shadows and fibrotic stripes. Pathological examination showed the fibrosis pulmonary alveolar wall thickened obviously. CONCLUSIONS The clinical animal model of chronic radiation-induced lung injury which corresponds to practical conditions in clinic can be successfully established.
Collapse
Affiliation(s)
- Zhen-Zong Du
- Cardio-Thoracic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, Guangxi, China
| | | | | | | | | | | |
Collapse
|