51
|
Yadav P, Shankar BS. Radio resistance in breast cancer cells is mediated through TGF-β signalling, hybrid epithelial-mesenchymal phenotype and cancer stem cells. Biomed Pharmacother 2019; 111:119-130. [DOI: 10.1016/j.biopha.2018.12.055] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 12/11/2018] [Accepted: 12/14/2018] [Indexed: 12/20/2022] Open
|
52
|
Liu Z, Liang X, Li X, Liu X, Zhu M, Gu Y, Zhou P. MiRNA-21 functions in ionizing radiation-induced epithelium-to-mesenchymal transition (EMT) by downregulating PTEN. Toxicol Res (Camb) 2019; 8:328-340. [PMID: 31160967 DOI: 10.1039/c9tx00019d] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 02/26/2019] [Indexed: 12/19/2022] Open
Abstract
Radiation-induced pulmonary fibrosis (RIPF) results from thoracic radiotherapy and severely limits the use of radiotherapy. Recent studies suggest that epithelium-to-mesenchymal transition (EMT) contributes to pulmonary fibrosis. Although miRNA dysregulation participates in a variety of pathophysiologic processes, their roles in fibrotic lung diseases and EMT are unclear. In this study, we aimed to identify key miRNAs involved in this process using a mouse model of RIPF previously established by irradiation with a single dose (20 Gy) of 60Co γ-rays. At 2-weeks post-irradiation, a set of significantly upregulated miRNAs was identified in lung tissue by miRNA array analysis. This included miR-21, which has been reported to contribute to the pulmonary fibrotic response induced by stereotactic body radiotherapy. Here, we showed that miR-21 expression increased in parallel with EMT progression in the lungs of irradiated mice. Ectopic miR-21 expression promoted EMT progression in lung epithelial cells. Furthermore, downregulation of miR-21 expression by transfection of its inhibitor inhibited ionizing radiation (IR)-induced EMT. Knockdown of PTEN, which is the functional target of miR-21, reversed the attenuation of IR-induced EMT mediated by miR-21 downregulation. Radiation treatment decreased PTEN expression and increased Akt phosphorylation; these effects were abolished by the miR-21 inhibitor. MiR-21 overexpression in lung epithelial cell also downregulated PTEN expression and upregulated Akt phosphorylation. In conclusion, we have demonstrated that miR-21 functions as a key regulator of IR-induced EMT in lung epithelial cells via the PTEN/Akt pathway. Targeting miR-21 is implicated as a novel therapeutic strategy for the prevention of RIPF.
Collapse
Affiliation(s)
- Zheng Liu
- School of Public Health , University of South China , Hengyang , Hunan Province 421001 , P. R. China . ; .,Beijing Key Laboratory for Radiobiology , Beijing Institute of Radiation Medicine , Beijing 100850 , P. R. China
| | - Xin Liang
- Graduate School , Anhui Medical University , Hefei , Anhui province 230032 , P. R. China
| | - Xueping Li
- School of Life Science , Shihezi University , Shihezi , Xinjiang Province 832003 , P. R. China
| | - Xiaodan Liu
- Beijing Key Laboratory for Radiobiology , Beijing Institute of Radiation Medicine , Beijing 100850 , P. R. China
| | - Maoxiang Zhu
- Beijing Key Laboratory for Radiobiology , Beijing Institute of Radiation Medicine , Beijing 100850 , P. R. China
| | - Yongqing Gu
- School of Public Health , University of South China , Hengyang , Hunan Province 421001 , P. R. China . ; .,Beijing Key Laboratory for Radiobiology , Beijing Institute of Radiation Medicine , Beijing 100850 , P. R. China
| | - Pingkun Zhou
- School of Public Health , University of South China , Hengyang , Hunan Province 421001 , P. R. China . ; .,Beijing Key Laboratory for Radiobiology , Beijing Institute of Radiation Medicine , Beijing 100850 , P. R. China
| |
Collapse
|
53
|
Zhong Y, Lin Z, Lu J, Lin X, Xu W, Wang N, Huang S, Wang Y, Zhu Y, Chen Z, Lin S. CCL2-CCL5/CCR4 contributed to radiation-induced epithelial-mesenchymal transition of HPAEpiC cells via the ERK signaling pathways. Am J Transl Res 2019; 11:733-743. [PMID: 30899375 PMCID: PMC6413271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 12/23/2018] [Indexed: 06/09/2023]
Abstract
Radiation-induced lung toxicity, including radiation pneumonitis and pulmonary fibrosis, often occurs in patients receiving radiation therapy. Epithelial-mesenchymal transition (EMT) of alveolar epithelial cells (AECs) plays critical roles in radiation-induced lung toxicity. In the present study, RNA sequencing was applied to examine the whole transcriptomes of human pulmonary AEC cells (HPAEpiC) with or without radiation treatment. We found that cytokine, chemokine and cell adhesion signaling pathways were enriched in radiation-treated cells. CCL2 (C-C Motif Chemokine Ligand 2), CCL5 and CCR4 (C-C Motif Chemokine Receptor 4) were among the top enriched genes in chemokine signaling pathway. The upregulation of CCL2, CCL5 and CCR4 in response to irradiation was confirmed at both mRNA and protein levels by real-time PCR, western blotting and enzyme-linked immunosorbent assay analyses. Ophiopogonin B, a bioactive ingredient of Radix Ophiopogon japonicas, was found to attenuate radiation-induced EMT in HPAEpiC cells as demonstrated by the alteration in cell morphology, and the expression of E-cadherin and Vimentin. Ophiopogonin B could also reduce radiation-induced expression of CCL2, CCL5, CCR4 and phosphorylated ERK (p-ERK). Moreover, CCR4 knockdown, U0126 (a MEK/ERK inhibitor) or ophiopogonin B also partially blocked the EMT promoting effects of CCL2 and CCL5. Our data suggested CCL2, CCL5 and CCR4 may be potential therapeutic targets for radiation-induced lung toxicity. Ophiopogonin B, which could down-regulate CCL2, CCL5 and CCR4, may be a useful radioprotective agent.
Collapse
Affiliation(s)
- Yazhen Zhong
- Oncology Department, Hangzhou Hospital of Traditional Chinese Medicine, GuangXing Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhou, China
| | - Zechen Lin
- Fourth Clinical Medical College, Hangzhou Cancer HospitalHangzhou, China
| | - Jinhua Lu
- Department of Oncology Comprehensive Treatment, Hangzhou Cancer HospitalHangzhou, China
| | - Xianlei Lin
- Oncology Department, Hangzhou Hospital of Traditional Chinese Medicine, GuangXing Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhou, China
| | - Wei Xu
- Department of Integrated Chinese and Western Medicine, Hangzhou Cancer Hospital, Hangzhou Hospital Affiliated to Nanjing Medical UniversityHangzhou, China
| | - Nan Wang
- The First People’s Hospital of Xiaoshan HangzhouHangzhou, China
| | - Siyu Huang
- Fourth Clinical Medical College, Hangzhou Cancer HospitalHangzhou, China
| | - Yuanyuan Wang
- Fourth Clinical Medical College, Hangzhou Cancer HospitalHangzhou, China
| | - Yuan Zhu
- Fourth Clinical Medical College, Hangzhou Cancer HospitalHangzhou, China
| | - Zhu Chen
- Department of Oncology Comprehensive Treatment, Hangzhou Cancer HospitalHangzhou, China
| | - Shengyou Lin
- Oncology Department, Hangzhou Hospital of Traditional Chinese Medicine, GuangXing Hospital Affiliated to Zhejiang Chinese Medical UniversityHangzhou, China
| |
Collapse
|
54
|
Zhang E, Yang Y, Chen S, Peng C, Lavin MF, Yeo AJ, Li C, Liu X, Guan Y, Du X, Du Z, Shao H. Bone marrow mesenchymal stromal cells attenuate silica-induced pulmonary fibrosis potentially by attenuating Wnt/β-catenin signaling in rats. Stem Cell Res Ther 2018; 9:311. [PMID: 30428918 PMCID: PMC6234553 DOI: 10.1186/s13287-018-1045-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/27/2018] [Accepted: 10/14/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pulmonary fibrosis induced by silica dust is an irreversible, chronic, and fibroproliferative lung disease with no effective treatment at present. Previous studies have shown that early intervention with bone marrow mesenchymal stem/stromal cells (BMSCs) has positive effect on anti-pulmonary fibrosis caused by silica dust. However, early intervention using BMSCs is not practical, and the therapeutic effects of BMSCs advanced intervention on pulmonary fibrosis have rarely been reported. In this study, we investigated the effects of advanced transplantation (on the 28th day after exposure to silica suspension) of BMSCs on an established rat model of pulmonary fibrosis. METHODS Sprague Dawley (SD) rats were randomly divided into four groups including (1) control group (n = 6) which were normally fed, (2) silica model group (n = 6) which were exposed to silica suspension (1 mL of 50 mg/mL/rat), (3) BMSC transplantation group (n = 6) which received 1 mL BMSC suspension (2 × 106 cells/mL) by tail vein injection on the 28th day after exposure to silica suspension, and (4) BMSC-CM (conditioned medium) transplantation group (n = 6) which received CM from the same cell number by tail vein injection on the 28th day after exposure to silica suspension. On the 56th day after exposure to silica suspension, we used computed tomography (CT), hematoxylin and eosin (H&E), and Masson's trichrome staining to evaluate the changes in lung tissue. We examined the expression of epithelial-mesenchymal transition (EMT) and Wnt/β-catenin pathway-related proteins in lung tissue using immunohistochemistry and western blotting. RESULTS Successful construction of a pulmonary fibrosis model was confirmed by H&E and Masson's trichrome staining on the 28th day after exposure to silica suspension. On the 56th day after exposure, pulmonary CT examination showed a relieving effect of BMSCs on silica-induced pulmonary fibrosis which was confirmed by H&E and Masson's trichrome staining. Treatment of BMSCs increased the expression of epithelial marker proteins including E-cadherin (E-cad) and cytokeratin19 (CK19) and reduced the expression of fibrosis marker proteins including Vimentin (Vim) and α-Smooth actin (α-SMA) after exposure to silica suspension. Furthermore, we found that Wnt/β-catenin signaling pathway is abnormally activated in silica-induced pulmonary fibrosis, and exogenous transplantation of BMSCs may attenuate their expression. CONCLUSIONS BMSC transplantation inhibits the EMT to alleviate silica-induced pulmonary fibrosis in rats and the anti-fibrotic effect potentially by attenuating Wnt/β-catenin signaling. ᅟ: ᅟ.
Collapse
Affiliation(s)
- Enguo Zhang
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China.,School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Ye Yang
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China.,School of Medicine and Life Sciences, University of Jinan-Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Shangya Chen
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China
| | - Cheng Peng
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China.,Queensland Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, Brisbane, Queensland, Australia
| | - Martin F Lavin
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China.,University of Queensland Centre for Clinical Research (UQCCR), The University of Queensland, Brisbane, Queensland, Australia
| | - Abrey J Yeo
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China.,University of Queensland Centre for Clinical Research (UQCCR), The University of Queensland, Brisbane, Queensland, Australia
| | - Chao Li
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China
| | - Xiaoshan Liu
- Department of Radiology, Shandong Tumor Hospital, Shandong Academy of Medical Sciences, Jinan, Shandong, People's Republic of China
| | - Yingjun Guan
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China
| | - Xinjing Du
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China
| | - Zhongjun Du
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China.
| | - Hua Shao
- Department of Toxicology, Shandong Academy of Occupational Health and Occupational Medicine, Shandong Academy of Medical Sciences, No 18877 Jingshi Road, Lixia District Jinan, Jinan, 250062, Shandong, People's Republic of China.
| |
Collapse
|
55
|
Sunil Gowda SN, Rajasowmiya S, Vadivel V, Banu Devi S, Celestin Jerald A, Marimuthu S, Devipriya N. Gallic acid-coated sliver nanoparticle alters the expression of radiation-induced epithelial-mesenchymal transition in non-small lung cancer cells. Toxicol In Vitro 2018; 52:170-177. [PMID: 29928970 DOI: 10.1016/j.tiv.2018.06.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/15/2018] [Accepted: 06/16/2018] [Indexed: 01/03/2023]
Abstract
BACKGROUND Radiotherapy is the most widely used treatment method for treating cancer with or without surgery and chemotherapy. In lung cancer, it is one of the important treatment steps in excising the tumor from the lung tissue; unfortunately, radiation can induce epithelial- mesenchymal transition (EMT), a typical physiological process in which cuboidal shaped epithelial cell loses its phenotype and acquires mesenchymal-like phenotype thus, increases the metastasis progression in the body. To prevent EMT mediated metastasis, we aimed to 1) synthesize silver nanoparticles by using Gallic acid, a potential antioxidant which acts as stabilizing and reducing agent in the form of silver nanoparticle (GA-AgNPs) 2) to analyze its effect on EMT markers during radiation-induced EMT in A549 cells. METHODS A549 cells were irradiated with 8Gy (X-ray) and treated with GA-AgNPs at a fixed concentration under in vitro condition. GA-AgNPs were prepared and characterized for absorption, potential stability, size and morphology by UV-Visible spectrophotometer, Zeta potential and Transmission electron microscopy respectively. After irradiation, the morphology changes were observed using an inverted microscope, the gene and protein expression of EMT markers were analyzed by RT-PCR and western blotting. RESULTS/CONCLUSION GA-AgNPs are in nano size with fair stability. The synthesized nanoparticles suppressed the EMT markers including Vimentin, N-cadherin, Snail-1 and increased E-cadherin expression which might inhibit cancer cells to acquire radio resistant metastasis potential.
Collapse
Affiliation(s)
- S N Sunil Gowda
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - S Rajasowmiya
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - Vellingiri Vadivel
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - S Banu Devi
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - A Celestin Jerald
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India
| | - S Marimuthu
- Vishnu Cancer Center, Thanjavur, Tamil Nadu, India
| | - N Devipriya
- School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, India.
| |
Collapse
|
56
|
Aoto K, Ito K, Aoki S. Complex formation between platelet-derived growth factor receptor β and transforming growth factor β receptor regulates the differentiation of mesenchymal stem cells into cancer-associated fibroblasts. Oncotarget 2018; 9:34090-34102. [PMID: 30344924 PMCID: PMC6183337 DOI: 10.18632/oncotarget.26124] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/04/2018] [Indexed: 11/25/2022] Open
Abstract
Cancer-associated fibroblasts (CAFs) have recently gained attention as potent targets in cancer therapy because they are a crucial component of the tumor microenvironment and promote the growth and invasion of cancer cells. CAFs differentiate from fibroblasts, mesenchymal stem cells (MSCs), epithelial cells, and other cell types in response to transforming growth factor β (TGFβ) stimulation. The drugs tranilast, imatinib, and pirfenidone reportedly inhibit the differentiation of such cells into CAFs; however, it is unclear how they regulate TGFβ signaling. Here, we differentiated MSCs into CAFs in vitro and investigated which drugs suppressed this differentiation. Based on these results, we focused on platelet-derived growth factor (PDGF) receptor β (PDGFRβ) as a key molecule in the initiation of TGFβ signaling. PDGFRβ transmitted TGFβ signaling in MSCs by forming a complex with TGFβ receptor (TGFβR) independently of stimulation with its well-known ligand PDGF. Inhibitors of the differentiation of MSCs into CAFs attenuated complex formation between PDGFRβ and TGFβR. Moreover, PDGF stimulated PDGFRβ to a lesser extent in CAFs than in MSCs. This study indicates that PDGFRβ and TGFβ-TGFβR signaling cooperatively promote the differentiation of MSCs into CAFs in tumor microenvironments independently of canonical PDGF-PDGFR signaling. We propose that blockade of the interaction between PDGFRβ and TGFβR is a potential strategy to prevent TGFβ-mediated differentiation of MSCs into CAFs.
Collapse
Affiliation(s)
- Kaori Aoto
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba-city, Chiba 260-8675, Japan
| | - Kousei Ito
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba-city, Chiba 260-8675, Japan
| | - Shigeki Aoki
- Laboratory of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Chiba University, Inohana 1-8-1, Chuo-ku, Chiba-city, Chiba 260-8675, Japan
| |
Collapse
|
57
|
Zhang H, Song Y, Zhou C, Bai Y, Yuan D, Pan Y, Shao C. Blocking Endogenous H 2S Signaling Attenuated Radiation-Induced Long-Term Metastasis of Residual HepG2 Cells through Inhibition of EMT. Radiat Res 2018; 190:374-384. [PMID: 30016218 DOI: 10.1667/rr15074.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Recurrence and metastasis of hepatocellular carcinoma (HCC) after radiotherapy are frequently observed in clinical practice. To date, the involved mechanism, endogenous hydrogen sulfide (H2S), has not been well understood and warrants investigation. Here we demonstrated that both single-dose and fractionated irradiation enhanced metastasis of HCC cells both in vitro and in vivo at 20-60 days postirradiation. In particular, a gain in epithelial-mesenchymal transition (EMT) and mesenchymal features was observed. Further experiments revealed that endogenous H2S signaling was constitutively activated after irradiation. Knockdown of cystathionine-γ-lyase (CSE) or cystathionine-β-synthase (CBS), two main H2S-producing proteins, significantly diminished the increased expressions of EMT-related proteins induced by radiation through the p38MAPK pathway, leading to impaired invasion and metastasis of the residual HepG2 cells and their xenograft tumors. Moreover, blocking of the H2S pathway increased the radiosensitivity of the HepG2 xenograft tumor. Collectively, our results strongly suggest that endogenous H2S/CSE contributes to the long-term cell invasion and tumor metastasis induced by fractionated exposures and therefore, could become an attractive therapeutic target of HCC to eliminate radiotherapy-induced adverse effects.
Collapse
Affiliation(s)
- Hang Zhang
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Yimeng Song
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Cuiping Zhou
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Yang Bai
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Dexiao Yuan
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Yan Pan
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| | - Chunlin Shao
- Institute of Radiation Medicine, Fudan University, Shanghai 200032, China
| |
Collapse
|
58
|
Akter H, Yoon JH, Yoo YS, Kang MJ. Validation of Neurotensin Receptor 1 as a Therapeutic Target for Gastric Cancer. Mol Cells 2018; 41:591-602. [PMID: 29794962 PMCID: PMC6030244 DOI: 10.14348/molcells.2018.0025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/13/2018] [Accepted: 03/20/2018] [Indexed: 12/15/2022] Open
Abstract
Gastric cancer is the fifth most common type of malignancy worldwide, and the survival rate of patients with advanced-stage gastric cancer is low, even after receiving chemotherapy. Here, we validated neurotensin receptor 1 (NTSR1) as a potential therapeutic target in gastric cancer. We compared NTSR1 expression levels in sixty different gastric cancer-tissue samples and cells, as well as in other cancer cells (lung, breast, pancreatic, and colon), by assessing NTSR1 expression via semi-quantitative real-time reverse transcription polymerase chain reaction, immunocytochemistry and western blot. Following neurotensin (NT) treatment, we analyzed the expression and activity of matrix metalloproteinase-9 (MMP-9) and further determined the effects on cell migration and invasion via wound-healing and transwell assays. Our results revealed that NTSR1 mRNA levels were higher in gastric cancer tissues than non-cancerous tissues. Both of NTSR1 mRNA levels and expression were higher in gastric cancer cell lines relative to levels observed in other cancer-cell lines. Moreover, NT treatment induced MMP-9 expression and activity in all cancer cell lines, which was significantly decreased following treatment with the NTSR1 antagonist SR48692 or small-interfering RNA targeting NTSR1. Furthermore, NT-mediated metastases was confirmed by observing epithelial-mesenchymal transition markers SNAIL and E-cadherin in gastric cancer cells. NT-mediated invasion and migration of gastric cancer cells were reduced by NTSR1 depletion through the Erk signaling. These findings strongly suggested that NTR1 constitutes a potential therapeutic target for the inhibition of gastric cancer invasion and metastasis.
Collapse
Affiliation(s)
- Hafeza Akter
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792,
Korea
- Department of Biological Chemistry, University of Science and Technology, Daejeon 34113,
Korea
| | - Jung Hwan Yoon
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 06591,
Korea
| | - Young Sook Yoo
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792,
Korea
| | - Min-Jung Kang
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul 02792,
Korea
- Department of Biological Chemistry, University of Science and Technology, Daejeon 34113,
Korea
| |
Collapse
|
59
|
Ren J, Liu T, Han Y, Wang Q, Chen Y, Li G, Jiang L. GSK-3β inhibits autophagy and enhances radiosensitivity in non-small cell lung cancer. Diagn Pathol 2018; 13:33. [PMID: 29793508 PMCID: PMC5968472 DOI: 10.1186/s13000-018-0708-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/06/2018] [Indexed: 12/20/2022] Open
Abstract
Background Radiotherapy is one of the most common and effective treatment methods for cancer, and improving the radiosensitivity of tumor tissues during the treatment process is vital. We report the mechanisms of glycogen synthase kinase 3 (GSK-3) β-regulated autophagy and the effects of autophagy on radiosensitivity in non-small cell lung cancer (NSCLC). Method Immunohistochemical staining was performed to determine GSK-3β tissue expression in 89 NSCLC patients with follow-up data and the expression status of GSK-3β and autophagy in NSCLC tissues after X-ray radiotherapy. Western blots were used to quantitate changes in autophagy-related protein expression after A549 cells were treated with GSK-3β inhibitors and after H460 cells were transfected with GSK-3β mutants with different activities and X-ray irradiated. Clonogenic assays were used to measure the effect of autophagy on cellular proliferation. Results GSK-3β expression positively correlated with NSCLC differentiation (P < 0.05), and GSK-3β negativity was associated with a better prognosis in 89 NSCLC patients. After X-ray irradiation, the expression levels of GSK-3β and p62 were decreased in NSCLC tissues, and the expression levels of the autophagy-related protein LC3 were increased. A549 and H460 cells were selected as representative GSK-3β-high and GSK-3β-low expression cell lines. After transfecting H460 cells with different GSK-3β mutants [wild type GSK-3β (GSK-3β-WT), constitutively active GSK-3β (GSK-3β-S9A), and catalytically inactive GSK-3β (GSK-3β-K85R)] and subjecting these cells to X-ray irradiation, AMPK and LC3 expression levels decreased, and p62 expression levels increased. These effects were particularly significant for the GSK-3β-S9A mutant. In A549 cells, after GSK-3β inhibition and X-ray irradiation, AMPK and LC3 protein expression levels increased. Moreover, when autophagy was inhibited, cell proliferation decreased. Conclusion Our studies revealed that GSK-3β expression is associated with NSCLC differentiation, and patients with GSK-3β-negative tumors had a better prognosis. X-ray irradiation inhibited GSK-3β expression and promoted autophagy. Therefore, GSK-3β inhibits autophagy and enhances the radiosensitivity of NSCLC cells.
Collapse
Affiliation(s)
- Jialin Ren
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Science, China Medical University, Shenyang, 110001, China
| | - Tingting Liu
- Department of Surgery, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Yang Han
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Science, China Medical University, Shenyang, 110001, China.
| | - Qiongzi Wang
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Science, China Medical University, Shenyang, 110001, China
| | - Yanzhi Chen
- Department of Radiotherapy, Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Guang Li
- Department of Radiotherapy, First Affiliated Hospital, China Medical University, Shenyang, China
| | - Lihong Jiang
- Department of Pathology, General Hospital of Liaohe Oilfield, Panjin, China
| |
Collapse
|
60
|
Zhang Z, Luo Z, Min W, Zhang L, Wu Y, Hu X. An anti-cancer WxxxE-containing azurin polypeptide inhibits Rac1-dependent STAT3 and ERK/GSK-3β signaling in breast cancer cells. Oncotarget 2018; 8:43091-43103. [PMID: 28549350 PMCID: PMC5522130 DOI: 10.18632/oncotarget.17759] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/28/2017] [Indexed: 12/15/2022] Open
Abstract
In our previous study, we characterized a mycoplasmal small GTPase-like polypeptide of 240 amino acids that possesses an N-terminal WVLGE sequence. The N-terminal WVLGE sequence promotes activation of Rac1 and subsequent host cancer cell proliferation. To investigate the function of the WxxxE motif in the interaction with Rac1 and host tumor progression, we synthesized a 35-amino acid WVLGE-containing polypeptide derived from a cell-penetrating peptide derived from the azurin protein. We verified that the WVLGE-containing polypeptide targeted MCF-7 cells rather than MCF-10A cells. However, the WVLGE-containing polypeptide inhibited activation of Rac1 and induced cellular phenotypes that resulted from inhibition of Rac1. In addition, the WVLGE-containing polypeptide down-regulated phosphorylation of the STAT3 and ERK/GSK-3β signaling pathways, and this effect was abolished by either stimulation or inhibition of Rac1 activity. We also found that the WVLGE-containing polypeptide has a Rac1-dependent potential to suppress breast cancer growth in vitro and in vivo. We suggest that by acting as a Rac1 inhibitor, this novel polypeptide may be useful for the treatment of breast cancer.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Breast and Thyroid Surgery, Division of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Zhiyong Luo
- Department of Breast and Thyroid Surgery, Division of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Wenpu Min
- The First People's Hospital of Jingzhou, Jingzhou, People's Republic of China
| | - Lin Zhang
- Department of Breast and Thyroid Surgery, Division of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Yaqun Wu
- Department of Breast and Thyroid Surgery, Division of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Xiaopeng Hu
- Department of Breast and Thyroid Surgery, Division of General Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
61
|
Xiu G, Sui X, Wang Y, Zhang Z. FOXM1 regulates radiosensitivity of lung cancer cell partly by upregulating KIF20A. Eur J Pharmacol 2018; 833:79-85. [PMID: 29704495 DOI: 10.1016/j.ejphar.2018.04.021] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/17/2018] [Accepted: 04/20/2018] [Indexed: 12/25/2022]
Abstract
Forkhead box protein M1 (FOXM1), an important regulator of tumorigenesis in various human tumors, has recently been reported to play a role in the modulation of radiosensitivity in glioma and breast cancer cells. The present study aimed to investigate the effects of FOXM1 on radiotherapy resistance in human lung cancer and to explore the related molecular mechanisms. The results revealed that FOXM1 expression was upregulated in A549 and H1299 cells after IR (Ionizing radiation). FOXM1 inhibition impeded survival fractions, impeded proliferation, and triggered apoptosis after IR. Moreover, the silencing of FOXM1 dampened cell migration, invasion, and EMT (epithelial-mesenchyman transition) in A549 and H1299 cells treated by IR. In addition, KIF20A was also highly expressed in IR-treated A549 cells and downregulated by FOXM1 inhibition. Knockdown of KIF20A inhibited the survival fraction. Reintroduction of KIF20A partly reversed the effects of FOXM1 on the proliferation, apoptosis, and metastasis of A549 cells. Taken together, these results indicated that FOXM1 might enhance radioresistance partly through the induction of KIF20A expression.
Collapse
Affiliation(s)
- Guanghong Xiu
- No.1 Radiotherapy Department, Yantaishan Hospital, Yantai City, China.
| | - Xiujie Sui
- No.1 Radiotherapy Department, Yantaishan Hospital, Yantai City, China
| | - Yirong Wang
- No.1 Radiotherapy Department, Yantaishan Hospital, Yantai City, China
| | - Ze Zhang
- No.1 Radiotherapy Department, Yantaishan Hospital, Yantai City, China
| |
Collapse
|
62
|
Chen X, Zhang L, Jiang Y, Song L, Liu Y, Cheng F, Fan X, Cao X, Gong A, Wang D, Zhu H. Radiotherapy-induced cell death activates paracrine HMGB1-TLR2 signaling and accelerates pancreatic carcinoma metastasis. J Exp Clin Cancer Res 2018; 37:77. [PMID: 29615080 PMCID: PMC5883315 DOI: 10.1186/s13046-018-0726-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/06/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Dying cells after irradiation could promote the repopulation of surviving cancer cells leading to tumor recurrence. We aim to define the role of dying cells in promoting pancreatic cancer cells metastasis following radiotherapy. METHODS Using the transwell system as the in vitro co-culture model, a small number of untreated pancreatic cancer cells were seeded in the upper chamber, while a larger number of lethally treated pancreatic cancer cells were seeded in the lower chamber. A series of experiments were conducted to investigate the role of dying-cell-derived HMGB1 on the invasion of pancreatic cancer in vitro and cancer metastasis in vivo. We then designed shRNA knockdown and Western blot assays to detect signaling activity. RESULTS We found that dying pancreatic cancer cells significantly promote the invasion of pancreatic cancer cells in vitro and cancer metastasis in vivo. HMGB1 gene knockdown attenuated the migration-stimulating effect of irradiated, dying cells on living pancreatic cancer cells. Finally, we showed that dying-cell-derived HMGB1 functions in a paracrine manner to affect cancer-cell migration dependent on acquiring an epithelial-mesenchymal transition (EMT) phenotype and PI3K/pAkt activation. This process is mediated by the receptor for TLR2. CONCLUSION Our study indicates that, during radiotherapy, dying pancreatic cancer cells activate paracrine signaling events that promote the mobility of surviving tumor cells. We suggest a strategy to inhibit HMGB1 for preventing pancreatic carcinoma relapse and metastasis.
Collapse
Affiliation(s)
- Xuelian Chen
- The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Lirong Zhang
- The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yujie Jiang
- The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Lian Song
- The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yanfang Liu
- The First People's Hospital of Zhenjiang, Zhenjiang, 212001, China
| | - Fang Cheng
- Faculty of Science and Engineering, Åbo Akademi University and Turku Centre for Biotechnology, -20520, Turku, FI, Finland
| | - Xin Fan
- The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Xiongfeng Cao
- The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Aihua Gong
- School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Dongqing Wang
- The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
- Department of Radiology, The Affiliated Hospital of Jiangsu University, Jiangsu University, 438 Jiefang Road, Zhenjiang, Jiangsu Province, 212013, China.
| | - Haitao Zhu
- The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
- Department of Radiology, The Affiliated Hospital of Jiangsu University, Jiangsu University, 438 Jiefang Road, Zhenjiang, Jiangsu Province, 212013, China.
| |
Collapse
|
63
|
Fei Y, Xiong Y, Shen X, Zhao Y, Zhu Y, Wang L, Liang Z. Cathepsin L promotes ionizing radiation-induced U251 glioma cell migration and invasion through regulating the GSK-3β/CUX1 pathway. Cell Signal 2018; 44:62-71. [DOI: 10.1016/j.cellsig.2018.01.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 12/20/2017] [Accepted: 01/09/2018] [Indexed: 12/13/2022]
|
64
|
Meel MH, Schaper SA, Kaspers GJL, Hulleman E. Signaling pathways and mesenchymal transition in pediatric high-grade glioma. Cell Mol Life Sci 2018; 75:871-887. [PMID: 29164272 PMCID: PMC5809527 DOI: 10.1007/s00018-017-2714-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/24/2017] [Accepted: 11/14/2017] [Indexed: 12/16/2022]
Abstract
Pediatric high-grade gliomas (pHGG), including diffuse intrinsic pontine gliomas (DIPG), are the most lethal types of cancer in children. In recent years, it has become evident that these tumors are driven by epigenetic events, mainly mutations involving genes encoding Histone 3, setting them apart from their adult counterparts. These tumors are exceptionally resistant to chemotherapy and respond only temporarily to radiotherapy. Moreover, their delicate location and diffuse growth pattern make complete surgical resection impossible. In many other forms of cancer, chemo- and radioresistance, in combination with a diffuse, invasive phenotype, are associated with a transcriptional program termed the epithelial-to-mesenchymal transition (EMT). Activation of this program allows cancer cells to survive individually, invade surrounding tissues and metastasize. It also enables them to survive exposure to cytotoxic therapy, including chemotherapeutic drugs and radiation. We here suggest that EMT plays an important, yet poorly understood role in the biology and therapy resistance of pHGG and DIPG. This review summarizes the current knowledge on the major signal transduction pathways and transcription factors involved in the epithelial-to-mesenchymal transition in cancer in general and in pediatric HGG and DIPG in particular. Despite the fact that the mesenchymal transition has not yet been specifically studied in pHGG and DIPG, activation of pathways and high levels of transcription factors involved in EMT have been described. We conclude that the mesenchymal transition is likely to be an important element of the biology of pHGG and DIPG and warrants further investigation for the development of novel therapeutics.
Collapse
Affiliation(s)
- Michaël H Meel
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Sophie A Schaper
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Gertjan J L Kaspers
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Uppsalalaan 8, 3584CT, Utrecht, The Netherlands
| | - Esther Hulleman
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands.
| |
Collapse
|
65
|
A positive feedback loop of IL-17B-IL-17RB activates ERK/β-catenin to promote lung cancer metastasis. Cancer Lett 2018; 422:44-55. [PMID: 29496538 DOI: 10.1016/j.canlet.2018.02.037] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/09/2018] [Accepted: 02/23/2018] [Indexed: 11/20/2022]
Abstract
Inflammation contributes to the development and progression of cancer. Interleukin-17 (IL-17) is an inflammatory cytokine that functions in inflammation and cancer, as well as several other cellular processes. In this study, we investigated the roles and the prognostic value of IL-17 and the IL-17 receptor (IL-17R) in lung cancer. Gene expression microarray analysis followed by Kaplan-Meier survival curve showed that IL-17B was associated with poor patient survival, and IL-17B receptor (IL-17RB) was up-regulated in lung cancer tissue compared with normal tissue. Expression of IL-17RB was associated with lymph node metastasis and distant metastasis, as well as poor patient survival. IL-17RB overexpression significantly increased cancer cell invasion/migration and metastasis in vitro and in vivo. IL-17RB induced ERK phosphorylation, resulting in GSK3β inactivation and leading to β-catenin up-regulation. IL-17RB also participated in IL-17B synthesis via the ERK pathway. IL-17RB activation is required for IL-17B-mediated ERK phosphorylation. Taken together, IL-17B-IL-17RB signaling and ERK participate in a positive feedback loop that enhances invasion/migration ability in lung cancer cell lines. IL-17RB may therefore serve as an independent prognostic factor and a therapeutic target for lung cancer.
Collapse
|
66
|
Shagieva G, Domnina L, Makarevich O, Chernyak B, Skulachev V, Dugina V. Depletion of mitochondrial reactive oxygen species downregulates epithelial-to-mesenchymal transition in cervical cancer cells. Oncotarget 2018; 8:4901-4913. [PMID: 27902484 PMCID: PMC5354879 DOI: 10.18632/oncotarget.13612] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/14/2016] [Indexed: 01/11/2023] Open
Abstract
In the course of cancer progression, epithelial cells often acquire morphological and functional characteristics of mesenchymal cells, a process known as epithelial-to-mesenchymal transition (EMT). EMT provides epithelial cells with migratory, invasive, and stem cell capabilities. Reactive oxygen species produced by mitochondria (mtROS) could be of special importance for pro-tumorigenic signaling and EMT. In our study, we used mitochondria-targeted antioxidant SkQ1 to lower the mtROS level and analyze their role in the regulation of the actin cytoskeleton, adhesion junctions, and signaling pathways critical for tumorigenesis of cervical carcinomas. A decrease in mtROS was found to induce formation of β-cytoplasmic actin stress fibers and circumferential rings in cervical cancer SiHa and Ca-Ski cells. It was accompanied by an upregulation of E-cadherin in SiHa cells and a downregulation of N-cadherin in Ca-Ski cells. In SiHa cells, an increase in E-cadherin expression was accompanied by a reduction of Snail, E-cadherin negative regulator. A stimulation of mtROS by epidermal growth factor (EGF) caused a Snail upregulation in SiHa cells that could be downregulated by SkQ1. SkQ1 caused a decrease in activation of extracellular-signal-regulated kinases 1 and 2 (ERK1/2) in SiHa and Ca-Ski. EGF produced an opposite effect. Incubation with SkQ1 suppressed EGF-induced p-ERK1/2 upregulation in SiHa, but not in Ca-Ski cells. Thus, we showed that scavenging of mtROS by SkQ1 initiated reversal of EMT and suppressed proliferation of cervical cancer cells.
Collapse
Affiliation(s)
- Galina Shagieva
- Department of Mathematical Methods in Biology, Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Lidiya Domnina
- Department of Mathematical Methods in Biology, Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Olga Makarevich
- Faculty of Basic Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Boris Chernyak
- Department of Bioenergetics, Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Vladimir Skulachev
- Department of Bioenergetics, Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Vera Dugina
- Department of Mathematical Methods in Biology, Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
67
|
Irradiated Human Umbilical Vein Endothelial Cells Undergo Endothelial-Mesenchymal Transition via the Snail/miR-199a-5p Axis to Promote the Differentiation of Fibroblasts into Myofibroblasts. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4135806. [PMID: 29619372 PMCID: PMC5830288 DOI: 10.1155/2018/4135806] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/09/2018] [Accepted: 01/17/2018] [Indexed: 01/07/2023]
Abstract
Radiation induced pulmonary fibrosis (RIPF) is one of the major side effects of radiotherapy for lung cancer. Previous studies have shown that endothelial cells and activated myofibroblasts play a key role in RIPF. However, the interaction between irradiated endothelial cells and activation of myofibroblasts has not been reported. The aim of the present study was to examine whether irradiated endothelial cells would affect the differentiation of fibroblasts into myofibroblasts in the process of RIPF. In the current study, we used a coculture system that allowed direct contact between human fetal lung fibroblasts (MRC-5) and irradiated human umbilical vein endothelial cells (HUVECs). After 24 or 48 h, cells were sorted by flow cytometry. Radiation induced endothelial-mesenchymal transition (EndMT) by significantly increasing the expression of Snail and vimentin and reducing the expression of CD31 in HUVECs. In addition, irradiation of HUVECs induced the expression of collagen type I and α-smooth muscle actin (α-SMA) in MRC-5 cells. Further investigation indicated that irradiation of HUVECs induced the differentiation of fibroblasts into myofibroblasts through the Snail/miR-199a-5p axis. We conclude that irradiated endothelial cells undergo EndMT to promote differentiation of fibroblasts into myofibroblasts via the Snail/miR-199a-5p axis.
Collapse
|
68
|
Bai L, Chang HM, Cheng JC, Chu G, Leung PCK, Yang G. Lithium chloride inhibits StAR and progesterone production through GSK-3β and ERK1/2 signaling pathways in human granulosa-lutein cells. Mol Cell Endocrinol 2018; 461:89-99. [PMID: 28867214 DOI: 10.1016/j.mce.2017.08.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 01/20/2023]
Abstract
Lithium chloride (LiCl) is a widely-used medication to treat neurological disorders that has undesirable side effects on the female reproductive system. It has been show that LiCl can inhibit ovarian folliculogenesis, promote follicle atresia and suppress steroid hormone production in rodents. However, the effects of LiCl on human ovarian steroidogenesis remain completely unknown. In this study, both primary and immortalized human granulosa-lutein (hGL) cells were used to investigate the effects of LiCl on progesterone production and its related enzyme expression as well as the underlying mechanisms. Our results showed that LiCl significantly down-regulated the steroidogenic acute regulatory protein (StAR) expression and subsequent progesterone production in hGL cells. Additionally, LiCl induced the phosphorylation of GSK-3β and ERK1/2 but not AKT or CREB. Knockdown of endogenous GSK-3β or inhibition of ERK1/2 partially reversed LiCl-induced down-regulation of StAR. Furthermore, by using dual inhibition approaches, the results showed that both GSK-3β and ERK1/2 signaling mediated the regulatory effect of LiCl on StAR expression. Our findings deepen our understanding of the pathological effects and the underlying molecular mechanisms of how lithium might affect the female reproductive system.
Collapse
Affiliation(s)
- Long Bai
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China; Department of Obstetrics and Gynaecology, University of British Columbia, BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynaecology, University of British Columbia, BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Jung-Chien Cheng
- Department of Obstetrics and Gynaecology, University of British Columbia, BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada
| | - Guiyan Chu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, University of British Columbia, BC Children's Hospital Research Institute, Vancouver, British Columbia V5Z 4H4, Canada.
| | - Gongshe Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
69
|
Peng X, He J, Zhao J, Wu Y, Shi X, Du L, Nong M, Zong S, Zeng G. Polygonatum Sibiricum Polysaccharide Promotes Osteoblastic Differentiation Through the ERK/GSK-3β/β-Catenin Signaling Pathway In Vitro. Rejuvenation Res 2018. [PMID: 28629266 DOI: 10.1089/rej.2017.1956] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Xiaoming Peng
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jichen He
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinmin Zhao
- Department of Osteopathia, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Research Center for Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Yunle Wu
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiongzhi Shi
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li Du
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Mengni Nong
- College of Public Hygiene of Guangxi Medical University, Nanning, China
| | - Shaohui Zong
- Department of Spine Osteopathia, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Research Center for Regenerative Medicine, Guangxi Medical University, Nanning, China
| | - Gaofeng Zeng
- Research Center for Regenerative Medicine, Guangxi Medical University, Nanning, China
- College of Public Hygiene of Guangxi Medical University, Nanning, China
| |
Collapse
|
70
|
Kim RK, Kaushik N, Suh Y, Yoo KC, Cui YH, Kim MJ, Lee HJ, Kim IG, Lee SJ. Radiation driven epithelial-mesenchymal transition is mediated by Notch signaling in breast cancer. Oncotarget 2018; 7:53430-53442. [PMID: 27462787 PMCID: PMC5288197 DOI: 10.18632/oncotarget.10802] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/13/2016] [Indexed: 12/26/2022] Open
Abstract
Epithelial to mesenchymal transition (EMT) is developmental process associated with cancer metastasis. Here, we found that breast carcinoma cells adopt epithelial-to-mesenchymal transition (EMT) in response to fractionated-radiation. Importantly, we show that Notch signaling is highly activated in fractionally-irradiated tumors as compared to non-irradiated tumors that are accompanied by an EMT. Moreover, we uncovered the mechanism of Notch-driven EMT, in which Notch enhanced EMT through IL-6/JAK/STAT3 signaling axis in mammary tumor cells. Collectively, we present converging evidence from our studies that Notch2 is a critical mediator of radiation-induced EMT and responsible for induced malignant tumor growth.
Collapse
Affiliation(s)
- Rae-Kwon Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Korea
| | - Neha Kaushik
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Korea
| | - Yongjoon Suh
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Korea
| | - Ki-Chun Yoo
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Korea
| | - Yan-Hong Cui
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Korea
| | - Min-Jung Kim
- Laboratory of Radiation Exposure and Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Hae-June Lee
- Division of Radiation Effect, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - In-Gyu Kim
- Department of Radiation Biology, Environmental Radiation Research Group, Korea Atomic Energy Research Institute, Daejeon, Korea
| | - Su-Jae Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, Korea
| |
Collapse
|
71
|
Response to stereotactic ablative radiotherapy in a novel orthotopic model of non-small cell lung cancer. Oncotarget 2017; 9:1630-1640. [PMID: 29416719 PMCID: PMC5788587 DOI: 10.18632/oncotarget.22727] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 10/28/2017] [Indexed: 12/12/2022] Open
Abstract
Stereotactic ablative radiotherapy (SABR) is the main treatment for inoperable early-stage non-small cell lung cancer (NSCLC). Despite the widespread use of SABR, the biological determinants of response to SABR remain poorly investigated. We developed an orthotopic NSCLC animal model to study the response to clinically-relevant doses of SABR. Image-guided intra-thoracic injection of NSCLC cells was performed in the right lung of nude rats. A highly conformal dose of 34 Gy was delivered in a single fraction using clinical photon energies. Animals were sacrificed 10–60 days post treatment. Lung tumors were assessed for tumor differentiation, proliferation and invasiveness. An analysis of 770 cancer-related genes was performed on tumor-derived cell lines from treated animals at early and late time points after SABR. The majority of animals receiving SABR demonstrated complete response (67%), while 33% demonstrated local failure. 50% of animals with complete response failed distantly. Analysis of cancer-related genes revealed significant differences between tumors treated with SABR and untreated tumors. SABR significantly modulated expression of genes involved in adhesion, migration and angiogenesis. In particular, interleukin-8 (IL8) which plays a critical role in promoting tumor invasion was found to be secreted at high levels after SABR. In vitro invasion assays confirmed SABR-induced invasion and demonstrated induction of IL-8 secretion in multiple NSCLC cell lines. Our findings underscore the importance of developing targeted therapies that can circumvent the pro-invasive effects of SABR in NSCLC.
Collapse
|
72
|
Lavigne J, Soysouvanh F, Buard V, Tarlet G, Guipaud O, Paget V, Milliat F, François A. Conditional Plasminogen Activator Inhibitor Type 1 Deletion in the Endothelial Compartment Has No Beneficial Effect on Radiation-Induced Whole-Lung Damage in Mice. Int J Radiat Oncol Biol Phys 2017; 99:972-982. [DOI: 10.1016/j.ijrobp.2017.07.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/21/2017] [Accepted: 07/06/2017] [Indexed: 12/13/2022]
|
73
|
Nagarajan D, Wang L, Zhao W, Han X. Trichostatin A inhibits radiation-induced epithelial-to-mesenchymal transition in the alveolar epithelial cells. Oncotarget 2017; 8:101745-101759. [PMID: 29254201 PMCID: PMC5731911 DOI: 10.18632/oncotarget.21664] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 08/29/2017] [Indexed: 12/29/2022] Open
Abstract
Radiation-induced pneumonitis and fibrosis are major complications following thoracic radiotherapy. Epithelial-to-mesenchymal transition (EMT) plays an important role in tissue injury leading to organ fibrosis, including lung. Our previous studies have reported that radiation can induce EMT in the type II alveolar epithelial cells in both in vitro and in vivo. HDAC inhibitors are a new family of anti-cancer agents currently being used in several clinical trials. In addition to their intrinsic anti-tumor properties, HDAC inhibition is also important in other human diseases, including fibrosis and radiation-induced damage. In this study, we evaluated the effect of Trichostatin A (TSA), a HDAC inhibitor, on radiation-induced EMT in type II alveolar epithelial cells (RLE-6TN). Pre-treatment of RLE-6TN cells with TSA inhibited radiation-induced EMT-like morphological alterations including elevated protein level of α-SMA and Snail, reduction of E-cadherin expression, enhanced phosphorylation of GSK3β and ERK1/2, increased generation of ROS. Radiation enhanced the protein level of TGF-β1, which was blocked by N-acetylcysteine, an antioxidant. Treating cells with SB-431542, TGF-β1 type I receptor inhibitor, diminished radiation-induced alterations in the protein levels of p-GSK-3β, Snail-1 and α-SMA, suggesting a regulatory role of TGF-β1 in EMT. Pre-incubation of cells with TSA showed significant decrease in the level of TGF-β1 compared to radiation control. Collectively, these results demonstrate that i] radiation-induced EMT in RLE-6TN cells is mediated by ROS/MEK/ERK and ROS/TGF-β1 signaling pathways and ii] the inhibitory role of TSA in radiation-induced EMT appears to be due, at least in part, to its action of blocking ROS and TGF-β1 signaling.
Collapse
Affiliation(s)
- Devipriya Nagarajan
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,School of Chemical & Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | - Lei Wang
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Weiling Zhao
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC, USA.,Tangshan People's Hospital, Tangshan, Hebei, China
| | - Xiaochen Han
- Tangshan People's Hospital, Tangshan, Hebei, China
| |
Collapse
|
74
|
Nagaraja SS, Krishnamoorthy V, Raviraj R, Paramasivam A, Nagarajan D. Effect of Trichostatin A on radiation induced epithelial-mesenchymal transition in A549 cells. Biochem Biophys Res Commun 2017; 493:1534-1541. [PMID: 28993195 DOI: 10.1016/j.bbrc.2017.10.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 10/05/2017] [Indexed: 12/11/2022]
Abstract
Radiotherapy is used to treat tumors of different origins and nature, but often lead to development of radioresistance and metastasis of cells. Interestingly, radiation induces epithelial-mesenchymal transition (EMT), a process by which epithelial cells undergo mesenchymal phenotype and stimulates tumor progression capability. Our study investigated the effect of Trichostatin A (TSA), a natural derivate isolated from Streptomyces, upon radiation-induced lung EMT and we tried to understand the role of signaling molecules in irradiated lung cancer cells (A549). The cells were categorized into four groups: untreated control, radiation alone (R; 8Gy, X-ray), radiation combined with TSA (R + T) and TSA (100nM). Radiation-induced lung EMT were evidenced by decreased expression of epithelial marker like E-cadherin, Zona occluden1 (ZO-1) and increased expression of N-cadherin and Vimentin. The Snail protein, a master regulator of EMT, was observed to be elevated after radiation treatment. In addition, TGF-β1 signaling (smad2, 3, and 4) proteins were activated upon irradiation. Western blot data were supported by the altered m-RNA expression of E-cadherin, TGF-β and Snail genes and this effect were reversed by TSA treatment. In addition to this, as supportive evidence, we performed docking studies between snail protein and TSA using Auto docking software and results suggested that less binding energy was needed for the putative binding of TSA on C-terminal domain of Snail protein. Based on our report, we suggest that TSA can effectively inhibit radiation-induced EMT (i) by altering epithelial and mesenchymal markers (ii) by modulating signaling molecules of TGFβ1 pathway (iii) by inhibiting cancer cell migratory potential in A549 cells (iv)by effectively binding to Snail which is an enhancer of EMT.
Collapse
Affiliation(s)
| | - Vishnuvarthan Krishnamoorthy
- Radiation Biology Lab, Anusandhan Kendra-II, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India
| | - Raghavi Raviraj
- Radiation Biology Lab, Anusandhan Kendra-II, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India
| | - Alagudinesh Paramasivam
- Radiation Biology Lab, Anusandhan Kendra-II, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India
| | - Devipriya Nagarajan
- Radiation Biology Lab, Anusandhan Kendra-II, School of Chemical and Biotechnology, SASTRA University, Thanjavur, India.
| |
Collapse
|
75
|
Vallée A, Lecarpentier Y, Guillevin R, Vallée JN. Interactions between TGF-β1, canonical WNT/β-catenin pathway and PPAR γ in radiation-induced fibrosis. Oncotarget 2017; 8:90579-90604. [PMID: 29163854 PMCID: PMC5685775 DOI: 10.18632/oncotarget.21234] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/17/2017] [Indexed: 12/16/2022] Open
Abstract
Radiation therapy induces DNA damage and inflammation leading to fibrosis. Fibrosis can occur 4 to 12 months after radiation therapy. This process worsens with time and years. Radiation-induced fibrosis is characterized by fibroblasts proliferation, myofibroblast differentiation, and synthesis of collagen, proteoglycans and extracellular matrix. Myofibroblasts are non-muscle cells that can contract and relax. Myofibroblasts evolve towards irreversible retraction during fibrosis process. In this review, we discussed the interplays between transforming growth factor-β1 (TGF-β1), canonical WNT/β-catenin pathway and peroxisome proliferator-activated receptor gamma (PPAR γ) in regulating the molecular mechanisms underlying the radiation-induced fibrosis, and the potential role of PPAR γ agonists. Overexpression of TGF-β and canonical WNT/β-catenin pathway stimulate fibroblasts accumulation and myofibroblast differentiation whereas PPAR γ expression decreases due to the opposite interplay of canonical WNT/β-catenin pathway. Both TGF-β1 and canonical WNT/β-catenin pathway stimulate each other through the Smad pathway and non-Smad pathways such as phosphatidylinositol 3-kinase/serine/threonine kinase (PI3K/Akt) signaling. WNT/β-catenin pathway and PPAR γ interact in an opposite manner. PPAR γ agonists decrease β-catenin levels through activation of inhibitors of the WNT pathway such as Smad7, glycogen synthase kinase-3 (GSK-3 β) and dickkopf-related protein 1 (DKK1). PPAR γ agonists also stimulate phosphatase and tensin homolog (PTEN) expression, which decreases both TGF-β1 and PI3K/Akt pathways. PPAR γ agonists by activating Smad7 decrease Smads pathway and then TGF-β signaling leading to decrease radiation-induced fibrosis. TGF-β1 and canonical WNT/β-catenin pathway promote radiation-induced fibrosis whereas PPAR γ agonists can prevent radiation-induced fibrosis.
Collapse
Affiliation(s)
- Alexandre Vallée
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of Poitiers, Poitiers, France.,Laboratory of Mathematics and Applications (LMA), UMR CNRS 7348, University of Poitiers, Poitiers, France
| | - Yves Lecarpentier
- Centre de Recherche Clinique, Grand Hôpital de l'Est Francilien (GHEF), Meaux, France
| | - Rémy Guillevin
- DACTIM, UMR CNRS 7348, University of Poitiers et CHU de Poitiers, Poitiers, France
| | - Jean-Noël Vallée
- Laboratory of Mathematics and Applications (LMA), UMR CNRS 7348, University of Poitiers, Poitiers, France.,CHU Amiens Picardie, University of Picardie Jules Verne (UPJV), Amiens, France
| |
Collapse
|
76
|
Lu J, Zhong Y, Lin Z, Lin X, Chen Z, Wu X, Wang N, Zhang H, Huang S, Zhu Y, Wang Y, Lin S. Baicalin alleviates radiation-induced epithelial-mesenchymal transition of primary type II alveolar epithelial cells via TGF-β and ERK/GSK3β signaling pathways. Biomed Pharmacother 2017; 95:1219-1224. [PMID: 28931214 DOI: 10.1016/j.biopha.2017.09.037] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/18/2017] [Accepted: 09/10/2017] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Radiation therapy is commonly used to treat thoracic malignancies. However, it may lead to severe lung pneumonitis and ultimately fibrosis. Irradiation has been reported to increase epithelial-mesenchymal transition (EMT) of type II alveolar epithelial cells (AEC), which play an important role in pulmonary fibrosis. The transforming growth factor-β (TGF-β) and ERK/glycogen synthase kinase 3β (GSK3β) pathways are critically involved in radiation-induced EMT. In the present study, we investigated whether baicalin was a novel therapeutic candidate for radiation-induced EMT in type II AEC. METHODS Primary type II AEC were isolated and treated with 60Co γ-rays and a series doses of baicalin (2μM, 10μM and 50μM). The ultrastructure and morphology changes were observed by transmission electron microscopy and optical microscopy, respectively. Protein expression was determined by western blotting analysis. Immunofluorescence staining was performed to detect the nuclear translocation of Snail. RESULTS After irradiation, type II AEC displayed a mesenchymal-like morphology accompanied by a decrease in E-cadherin expression, an increase in the expression of Vimentin and α-SMA. Nuclear translocation of Snail, the activation of TGF-β/Smad pathway, and the inactivation of GSK3β were prominent in radiation-treated cells. Baicalin significantly attenuated the effects of radiation on type II AEC. CONCLUSIONS Baicalin may a useful radioprotective agent through suppressing the EMT of type II AEC.
Collapse
Affiliation(s)
- Jinhua Lu
- Hangzhou Cancer Hospital, Hangzhou Hospital affiliated to Nanjing Medical University, Hangzhou Cancer Center, Hangzhou, Zhejiang, 310000, PR China
| | - Yazhen Zhong
- Hangzhou Cancer Hospital, Hangzhou Hospital affiliated to Nanjing Medical University, Hangzhou Cancer Center, Hangzhou, Zhejiang, 310000, PR China
| | - Zechen Lin
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310000, PR China
| | - Xianlei Lin
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310000, PR China
| | - Zhaohui Chen
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310000, PR China
| | - Xuping Wu
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310000, PR China
| | - Nan Wang
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310000, PR China
| | - Haiqiao Zhang
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310000, PR China
| | - Siyu Huang
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310000, PR China
| | - Yuan Zhu
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310000, PR China
| | - Yuanyuan Wang
- Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, 310000, PR China
| | - Shengyou Lin
- Hangzhou Cancer Hospital, Hangzhou Hospital affiliated to Nanjing Medical University, Hangzhou Cancer Center, Hangzhou, Zhejiang, 310000, PR China.
| |
Collapse
|
77
|
β-catenin, Twist and Snail: Transcriptional regulation of EMT in smokers and COPD, and relation to airflow obstruction. Sci Rep 2017; 7:10832. [PMID: 28883453 PMCID: PMC5589881 DOI: 10.1038/s41598-017-11375-x] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 08/23/2017] [Indexed: 12/20/2022] Open
Abstract
COPD is characterised by poorly reversible airflow obstruction usually due to cigarette smoking. The transcription factor clusters of β-catenin/Snail1/Twist has been implicated in the process of epithelial mesenchymal transition (EMT), an intermediate between smoking and airway fibrosis, and indeed lung cancer. We have investigated expression of these transcription factors and their "cellular localization" in bronchoscopic airway biopsies from patients with COPD, and in smoking and non-smoking controls. An immune-histochemical study compared cellular protein expression of β-catenin, Snail1 and Twist, in these subject groups in 3 large airways compartment: epithelium (basal region), reticular basement membrane (Rbm) and underlying lamina propria (LP). β-catenin and Snail1 expression was generally high in all subjects throughout the airway wall with marked cytoplasmic to nuclear shift in COPD (P < 0.01). Twist expression was generalised in the epithelium in normal but become more basal and nuclear with smoking (P < 0.05). In addition, β-catenin and Snail1 expression, and to lesser extent of Twist, was related to airflow obstruction and to expression of a canonical EMT biomarker (S100A4). The β-catenin-Snail1-Twist transcription factor cluster is up-regulated and nuclear translocated in smokers and COPD, and their expression is closely related to both EMT activity and airway obstruction.
Collapse
|
78
|
Cai HD, Su SL, Qian DW, Guo S, Tao WW, Cong XD, Tang R, Duan JA. Renal protective effect and action mechanism of Huangkui capsule and its main five flavonoids. JOURNAL OF ETHNOPHARMACOLOGY 2017; 206:152-159. [PMID: 28408246 DOI: 10.1016/j.jep.2017.02.046] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 02/12/2017] [Accepted: 02/20/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The flower of Abelmoschus manihot (Linn.) Medicus (A. manihot), as a traditional Chinese Herbal medicine, was used widely in China with efficacy of inducing diuresis for treating strangurtia, and subdhing swelling and detoxicating. It has been reported that Huangkui capsule, prepared by the extract of the flower of A. manihot, can reduce the content of urinary protein, serum creatinine and serum urea nitrogen in nephropathy rats and processes renoprotective activity, while the action mechanism need to illuminate deeply. AIMS OF THE STUDY In this study, we investigated the protection effect of Huangkui capsule on tubulointerstitial fibrosis in chronic renal failure (CRF) rats and its mechanism against high glucose-induced epithelial to mesenchymal transition (EMT) in renal tubular epithelial cells (HK-2) of its bioactive components. MATERIALS AND METHODS The animals were divided into normal group, CRF model group and Huangkui capsule-treated group. Hematoxylin eosin (HE) staining and Masson staining were applied to observe pathological changes in renal tissue of different groups. Biochemical indicators including serum urea nitrogen (BUN), urine protein (UP) and serum creatinine (Scr) were measured according to the manufacturer's instructions of kits. HK-2 cell damaged model was established to access the protection effect and action mechanism of five main flavonoids from Huangkui capsule. The experimental cells were divided into eight groups: control group, model group, positive drug group and five main flavonoids treated groups. The dichlorodihydrofluorescein diacetate (DCFH-DA) assay was used to determine the reactive oxygen species (ROS) in different groups. Western blot was applied to analyze the expression of pathogenesis-related proteins in different groups. RESULTS The results stated that Huangkui capsule significantly inhibited the elevation of Scr, BUN, UP, the expression of α-smooth muscle actin (α-SMA), phosphorylation-extracellular signal-regulated kinase (p-ERK1/2), NADPH Oxidase 1, NADPH Oxidase 2 and NADPH Oxidase 4 in adenine-induced CRF rats. The main bioactive components of quercetin (QT), hyperoside (HY), isoquercitrin (IQT), gossypetin-8-O-β-D-glucuronide (GG) and quercetin-3'-O-glucoside (QG) at the dosage of 100µM, like NADPH oxidase inhibitor diphenyleneiodonium, exhibited a significant effect on inhibiting the expression of α-SMA, p-ERK1/2, NADPH Oxidase 1, NADPH Oxidase 2 and NADPH Oxidase 4 in high glucose-induced HK-2 cells, especially GG. CONCLUSIONS These results demonstrated that Huangkui capsule and the flavonoids components prevent tubulointerstitial fibrosis in CRF rat involvement in the action mechanism of inhibiting NADPH oxidase/ROS/ERK pathway.
Collapse
Affiliation(s)
- Hong-Die Cai
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Shu-Lan Su
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Da-Wei Qian
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Sheng Guo
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Wei-Wei Tao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; Center for Translational Systems Biology and Neuroscience, Laboratory of Integrative Biomedicine of Brain Diseases, School of Basic Biomedical Science, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Xu Dong Cong
- SZYY Group Pharmaceutical Limited, Jiangyan 225500, China
| | - Renmao Tang
- SZYY Group Pharmaceutical Limited, Jiangyan 225500, China.
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
79
|
Abstract
The endothelial-mesenchymal transition (EndMT) is a fundamental cellular mechanism that occurs under both physiological and pathological conditions and includes the fibrotic stages of numerous organs, namely, the skin, kidneys, heart, lungs and liver. Endothelial cells that undergo EndMT are one of the main source of (myo)fibroblasts in fibrotic tissues. A critical step in cellular transdifferentiation is morphological change, which is engineered by the reorganization of cytoskeletal elements such as microtubules. These dynamic structures consist of αβ-tubulin heterodimers that are also involved in cellular movement and intracellular trafficking, processes modulated during EndMT. One fundamental mechanism that underlies microtubule stabilization is the regulation of the levels of α and β-tubulin. However, little is known about the roles of specific tubulin isotypes in the development of EndMT-based diseases. This study provides the first evidence that the upregulation of TUBB3 and TUBB4 is coupled with increased cell migration in EndMT-induced HMEC-1 cells. Immunochemical analysis reveals that these tubulins are upregulated in the early stages of EndMT, and siRNA analysis indicates that they are engaged in the generation of mesenchymal behavior via the enhancement of cell migration. This modulation seems to be especially important in wound healing. Finally, cell surface analysis reveals that TUBB3 and TUBB4 are necessary for the transport and proper localization of N-cadherin within the plasma membrane. We believe that our results will be valuable for the development of effective new anti-fibrotic therapies.
Collapse
|
80
|
Deng G, Liang N, Xie J, Luo H, Qiao L, Zhang J, Wang D, Zhang J. Pulmonary toxicity generated from radiotherapeutic treatment of thoracic malignancies. Oncol Lett 2017; 14:501-511. [PMID: 28693198 PMCID: PMC5494764 DOI: 10.3892/ol.2017.6268] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 03/14/2017] [Indexed: 02/06/2023] Open
Abstract
Radiation-induced lung injury (RILI) remains a major obstacle for thoracic radiotherapy for the treatment of lung cancer, esophageal cancer and lymphoma. It is the principal dose-limiting complication, and can markedly impair the therapeutic ratio as well as a patient's quality of life. The current review presents the relevant concepts associated with RILI, including the pathogenic mechanisms and the potential treatment strategies, so as to achieve a general understanding of this issue. RILI comprises an acute radiation pneumonitis phase and subsequent late lung fibrosis. The established assessment criteria are clinical manifestations, imaging changes and the necessity for medical assistance. Risk factors are also considered in order to optimize treatment planning. Due to the underlying molecular mechanisms of RILI, the present review also discusses several targeted pharmacological approaches for its treatment, as well as corticosteroid therapy.
Collapse
Affiliation(s)
- Guodong Deng
- Department of Radiation Oncology, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Ning Liang
- Department of Radiation Oncology, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Jian Xie
- Department of Radiation Oncology, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Hui Luo
- Department of Radiation Oncology, Henan Cancer Hospital, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Lili Qiao
- Department of Oncology, The Fifth People's Hospital of Jinan, Jinan, Shandong 250022, P.R. China
| | - Jingxin Zhang
- Division of Oncology, Graduate School, Weifang Medical College, Weifang, Shandong 261053, P.R. China
| | - Dawei Wang
- Department of Radiology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China
| | - Jiandong Zhang
- Department of Radiation Oncology, Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
81
|
Wei L, Zhang J, Yang ZL, You H. Extracellular superoxide dismutase increased the therapeutic potential of human mesenchymal stromal cells in radiation pulmonary fibrosis. Cytotherapy 2017; 19:586-602. [PMID: 28314668 DOI: 10.1016/j.jcyt.2017.02.359] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 01/31/2017] [Accepted: 02/16/2017] [Indexed: 01/22/2023]
Abstract
BACKGROUND AIMS Pulmonary fibrosis induced by irradiation is a significant problem of radiotherapy in cancer patients. Extracellular superoxide dismutase (SOD3) is found to be predominantly and highly expressed in the extracellular matrix of lung and plays a pivotal role against oxidative damage. Early administration of mesenchymal stromal cells (MSCs) has been demonstrated to reduce fibrosis of damaged lung. However, injection of MSCs at a later stage would be involved in fibrosis development. The present study aimed to determine whether injection of human umbilical cord-derived MSCs (UC-MSCs) over-expressing SOD3 at the established fibrosis stage would have beneficial effects in a mice model of radiation pulmonary fibrosis. METHODS Herein, pulmonary fibrosis in mice was induced using Cobalt-60 (60Co) irradiator with 20 Gy, followed by intravenous injection of UC-MSCs, transduced or not to express SOD3 at 2 h (early delivery) and 60 day (late delivery) post-irradiation, respectively. RESULTS Our results demonstrated that the early administration of UC-MSCs could attenuate the microscopic damage, reduce collagen deposition, inhibit (myo)fibroblast proliferation, reduce inflammatory cell infiltration, protect alveolar type II (AE2) cell injury, prevent oxidative stress and increase antioxidant status, and reduce pro-fibrotic cytokine level in serum. Furthermore, the early treatment with SOD3-infected UC-MSCs resulted in better improvement. However, we failed to observe the therapeutic effects of UC-MSCs, transduced to express SOD3, during established fibrosis. CONCLUSION Altogether, our results demonstrated that the early treatment with UC-MSCs alone significantly reduced radiation pulmonary fibrosis in mice through paracrine effects, with further improvement by administration of SOD3-infected UC-MSCs, suggesting that SOD3-infected UC-MSCs may be a potential cell-based gene therapy to treat clinical radiation pulmonary fibrosis.
Collapse
Affiliation(s)
- Li Wei
- Key Laboratory of Birth Defects and Reproductive Health of National Health and Family Planning Commission, Chongqing Population and Family Planning Science and Technology Research Institute, Chongqing, China
| | - Jing Zhang
- Oncology Department, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Zai-Liang Yang
- Department of Breast and Thyroid, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China.
| | - Hua You
- Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China.
| |
Collapse
|
82
|
Lee SY, Jeong EK, Ju MK, Jeon HM, Kim MY, Kim CH, Park HG, Han SI, Kang HS. Induction of metastasis, cancer stem cell phenotype, and oncogenic metabolism in cancer cells by ionizing radiation. Mol Cancer 2017; 16:10. [PMID: 28137309 PMCID: PMC5282724 DOI: 10.1186/s12943-016-0577-4] [Citation(s) in RCA: 385] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 12/25/2016] [Indexed: 12/12/2022] Open
Abstract
Radiation therapy is one of the major tools of cancer treatment, and is widely used for a variety of malignant tumours. Radiotherapy causes DNA damage directly by ionization or indirectly via the generation of reactive oxygen species (ROS), thereby destroying cancer cells. However, ionizing radiation (IR) paradoxically promotes metastasis and invasion of cancer cells by inducing the epithelial-mesenchymal transition (EMT). Metastasis is a major obstacle to successful cancer therapy, and is closely linked to the rates of morbidity and mortality of many cancers. ROS have been shown to play important roles in mediating the biological effects of IR. ROS have been implicated in IR-induced EMT, via activation of several EMT transcription factors—including Snail, HIF-1, ZEB1, and STAT3—that are activated by signalling pathways, including those of TGF-β, Wnt, Hedgehog, Notch, G-CSF, EGFR/PI3K/Akt, and MAPK. Cancer cells that undergo EMT have been shown to acquire stemness and undergo metabolic changes, although these points are debated. IR is known to induce cancer stem cell (CSC) properties, including dedifferentiation and self-renewal, and to promote oncogenic metabolism by activating these EMT-inducing pathways. Much accumulated evidence has shown that metabolic alterations in cancer cells are closely associated with the EMT and CSC phenotypes; specifically, the IR-induced oncogenic metabolism seems to be required for acquisition of the EMT and CSC phenotypes. IR can also elicit various changes in the tumour microenvironment (TME) that may affect invasion and metastasis. EMT, CSC, and oncogenic metabolism are involved in radioresistance; targeting them may improve the efficacy of radiotherapy, preventing tumour recurrence and metastasis. This study focuses on the molecular mechanisms of IR-induced EMT, CSCs, oncogenic metabolism, and alterations in the TME. We discuss how IR-induced EMT/CSC/oncogenic metabolism may promote resistance to radiotherapy; we also review efforts to develop therapeutic approaches to eliminate these IR-induced adverse effects.
Collapse
Affiliation(s)
- Su Yeon Lee
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Pusan, 609-735, Korea
| | - Eui Kyong Jeong
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Pusan, 609-735, Korea
| | - Min Kyung Ju
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Pusan, 609-735, Korea
| | - Hyun Min Jeon
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Pusan, 609-735, Korea
| | - Min Young Kim
- Research Center, Dongnam Institute of Radiological and Medical Science (DIRAMS), Pusan, 619-953, Korea
| | - Cho Hee Kim
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Pusan, 609-735, Korea.,DNA Identification Center, National Forensic Service, Seoul, 158-707, Korea
| | - Hye Gyeong Park
- Nanobiotechnology Center, Pusan National University, Pusan, 609-735, Korea
| | - Song Iy Han
- The Division of Natural Medical Sciences, College of Health Science, Chosun University, Gwangju, 501-759, Korea
| | - Ho Sung Kang
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Pusan, 609-735, Korea.
| |
Collapse
|
83
|
Li F, Zhou K, Gao L, Zhang B, Li W, Yan W, Song X, Yu H, Wang S, Yu N, Jiang Q. Radiation induces the generation of cancer stem cells: A novel mechanism for cancer radioresistance. Oncol Lett 2016; 12:3059-3065. [PMID: 27899964 PMCID: PMC5103903 DOI: 10.3892/ol.2016.5124] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 08/19/2016] [Indexed: 12/13/2022] Open
Abstract
Radioresistance remains a major obstacle for the radiotherapy treatment of cancer. Previous studies have demonstrated that the radioresistance of cancer is due to the existence of intrinsic cancer stem cells (CSCs), which represent a small, but radioresistant cell subpopulation that exist in heterogeneous tumors. By contrast, non-stem cancer cells are considered to be radiosensitive and thus, easy to kill. However, recent studies have revealed that under conditions of radiation-induced stress, theoretically radiosensitive non-stem cancer cells may undergo dedifferentiation subsequently obtaining the phenotypes and functions of CSCs, including high resistance to radiotherapy, which indicates that radiation may directly result in the generation of novel CSCs from non-stem cancer cells. These findings suggest that in addition to intrinsic CSCs, non-stem cancer cells may also contribute to the relapse and metastasis of cancer following transformation into CSCs. This review aims to investigate the radiation-induced generation of CSCs, its association with epithelial-mesenchymal transition and its significance with regard to the radioresistance of cancer.
Collapse
Affiliation(s)
- Fengsheng Li
- Central Laboratories, The Second Artillery General Hospital, Beijing 100088, P.R. China
| | - Kunming Zhou
- Central Laboratories, The Second Artillery General Hospital, Beijing 100088, P.R. China
| | - Ling Gao
- Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, China Center for Disease Control and Prevention, Beijing 100088, P.R. China
| | - Bin Zhang
- Department of Colorectal Disease Surgery, The Second Artillery General Hospital, Beijing 100088, P.R. China
| | - Wei Li
- Central Laboratories, The Second Artillery General Hospital, Beijing 100088, P.R. China
| | - Weijuan Yan
- Central Laboratories, The Second Artillery General Hospital, Beijing 100088, P.R. China
| | - Xiujun Song
- Central Laboratories, The Second Artillery General Hospital, Beijing 100088, P.R. China
| | - Huijie Yu
- Central Laboratories, The Second Artillery General Hospital, Beijing 100088, P.R. China
| | - Sinian Wang
- Central Laboratories, The Second Artillery General Hospital, Beijing 100088, P.R. China
| | - Nan Yu
- Central Laboratories, The Second Artillery General Hospital, Beijing 100088, P.R. China
| | - Qisheng Jiang
- Central Laboratories, The Second Artillery General Hospital, Beijing 100088, P.R. China
| |
Collapse
|
84
|
Kolahian S, Fernandez IE, Eickelberg O, Hartl D. Immune Mechanisms in Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2016; 55:309-22. [DOI: 10.1165/rcmb.2016-0121tr] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
85
|
Peroxisome Proliferator-Activated Receptor-γ Is Critical to Cardiac Fibrosis. PPAR Res 2016; 2016:2198645. [PMID: 27293418 PMCID: PMC4880703 DOI: 10.1155/2016/2198645] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Revised: 04/16/2016] [Accepted: 04/26/2016] [Indexed: 02/06/2023] Open
Abstract
Peroxisome proliferator-activated receptor-γ (PPARγ) is a ligand-activated transcription factor belonging to the nuclear receptor superfamily, which plays a central role in regulating lipid and glucose metabolism. However, accumulating evidence demonstrates that PPARγ agonists have potential to reduce inflammation, influence the balance of immune cells, suppress oxidative stress, and improve endothelial function, which are all involved in the cellular and molecular mechanisms of cardiac fibrosis. Thus, in this review we discuss the role of PPARγ in various cardiovascular conditions associated with cardiac fibrosis, including diabetes mellitus, hypertension, myocardial infarction, heart failure, ischemia/reperfusion injury, atrial fibrillation, and several other cardiovascular disease (CVD) conditions, and summarize the developmental status of PPARγ agonists for the clinical management of CVD.
Collapse
|
86
|
Zhao L, Jiang R, Xu M, Zhu P, Mo XM, Wang N, Chen GG, Liu ZM. Concomitant high expression of BRAFV600E, P-cadherin and cadherin 6 is associated with High TNM stage and lymph node metastasis in conventional papillary thyroid carcinoma. Clin Endocrinol (Oxf) 2016; 84:748-55. [PMID: 26285159 DOI: 10.1111/cen.12878] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 08/10/2015] [Accepted: 08/12/2015] [Indexed: 11/28/2022]
Abstract
CONTEXT AND OBJECTIVE BRAFV600E mutation is the most common activating mutation associated with aggressive behaviours in human tumours including conventional papillary thyroid carcinoma (cPTC). P-cadherin and cadherin 6 have been shown to be mesenchymal-associated cadherins and promote cancer cell invasion and metastasis. The purpose of this study was to examine BRAFV600E, P-cadherin and cadherin 6 expressions in cPTC and to assess the association of their expression with clinicopathological indicators. METHODS BRAFV600E, P-cadherin and cadherin 6 protein expressions in 80 cPTCs, 61 nodular hyperplasia and 76 normal thyroid tissues were examined by immunohistochemistry. The correlation of their protein expression with clinicopathological indicators of cPTC was statistically analysed. RESULTS Protein expression of BRAFV600E, P-cadherin and cadherin 6 was upregulated in cPTC. High protein expression of BRAFV600E, P-cadherin and cadherin 6 was significantly correlated with high TNM stage and lymph node metastasis (LNM) (P < 0·001). Furthermore, BRAFV600E, P-cadherin and cadherin 6 protein expressions were correlated with one another. BRAFV600E high expression combined with both P-cadherin and cadherin-6 high expressions had stronger correlation with high TNM stage and LNM when compared with BRAFV600E high expression combined with either P-cadherin or cadherin-6 high expression (P = 0·042, 0·017 for TNM stage and P = 0·003, 0·006 for LNM, respectively) and only BRAFV600E high expression (P < 0·001 for both TNM stage and LNM). CONCLUSIONS Concomitant high expression of BRAFV600E, P-cadherin and cadherin 6 is strongly associated with high TNM stage and LNM in cPTC.
Collapse
Affiliation(s)
- Le Zhao
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Rong Jiang
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Man Xu
- Department of Pathology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Ping Zhu
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Xiao-Mei Mo
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - Ni Wang
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| | - George G Chen
- Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin N.T., Hong Kong, China
| | - Zhi-Min Liu
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, China
| |
Collapse
|
87
|
Li S, Lu J, Chen Y, Xiong N, Li L, Zhang J, Yang H, Wu C, Zeng H, Liu Y. MCP-1-induced ERK/GSK-3β/Snail signaling facilitates the epithelial-mesenchymal transition and promotes the migration of MCF-7 human breast carcinoma cells. Cell Mol Immunol 2016; 14:621-630. [PMID: 26996066 DOI: 10.1038/cmi.2015.106] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/28/2015] [Accepted: 11/29/2015] [Indexed: 01/08/2023] Open
Abstract
Monocyte chemoattractant protein-1 (MCP-1) is a chemotactic cytokine that can bind to its receptor cysteine-cysteine chemokine receptor 2 (CCR2) and plays an important role in breast cancer cell metastasis. However, the molecular mechanisms underlying MCP-1-induced alterations in cellular functions during tumor progression are poorly understood. Here, we showed that MCP-1 stimulated the epithelial-mesenchymal transition (EMT) and induced the tumorigenesis of breast cancer cells by downregulating E-cadherin, upregulating vimentin and fibronectin, activating matrix metallopeptidase-2 (MMP-2), and promoting migration and invasion. Moreover, MCP-1 treatment reduced glycogen synthase kinase-3β (GSK-3β) activity via the MEK/ERK-mediated phosphorylation of serine-9 in MCF-7 cells. The inhibition of MEK/ERK by U0126 attenuated the MCP-1-induced phosphorylation of GSK-3β and decreased the expression of Snail, an EMT-related transcription factor, leading to the inhibition of MCF-7 cell migration and invasion. Inactivation of GSK-3β by LiCl (lithium chloride) treatment notably increased MMP-2 activity, vascular endothelial growth factor expression and EMT of MCF-7 cells. These findings revealed that MCP-1-induced EMT and migration are mediated by the ERK/GSK-3β/Snail pathway, and identified a potential novel target for therapeutic intervention in breast cancer.
Collapse
Affiliation(s)
- Shun Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Juan Lu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Yu Chen
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Niya Xiong
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Li Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Jing Zhang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Hong Yang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China.,Center for Information in Biomedicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Chunhui Wu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China.,Center for Information in Biomedicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Hongjuan Zeng
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China.,Center for Information in Biomedicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| | - Yiyao Liu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China.,Center for Information in Biomedicine, University of Electronic Science and Technology of China, Chengdu 610054, Sichuan, China
| |
Collapse
|
88
|
Díaz VM, de Herreros AG. F-box proteins: Keeping the epithelial-to-mesenchymal transition (EMT) in check. Semin Cancer Biol 2016; 36:71-9. [DOI: 10.1016/j.semcancer.2015.10.003] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 10/01/2015] [Accepted: 10/17/2015] [Indexed: 12/22/2022]
|
89
|
Focal exposure of limited lung volumes to high-dose irradiation down-regulated organ development-related functions and up-regulated the immune response in mouse pulmonary tissues. BMC Genet 2016; 17:29. [PMID: 26818610 PMCID: PMC4729165 DOI: 10.1186/s12863-016-0338-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 01/20/2016] [Indexed: 11/25/2022] Open
Abstract
Background Despite the emergence of stereotactic body radiotherapy (SBRT) for treatment of medically inoperable early-stage non-small-cell lung cancer patients, the molecular effects of focal exposure of limited lung volumes to high-dose radiation have not been fully characterized. This study was designed to identify molecular changes induced by focal high-dose irradiation using a mouse model of SBRT. Results Central areas of the mouse left lung were focally-irradiated (3 mm in diameter) with a single high-dose of radiation (90 Gy). Temporal changes in gene expression in the irradiated and non-irradiated neighboring lung regions were analyzed by microarray. For comparison, the long-term effect (12 months) of 20 Gy radiation on a diffuse region of lung was also measured. The majority of genes were down-regulated in the focally-irradiated lung areas at 2 to 3 weeks after irradiation. This pattern of gene expression was clearly different than gene expression in the diffuse region of lungs exposed to low-dose radiation. Ontological and pathway analyses indicated these down-regulated genes were mainly associated with organ development. Although the number was small, genes that were up-regulated after focal irradiation were associated with immune-related functions. The temporal patterns of gene expression and the associated biological functions were also similar in non-irradiated neighboring lung regions, although statistical significance was greatly reduced when compared with those from focally-irradiated areas of the lung. From network analysis of temporally regulated genes, we identified inter-related modules associated with diverse functions, including organ development and the immune response, in both the focally-irradiated regions and non-irradiated neighboring lung regions. Conclusions Focal exposure of lung tissue to high-dose radiation induced expression of genes associated with organ development and the immune response. This pattern of gene expression was also observed in non-irradiated neighboring areas of lung tissue, indicating a global lung response to focal high-dose irradiation. Electronic supplementary material The online version of this article (doi:10.1186/s12863-016-0338-9) contains supplementary material, which is available to authorized users.
Collapse
|
90
|
Ngo HKC, Lee HG, Piao JY, Zhong X, Lee HN, Han HJ, Kim W, Kim DH, Cha YN, Na HK, Surh YJ. Helicobacter pyloriinduces Snail expression through ROS-mediated activation of Erk and inactivation of GSK-3β in human gastric cancer cells. Mol Carcinog 2016; 55:2236-2246. [DOI: 10.1002/mc.22464] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 01/03/2016] [Accepted: 01/08/2016] [Indexed: 01/03/2023]
Affiliation(s)
- Hoang-Kieu-Chi Ngo
- Tumor Microenvironment Global Core Research Center; College of Pharmacy; Seoul National University; Seoul South Korea
| | - Hee Geum Lee
- Tumor Microenvironment Global Core Research Center; College of Pharmacy; Seoul National University; Seoul South Korea
| | - Juan-Yu Piao
- Tumor Microenvironment Global Core Research Center; College of Pharmacy; Seoul National University; Seoul South Korea
| | - Xiancai Zhong
- Tumor Microenvironment Global Core Research Center; College of Pharmacy; Seoul National University; Seoul South Korea
| | - Ha-Na Lee
- Tumor Microenvironment Global Core Research Center; College of Pharmacy; Seoul National University; Seoul South Korea
| | - Hyeong-Jun Han
- Tumor Microenvironment Global Core Research Center; College of Pharmacy; Seoul National University; Seoul South Korea
| | - Wonki Kim
- Tumor Microenvironment Global Core Research Center; College of Pharmacy; Seoul National University; Seoul South Korea
| | - Do-Hee Kim
- Tumor Microenvironment Global Core Research Center; College of Pharmacy; Seoul National University; Seoul South Korea
| | - Young-Nam Cha
- Department of Pharmacology; College of Medicine; Inha University; Incheon South Korea
| | - Hye-Kyung Na
- Department of Food and Nutrition; Sungshin Women's University; Seoul South Korea
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Center; College of Pharmacy; Seoul National University; Seoul South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences; Graduate School of Convergence Science and Technology; Seoul National University; Seoul South Korea
- Cancer Research Institute; Seoul National University; Seoul South Korea
| |
Collapse
|
91
|
Wei L, Yao Y, Zhao K, Huang Y, Zhou Y, Zhao L, Guo Q, Lu N. Oroxylin A inhibits invasion and migration through suppressing ERK/GSK-3β signaling in snail-expressing non-small-cell lung cancer cells. Mol Carcinog 2016; 55:2121-2134. [PMID: 26741501 DOI: 10.1002/mc.22456] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 11/30/2015] [Accepted: 12/15/2015] [Indexed: 12/17/2022]
Abstract
Snail is closely linked to tumor invasion, metastasis, and recurrence and indicates prognosis of patients suffering from cancer. Overexpression of Snail increases motility and invasiveness of cancer cells, which has become target for anti-metastatic treatment. Oroxylin A, a natural compound extracted from Scutellaria radix, has been reported to inhibit invasion and migration in breast cancer. In this study, we investigated the anti-invasive effect of oroxylin A on lung cells and uncovered its underlying mechanism. The results suggested that oroxylin A could inhibit migration and invasion in Snail-expressing 95-D, and A549 cells whereas it had little effect on non-expressing GLC-82 cells. Furthermore, enhanced Snail expression after transfection of Snail vector in GLC-82 cells is decreased by oroxylin A. Snail can also induce epithelial-mesenchymal transition. We found oroxylin A could reverse TGFβ1-induced epithelial-mesenchymal transition by inhibiting Snail expression. As a result, oroxylin A up-regulated E-cadherin expression and down-regulated vimentin, MMP-9, and CD44v6 expression, which could lead to the inhibition of tumor migration and invasion. Mechanically, we demonstrated that oroxylin A suppressed activation of ERK instead of AKT pathway and then promoted activation of GSK-3β to reduce Snail protein content. Finally, we established transplanted, metastatic, and orthotopic models of A549 cells, and found that oroxylin A inhibited the growth and lung metastasis of A549 cells in vivo. Taken together, we proposed that oroxylin A might be a promising candidate targeting tumor metastasis. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Libin Wei
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, P.R. China
| | - Yuyuan Yao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, P.R. China
| | - Kai Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, P.R. China
| | - Yujie Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, P.R. China
| | - Yuxin Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, P.R. China
| | - Li Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, P.R. China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, P.R. China
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, P.R. China
| |
Collapse
|
92
|
Yuan CT, Li XX, Cheng QJ, Wang YH, Wang JH, Liu CL. MiR-30a regulates the atrial fibrillation-induced myocardial fibrosis by targeting snail 1. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:15527-15536. [PMID: 26884822 PMCID: PMC4730035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 11/28/2015] [Indexed: 06/05/2023]
Abstract
OBJECTIVE Our study aims at assessing the association between miR-30a along with its target gene snail 1 and atrial fibrillation (AF)-induced myocardial fibrosis. METHODS Ang II was used to up-regulate cardiac fibroblasts fibrosis in vitro, and then the cardiac fibroblasts were divided into the mimics group (mimics miR-30a), inhibitors group (inhibitors miR-30a), NC group (transfected miR-30a, negative control) and blank control group (non-transfected cells). Two-group (sham operated group and rapid pacing group) AF rabbit models were constructed according to whether rapid pacing was presented in the subject. Then the establishment of rabbit models was examined using histopathology after Masson staining. The mRNA and protein expression levels of snail 1 and periostin in cardiac fibroblasts and myocardial tissues were detected using the method of RT-PCR and Western blot, respectively. RESULTS In vitro, our experiment showed that overexpression of miR-30a in cardiac fibroblasts contribute to a significant decrease in the average expression level of snail 1 and periostin (P < 0.05) whereas inhibition of miR-30a significantly increased the average expression level of snail 1 and periostin (P < 0.05). In vivo, the average expression level of miR-30a significantly decreased in myocardial tissues with an increased degree of myocardial fibrosis, while the snail 1 and periostin expression level significantly increased during a certain period of time (P < 0.05). CONCLUSION Our results suggest that miR-30a target snail 1 protein may be related to AF-induced myocardial fibrosis. The average expression levels of snail 1 increased significantly in both myocardial cells and tissues, while miR-30a could inhibit the expression of snail 1. Thus, we speculate that miR-30a and snail 1 may be potential therapeutic targets for curing AF-induced myocardial fibrosis.
Collapse
Affiliation(s)
- Chuan-Tao Yuan
- Department of Pathology, Affiiated Hospital of Jining Medical UniversityShandong, China
| | - Xiao-Xia Li
- Department of Pathology, Affiiated Hospital of Jining Medical UniversityShandong, China
| | - Qian-Jin Cheng
- Department of Cardiovascular, Affiiated Hospital of Jining Medical UniversityShandong, China
| | - Yan-Hui Wang
- Department of CT, The Affiiated Hospital of Jining Medical UniversityJining, Shandong Province, China
| | - Jie-Huan Wang
- Department of Cardiovascular, Affiiated Hospital of Jining Medical UniversityShandong, China
| | - Chao-Liang Liu
- Department of Cardiovascular, Affiiated Hospital of Jining Medical UniversityShandong, China
| |
Collapse
|
93
|
Long X, Ye Y, Zhang L, Liu P, Yu W, Wei F, Ren X, Yu J. IL-8, a novel messenger to cross-link inflammation and tumor EMT via autocrine and paracrine pathways (Review). Int J Oncol 2015; 48:5-12. [PMID: 26548401 DOI: 10.3892/ijo.2015.3234] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 10/15/2015] [Indexed: 02/06/2023] Open
Abstract
The epithelial-mesenchymal transition (EMT) is a process through which epithelial cells trans-differentiate and acquire an aggressive mesenchymal phenotype. In tumor cells, EMT is a vital step of tumor progression and metastasis. Amid the increasing interest in tumor EMT, only a few studies focused on the soluble mediators secreted by tumor cells passing through this phenotypic switch. In this review, we focus on the essential role of interleukin-8 (IL-8) signaling for the acquisition and maintenance of tumor EMT via direct and indirect mechanisms. Besides the autocrine loop between IL-8 and tumor cells that have gone through EMT, IL-8 could potentiate adjacent epithelial tumor cells into a mesenchymal phenotype via a paracrine mode. Moreover, understanding the role of IL-8 in EMT will provide insight into the pathogenesis of tumor progression and may facilitate the development of an effective strategy for the prevention and treatment of metastatic cancer.
Collapse
Affiliation(s)
- Xinxin Long
- Department of Immunology, Key Laboratory of Cancer Immunology and Biotherapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Yingnan Ye
- Biotherapy Center, Key Laboratory of Cancer Immunology and Biotherapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Lijie Zhang
- Department of Immunology, Key Laboratory of Cancer Immunology and Biotherapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Pengpeng Liu
- Biotherapy Center, Key Laboratory of Cancer Immunology and Biotherapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Wenwen Yu
- Department of Immunology, Key Laboratory of Cancer Immunology and Biotherapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Feng Wei
- Department of Immunology, Key Laboratory of Cancer Immunology and Biotherapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Xiubao Ren
- Department of Immunology, Key Laboratory of Cancer Immunology and Biotherapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| | - Jinpu Yu
- Department of Immunology, Key Laboratory of Cancer Immunology and Biotherapy, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, P.R. China
| |
Collapse
|
94
|
Yue H, Hu K, Liu W, Jiang J, Chen Y, Wang R. Role of matrix metalloproteinases in radiation-induced lung injury in alveolar epithelial cells of Bama minipigs. Exp Ther Med 2015; 10:1437-1444. [PMID: 26622503 DOI: 10.3892/etm.2015.2658] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 07/07/2015] [Indexed: 02/06/2023] Open
Abstract
Radiation-induced lung injury (RILI) is a common complication associated with thoracic radiotherapy. The aim of the present study was to investigate the effects of a single 15-Gy dose of right-thoracic lung irradiation on the expression levels of matrix metalloproteinases (MMPs) and other proteins in the alveolar epithelial type II (AE2) cells of Bama minipigs. All minipigs received either right-thoracic irradiation or sham irradiation under anesthesia, and were sacrificed at 4, 8, 12 or 24 weeks after irradiation. Collagen deposition was measured using Massons trichrome staining. Surfactant protein A (SP-A), transforming growth factor β1 (TGFβ1), MMP2, MMP9, vimentin and E-cadherin protein expression levels were evaluated using western blot analysis, and the MMP2 and MMP9 gelatinase activities were tested using gelatin zymography. SP-A and α-smooth muscle actin (α-SMA) co-localization was visualized using double immunofluorescence staining. At each time-point following irradiation, a significant increase in TGFβ1, α-SMA, MMP2, MMP9 and vimentin protein expression levels and MMP2 and MMP9 gelatinase activity were observed in the irradiated lungs compared with the sham-irradiated controls. By contrast, SP-A and E-cadherin protein expression levels decreased in a time-dependent manner post-irradiation. SP-A and α-SMA co-localization was observed in irradiated alveolar epithelial cells. These data demonstrate that E-cadherin, SP-A, MMP2 and MMP9 may function as sensitive predictors of RILI. Epithelial-mesenchymal transition (EMT) occurs in the irradiated lungs of Bama minipigs, and MMP2 and MMP9 may contribute to EMT in AE2 cells by regulating TGFβ1. Therefore, EMT may serve a crucial function in the development of RILI.
Collapse
Affiliation(s)
- Haiying Yue
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Kai Hu
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Wenqi Liu
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Jie Jiang
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yuhua Chen
- Department of Pathology, Guangxi Province Maternity and Child Care Hospital, Nanning, Guangxi 530002, P.R. China
| | - Rensheng Wang
- Department of Radiotherapy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
95
|
Wang YY, Zhang CY, Ma YQ, He ZX, Zhe H, Zhou SF. Therapeutic effects of C-28 methyl ester of 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid (CDDO-Me; bardoxolone methyl) on radiation-induced lung inflammation and fibrosis in mice. Drug Des Devel Ther 2015; 9:3163-78. [PMID: 26124639 PMCID: PMC4482372 DOI: 10.2147/dddt.s80958] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The C-28 methyl ester of 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid (CDDO-Me), one of the synthetic triterpenoids, has been found to have potent anti-inflammatory and anticancer properties in vitro and in vivo. However, its usefulness in mitigating radiation-induced lung injury (RILI), including radiation-induced lung inflammation and fibrosis, has not been tested. The aim of this study was to explore the therapeutic effect of CDDO-Me on RILI in mice and the underlying mechanisms. Herein, we found that administration of CDDO-Me improved the histopathological score, reduced the number of inflammatory cells and concentrations of total protein in bronchoalveolar lavage fluid, suppressed secretion and expression of proinflammatory cytokines, including transforming growth factor-β and interleukin-6, elevated expression of the anti-inflammatory cytokine interleukin-10, and downregulated the mRNA level of profibrotic genes, including for fibronectin, α-smooth muscle actin, and collagen I. CDDO-Me attenuated radiation-induced lung inflammation. CDDO-Me also decreased the Masson's trichrome stain score, hydroxyproline content, and mRNA level of profibrotic genes, and blocked radiation-induced collagen accumulation and fibrosis. Collectively, these findings suggest that CDDO-Me ameliorates radiation-induced lung inflammation and fibrosis, and this synthetic triterpenoid is a promising novel therapeutic agent for RILI. Further mechanistic, efficacy, and safety studies are warranted to elucidate the role of CDDO-Me in the management of RILI.
Collapse
Affiliation(s)
- Yan-Yang Wang
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Guiyang, People’s Republic of China
| | - Cui-Ying Zhang
- Graduate School, Ningxia Medical University, Guiyang, People’s Republic of China
| | - Ya-Qiong Ma
- Department of Pathology, General Hospital of Ningxia Medical University, Guiyang, People’s Republic of China
| | - Zhi-Xu He
- Guizhou Provincial Key Laboratory for Regenerative Medicine, Stem Cell and Tissue Engineering Research Center and Sino-US Joint Laboratory for Medical Sciences, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Hong Zhe
- Department of Radiation Oncology, General Hospital of Ningxia Medical University, Guiyang, People’s Republic of China
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, FL, USA
| |
Collapse
|
96
|
He E, Pan F, Li G, Li J. Fractionated Ionizing Radiation Promotes Epithelial-Mesenchymal Transition in Human Esophageal Cancer Cells through PTEN Deficiency-Mediated Akt Activation. PLoS One 2015; 10:e0126149. [PMID: 26000878 PMCID: PMC4441389 DOI: 10.1371/journal.pone.0126149] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 03/29/2015] [Indexed: 01/10/2023] Open
Abstract
In some esophageal cancer patients, radiotherapy may not prevent distant metastasis thus resulting in poor survival. The underlying mechanism of metastasis in these patients is not well established. In this study, we have demonstrated that ionizing radiation may induce epithelial-mesenchymal transition (EMT) accompanied with increased cell migration and invasion, through downregulation of phosphatase and tensin homolog (PTEN), and activation of Akt/GSK-3β/Snail signaling. We developed a radioresistant (RR) esophageal squamous cancer cell line, KYSE-150/RR, by fractionated ionizing radiation (IR) treatment, and confirmed its radioresistance using a clonogenic survival assay. We found that the KYSE-150/RR cell line displayed typical morphological and molecular characteristics of EMT. In comparison to the parental cells, KYSE-150/RR cells showed an increase in post-IR colony survival, migration, and invasiveness. Furthermore, a decrease in PTEN in KYSE-150/RR cells was observed. We postulated that over-expression of PTEN may induce mesenchymal-epithelial transition in KYSE-150/RR cells and restore IR-induced increase of cell migration. Mechanistically, fractionated IR inhibits expression of PTEN, which leads to activation of Akt/GSK-3β signaling and is associated with the elevated levels of Snail protein, a transcription factor involved in EMT. Correspondingly, treatment with LY294002, a phosphatidylinositol-3-kinase inhibitor, mimicked PTEN overexpression effect in KYSE-150/RR cells, further suggesting a role for the Akt/GSK-3β/Snail signaling in effects mediated through PTEN. Together, these results strongly suggest that fractionated IR-mediated EMT in KYSE-150/RR cells is through PTEN-dependent pathways, highlighting a direct proinvasive effect of radiation treatment on tumor cells.
Collapse
Affiliation(s)
- Enhui He
- Nankai University School of Medicine, Tianjin, China
- Chinese PLA General Hospital and Chinese PLA Medical School, Beijing, China
- Beijing Friendship Hospital, affiliated with Capital Medical University, Beijing, China
| | - Fei Pan
- Nankai University School of Medicine, Tianjin, China
| | - Guangchao Li
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Jingjing Li
- Chinese PLA General Hospital and Chinese PLA Medical School, Beijing, China
- Beijing Friendship Hospital, affiliated with Capital Medical University, Beijing, China
- * E-mail:
| |
Collapse
|
97
|
Kuo YC, Wu HT, Hung JJ, Chou TY, Teng SC, Wu KJ. Nijmegen breakage syndrome protein 1 (NBS1) modulates hypoxia inducible factor-1α (HIF-1α) stability and promotes in vitro migration and invasion under ionizing radiation. Int J Biochem Cell Biol 2015; 64:229-38. [PMID: 25959252 DOI: 10.1016/j.biocel.2015.04.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 04/06/2015] [Accepted: 04/28/2015] [Indexed: 10/23/2022]
Abstract
Hypoxia-inducible factor (HIF) is a heterodimer transcription factor complex that monitors the cellular response to the oxygen levels in cells. Hypoxia-inducible factor-1α (HIF-1α) has been shown to be stabilized by ionizing radiation (IR) and its stabilization promotes tumor progression and metastasis. Nijmegen breakage syndrome protein 1 (NBS1), a component of the MRE11-RAD50-NBS1 complex, plays an important role in the cellular response to DNA damage but its overexpression contributes to transformation and has been found to correlate with metastasis. However, whether NBS1 participates in IR-induced metastasis needs to be further determined. The aim of this study is to investigate whether radiation-induced HIF-1α stabilization is regulated by NBS1 and thereby promotes tumor cell migration/invasion. Here, we show that both NBS1 and HIF-1α expression are up-regulated after exposure to IR, and NBS1 increases HIF-1α expression at the protein level. In addition, IR treatment promotes the epithelial-mesenchymal transition (EMT) and in vitro cell migration and invasion activity, which could be abolished by suppression of NBS1. Furthermore, NBS1 directly interacts with HIF-1α and reduces the ubiquitination of HIF-1α⋅ Co-expression of HIF-1α and NBS1 in primary tumors of patients with lung adenocarcinoma correlates with a worse prognosis. These results provide a new function of NBS1 in stabilizing HIF-1α under IR, which leads to enhanced cancer cell migration and invasion.
Collapse
Affiliation(s)
- Yi-Chih Kuo
- Institute of Biochemistry & Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan
| | - Han-Tsang Wu
- Research Center for Tumor Medical Science, Graduate Institute of Cancer Biology, China Medical University, Taichung 404, Taiwan
| | - Jung-Jyh Hung
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan; Division of Thoracic Surgery, Dept. of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Teh-Ying Chou
- Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan; Dept of Pathology, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Shu-Chun Teng
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Kou-Juey Wu
- Institute of Biochemistry & Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan; Research Center for Tumor Medical Science, Graduate Institute of Cancer Biology, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
98
|
Dong LH, Jiang YY, Liu YJ, Cui S, Xia CC, Qu C, Jiang X, Qu YQ, Chang PY, Liu F. The anti-fibrotic effects of mesenchymal stem cells on irradiated lungs via stimulating endogenous secretion of HGF and PGE2. Sci Rep 2015; 5:8713. [PMID: 25736907 PMCID: PMC4348621 DOI: 10.1038/srep08713] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 02/02/2015] [Indexed: 12/11/2022] Open
Abstract
Radiation-induced pulmonary fibrosis is a common disease and has a poor prognosis owing to the progressive breakdown of gas exchange regions in the lung. Recently, a novel strategy of administering mesenchymal stem cells for pulmonary fibrosis has achieved high therapeutic efficacy. In the present study, we attempted to use human adipose tissue-derived mesenchymal stem cells to prevent disease in Sprague-Dawley rats that received semi-thoracic irradiation (15 Gy). To investigate the specific roles of mesenchymal stem cells in ameliorating radiation-induced pulmonary fibrosis, we treated control groups of irradiated rats with human skin fibroblasts or phosphate-buffered saline. After mesenchymal stem cells were infused, host secretions of hepatocyte growth factor (HGF) and prostaglandin E2 (PGE2) were elevated compared with those of the controls. In contrast, tumour necrosis factor-alpha (TNF-α) and transforming growth factor-beta1 (TGF-β1) levels were decreased after infusion of mesenchymal stem cells. Consequently, the architecture of the irradiated lungs was preserved without marked activation of fibroblasts or collagen deposition within the injured sites. Moreover, mesenchymal stem cells were able to prevent the irradiated type II alveolar epithelial cells from undergoing epithelial-mesenchymal transition. Collectively, these data confirmed that mesenchymal stem cells have the potential to limit pulmonary fibrosis after exposure to ionising irradiation.
Collapse
Affiliation(s)
- Li-Hua Dong
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130000, China
| | - Yi-Yao Jiang
- Department of Cardiac Surgery, TEDA International Cardiovascular Hospital, Tianjin 300000, China
| | - Yong-Jun Liu
- Tianjin Allian Stem Cell Techonology CO., LTD, Tianjin 300308, China
| | - Shuang Cui
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130000, China
| | - Cheng-Cheng Xia
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130000, China
| | - Chao Qu
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130000, China
| | - Xin Jiang
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130000, China
| | - Ya-Qin Qu
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130000, China
| | - Peng-Yu Chang
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun 130000, China
| | - Feng Liu
- Nephrology department, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| |
Collapse
|
99
|
He T, Guan X, Wang S, Xiao T, Yang K, Xu X, Wang J, Zhao J. Resveratrol prevents high glucose-induced epithelial-mesenchymal transition in renal tubular epithelial cells by inhibiting NADPH oxidase/ROS/ERK pathway. Mol Cell Endocrinol 2015; 402:13-20. [PMID: 25540919 DOI: 10.1016/j.mce.2014.12.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 12/11/2014] [Accepted: 12/11/2014] [Indexed: 11/28/2022]
Abstract
Resveratrol (RSV) is reported to have renoprotective activity against diabetic nephropathy, while the mechanisms underlying its function have not been fully elucidated. In this study, we investigate the effect and related mechanism of RSV against high glucose-induced epithelial to mesenchymal transition (EMT) in human tubular epithelial cells (HK-2). A typical EMT is induced by high glucose in HK-2 cells, accompanied by increased levels of reactive oxygen species (ROS). RSV exhibits a strong ability to inhibit high glucose-induced EMT by decreasing intracellular ROS levels via down-regulation of NADPH oxidase subunits NOX1 and NOX4. The activation of extracellular signal-regulated kinase (ERK1/2) is found to be involved in high glucose-induced EMT in HK-2 cells. RSV, like NADPH oxidase inhibitor diphenyleneiodonium, can block ERK1/2 activation induced by high glucose. Our results demonstrate that RSV is a potent agent against high glucose-induced EMT in renal tubular cells via inhibition of NADPH oxidase/ROS/ERK1/2 pathway.
Collapse
Affiliation(s)
- Ting He
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Xu Guan
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Song Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Third Military Medical University, Chongqing 400038, China
| | - Tangli Xiao
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Ke Yang
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Xinli Xu
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Junping Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury of PLA, Third Military Medical University, Chongqing 400038, China
| | - Jinghong Zhao
- Department of Nephrology, Institute of Nephrology of Chongqing and Kidney Center of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| |
Collapse
|
100
|
CXCR7 signaling induced epithelial-mesenchymal transition by AKT and ERK pathways in epithelial ovarian carcinomas. Tumour Biol 2014; 36:1679-83. [PMID: 25359618 DOI: 10.1007/s13277-014-2768-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 10/22/2014] [Indexed: 10/24/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) plays an important role in oncogenesis, through which cancer cells acquire an invasion and metastasis capacity. Notably, the chemokine receptor CXCR7 and its ligands CCL19 can also facilitate lymph node metastasis in epithelial ovarian carcinomas. Here, we assumed that CXCR7 might be involved in the EMT process of epithelial ovarian carcinomas. In our study, CXCR7 activation and inhibition in SKOV3 were induced with exogenous CCL19 and CXCR7 small interfering RNA (CXCR7 siRNA), respectively. AKT and ERK protein of CXCR7 pathways as well as biomarkers (vimentin, snail, N-cadherin, and E-cadherin) of EMT were detected using the Western blot. Our results showed that CCL19 can induce AKT and ERK phosphorylation in a dose-dependent fashion; however, CXCR7 siRNA efficaciously suppressed CCL19-induced AKT and ERK phosphorylation in comparison with control siRNA. Importantly, CCL19 alone treatment can upregulate the expression of vimentin, snail, and N-cadherin of SKOV3 and downregulate the expression of E-cadherin. Conversely, knockdown of CXCR7 did not reveal any changes compared with CCL19 and the control. In conclusion, these findings demonstrate that EMT can be regulated by the CCL19/CXCR7 axis in epithelial ovarian carcinomas and then involved in the tumor cell invasion and metastasis process via activation of AKT and ERK pathways. Our study lays a new foundation for the treatment of epithelial ovarian carcinomas through antagonizing CXCR7.
Collapse
|