1
|
Davis PJ, Mousa SA, Lin HY. Nongenomic Actions of Thyroid Hormone: The Integrin Component. Physiol Rev 2020; 101:319-352. [PMID: 32584192 DOI: 10.1152/physrev.00038.2019] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The extracellular domain of plasma membrane integrin αvβ3 contains a cell surface receptor for thyroid hormone analogues. The receptor is largely expressed and activated in tumor cells and rapidly dividing endothelial cells. The principal ligand for this receptor is l-thyroxine (T4), usually regarded only as a prohormone for 3,5,3'-triiodo-l-thyronine (T3), the hormone analogue that expresses thyroid hormone in the cell nucleus via nuclear receptors that are unrelated structurally to integrin αvβ3. At the integrin receptor for thyroid hormone, T4 regulates cancer and endothelial cell division, tumor cell defense pathways (such as anti-apoptosis), and angiogenesis and supports metastasis, radioresistance, and chemoresistance. The molecular mechanisms involve signal transduction via mitogen-activated protein kinase and phosphatidylinositol 3-kinase, differential expression of multiple genes related to the listed cell processes, and regulation of activities of other cell surface proteins, such as vascular growth factor receptors. Tetraiodothyroacetic acid (tetrac) is derived from T4 and competes with binding of T4 to the integrin. In the absence of T4, tetrac and chemically modified tetrac also have anticancer effects that culminate in altered gene transcription. Tumor xenografts are arrested by unmodified and chemically modified tetrac. The receptor requires further characterization in terms of contributions to nonmalignant cells, such as platelets and phagocytes. The integrin αvβ3 receptor for thyroid hormone offers a large panel of cellular actions that are relevant to cancer biology and that may be regulated by tetrac derivatives.
Collapse
Affiliation(s)
- Paul J Davis
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, New York; Department of Medicine, Albany Medical College, Albany, New York; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan; and Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, New York; Department of Medicine, Albany Medical College, Albany, New York; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan; and Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| | - Hung-Yun Lin
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, New York; Department of Medicine, Albany Medical College, Albany, New York; Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan; Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan; and Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
2
|
Yang B, Pan CS, Li Q, Yang Z, Long FX, Fan JY, Wang CS, Han JY, Tang DX. Inhibitory effects of Chanling Gao on the proliferation and liver metastasis of transplanted colorectal cancer in nude mice. PLoS One 2019; 14:e0201504. [PMID: 30789971 PMCID: PMC6383928 DOI: 10.1371/journal.pone.0201504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 01/31/2019] [Indexed: 12/20/2022] Open
Abstract
This study aimed to explore the efficacy and mechanism of Chanling Gao (CLG), a compound Chinese medicine, on colorectal cancer (CRC). A model of transplanted CRC was established in nude mice. The mice were treated 7 days after CRC transplantation with either Capecitabine or CLG for 3 weeks. On the 28th day after the operation, CRC growth and liver metastasis were assessed by morphology, the changes in the expression of HIF-1α (hypoxia inducible factor-1α), stromal cell-derived factor-1 alpha (SDF-1α), CXCR4 (C-X-C chemokine receptor type 4), PI3K, and Akt in the transplanted tumor and SDF-1α and CXCR4 in the liver were detected by Western blot and immunohistochemistry. The protein contents of vascular endothelial growth factor (VEGF), matrix metalloproteinase (MMP)-2, and collagen IV in the serum and transplanted tumor and SDF-1α and CXCR4 in liver tissues were detected by enzyme-linked immunosorbent assay. In the Capecitabine and high dose CLG groups, the growth and liver metastasis of CRC were significantly inhibited, the protein levels of HIF-1α, SDF-1α, CXCR4, MMP-2, VEGF, PI3K, Akt, P-PI3K and P-Akt in the transplanted tumor were lower, while the content of collagen IV in the transplanted tumor was higher, than in Model group. A high dose of CLG inhibited the growth of transplanted tumor and liver metastasis of CRC in nude mice, probably by inhibiting the HIF-1α/SDF-1α-CXCR4/PI3K-Akt signaling pathway reducing the synthesis and release of VEGF and degradation of collagen IV.
Collapse
Affiliation(s)
- Bing Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Chun-Shui Pan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
| | - Quan Li
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
| | - Zhu Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Feng-Xi Long
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Jing-Yu Fan
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
| | - Chuan-She Wang
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jing-Yan Han
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Dong-Xin Tang
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Department of Oncology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
3
|
Lyu Y, Xu WQ, Sun LJ, Pan XY, Zhang J, Wang YS. Effect of integrin α5β1 inhibition on SDF-l/CXCR4-mediated choroidal neovascularization. Int J Ophthalmol 2018; 11:726-735. [PMID: 29862169 DOI: 10.18240/ijo.2018.05.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 02/28/2018] [Indexed: 11/23/2022] Open
Abstract
AIM To investigate the roles of integrins in choroidal neovascularization (CNV) and their associations with the stromal cell-derived factor-1 (SDF-1)/CXCR4 axis. METHODS CNV lesions were induced in mice using laser photocoagulation. After CNV induction, all animals were randomly assigned to: control, SDF-1, SDF-1+age-related macular degeneration (AMD) 3100 (CXCR4 inhibitor), and SDF-1+ATN161 (integrin α5β1 inhibitor) groups; their effects on CNV progression were observed using hematoxylin eosin (HE) staining, fundus fluorescein angiography (FFA) grading and optical coherence tomography (OCT), and their effects on CXCR4/integrin α5 expression were evaluated using Western blot and double immunofluorescence staining. Hypoxia-exposed endothelial cells (ECs) were used to simulate CNV in vitro, they were treated with SDF-1, combined with CXCR4 siRNA/AMD3100 or ATN161, and expression of integrin α5, cell migration and tube formation were analyzed. RESULTS Integrin subunit α5 increased at 3rd and 7th day and decreased at 14th day in CNV mice, with no significant change of β1-integrin. CXCR4 expression in CNV mice had persistent increase within 14d after induction. SDF-1 treatment significantly promoted the CNV progression during 3-14d. The mean CNV length in AMD3100 and ATN161 group at day 7 was 270.13 and 264.23 µm in HE images, significantly lower than the mean length in SDF-1 (345.70 µm) group. AMD3100 and ATN161 also significantly reduced thickness and leakage of CNV induced by SDF-1. Mean integrin α5 positive area in SDF-1 group reached 2.31×104 µm2, significantly higher than control (1.25×104 µm2), which decreased to 1.78×104 µm2 after AMD3100 treatment. About 61.36% of ECs in CNV lesions expressed α5 in SDF-1 group, which significantly decreased to 43.12% after AMD3100 treatment. In vitro, integrin α5 peaked by 6 folds after 6h of hypoxia exposure and CXCR4 gradually increased by up to 2.3 folds after 24h of hypoxia. Approximately 25.12% of ECs expressed integrin α5 after SDF-1 stimulation, which decreased to 7.2%-9.5% after si-CXCR4 or AMD3100 treatment. ATN161 exerted an inhibitory effect comparable to that of si-CXCR4 on EC migration and tube formation in the presence of SDF-1. CONCLUSION SDF-1/CXCR4 signaling induces integrin α5β1 expression in ECs to promote CNV.
Collapse
Affiliation(s)
- Yang Lyu
- Department of Ophthalmology, Eye Institute of China PLA, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.,Department of Ophthalmology, General Hospital of Lanzhou Military Command, Lanzhou 730050, Gansu Province, China
| | - Wen-Qin Xu
- Department of Ophthalmology, Eye Institute of China PLA, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Li-Juan Sun
- Department of Ophthalmology, Eye Institute of China PLA, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Xiao-Yan Pan
- Department of Ophthalmology, Eye Institute of China PLA, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Jian Zhang
- Department of Biochemistry and Molecular Biology, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Yu-Sheng Wang
- Department of Ophthalmology, Eye Institute of China PLA, Xijing Hospital, the Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| |
Collapse
|
4
|
Kim ES, Choi YE, Hwang SJ, Han YH, Park MJ, Bae IH. IL-4, a direct target of miR-340/429, is involved in radiation-induced aggressive tumor behavior in human carcinoma cells. Oncotarget 2018; 7:86836-86856. [PMID: 27895317 PMCID: PMC5349958 DOI: 10.18632/oncotarget.13561] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 11/07/2016] [Indexed: 12/12/2022] Open
Abstract
Radiotherapy induces the production of cytokines, thereby increasing aggressive tumor behavior. This radiation effect results in the failure of radiotherapy and increases the mortality rate in patients. We found that interleukin-4 (IL-4) and IL-4Rα (IL-4 receptor) are highly expressed in various human cancer cells subsequent to radiation treatment. In addition, IL-4 is highly overexpressed in metastatic carcinoma tissues compared with infiltrating carcinoma tissues. High expression of IL-4 in patients with cancer is strongly correlated with poor survival. The results of this study suggest that radiation-induced IL-4 contributes to tumor progression and metastasis. Radiation-induced IL-4 was associated with tumorigenicity and metastasis. IL-4 expression was downregulated by miR-340 and miR-429, which were decreased by ionizing radiation (IR). Radiation-regulated miR-340/429-IL4 signaling increased tumorigenesis and metastasis by inducing the production of Sox2, Vimentin, VEGF, Ang2, and MMP-2/9 via activating JAK, JNK, β-catenin, and Stat6 in vitro and in vivo. Our study presents a conceptual advance in our understanding of the modification of tumor microenvironment by radiation and suggests that combining radiotherapy with genetic therapy to inhibit IL-4 may be a promising strategy for preventing post-radiation recurrence and metastasis in patients.
Collapse
Affiliation(s)
- Eun Sook Kim
- Division of Basic Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| | - Young Eun Choi
- Division of Basic Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| | - Su Jin Hwang
- Division of Basic Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| | - Young-Hoon Han
- Division of Basic Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| | - Myung-Jin Park
- Division of Basic Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| | - In Hwa Bae
- Division of Basic Radiation Bioscience, Korea Institute of Radiological & Medical Sciences, Seoul, Republic of Korea
| |
Collapse
|
5
|
Malric L, Monferran S, Gilhodes J, Boyrie S, Dahan P, Skuli N, Sesen J, Filleron T, Kowalski-Chauvel A, Cohen-Jonathan Moyal E, Toulas C, Lemarié A. Interest of integrins targeting in glioblastoma according to tumor heterogeneity and cancer stem cell paradigm: an update. Oncotarget 2017; 8:86947-86968. [PMID: 29156849 PMCID: PMC5689739 DOI: 10.18632/oncotarget.20372] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/23/2017] [Indexed: 12/22/2022] Open
Abstract
Glioblastomas are malignant brain tumors with dismal prognosis despite standard treatment with surgery and radio/chemotherapy. These tumors are defined by an important cellular heterogeneity and notably contain a particular subpopulation of Glioblastoma-initiating cells, which recapitulate the heterogeneity of the original Glioblastoma. In order to classify these heterogeneous tumors, genomic profiling has also been undertaken to classify these heterogeneous tumors into several subtypes. Current research focuses on developing therapies, which could take into account this cellular and genomic heterogeneity. Among these targets, integrins are the subject of numerous studies since these extracellular matrix transmembrane receptors notably controls tumor invasion and progression. Moreover, some of these integrins are considered as membrane markers for the Glioblastoma-initiating cells subpopulation. We reviewed here integrin expression according to glioblastoma molecular subtypes and cell heterogeneity. We discussed their roles in glioblastoma invasion, angiogenesis, therapeutic resistance, stemness and microenvironment modulations, and provide an overview of clinical trials investigating integrins in glioblastomas. This review highlights that specific integrins could be identified as selective glioblastoma cells markers and that their targeting represents new diagnostic and/or therapeutic strategies.
Collapse
Affiliation(s)
- Laure Malric
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France
| | - Sylvie Monferran
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France.,Faculty of Pharmaceutical Sciences, University of Toulouse III Paul Sabatier, Toulouse, France
| | - Julia Gilhodes
- Department of Biostatistics, IUCT-Oncopole, Toulouse, France
| | - Sabrina Boyrie
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France
| | - Perrine Dahan
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France
| | - Nicolas Skuli
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France.,Department of Neurosurgery, Johns Hopkins University, Baltimore, Maryland, USA
| | - Julie Sesen
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France
| | - Thomas Filleron
- Department of Biostatistics, IUCT-Oncopole, Toulouse, France
| | | | - Elizabeth Cohen-Jonathan Moyal
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France.,Department of Radiotherapy, IUCT-Oncopole, Toulouse, France
| | - Christine Toulas
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France.,Laboratory of Oncogenetic, IUCT-Oncopole, Toulouse, France
| | - Anthony Lemarié
- INSERM U1037, Center for Cancer Research of Toulouse, Toulouse, France.,Faculty of Pharmaceutical Sciences, University of Toulouse III Paul Sabatier, Toulouse, France
| |
Collapse
|
6
|
Canella A, Welker AM, Yoo JY, Xu J, Abas FS, Kesanakurti D, Nagarajan P, Beattie CE, Sulman EP, Liu J, Gumin J, Lang FF, Gurcan MN, Kaur B, Sampath D, Puduvalli VK. Efficacy of Onalespib, a Long-Acting Second-Generation HSP90 Inhibitor, as a Single Agent and in Combination with Temozolomide against Malignant Gliomas. Clin Cancer Res 2017; 23:6215-6226. [PMID: 28679777 DOI: 10.1158/1078-0432.ccr-16-3151] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 05/14/2017] [Accepted: 06/28/2017] [Indexed: 01/09/2023]
Abstract
Purpose: HSP90, a highly conserved molecular chaperone that regulates the function of several oncogenic client proteins, is altered in glioblastoma. However, HSP90 inhibitors currently in clinical trials are short-acting, have unacceptable toxicities, or are unable to cross the blood-brain barrier (BBB). We examined the efficacy of onalespib, a potent, long-acting novel HSP90 inhibitor as a single agent and in combination with temozolomide (TMZ) against gliomas in vitro and in vivoExperimental Design: The effect of onalespib on HSP90, its client proteins, and on the biology of glioma cell lines and patient-derived glioma-initiating cells (GSC) was determined. Brain and plasma pharmacokinetics of onalespib and its ability to inhibit HSP90 in vivo were assessed in non-tumor-bearing mice. Its efficacy as a single agent or in combination with TMZ was assessed in vitro and in vivo using zebrafish and patient-derived GSC xenograft mouse glioma models.Results: Onalespib-mediated HSP90 inhibition depleted several survival-promoting client proteins such as EGFR, EGFRvIII, and AKT, disrupted their downstream signaling, and decreased the proliferation, migration, angiogenesis, and survival of glioma cell lines and GSCs. Onalespib effectively crossed the BBB to inhibit HSP90 in vivo and extended survival as a single agent in zebrafish xenografts and in combination with TMZ in both zebrafish and GSC mouse xenografts.Conclusions: Our results demonstrate the long-acting effects of onalespib against gliomas in vitro and in vivo, which combined with its ability to cross the BBB support its development as a potential therapeutic agent in combination with TMZ against gliomas. Clin Cancer Res; 23(20); 6215-26. ©2017 AACR.
Collapse
Affiliation(s)
- Alessandro Canella
- Division of Neuro-oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Department of Neurosurgery and the Dardinger Laboratory for Neuro-Oncology and Neurosciences, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Alessandra M Welker
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Ji Young Yoo
- Department of Neurosurgery and the Dardinger Laboratory for Neuro-Oncology and Neurosciences, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Jihong Xu
- Division of Neuro-oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Department of Neurosurgery and the Dardinger Laboratory for Neuro-Oncology and Neurosciences, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Fazly S Abas
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Divya Kesanakurti
- Division of Neuro-oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Department of Neurosurgery and the Dardinger Laboratory for Neuro-Oncology and Neurosciences, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Prabakaran Nagarajan
- Division of Neuro-oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Department of Neurosurgery and the Dardinger Laboratory for Neuro-Oncology and Neurosciences, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Christine E Beattie
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Erik P Sulman
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joseph Liu
- Department of Neurosurgery and the Dardinger Laboratory for Neuro-Oncology and Neurosciences, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Joy Gumin
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Metin N Gurcan
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Balveen Kaur
- Department of Neurosurgery and the Dardinger Laboratory for Neuro-Oncology and Neurosciences, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Deepa Sampath
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Vinay K Puduvalli
- Division of Neuro-oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio. .,Department of Neurosurgery and the Dardinger Laboratory for Neuro-Oncology and Neurosciences, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| |
Collapse
|
7
|
Tan W, Zhu S, Cao J, Zhang L, Li W, Liu K, Zhong J, Shang C, Chen Y. Inhibition of MMP-2 Expression Enhances the Antitumor Effect of Sorafenib in Hepatocellular Carcinoma by Suppressing the PI3K/AKT/mTOR Pathway. Oncol Res 2017; 25:1543-1553. [PMID: 28276313 PMCID: PMC7841021 DOI: 10.3727/096504017x14886444100783] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Sorafenib has been globally approved as the standard treatment for patients with advanced hepatocellular carcinoma (HCC). However, the response rate of HCC patients to sorafenib is limited because of tumor recurrence and metastasis. Therefore, seeking combined therapeutic strategies with sorafenib is necessary to improve the antitumor efficiency. Here we demonstrated that expression of MMP-2 is positively correlated with the migration ability of HCC cells. Cells with a higher MMP-2 expression (SK-HEP-1 cells) were less sensitive to sorafenib than those with lower MMP-2 expression (HepG2 cells). Cotreatment of cells with SB-3CT and sorafenib more strongly inhibited migration ability than with sorafenib treatment alone in both HCC cells with high and low expression of MMP-2. In vivo cell metastasis experiments confirmed the synergistic effects of sorafenib and SB-3CT in reducing lung metastasis of SK-HEP-1 cells. Mechanistically, we showed that the synergistic antitumor effect may be attributed to inhibition of the PI3K/AKT/mTOR signaling pathway, but not the RAF/MEK/ERK signaling pathway. With these results taken together, the current study demonstrates that inhibiting MMP-2 expression can enhance the antitumor effect of sorafenib in HCC cells with a high MMP-2 expression, which may provide a novel strategy to improve therapeutic efficiency in HCC.
Collapse
|
8
|
Yang Q, Wu H, Wang H, Li Y, Zhang L, Zhu L, Wang W, Zhou J, Fu Y, Chen S, Wu Q, Chen C, Zhou C. N-terminal polypeptide derived from vMIP-II exerts its antitumor activity by inhibiting the CXCR4 pathway in human glioma. Int J Oncol 2017; 50:1160-1174. [PMID: 28350074 PMCID: PMC5363877 DOI: 10.3892/ijo.2017.3906] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 02/28/2017] [Indexed: 11/06/2022] Open
Abstract
Emerging evidence demonstrates that the stromal derived factor-1 (SDF-1α)/CXCR4 axis is associated with tumor aggressiveness and metastasis, including glioma, the most common brain cancer. In the present study, we demonstrated that a novel designed peptide NT21MP of viral macrophage inflammatory protein II, targeting CXCR4 inhibits SDF-1α-induced activation in glioma. The effects of NT21MP on CXCR4 expression, cell survival and migration were assessed on the human glioma cell line U251 and SHG-44 exposed to SDF-1α, by western blotting, MTT assay, flow cytometry and transwell migration assay. Our results illustrated that NT21MP inhibited SDF-1α induced proliferation, migration and invasion by upregulated pro-apoptotic genes (Bak1 and caspase-3) and downregulated Bcl-2/Bax as well as cell cycle regulators (cyclin D1 and CDK4) to arrest cell cycle in G0/G1 phase and promote apoptosis. By RT-qPCR and immunofluorescence we found that CXCR4 was highly expressed in SHG-44 cells. Our results from wound healing and transwell invasion assays indicated silencing of CXCR4 significantly inhibited the SDF-1α‑induced migration and invasion; similarly, flow cytometry showed that treatment with si-CXCR4 affected cell cycle and induced cell apoptosis in SHG-44. However, these effects were significantly weakened by NT21MP. In conclusion, the present study indicates that NT21MP plays a regulatory role in the SDF-1α/CXCR4 axis and further manages the invasion, migration, apoptosis and cell cycle of glioma cells. Thus, NT21MP might represent a novel therapeutic approach against glioma.
Collapse
Affiliation(s)
- Qingling Yang
- Hefei National Laboratory for Physical Sciences at Microscale and the Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, P.R. China
| | - Haihua Wu
- Clinical Testing and Diagnose Experimental Center of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Haifeng Wang
- Clinical Testing and Diagnose Experimental Center of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Yu Li
- Clinical Testing and Diagnose Experimental Center of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Lingyu Zhang
- Clinical Testing and Diagnose Experimental Center of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Lihua Zhu
- Clinical Testing and Diagnose Experimental Center of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Wenrui Wang
- Department of Biotechnology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Jihong Zhou
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Yingxiao Fu
- Department of Bioscience, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Sulian Chen
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Qiong Wu
- Department of Medical Oncology, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Changjie Chen
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Congzhao Zhou
- Hefei National Laboratory for Physical Sciences at Microscale and the Innovation Center for Cell Signaling Network, School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, P.R. China
| |
Collapse
|
9
|
Shen YG, Feng W, Xu YJ, Jiao NN, Sun DQ, Qu WD, Tang Q, Xiong W, Tang Y, Xia Y, Cai QY, Liu DX, Zhang X, Xu G, Liang GY. Effects of RNA silencing of matrix metalloproteinase-2 on the growth of esophageal carcinoma cells in vivo. Oncol Lett 2016; 13:1119-1124. [PMID: 28454222 PMCID: PMC5403388 DOI: 10.3892/ol.2016.5542] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 09/22/2016] [Indexed: 12/14/2022] Open
Abstract
Esophageal carcinoma is one of the most common malignancies in China. Previous studies reported that matrix metalloproteinases (MMPs) have important roles in the progression and invasion of numerous types of solid tumors. Among the MMPs, MMP-2 has been closely associated with tumor growth and invasion. In the present study, a short hairpin RNA (shRNA) lentiviral expression vector targeting the MMP-2 gene was constructed in order to observe the inhibitory effect of MMP-2 gene silencing on the growth of the KYSE150 esophageal carcinoma cell line in vivo. Three small hairpin RNA sequences targeting MMP-2 were designed and cloned into lentiviral vectors. Following transfection of the lentiviral vectors into KTSE150 cells, MMP-2 mRNA and protein expression levels were examined by reverse transcription-quantitative polymerase chain reaction and western blotting, and the growth rate of cells was analyzed by MTT assays. Subsequently, tumor growth was assessed in nude mice. Lentivirus-mediated RNA interference effectively inhibited the expression of MMP-2 mRNA and protein in KYSE150 esophageal carcinoma cells, and suppressed the growth of esophageal carcinoma cells in vivo. The results of the present study suggested that lentivirus-mediated gene therapy targeting MMP-2 may be an attractive strategy for the treatment of esophageal carcinoma and justifies the performance of further studies on the application of lentivirus vectors to cancer gene therapy.
Collapse
Affiliation(s)
- Yu-Guang Shen
- Department of Thoracic and Cardiovascular Surgery, The First People's Hospital of Zunyi, Zunyi, Guizhou 563003, P.R. China
| | - Wen Feng
- Department of Pathology, Henan Tumor Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yi-Jun Xu
- Thoracic Department, Tianjin Chest Hospital, Tianjin 300051, P.R. China
| | - Na-Na Jiao
- Department of Thoracic and Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical College, Guizhou 563003, P.R. China
| | - Da-Qiang Sun
- Thoracic Department, Tianjin Chest Hospital, Tianjin 300051, P.R. China
| | - Wen-Dong Qu
- Department of Thoracic and Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical College, Guizhou 563003, P.R. China
| | - Quan Tang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical College, Guizhou 563003, P.R. China
| | - Wei Xiong
- Department of Thoracic and Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical College, Guizhou 563003, P.R. China
| | - Yang Tang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical College, Guizhou 563003, P.R. China
| | - Yu Xia
- Department of Thoracic and Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical College, Guizhou 563003, P.R. China
| | - Qing-Yong Cai
- Department of Thoracic and Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical College, Guizhou 563003, P.R. China
| | - Da-Xing Liu
- Department of Thoracic and Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical College, Guizhou 563003, P.R. China
| | - Xun Zhang
- Thoracic Department, Tianjin Chest Hospital, Tianjin 300051, P.R. China
| | - Gang Xu
- Department of Thoracic and Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical College, Guizhou 563003, P.R. China
| | - Gui-You Liang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical College, Guizhou 563003, P.R. China
| |
Collapse
|
10
|
Liang X, Huang X, Zhou Y, Jin R, Li Q. Mechanical Stretching Promotes Skin Tissue Regeneration via Enhancing Mesenchymal Stem Cell Homing and Transdifferentiation. Stem Cells Transl Med 2016; 5:960-9. [PMID: 27130223 DOI: 10.5966/sctm.2015-0274] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 02/23/2016] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED Skin tissue expansion is a clinical procedure for skin regeneration to reconstruct cutaneous defects that can be accompanied by severe complications. The transplantation of mesenchymal stem cells (MSCs) has been proven effective in promoting skin expansion and helping to ameliorate complications; however, systematic understanding of its mechanism remains unclear. MSCs from luciferase-Tg Lewis rats were intravenously transplanted into a rat tissue expansion model to identify homing and transdifferentiation. To clarify underlying mechanisms, a systematic approach was used to identify the differentially expressed genes between mechanically stretched human MSCs and controls. The biological significance of these changes was analyzed through bioinformatic methods. We further investigated genes and pathways of interest to disclose their potential role in mechanical stretching-induced skin regeneration. Cross sections of skin samples from the expanded group showed significantly more luciferase(+) and stromal cell-derived factor 1α (SDF-1α)(+), luciferase(+)keratin 14(+), and luciferase(+)CD31(+) cells than the control group, indicating MSC transdifferentiation into epidermal basal cells and endothelial cells after SDF-1α-mediated homing. Microarray analysis suggested upregulation of genes related to hypoxia, vascularization, and cell proliferation in the stretched human MSCs. Further investigation showed that the homing of MSCs was blocked by short interfering RNA targeted against matrix metalloproteinase 2, and that mechanical stretching-induced vascular endothelial growth factor A upregulation was related to the Janus kinase/signal transducer and activator of transcription (Jak-STAT) and Wnt signaling pathways. This study determines that mechanical stretching might promote skin regeneration by upregulating MSC expression of genes related to hypoxia, vascularization, and cell proliferation; enhancing transplanted MSC homing to the expanded skin; and transdifferentiation into epidermal basal cells and endothelial cells. SIGNIFICANCE Skin tissue expansion is a clinical procedure for skin regeneration to cover cutaneous defects that can be accompanied by severe complications. The transplantation of mesenchymal stem cells (MSCs) has been proven effective in promoting skin expansion and ameliorating complications. This study, which sought to provide a systematic understanding of the mechanism, determined that mechanical stretching could upregulate MSC expression of genes related to hypoxia, vascularization, and cell proliferation; enhance transplanted MSC homing to the expanded skin tissue; and promote their transdifferentiation into epidermal basal cells and endothelial cells.
Collapse
Affiliation(s)
- Xiao Liang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Xiaolu Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yiwen Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Rui Jin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
11
|
Huber SM, Butz L, Stegen B, Klumpp L, Klumpp D, Eckert F. Role of ion channels in ionizing radiation-induced cell death. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2015; 1848:2657-64. [DOI: 10.1016/j.bbamem.2014.11.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 10/30/2014] [Accepted: 11/05/2014] [Indexed: 02/05/2023]
|
12
|
Sun Q, Mu L, Qiao W, Li H, Tang J, Wang C, Hu W, Zhao T, Dong B, Song Y, Liu X. Inhibition of SHP-2 promotes radiosensitivity in glioma. Mol Med Rep 2015; 12:3563-3568. [PMID: 26004555 DOI: 10.3892/mmr.2015.3829] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 04/20/2015] [Indexed: 11/06/2022] Open
Abstract
As a phosphatase, SHP-2 has been identified to be involved in regulating several cell functions, including growth, division, adhesion and motility. Therefore, SHP‑2 may affect the response of glioma to radiotherapy, such as via enhancing angiogenesis. The present study aimed to investigate the function of SHP‑2, a protein tyrosine phosphatase, in the radiosensitivity of glioma. U251, U87 and SHG44 glioma cell lines were transfected with small interfering (si)RNA against SHP‑2 and cell proliferation was assessed using a cell counting kit 8 assay, cell apoptosis was assessed by fluorescence‑activated cell sorting and immunoblotting, cell invasion was determined by an invasion assay, and the vasculogenic mimicry capacity was assessed by a tube formation assay. SHP‑2 siRNA transfection reduced the proliferation and increased apoptosis in the glioma cell lines. Downregulation of SHP‑2 suppressed glioma cell invasion and vasculogenic mimicry. These results demonstrated that no significant difference was observed between glioma tissues and normal brain tissues, however, silencing of SHP‑2 inhibited cell proliferation, invasion and vasculogenic mimicry in the glioma cell lines. SHP‑2 may be a novel therapeutic target for glioma.
Collapse
Affiliation(s)
- Quanye Sun
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Luyan Mu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Wanchen Qiao
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Hui Li
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Jiabin Tang
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Ce Wang
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Wei Hu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Tianshu Zhao
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Baijing Dong
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yuwen Song
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xiaoqian Liu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
13
|
Fu ZC, Wang FM, Cai JM. Gene expression changes in residual advanced cervical cancer after radiotherapy: indicators of poor prognosis and radioresistance? Med Sci Monit 2015; 21:1276-87. [PMID: 25940978 PMCID: PMC4432617 DOI: 10.12659/msm.893689] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Background Different sensitivity of advanced cervical cancer to irradiation can decrease effectiveness of radiotherapy in some cases. We attempted to identify the differentially expressed genes in residual cervical cancer after radiotherapy that might be associated with poor prognosis and radioresistance. Material/Methods Differential genes expression was identified by an oligonucleotide microarray in cervical cancer tissues before radiation and after a 50-Gy dose of radiation. The microarray results were validated by quantitative real-time PCR. CXCL12 was validated by immunohistochemistry in paraffin-embedded cervical cancer tissues before radiotherapy. The relationship between the differentiated gene and prognosis was validated by survival analysis. Results Hierarchic cluster analysis identified 238 differentiated genes that exhibited ≥3.0-fold change and p<0.05. We found 111 genes that were in persistent up-regulation and 127 in persistent down-regulation after a 50-Gy dose of radiation when compared with the control group. These genes were involved in processes such as cell growth and death, cell-apoptosis, cell cycle regulation, cell signaling, DNA synthesis and repair, and cell adhesion. High differential expression of CXCL12, CD74, FGF7, COL14A1, PRC1, and RAD54L genes was validated by quantitative PCR before and after radiotherapy. Survival analysis results showed that the high expression of CXCL12 was closely related to poor prognosis. Conclusions The higher expression of CXCL12 might be informative regarding poor prognosis in patients undergoing radical radiotherapy. The differentially expressed genes identified in our study might provide a new method for diagnosis and treatment of radioresistance in cervical cancer.
Collapse
Affiliation(s)
- Zhi-chao Fu
- Department of Radiotherapy, Fu Zhou General Hospital, Fuzhou, Fujian, China (mainland)
| | - Feng-mei Wang
- Department of Obstetrics and Gynecology, Fu Zhou General Hospital, Fuzhou, Fujian, China (mainland)
| | - Jian-ming Cai
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China (mainland)
| |
Collapse
|
14
|
Pham K, Luo D, Siemann DW, Law BK, Reynolds BA, Hothi P, Foltz G, Harrison JK. VEGFR inhibitors upregulate CXCR4 in VEGF receptor-expressing glioblastoma in a TGFβR signaling-dependent manner. Cancer Lett 2015; 360:60-7. [PMID: 25676691 DOI: 10.1016/j.canlet.2015.02.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 02/03/2015] [Accepted: 02/03/2015] [Indexed: 11/16/2022]
Abstract
The failure of standard treatment for patients diagnosed with glioblastoma (GBM) coupled with the highly vascularized nature of this solid tumor has led to the consideration of agents targeting VEGF or VEGFRs, as alternative therapeutic strategies for this disease. Despite modest achievements in survival obtained with such treatments, failure to maintain an enduring survival benefit and more invasive relapsing tumors are evident. Our study suggests a potential mechanism by which anti-VEGF/VEGFR therapies regulate the enhanced invasive phenotype through a pathway that involves TGFβR and CXCR4. VEGFR signaling inhibitors (Cediranib and Vandetanib) elevated the expression of CXCR4 in VEGFR-expressing GBM cell lines and tumors, and enhanced the in vitro migration of these lines toward CXCL12. The combination of VEGFR inhibitor and CXCR4 antagonist provided a greater survival benefit to tumor-bearing animals. The upregulation of CXCR4 by VEGFR inhibitors was dependent on TGFβ/TGFβR, but not HGF/MET, signaling activity, suggesting a mechanism of crosstalk among VEGF/VEGFR, TGFβ/TGFβR, and CXCL12/CXCR4 pathways in the malignant phenotype of recurrent tumors after anti-VEGF/VEGFR therapies. Thus, the combination of VEGFR, CXCR4, and TGFβR inhibitors could provide an alternative strategy to halt GBM progression.
Collapse
Affiliation(s)
- Kien Pham
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Defang Luo
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Dietmar W Siemann
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA; Department of Radiation Oncology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brian K Law
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Brent A Reynolds
- Department of Neurosurgery, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Parvinder Hothi
- The Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA 98122, USA
| | - Gregory Foltz
- The Ben and Catherine Ivy Center for Advanced Brain Tumor Treatment, Swedish Neuroscience Institute, Seattle, WA 98122, USA
| | - Jeffrey K Harrison
- Department of Pharmacology & Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
15
|
Nambiar DK, Rajamani P, Singh RP. Silibinin attenuates ionizing radiation-induced pro-angiogenic response and EMT in prostate cancer cells. Biochem Biophys Res Commun 2014; 456:262-8. [PMID: 25446081 DOI: 10.1016/j.bbrc.2014.11.069] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 11/18/2014] [Indexed: 01/31/2023]
Abstract
Radiotherapy of is well established and frequently utilized in prostate cancer (PCa) patients. However, recurrence following therapy and distant metastases are commonly encountered problems. Previous studies underline that, in addition to its therapeutic effects, ionizing radiation (IR) increases the vascularity and invasiveness of surviving radioresistant cancer cells. This invasive phenotype of radioresistant cells is an upshot of IR-induced pro-survival and mitogenic signaling in cancer as well as endothelial cells. Here, we demonstrate that a plant flavonoid, silibinin can radiosensitize endothelial cells by inhibiting expression of pro-angiogenic factors. Combining silibinin with IR not only strongly down-regulated endothelial cell proliferation, clonogenicity and tube formation ability rather it strongly (p<0.001) reduced migratory and invasive properties of PCa cells which were otherwise marginally affected by IR treatment alone. Most of the pro-angiogenic (VEGF, iNOS), migratory (MMP-2) and EMT promoting proteins (uPA, vimentin, N-cadherin) were up-regulated by IR in PCa cells. Interestingly, all of these invasive and EMT promoting actions of IR were markedly decreased by silibinin. Further, we found that potentiated effect was an end result of attenuation of IR-activated mitogenic and pro-survival signaling, including Akt, Erk1/2 and STAT-3, by silibinin.
Collapse
Affiliation(s)
- Dhanya K Nambiar
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India; School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Paulraj Rajamani
- School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Rana P Singh
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India; School of Life Sciences, Central University of Gujarat, Gandhinagar, India.
| |
Collapse
|
16
|
Zheng S, Shi L, Zhang Y, He T. Expression of SNCG, MAP2, SDF-1 and CXCR4 in gastric adenocarcinoma and their clinical significance. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:6606-6615. [PMID: 25400739 PMCID: PMC4230097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 09/13/2014] [Indexed: 06/04/2023]
Abstract
OBJECTIVES The purpose of the study was to detect the expression of SNCG, MAP2, SDF-1 and CXCR4 in gastric adenocarcinoma, and to evaluate their roles in the carcinogenesis of gastric adenocarcinoma, development, invasion and metastasis as well as their clinical significance. METHODS The expression of SNCG, MAP2, SDF-1 and CXCR4 was detected by SP immunohistochemical method in 225 cases of gastric adenocarcinoma and 105 cases of nonneoplastic adjacent gastric tissue. The expression of SNCG, MAP2, SDF-1 and CXCR4 mRNA was also detected by RT-PCR method in 50 cases of gastric adenocarcinoma and 30 cases of nonneoplastic adjacent gastric tissue. RESULTS The expression of SNCG, MAP2, SDF-1 and CXCR4 in the gastric adenocarcinoma was remarkably higher than those in the nonneoplastic adjacent gastric tissue (P < 0.01); The positive expression of SNCG and MAP2 was correlated with the depth of tumor invasion and the metastasis of lymph nodes (P < 0.05), and that of SDF-1 and CXCR4 was correlated with the metastasis of lymph nodes (P < 0.05). CONCLUSIONS SNCG, MAP2, SDF-1 and CXCR4 may play an important role in the carcinogenesis, progression, invasion and metastasis of gastric adenocarcinoma. However, it still needs more exploration whether they can serve as promising therapeutic targets of gastric adenocarcinoma.
Collapse
Affiliation(s)
- Shufang Zheng
- Department of Pathology, Affiliated Hospital of Logistics College of The Chinese People’s Armed Police Force220 Chenglin Road, Hedong District, Tianjin 300162, People’s Republic of China
| | - Lifang Shi
- Department of Pathology, Affiliated Hospital of Logistics College of The Chinese People’s Armed Police Force220 Chenglin Road, Hedong District, Tianjin 300162, People’s Republic of China
| | - Yi Zhang
- Postgraduate Training Center for Liaoning Medical College in Affiliated Hospital of Logistics College, The Chinese People’s Armed Police Force220 Chenglin Road, Hedong District, Tianjin 300162, People’s Republic of China
| | - Tao He
- Department of Pathology, Affiliated Hospital of Logistics College of The Chinese People’s Armed Police Force220 Chenglin Road, Hedong District, Tianjin 300162, People’s Republic of China
| |
Collapse
|
17
|
Multhoff G, Radons J, Vaupel P. Critical role of aberrant angiogenesis in the development of tumor hypoxia and associated radioresistance. Cancers (Basel) 2014; 6:813-28. [PMID: 24717239 PMCID: PMC4074805 DOI: 10.3390/cancers6020813] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 03/18/2014] [Accepted: 03/21/2014] [Indexed: 12/02/2022] Open
Abstract
Newly formed microvessels in most solid tumors show an abnormal morphology and thus do not fulfil the metabolic demands of the growing tumor mass. Due to the chaotic and heterogeneous tumor microcirculation, a hostile tumor microenvironment develops, that is characterized inter alia by local hypoxia, which in turn can stimulate the HIF-system. The latter can lead to tumor progression and may be involved in hypoxia-mediated radioresistance of tumor cells. Herein, cellular and molecular mechanisms in tumor angiogenesis are discussed that, among others, might impact hypoxia-related radioresistance.
Collapse
Affiliation(s)
- Gabriele Multhoff
- Department of Radiotherapy and Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Straße 22, 81675 Munich, Germany.
| | - Jürgen Radons
- GmbH, Munich, Ismaningerstr. 22, 81675 Munich, Germany.
| | - Peter Vaupel
- Department of Radiotherapy and Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Straße 22, 81675 Munich, Germany.
| |
Collapse
|
18
|
Yu ZH, Liu T, Zhao YH, Huang YY, Gao YT. Cisplatin targets the stromal cell-derived factor-1-CXC chemokine receptor type 4 axis to suppress metastasis and invasion of ovarian cancer-initiating cells. Tumour Biol 2014; 35:4637-44. [PMID: 24408020 DOI: 10.1007/s13277-014-1607-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 01/02/2014] [Indexed: 12/13/2022] Open
Abstract
In ovarian cancer, CD44+/CD117+ stem cells, also known as cancer-initiating cells (CICs), are highly proliferative and invasive. Therefore, the CD44+/CD117+ subpopulation is thought to be an important target for novel therapeutic strategies. In this study, we investigated the effects of cisplatin (CDDP) on metastasis and invasion suppression of ovarian CICs by targeting the CXC chemokine receptor-4 (CXCR4) signaling pathway in vitro and in vivo. CD44+/CD117+ ovarian CICs were enriched from human primary ovarian tumor tissues and confirmed by flow cytometry sorting. A 3-(4,5-dimethylthiazol-2-yl)-2.5-dipheny-tetrazolium bromide (MTT) assay revealed significant inhibition of proliferation of ovarian CICs with increasing CDDP drug concentrations. Moreover, colony formation and transwell migration assays indicated that CDDP significantly suppressed the invasive capacity of ovarian CICs in vitro. The expression levels of stromal cell-derived factor (SDF)-1, CXCR4, matrix metalloproteinase (MMP) 2, and MMP9 mRNA and protein levels were significantly reduced in CDDP-treated cells compared to untreated ovarian CICs. Furthermore, xenograft experiments confirmed that CDDP suppressed the growth of xenograft tumors formed by ovarian CICs in vivo. In addition, CXCR4 agonist (diprotin A) treatment of ovarian CICs weakened the effects of CDDP and enhanced SDF-1-CXCR4 axis expression in ovarian CICs. Thus, the SDF-1-CXCR4 axis is an important mediator of proliferation and invasion in CXCR4-overexpressing ovarian cancer-initiating cells (OCICs). Furthermore, CDDP inhibits invasion and metastasis of OCICs by targeting SDF-1-CXCR4 axis expression.
Collapse
Affiliation(s)
- Zhi-hua Yu
- Shanghai Geriatric Institute of Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200031, China
| | | | | | | | | |
Collapse
|