1
|
Wang Y, Wang G, Xiang W, Liu X, Jiang M, Hu J. Proteasome activation is critical for cell death induced by inhibitors of polo-like kinase 1 (PLK1) in multiple cancers. Eur J Pharmacol 2024; 972:176558. [PMID: 38614382 DOI: 10.1016/j.ejphar.2024.176558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/15/2024]
Abstract
Inhibitors of polo-like kinase (PLK) are currently being evaluated as anticancer drugs. However, the molecular mechanism of PLK inhibitor-induced cell death is not fully understood. In this study, we found that GW843682X and BI2536, two inhibitors of PLK1, significantly induced cell death in multiple type cells. The induction of cell death was related to the preferring expression of PLK1. However, in human umbilical vascular endothelial cells (HUVEC) and human colorectal carcinoma cells, which expressed higher levels of both PLK1 and PLK2, PLK1 inhibitors induced very low levels of cell death. Clinical analysis reveals PLK1 presence in 26 of 30 NPC tumor tissues. In in vivo NPC lung metastasis nude mouse models, PLK1 inhibitors decreased NPC progress. Mechanistically, the PLK1 inhibitor did not activate p53, and the cell death was not reversed by p53 inhibition. Moreover, PLK1 inhibitor-induced cell death was PARP- and caspase-independent. Although PLK1 inhibitors induced down-regulation of calpain inhibitor calpastatin and calpain was activated by PLK1 inhibition, calpain blocking did not reverse cell death induced by PLK1 inhibitors, suggesting the non-involvement of calpain. Surprisingly, we found that PLK1 inhibitors induced the activation of proteasome, and the treatment of cells with PLK1 inhibitors reduced the levels of ubiquitinated proteins. And proteasome inhibitors reversed cell death induced by PLK1 inhibitors in various cell types in which PLK1 was preferentially expressed. Moreover, PLK1 inhibition reversed the degradation of proteins including p53, caspase 8, PARP and calpastatin. These results suggest that the activation of proteasome is critical for cell death induced by PLK1 inhibition.
Collapse
Affiliation(s)
- Yufei Wang
- Medical Research Center, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha, 410004, China
| | - Guihua Wang
- Department of Oncology, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha, 410004, China
| | - Wei Xiang
- Medical Research Center, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha, 410004, China
| | - Xueting Liu
- Medical Research Center, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha, 410004, China
| | - Manli Jiang
- Medical Research Center, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha, 410004, China
| | - Jinyue Hu
- Medical Research Center, Affiliated Changsha Central Hospital of Hengyang Medical School, University of South China, Changsha, 410004, China.
| |
Collapse
|
2
|
Xu P, Cai X, Guan X, Xie W. Sulfoconjugation of protein peptides and glycoproteins in physiology and diseases. Pharmacol Ther 2023; 251:108540. [PMID: 37777160 PMCID: PMC10842354 DOI: 10.1016/j.pharmthera.2023.108540] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023]
Abstract
Protein sulfoconjugation, or sulfation, represents a critical post-translational modification (PTM) process that involves the attachment of sulfate groups to various positions of substrates within the protein peptides or glycoproteins. This process plays a dynamic and complex role in many physiological and pathological processes. Here, we summarize the importance of sulfation in the fields of oncology, virology, drug-induced liver injury (DILI), inflammatory bowel disease (IBD), and atherosclerosis. In oncology, sulfation is involved in tumor initiation, progression, and migration. In virology, sulfation influences viral entry, replication, and host immune response. In DILI, sulfation is associated with the incidence of DILI, where altered sulfation affects drug metabolism and toxicity. In IBD, dysregulation of sulfation compromises mucosal barrier and immune response. In atherosclerosis, sulfation influences the development of atherosclerosis by modulating the accumulation of lipoprotein, and the inflammation, proliferation, and migration of smooth muscle cells. The current review underscores the importance of further research to unravel the underlying mechanisms and therapeutic potential of targeting sulfoconjugation in various diseases. A better understanding of sulfation could facilitate the emergence of innovative diagnostic or therapeutic strategies.
Collapse
Affiliation(s)
- Pengfei Xu
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430072, China
| | - Xinran Cai
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiuchen Guan
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing 100069, China
| | - Wen Xie
- Center for Pharmacogenetics and Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
3
|
Taheri F, Ebrahimi SO, Heidari R, Pour SN, Reiisi S. Mechanism and function of miR-140 in human cancers: A review and in silico study. Pathol Res Pract 2023; 241:154265. [PMID: 36509008 DOI: 10.1016/j.prp.2022.154265] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/27/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022]
Abstract
MicroRNA-140 (miR-140) acts as a tumor suppressor and plays a vital role in cell biological functions such as cell proliferation, apoptosis, and DNA repair. The expression of this miRNA has been shown to be considerably decreased in cancer tissues and cell lines compared with normal adjacent tissues. Consequently, aberrant expression of some miR-140 target genes can lead to the initiation and progression of various human cancers, such as breast cancer, gastrointestinal cancers, lung cancer, and prostate cancer. The dysregulation of the miR-140 network also affects cell proliferation, invasion, metastasis, and apoptosis of cancer cells by affecting various signaling pathways. Besides, up-regulation of miR-140 could enhance the efficacy of chemotherapeutic agents in different cancer. We aimed to cover most aspects of miR-140 function in cancer development and address its importance in different stages of cancer progression.
Collapse
Affiliation(s)
- Forough Taheri
- Department of Genetics, Sharekord Branch, Islamic Azad University, Sharekord, Iran
| | - Seyed Omar Ebrahimi
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran
| | - Razieh Heidari
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Somaye Nezamabadi Pour
- Department of Obstetrics and Gynecology, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| | - Somayeh Reiisi
- Department of Genetics, Faculty of Basic Sciences, Shahrekord University, Shahrekord, Iran.
| |
Collapse
|
4
|
Ling M, Liu Q, Wang Y, Liu X, Jiang M, Hu J. LCS-1 inhibition of superoxide dismutase 1 induces ROS-dependent death of glioma cells and degradates PARP and BRCA1. Front Oncol 2022; 12:937444. [PMID: 35978820 PMCID: PMC9376264 DOI: 10.3389/fonc.2022.937444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/11/2022] [Indexed: 11/15/2022] Open
Abstract
Gliomas are characterized by high morbidity and mortality, and have only slightly increased survival with recent considerable improvements for treatment. An innovative therapeutic strategy had been developed via inducing ROS-dependent cell death by targeting antioxidant proteins. In this study, we found that glioma tissues expressed high levels of superoxide dismutase 1 (SOD1). The expression of SOD1 was upregulated in glioma grade III and V tissues compared with that in normal brain tissues or glioma grade I tissues. U251 and U87 glioma cells expressed high levels of SOD1, low levels of SOD2 and very low levels of SOD3. LCS-1, an inhibitor of SOD1, increased the expression SOD1 at both mRNA and protein levels slightly but significantly. As expected, LCS-1 caused ROS production in a dose- and time-dependent manner. SOD1 inhibition also induced the gene expression of HO-1, GCLC, GCLM and NQO1 which are targeting genes of nuclear factor erythroid 2-related factor 2, suggesting the activation of ROS signal pathway. Importantly, LCS-1 induced death of U251 and U87 cells dose- and time-dependently. The cell death was reversed by the pretreatment of cells with ROS scavenges NAC or GSH. Furthermore, LCS-1 decreased the growth of xenograft tumors formed by U87 glioma cells in nude mice. Mechanistically, the inhibition of P53, caspases did not reverse LCS-1-induced cell death, indicating the failure of these molecules involving in cell death. Moreover, we found that LCS-1 treatment induced the degradation of both PARP and BRCA1 simultaneously, suggesting that LCS-1-induced cell death may be associated with the failure of DNA damage repair. Taking together, these results suggest that the degradation of both PARP and BRCA1 may contribute to cell death induced by SOD1 inhibition, and SOD1 may be a target for glioma therapy.
Collapse
Affiliation(s)
- Min Ling
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Qing Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yufei Wang
- Department of Clinical Laboratory, Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Medical Research Center, Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Xueting Liu
- Medical Research Center, Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Manli Jiang
- Medical Research Center, Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| | - Jinyue Hu
- Medical Research Center, Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- *Correspondence: Jinyue Hu,
| |
Collapse
|
5
|
Xia Y, Wang G, Jiang M, Liu X, Zhao Y, Song Y, Jiang B, Zhu D, Hu L, Zhang Z, Cao T, Wang JM, Hu J. A Novel Biological Activity of the STAT3 Inhibitor Stattic in Inhibiting Glutathione Reductase and Suppressing the Tumorigenicity of Human Cervical Cancer Cells via a ROS-Dependent Pathway. Onco Targets Ther 2021; 14:4047-4060. [PMID: 34262291 PMCID: PMC8275107 DOI: 10.2147/ott.s313507] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/17/2021] [Indexed: 12/16/2022] Open
Abstract
Introduction Glutathione reductase (GSR) provides reduced glutathione (GSH) to maintain redox homeostasis. Inhibition of GSR disrupts this balance, resulting in cell damage, which benefits cancer therapy. However, the effect of GSR inhibition on the tumorigenicity of human cervical cancer is not fully understood. Materials and Methods Tissue microarray analysis was employed to determine GSR expression in cervical cancer tissues by immunohistochemical staining. Cell death was measured with PI/FITC-annexin V staining. mRNA levels were measured via quantitative RT-PCR. Protein expression was measured by Western blotting and flow cytometry. STAT3 deletion was performed with CRISPR/Cas9 technology. GSR knockdown was achieved by RNA interference. Reactive oxygen species (ROS) levels were measured by DCF staining. GSR enzymatic activity was measured with a GSR assay kit. The effect of GSR inhibition on the growth of tumors formed by cervical cancer cells was investigated using a xenograft model. Results The expression of GSR was increased in human cervical cancer tissues, as shown by immunohistochemical staining. GSR knockdown by RNA interference in human cervical cancer cell lines resulted in cell death, suggesting the ability of GSR to maintain cancer cell survival. The STAT3 inhibitor 6-nitrobenzo[b]thiophene 1,1-dioxide (Stattic) also inhibited the enzymatic activity of GSR and induced the death of cervical cancer cells. More importantly, Stattic decreased the growth of xenograft tumors formed by cervical cancer cells in nude mice. Mechanistically, tumor cell death induced by Stattic-mediated GSR inhibition was ROS-dependent, since the ROS scavengers GSH and N-acetyl cysteine (NAC) reversed the effect of Stattic. In contrast, pharmacological and molecular inhibition of STAT3 did not induce the death of cervical cancer cells, suggesting a STAT3-independent activity of Stattic. Conclusion Stattic inhibits the enzymatic activity of GSR and induces STAT3-independent but ROS-dependent death of cervical cancer cells, suggesting its potential application as a therapeutic agent for human cervical cancers.
Collapse
Affiliation(s)
- Yuchen Xia
- Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, People's Republic of China.,Department of Oncology, Changsha Central Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Guihua Wang
- Department of Oncology, Changsha Central Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Manli Jiang
- Medical Research Center, Changsha Central Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Xueting Liu
- Medical Research Center, Changsha Central Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Yan Zhao
- Department of Pathology, Changsha Central Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Yinghui Song
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital, Hunan Normal University, Changsha, Hunan, People's Republic of China
| | - Binyuan Jiang
- Medical Research Center, Changsha Central Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Demao Zhu
- Department of Pathology, Changsha Central Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Ling Hu
- Department of Clinical Laboratory, Changsha Central Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Zhao Zhang
- Department of Clinical Laboratory, Changsha Central Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Ting Cao
- Department of Clinical Laboratory, Changsha Central Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Ji Ming Wang
- Laboratory of Cancer Immunometabolism, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, 21702, USA
| | - Jinyue Hu
- Medical Research Center, Changsha Central Hospital, University of South China, Changsha, Hunan, People's Republic of China
| |
Collapse
|
6
|
Fung AS, Kopciuk K, Dean ML, D’Silva A, Otsuka S, Klimowicz A, Hao D, Morris D, Bebb DG. CXCR4 expression in lung carcinogenesis: Evaluating gender-specific differences in survival outcomes based on CXCR4 expression in early stage non-small cell lung cancer patients. PLoS One 2021; 16:e0241240. [PMID: 33507926 PMCID: PMC7842928 DOI: 10.1371/journal.pone.0241240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/11/2020] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Evidence suggests that the expression of certain cytokine receptors increases with lung cancer evolution. Overexpression of the cytokine receptor CXCR4 is associated with poor outcomes in stage IV non-small cell lung cancer (NSCLC), with shorter survival in females with high CXCR4 expression. This study quantifies CXCR4 expression in early stage disease and evaluates its association with gender-specific recurrence-free (RFS) and overall survival (OS) in resected stage I-III NSCLC patients. METHODS Patient characteristics and clinical outcomes were obtained from the Glans-Look Lung Cancer (G-LLC) database for early stage NSCLC patients diagnosed between 2003-2006 at the Tom Baker Cancer Centre (TBCC). CXCR4 expression was quantified on tissue microarrays (TMA). Median RFS and OS were evaluated by gender using Kaplan-Meier analyses. CXCR4 expression and outcome data were analyzed using Cox proportional hazards (PH) and multi-state models (MSM). RESULTS 176 stage I-III NSCLC patients were identified. CXCR4 expression was lower in early stage NSCLC patients, with a mean CXCR4 expression of 1729 (SD 1083) compared to 2640 (SD 1541) in stage IV patients. On Kaplan-Meier, median RFS by gender was similar (male 52.8 months vs. female 54.5 months) as was median OS (male 80.9 months vs. female 89.0 months), and there was no significant difference in RFS (p = 0.60) or OS (p = 0.30) by gender and CXCR4 groups over follow-up. By multivariable analysis, CXCR4 expression was not prognostic for RFS (Hazard Ratio (HR) = 1.00, p = 0.73) or OS (HR = 1.00, p = 0.44), and no gender difference was observed. CONCLUSIONS CXCR4 expression increases with stage progression in NSCLC but is not prognostic in early stage NSCLC patients of either gender. Mechanisms by which CXCR4 expression increases during lung carcinogenesis warrant further exploration and testing in clinical trials.
Collapse
Affiliation(s)
- Andrea S. Fung
- Tom Baker Cancer Centre, Calgary, Alberta, Canada
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Oncology, Queen’s University, Kingston, Ontario, Canada
| | - Karen Kopciuk
- Tom Baker Cancer Centre, Calgary, Alberta, Canada
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michelle L. Dean
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Adrijana D’Silva
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Shannon Otsuka
- Tom Baker Cancer Centre, Calgary, Alberta, Canada
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Desiree Hao
- Tom Baker Cancer Centre, Calgary, Alberta, Canada
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Don Morris
- Tom Baker Cancer Centre, Calgary, Alberta, Canada
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - D. Gwyn Bebb
- Tom Baker Cancer Centre, Calgary, Alberta, Canada
- Department of Oncology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- * E-mail:
| |
Collapse
|
7
|
Cao Y, Xie L, Shi F, Tang M, Li Y, Hu J, Zhao L, Zhao L, Yu X, Luo X, Liao W, Bode AM. Targeting the signaling in Epstein-Barr virus-associated diseases: mechanism, regulation, and clinical study. Signal Transduct Target Ther 2021; 6:15. [PMID: 33436584 PMCID: PMC7801793 DOI: 10.1038/s41392-020-00376-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/30/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
Epstein–Barr virus-associated diseases are important global health concerns. As a group I carcinogen, EBV accounts for 1.5% of human malignances, including both epithelial- and lymphatic-originated tumors. Moreover, EBV plays an etiological and pathogenic role in a number of non-neoplastic diseases, and is even involved in multiple autoimmune diseases (SADs). In this review, we summarize and discuss some recent exciting discoveries in EBV research area, which including DNA methylation alterations, metabolic reprogramming, the changes of mitochondria and ubiquitin-proteasome system (UPS), oxidative stress and EBV lytic reactivation, variations in non-coding RNA (ncRNA), radiochemotherapy and immunotherapy. Understanding and learning from this advancement will further confirm the far-reaching and future value of therapeutic strategies in EBV-associated diseases.
Collapse
Affiliation(s)
- Ya Cao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China. .,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China. .,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China. .,Research Center for Technologies of Nucleic Acid-Based Diagnostics and Therapeutics Hunan Province, 410078, Changsha, China. .,Molecular Imaging Research Center of Central South University, 410008, Changsha, Hunan, China. .,National Joint Engineering Research Center for Genetic Diagnostics of Infectious Diseases and Cancer, 410078, Changsha, China. .,Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.
| | - Longlong Xie
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Feng Shi
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Min Tang
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China.,Molecular Imaging Research Center of Central South University, 410008, Changsha, Hunan, China
| | - Yueshuo Li
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Jianmin Hu
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Lin Zhao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Luqing Zhao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China
| | - Xinfang Yu
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China
| | - Xiangjian Luo
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, 410078, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, 410078, Changsha, China.,Molecular Imaging Research Center of Central South University, 410008, Changsha, Hunan, China
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital, Central South University, 410078, Changsha, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| |
Collapse
|
8
|
Hu J, Li Y, Li H, Shi F, Xie L, Zhao L, Tang M, Luo X, Jia W, Fan J, Zhou J, Gao Q, Qiu S, Wu W, Zhang X, Liao W, Bode AM, Cao Y. Targeting Epstein-Barr virus oncoprotein LMP1-mediated high oxidative stress suppresses EBV lytic reactivation and sensitizes tumors to radiation therapy. Theranostics 2020; 10:11921-11937. [PMID: 33204320 PMCID: PMC7667690 DOI: 10.7150/thno.46006] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 09/28/2020] [Indexed: 12/24/2022] Open
Abstract
Generating oxidative stress is a critical mechanism by which host cells defend against infection by pathogenic microorganisms. Radiation resistance is a critical problem in radiotherapy against cancer. Epstein-Barr virus (EBV) is a cancer-causing virus and its reactivation plays an important role in the development of EBV-related tumors. This study aimed to explore the inner relationship and regulatory mechanism among oxidative stress, EBV reactivation, and radioresistance and to identify new molecular subtyping models and treatment strategies to improve the therapeutic effects of radiotherapy. Methods: ROS, NADP+/NADPH, and GSSG/GSH were detected to evaluate the oxidative stress of cells. 8-OHdG is a reliable oxidative stress marker to evaluate the oxidative stress in patients. Its concentration in serum was detected using an ELISA method and in biopsies was detected using IHC. qPCR array was performed to evaluate the expression of essential oxidative stress genes. qPCR, Western blot, and IHC were used to measure the level of EBV reactivation in vitro and in vivo. A Rta-IgG ELISA kit and EBV DNA detection kit were used to analyze the reactivation of EBV in serum from NPC patients. NPC tumor tissue microarrays was used to investigate the prognostic role of oxidative stress and EBV reactivation. Radiation resistance was evaluated by a colony formation assay. Xenografts were treated with NAC, radiation, or a combination of NAC and radiation. EBV DNA load of tumor tissue was evaluated using an EBV DNA detection kit. Oxidative stress, EBV reactivation, and the apoptosis rate in tumor tissues were detected by using 8-OHdG, EAD, and TUNEL assays, respectively. Results: We found that EBV can induce high oxidative stress, which promotes its reactivation and thus leads to radioresistance. Basically, EBV caused NPC cells to undergo a process of 'Redox Resetting' to acquire a new redox status with higher levels of ROS accumulation and stronger antioxidant systems by increasing the expression of the ROS-producing enzyme, NOX2, and the cellular master antioxidant regulator, Nrf2. Also, EBV encoded driving protein LMP1 promotes EBV reactivation through production of ROS. Furthermore, high oxidative stress and EBV reactivation were positively associated with poor overall survival of patients following radiation therapy and were significant related to NPC patients' recurrence and clinical stage. By decreasing oxidative stress using an FDA approved antioxidant drug, NAC, sensitivity of tumors to radiation was increased. Additionally, 8-OHdG and EBV DNA could be dual prognostic markers for NPC patients. Conclusions: Oxidative stress mediates EBV reactivation and leads to radioresistance. Targeting oxidative stress can provide therapeutic benefits to cancer patients with radiation resistance. Clinically, we, for the first time, generated a molecular subtyping model for NPC relying on 8-OHdG and EBV DNA level. These dual markers could identify patients who are at a high risk of poor outcomes but who might benefit from the sequential therapy of reactive oxygen blockade followed by radiation therapy, which provides novel perspectives for the precise treatment of NPC.
Collapse
|
9
|
Zhao W, Xiang Y, Zhang Z, Liu X, Jiang M, Jiang B, Song Y, Hu J. Pharmacological inhibition of GSK3 promotes TNFα-induced GM-CSF via up-regulation of ERK signaling in nasopharyngeal carcinoma (NPC). Int Immunopharmacol 2020; 83:106447. [PMID: 32248019 DOI: 10.1016/j.intimp.2020.106447] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 10/24/2022]
Abstract
Granulocyte-macrophage colony stimulating factor (GM-CSF) functions to drive nasopharyngeal cancer (NPC) metastasis via recruitment and activation of macrophages. However, the source and the regulation of GM-CSF in tumor microenvironment of NPC are not fully understood. In this study, we found that TNFα induced GM-CSF production in NPC CNE1, CNE2, and 5-8F cells in time- and dose-dependent manners. GM-CSF production was tolerant, because the pre-treatment of NPC cells with TNFα down-regulated the GM-CSF production induced by TNFα re-treatment. TNFα activated glycogen synthase kinase-3 (GSK-3), which is an enzyme to regulate glycogen synthesis, and also is a critical downstream element of the PI3K/Akt to regulate cell survival. GSK3 inhibitors up-regulated TNFα-induced GM-CSF, and reversed GM-CSF tolerance induced by TNFα pre-treatment, suggesting that GSK3 activation down-regulated GM-CSF production. GM-CSF down-regulation was not related to ubiquitin-editing enzyme A20. The over-expression of A20 did not regulate GM-CSF production induced by TNFα. However, GSK3 inhibitors up-regulated ERK activation, which contributed to the production of GM-CSF induced by TNFα, suggesting that GSK3 negatively regulated TNFα-induced GM-CSF via down-regulation of ERK signaling. Taking together, these results suggested that GSK3 pathway may be a target for the regulation of TNFα-induced GM-CSF in the tumor microenvironment.
Collapse
Affiliation(s)
- Wang Zhao
- Department of Clinical Laboratory, Changsha Central Hospital, University of South China, Changsha 410004, China; Medical Research Center, Changsha Central Hospital, University of South China, Changsha 410004, China
| | - Yangen Xiang
- Department of Clinical Laboratory, Changsha Central Hospital, University of South China, Changsha 410004, China.
| | - Zhang Zhang
- Department of Pathology, Hunan Cancer Hospital, Affiliated Cancer Hospital of Xiangya Medical School of Central South University, Changsha 410013, China
| | - Xueting Liu
- Medical Research Center, Changsha Central Hospital, University of South China, Changsha 410004, China
| | - Manli Jiang
- Medical Research Center, Changsha Central Hospital, University of South China, Changsha 410004, China
| | - Binyuan Jiang
- Medical Research Center, Changsha Central Hospital, University of South China, Changsha 410004, China
| | - Yinghui Song
- Changsha Cancer Institute, Changsha Central Hospital, University of South China, Changsha 410004, China
| | - Jinyue Hu
- Medical Research Center, Changsha Central Hospital, University of South China, Changsha 410004, China; Changsha Cancer Institute, Changsha Central Hospital, University of South China, Changsha 410004, China.
| |
Collapse
|
10
|
Du Q, Tan Z, Shi F, Tang M, Xie L, Zhao L, Li Y, Hu J, Zhou M, Bode A, Luo X, Cao Y. PGC1α/CEBPB/CPT1A axis promotes radiation resistance of nasopharyngeal carcinoma through activating fatty acid oxidation. Cancer Sci 2019; 110:2050-2062. [PMID: 30945396 PMCID: PMC6550130 DOI: 10.1111/cas.14011] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 12/19/2022] Open
Abstract
The PPAR coactivator-1α (PGC1α) is an important transcriptional co-activator in control of fatty acid metabolism. Mitochondrial fatty acid oxidation (FAO) is the primary pathway for the degradation of fatty acids and promotes NADPH and ATP production. Our previous study demonstrated that upregulation of carnitine palmitoyl transferase 1 A (CPT1A), the key regulator of FAO, promotes radiation resistance of nasopharyngeal carcinoma (NPC). In this study, we found that high expression of PGC1α is associated with poor overall survival in NPC patients after radiation treatment. Targeting PGC1α could sensitize NPC cells to radiotherapy. Mechanically, PGC1α binds to CCAAT/enhancer binding protein β (CEBPB), a member of the transcription factor family of CEBP, to promote CPT1A transcription, resulting in activation of FAO. Our results revealed that the PGC1α/CEBPB/CPT1A/FAO signaling axis promotes radiation resistance of NPC. These findings indicate that the expression of PGC1α could be a prognostic indicator of NPC, and targeting FAO in NPC with high expression of PGC1α might improve the therapeutic efficacy of radiotherapy.
Collapse
Affiliation(s)
- Qianqian Du
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute School of Basic Medicine Science, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Zheqiong Tan
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute School of Basic Medicine Science, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Feng Shi
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute School of Basic Medicine Science, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Min Tang
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute School of Basic Medicine Science, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Longlong Xie
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute School of Basic Medicine Science, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Lin Zhao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute School of Basic Medicine Science, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Yueshuo Li
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute School of Basic Medicine Science, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Jianmin Hu
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute School of Basic Medicine Science, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Min Zhou
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute School of Basic Medicine Science, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China
| | - Ann Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota
| | - Xiangjian Luo
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute School of Basic Medicine Science, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China.,Molecular Imaging Research Center of Central South University, Changsha, China
| | - Ya Cao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.,Cancer Research Institute School of Basic Medicine Science, Xiangya School of Medicine, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis, Chinese Ministry of Health, Changsha, China.,Molecular Imaging Research Center of Central South University, Changsha, China.,Research Center for Technologies of Nucleic Acid-Based Diagnostics and Therapeutics Hunan Province, Changsha, China.,National Joint Engineering Research Center for Genetic Diagnostics of Infectious Diseases and Cancer, Changsha, China
| |
Collapse
|
11
|
Li J, He Y, Tan Z, Lu J, Li L, Song X, Shi F, Xie L, You S, Luo X, Li N, Li Y, Liu X, Tang M, Weng X, Yi W, Fan J, Zhou J, Qiang G, Qiu S, Wu W, Bode AM, Cao Y. Wild-type IDH2 promotes the Warburg effect and tumor growth through HIF1α in lung cancer. Am J Cancer Res 2018; 8:4050-4061. [PMID: 30128035 PMCID: PMC6096397 DOI: 10.7150/thno.21524] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 06/11/2018] [Indexed: 12/19/2022] Open
Abstract
Hotspot mutations of isocitrate dehydrogenase 1 and 2 (IDH1/2) have been studied in several cancers. However, the function of wild-type IDH2 in lung cancer and the mechanism of its contribution to growth of cancer cells remain unknown. Here, we explored the role and mechanism of wild-type IDH2 in promoting growth of lung cancer. Methods: Information regarding genomic and clinical application focusing on IDH2 in cancer was examined in several databases of more than 1,000 tumor samples. IDH2 expression was assessed by immunohistochemistry in tissues from lung cancer patients. The biological functions of IDH2 were evaluated by using cell-based assays and in vivo xenograft mouse models. Results: Here we reported that wild-type IDH2 is up-regulated and is an indicator of poor survival in lung cancer and several other cancers. Targeting IDH2 with shRNA resulted in decreased HIF1α expression, leading to the attenuation of lung cancer cell proliferation and tumor growth. Treatment of lung cancer cells with AGI-6780 (a small molecule inhibitor of IDH2), PX-478 (an inhibitor of HIF1α) or incubation with octyl-α-KG inhibited lung cancer cell proliferation. Conclusion: IDH2 promotes the Warburg effect and lung cancer cell growth, which is mediated through HIF1α activation followed by decreased α-KG. Therefore, IDH2 could possibly serve as a novel therapeutic target for lung cancer.
Collapse
|
12
|
Tan Z, Xiao L, Tang M, Bai F, Li J, Li L, Shi F, Li N, Li Y, Du Q, Lu J, Weng X, Yi W, Zhang H, Fan J, Zhou J, Gao Q, Onuchic JN, Bode AM, Luo X, Cao Y. Targeting CPT1A-mediated fatty acid oxidation sensitizes nasopharyngeal carcinoma to radiation therapy. Am J Cancer Res 2018; 8:2329-2347. [PMID: 29721083 PMCID: PMC5928893 DOI: 10.7150/thno.21451] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 02/28/2018] [Indexed: 12/17/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) has a particularly high prevalence in southern China, southeastern Asia and northern Africa. Radiation resistance remains a serious obstacle to successful treatment in NPC. This study aimed to explore the metabolic feature of radiation-resistant NPC cells and identify new molecular-targeted agents to improve the therapeutic effects of radiotherapy in NPC. Methods: Radiation-responsive and radiation-resistant NPC cells were used as the model system in vitro and in vivo. Metabolomics approach was used to illustrate the global metabolic changes. 13C isotopomer tracing experiment and Seahorse XF analysis were undertaken to determine the activity of fatty acid oxidation (FAO). qRT-PCR was performed to evaluate the expression of essential FAO genes including CPT1A. NPC tumor tissue microarray was used to investigate the prognostic role of CPT1A. Either RNA interference or pharmacological blockade by Etomoxir were used to inhibit CPT1A. Radiation resistance was evaluated by colony formation assay. Mitochondrial membrane potential, apoptosis and neutral lipid content were measured by flow cytometry analysis using JC-1, Annexin V and LipidTOX Red probe respectively. Molecular markers of mitochondrial apoptosis were detected by western blot. Xenografts were treated with Etomoxir, radiation, or a combination of Etomoxir and radiation. Mitochondrial apoptosis and lipid droplets content of tumor tissues were detected by cleaved caspase 9 and Oil Red O staining respectively. Liquid chromatography coupled with tandem mass spectrometry approach was used to identify CPT1A-binding proteins. The interaction of CPT1A and Rab14 were detected by immunoprecipitation, immunofluorescence and in situ proximity ligation analysis. Fragment docking and direct coupling combined computational protein-protein interaction prediction method were used to predict the binding interface. Fatty acid trafficking was measured by pulse-chase assay using BODIPY C16 and MitoTracker Red probe. Results: FAO was active in radiation-resistant NPC cells, and the rate-limiting enzyme of FAO, carnitine palmitoyl transferase 1 A (CPT1A), was consistently up-regulated in these cells. The protein level of CPT1A was significantly associated with poor overall survival of NPC patients following radiotherapy. Inhibition of CPT1A re-sensitized NPC cells to radiation therapy by activating mitochondrial apoptosis both in vitro and in vivo. In addition, we identified Rab14 as a novel CPT1A binding protein. The CPT1A-Rab14 interaction facilitated fatty acid trafficking from lipid droplets to mitochondria, which decreased radiation-induced lipid accumulation and maximized ATP production. Knockdown of Rab14 attenuated CPT1A-mediated fatty acid trafficking and radiation resistance. Conclusion: An active FAO is a vital signature of NPC radiation resistance. Targeting CPT1A could be a beneficial regimen to improve the therapeutic effects of radiotherapy in NPC patients. Importantly, the CPT1A-Rab14 interaction plays roles in CPT1A-mediated radiation resistance by facilitating fatty acid trafficking. This interaction could be an attractive interface for the discovery of novel CPT1A inhibitors.
Collapse
|
13
|
De-Colle C, Menegakis A, Mönnich D, Welz S, Boeke S, Sipos B, Fend F, Mauz PS, Tinhofer I, Budach V, Abu Jawad J, Stuschke M, Balermpas P, Rödel C, Grosu AL, Abdollahi A, Debus J, Belka C, Ganswindt U, Pigorsch S, Combs SE, Lohaus F, Linge A, Krause M, Baumann M, Zips D. SDF-1/CXCR4 expression is an independent negative prognostic biomarker in patients with head and neck cancer after primary radiochemotherapy. Radiother Oncol 2017; 126:125-131. [PMID: 29061496 DOI: 10.1016/j.radonc.2017.10.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 10/04/2017] [Accepted: 10/04/2017] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Preclinical and clinical data suggest that the chemokine pathway governed by SDF-1 and CXCR4 contributes to a resistant phenotype. This retrospective biomarker study aims to explore the specific prognostic value of SDF-1 and CXCR4 expression in locally advanced head and neck squamous cell carcinomas (HNSCC) treated with primary radiochemotherapy (RT-CT). MATERIAL AND METHODS Biopsies from 141 HNSCC tumours of the oral cavity, oropharynx and hypopharynx were evaluated for SDF-1 and CXCR4 expression by immunofluorescence. SDF-1 and CXCR4 expression was correlated with clinico-pathological characteristics and outcome after RT-CT. RESULTS Patients with tumours exhibiting overexpression of intracellular SDF-1 and CXCR4 have a higher risk for loco-regional relapse and a worse overall survival after RT-CT (multivariate analysis, hazard ratio 2.33, CI [1.18-4.62], p = 0.02 and hazard ratio 2.02, CI [1.13-3.59], p = 0.02, respectively). Similar results were observed when only the subgroup of HPV DNA negative patients were analysed (hazard ratio 2.23 and 2.16, p = 0.02 and p = 0.01, respectively). CONCLUSIONS Our data support the importance of SDF-1 and CXCR4 expression for loco-regional control and overall survival in HNSCC after primary radiochemotherapy. Prospective multivariate validation and further studies into CXCR4 inhibition to overcome radiation resistance are warranted.
Collapse
Affiliation(s)
- Chiara De-Colle
- Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany.
| | - Apostolos Menegakis
- Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany
| | - David Mönnich
- Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany; German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Tübingen, Germany
| | - Stefan Welz
- Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany
| | - Simon Boeke
- Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany
| | - Bence Sipos
- Department of Pathology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany
| | - Falko Fend
- Department of Pathology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany
| | - Paul-Stefan Mauz
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany
| | - Inge Tinhofer
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Berlin, Germany; Department of Radiooncology and Radiotherapy, Charité University Hospital, Berlin, Germany
| | - Volker Budach
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Berlin, Germany; Department of Radiooncology and Radiotherapy, Charité University Hospital, Berlin, Germany
| | - Jehad Abu Jawad
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Essen, Germany; Department of Radiotherapy, Medical Faculty, University of Duisburg-Essen, Germany
| | - Martin Stuschke
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Essen, Germany; Department of Radiotherapy, Medical Faculty, University of Duisburg-Essen, Germany
| | - Panagiotis Balermpas
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Frankfurt, Germany; Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Germany
| | - Claus Rödel
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Frankfurt, Germany; Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Germany
| | - Anca-Ligia Grosu
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Freiburg, Germany; Department of Radiation Oncology, University of Freiburg, Germany
| | - Amir Abdollahi
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), University of Heidelberg Medical School and German Cancer Research Center (DKFZ), Germany; Heidelberg Ion Therapy Center (HIT), Department of Radiation Oncology, University of Heidelberg Medical School, Germany; National Center for Tumor Diseases (NCT), University of Heidelberg Medical School and German Cancer Research Center (DKFZ), Germany; Translational Radiation Oncology, University of Heidelberg Medical School and German Cancer Research Center (DKFZ), Germany
| | - Jürgen Debus
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Heidelberg, Germany; Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), University of Heidelberg Medical School and German Cancer Research Center (DKFZ), Germany; Heidelberg Ion Therapy Center (HIT), Department of Radiation Oncology, University of Heidelberg Medical School, Germany; National Center for Tumor Diseases (NCT), University of Heidelberg Medical School and German Cancer Research Center (DKFZ), Germany; Clinical Cooperation Unit Radiation Oncology, University of Heidelberg Medical School and German Cancer Research Center (DKFZ), Germany
| | - Claus Belka
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Munich, Germany; Department of Radiotherapy and Radiation Oncology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Ute Ganswindt
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Munich, Germany; Department of Radiotherapy and Radiation Oncology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Steffi Pigorsch
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Munich, Germany; Department of Radiation Oncology, Technische Universität München, Germany.; Institute for Innovative radiation therapy in Helmholtz-Zentrum München, Germany
| | - Stephanie E Combs
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Munich, Germany; Department of Radiation Oncology, Technische Universität München, Germany.; Institute for Innovative radiation therapy in Helmholtz-Zentrum München, Germany
| | - Fabian Lohaus
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Germany
| | - Annett Linge
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; National Center for Tumor Diseases (NCT), partner site Dresden, Germany
| | - Mechthild Krause
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; National Center for Tumor Diseases (NCT), partner site Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Germany
| | - Michael Baumann
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Dresden, Germany; Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden - Rossendorf, Germany; National Center for Tumor Diseases (NCT), partner site Dresden, Germany; Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Zips
- Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Germany; German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK) partner site Tübingen, Germany
| |
Collapse
|
14
|
De-Colle C, Mönnich D, Welz S, Boeke S, Sipos B, Fend F, Mauz PS, Tinhofer I, Budach V, Jawad JA, Stuschke M, Balermpas P, Rödel C, Grosu AL, Abdollahi A, Debus J, Bayer C, Belka C, Pigorsch S, Combs SE, Lohaus F, Linge A, Krause M, Baumann M, Zips D, Menegakis A. SDF-1/CXCR4 expression in head and neck cancer and outcome after postoperative radiochemotherapy. Clin Transl Radiat Oncol 2017; 5:28-36. [PMID: 29594214 PMCID: PMC5833920 DOI: 10.1016/j.ctro.2017.06.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/06/2017] [Accepted: 06/10/2017] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Outcome after postoperative radiochemotherapy (RT-CT) for patients with advanced head and neck squamous cell carcinomas (HNSCC) remains unsatisfactory, especially among those with HPV negative tumours. Therefore, new biomarkers are needed to further define subgroups for individualised therapeutic approaches. Preclinical and first clinical observations showed that the chemokine receptor CXCR4 and its ligand SDF-1 (CXCL12) play an important role in tumour cell proliferation, survival, cancer progression, metastasis and treatment resistance. However, the data on the prognostic value of SDF-1/CXCR4 expression for HNSCC are conflicting. The aim of our hypothesis-generating study was to retrospectively explore the prognostic potential of SDF-1/CXCR4 in a well-defined cohort of HNSCC patients collected within the multicenter biomarker study of the German Cancer Consortium Radiation Oncology Group (DKTK-ROG). MATERIAL AND METHODS Patients with stage III and IVA HNSCC of the oral cavity, oropharynx and hypopharynx were treated with resection and adjuvant radiotherapy (RT) with ≥60 Gy and concurrent cisplatin-based chemotherapy (CT). Tissue micro-arrays (TMAs) from a total of 221 patients were generated from surgical specimens, 201 evaluated for the SDF-1 and CXCR4 expression by immunofluorescence and correlated with clinico-pathological and outcome data. RESULTS In univariate and multivariate analyses intracellular SDF-1 expression was associated with lower loco-regional control (LRC) in the entire patient group as well as in the HPV16 DNA negative subgroup. CXCR4 expression showed a trend for lower LRC in the univariate analysis which was not confirmed in the multivariate analysis. Neither for SDF-1 nor CXCR4 expression associations with distant metastasis free or overall survival were found. CONCLUSIONS Our exploratory data support the hypothesis that overexpression of intracellular SDF-1 is an independent negative prognostic biomarker for LRC after postoperative RT-CT in high-risk HNSCC. Prospective validation is warranted and further exploration of SDF-1/CXCR4 as a potential therapeutic target to overcome treatment resistance in HNSCC appears promising.
Collapse
Affiliation(s)
- Chiara De-Colle
- Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
| | - David Mönnich
- Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Tübingen, Germany
| | - Stefan Welz
- Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Simon Boeke
- Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Bence Sipos
- Department of Pathology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Falko Fend
- Department of Pathology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Paul-Stefan Mauz
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Inge Tinhofer
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Berlin, Germany
- Department of Radiooncology and Radiotherapy, Charité University Hospital, Berlin, Germany
| | - Volker Budach
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Berlin, Germany
- Department of Radiooncology and Radiotherapy, Charité University Hospital, Berlin, Germany
| | - Jehad Abu Jawad
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Essen, Germany
- Department of Radiotherapy, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Martin Stuschke
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Essen, Germany
- Department of Radiotherapy, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Panagiotis Balermpas
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Frankfurt, Germany
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Germany
| | - Claus Rödel
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Frankfurt, Germany
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Germany
| | - Anca-Ligia Grosu
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Freiburg, Germany
- Department of Radiation Oncology, University of Freiburg, Germany
| | - Amir Abdollahi
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), University of Heidelberg Medical School and German Cancer Research Center (DKFZ), Germany
- Heidelberg Ion Therapy Center (HIT), Department of Radiation Oncology, University of Heidelberg Medical School, Germany
- National Center for Tumor Diseases (NCT), University of Heidelberg Medical School and German Cancer Research Center (DKFZ), Germany
- Translational Radiation Oncology, University of Heidelberg Medical School and German Cancer Research Center (DKFZ), Germany
| | - Jürgen Debus
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Heidelberg, Germany
- Heidelberg Institute of Radiation Oncology (HIRO), National Center for Radiation Research in Oncology (NCRO), University of Heidelberg Medical School and German Cancer Research Center (DKFZ), Germany
- Heidelberg Ion Therapy Center (HIT), Department of Radiation Oncology, University of Heidelberg Medical School, Germany
- National Center for Tumor Diseases (NCT), University of Heidelberg Medical School and German Cancer Research Center (DKFZ), Germany
- Clinical Cooperation Unit Radiation Oncology, University of Heidelberg Medical School and German Cancer Research Center (DKFZ), Germany
| | - Christine Bayer
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Munich, Germany
| | - Claus Belka
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Munich, Germany
- Department of Radiotherapy and Radiation Oncology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Steffi Pigorsch
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Munich, Germany
- Department of Radiation Oncology, Technische Universität München, Institute for Innovative Radiation Therapy in Helmholtz-Zentrum München, Germany
| | - Stephanie E. Combs
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Munich, Germany
- Department of Radiation Oncology, Technische Universität München, Institute for Innovative Radiation Therapy in Helmholtz-Zentrum München, Germany
| | - Fabian Lohaus
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Annett Linge
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
| | - Mechthild Krause
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology – OncoRay, Germany
| | - Michael Baumann
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology – OncoRay, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Zips
- Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
- German Cancer Research Center (DKFZ), Heidelberg and German Cancer Consortium (DKTK), Tübingen, Germany
| | - Apostolos Menegakis
- Radiation Oncology, Medical Faculty and University Hospital, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
15
|
CXCR4 knockdown inhibits the growth and invasion of nasopharyngeal cancer stem cells. Oncol Lett 2017; 13:2253-2259. [PMID: 28454388 PMCID: PMC5403420 DOI: 10.3892/ol.2017.5694] [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: 08/27/2015] [Accepted: 11/28/2016] [Indexed: 12/23/2022] Open
Abstract
Nasopharyngeal cancer is a type of malignant tumor with a high rate of incidence. Cancer stem cells are regarded as one of the main causes for the formation and recurrence of nasopharyngeal cancer. CXC chemokine receptor type 4 (CXCR4) has been reported to perform an important role in cancer; however, the association between CXCR4 and nasopharyngeal cancer stem cells remains unclear. The present study explored the effect of CXCR4 on cellular viability, apoptosis and invasion of nasopharyngeal cancer stem cells. Results of the present study demonstrated that knockdown of CXCR4 inhibited the viability and invasion of nasopharyngeal cancer stem cells and promoted cellular apoptosis. Further studies have revealed that the anti-tumor effect of CXCR4 knockdown was associated with the inhibition of the protein kinase B signal. These results demonstrate that the knockdown of CXCR4 resulted in an anti-tumor effect in nasopharyngeal cancer stem cells. Therefore, CXCR4 may become a promising therapeutic target in the treatment of nasopharyngeal cancer.
Collapse
|
16
|
Mishan MA, Ahmadiankia N, Bahrami AR. CXCR4 and CCR7: Two eligible targets in targeted cancer therapy. Cell Biol Int 2016; 40:955-67. [PMID: 27248053 DOI: 10.1002/cbin.10631] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 05/26/2016] [Indexed: 12/12/2022]
Abstract
Cancer is one of the most common cause of death in the world with high negative emotional, economic, and social impacts. Conventional therapeutic methods, including chemotherapy and radiotherapy, have not proven satisfactory and relapse is common in most cases. Recent studies have focused on targeted therapy with more precise identification and targeted attacks to the cancer cells. For this purpose, chemokine receptors are proper targets and among them, CXCR4 and CCR7, with a crucial role in cancer metastasis, are being considered as desired candidates for investigation. In this review paper, the most important experimental results are highlighted on the potential targeted therapies based on CXCR4 and CCR7 chemokine receptors.
Collapse
Affiliation(s)
| | - Naghmeh Ahmadiankia
- Cancer Prevention Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | | |
Collapse
|
17
|
Tao H, Wei Y, Wang C, Yang K, Huang W, Liu H, Li B. Expression of chemokine receptor CXCR4 is closely correlated with clinical outcome in human nasopharyngeal carcinoma. Tumour Biol 2015; 37:6099-105. [PMID: 26611644 DOI: 10.1007/s13277-015-4464-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 11/17/2015] [Indexed: 12/26/2022] Open
Abstract
The CXC chemokine receptor 4 (CXCR4) has been reported to be involved in the development and progression of nasopharyngeal carcinoma (NPC). However, the role of CXCR4 in clinical outcome and prognosis of NPC patients remains controversial. In the present study, we investigated and reviewed the expression of CXCR4 in NPC tissues and then analyzed the definitive role of CXCR4 in clinical outcome and prognosis. Here, we found that the expression level of CXCR4 was significantly higher in NPC cancer specimens (61/98) than that in paired non-tumor tissues (p < 0.001). Together with our pathological analysis, statistic analysis revealed that CXCR4 expression was indeed closely correlated with UICC stage (p = 0.000), N stage (p = 0.019), and metastasis (p = 0.000). Most importantly, the systematic review combined with our survival and multivariate analysis that revealed high expression of CXCR4 was obviously associated with poor overall survival (OS) (p = 0.000) and progression-free survival (PFS) (p = 0.000) and can act as an independent prognostic factor in NPC patients. In conclusion, this study suggests that CXCR4 is highly activated and expressed in the development of NPC and may be recommended as an indicator in the diagnosis of NPC. Thus, targeting of CXCR4 gene or protein could be used as a potential therapy for NPC.
Collapse
Affiliation(s)
- Hengmin Tao
- Department of 6th Radiation Oncology, Shandong Cancer Hospital and Institute, No.440, Jiyan Road, Huaiyin District, 250117, Jinan, Shandong, China
- School of Medicine and Life Sciences, University of Jinan, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yumei Wei
- Department of 6th Radiation Oncology, Shandong Cancer Hospital and Institute, No.440, Jiyan Road, Huaiyin District, 250117, Jinan, Shandong, China
| | - Congan Wang
- Traditional Chinese Medicine (TCM) Orthopeadics Department, Affiliated Hospital of Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Kun Yang
- Accounting Office of Pharmacy Department, The Fourth People Hospital of Jinan, Jinan, Shandong, China
| | - Wei Huang
- Department of 6th Radiation Oncology, Shandong Cancer Hospital and Institute, No.440, Jiyan Road, Huaiyin District, 250117, Jinan, Shandong, China
| | - Haitao Liu
- Department of Radiation Oncology, Peking University 3rd Hospital, Beijing, 100191, China
| | - Baosheng Li
- Department of 6th Radiation Oncology, Shandong Cancer Hospital and Institute, No.440, Jiyan Road, Huaiyin District, 250117, Jinan, Shandong, China.
| |
Collapse
|
18
|
Goto M, Yoshida T, Yamamoto Y, Furukita Y, Inoue S, Fujiwara S, Kawakita N, Nishino T, Minato T, Yuasa Y, Yamai H, Takechi H, Seike J, Bando Y, Tangoku A. CXCR4 Expression is Associated with Poor Prognosis in Patients with Esophageal Squamous Cell Carcinoma. Ann Surg Oncol 2015; 24:832-840. [PMID: 26577115 DOI: 10.1245/s10434-015-4974-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Indexed: 01/29/2023]
Abstract
BACKGROUND Chemokines and their receptors are known to play important roles in the tumorigenesis of many malignancies. The chemokine CXCL12 and its receptors CXCR4 and CXCR7 were suggested to be involved in cancer invasion and metastasis. The aim of this retrospective study was to evaluate the prognostic impact of the expressions of CXCL12, CXCR4 and CXCR7 in patients with esophageal squamous cell carcinoma (ESCC). METHODS We used immunohistochemistry (IHC) and reverse transcriptase-polymerase chain reaction (RT-PCR) to evaluate the expressions of CXCL12, CXCR4, and CXCR7 in ESCC patients' tumor biopsy specimens obtained during preoperative endoscopy or surgery. These results were compared with the patients' clinicopathological parameters and survival. RESULTS IHC was conducted for 172 patients. High expression of CXCR4 in the cytoplasm and nuclei and that of CXCR7 were associated with poor cause-specific survival (CSS) (P= .002 and .010, respectively). The specimens from 52 of the 172 patients were examined by RT-PCR and quantitative real-time PCR. The expression levels of messenger RNA (mRNA) of CXCR4 and CXCR7 were significantly increased in the tumors compared with normal esophageal mucosae (P < .0001). The expression level of mRNA of CXCR4 was associated with poor recurrence-free survival and CSS (P = .012 and .038, respectively). CONCLUSIONS CXCR4 expression is associated with poor prognosis in patients with ESCC.
Collapse
Affiliation(s)
- Masakazu Goto
- Department of Thoracic, Endocrine Surgery and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Takahiro Yoshida
- Department of Thoracic, Endocrine Surgery and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Yota Yamamoto
- Sainokuni Higashiomiya Medical Center, Saitama, Japan
| | | | - Seiya Inoue
- Department of Thoracic, Endocrine Surgery and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | | | - Naoya Kawakita
- Department of Thoracic, Endocrine Surgery and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Takeshi Nishino
- Department of Thoracic, Endocrine Surgery and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Takuya Minato
- Shikoku Medical Center for Children and Adults, Kagawa, Japan
| | | | | | - Hirokazu Takechi
- Department of Thoracic, Endocrine Surgery and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Junichi Seike
- Department of Thoracic, Endocrine Surgery and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Yoshimi Bando
- Department of Molecular and Environmental Pathology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Akira Tangoku
- Department of Thoracic, Endocrine Surgery and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
19
|
Novel roles and therapeutic targets of Epstein-Barr virus-encoded latent membrane protein 1-induced oncogenesis in nasopharyngeal carcinoma. Expert Rev Mol Med 2015; 17:e15. [PMID: 26282825 DOI: 10.1017/erm.2015.13] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Epstein-Barr virus (EBV) was first discovered 50 years ago as an oncogenic gamma-1 herpesvirus and infects more than 90% of the worldwide adult population. Nasopharyngeal carcinoma (NPC) poses a serious health problem in southern China and is one of the most common cancers among the Chinese. There is now strong evidence supporting a role for EBV in the pathogenesis of NPC. Latent membrane protein 1 (LMP1), a primary oncoprotein encoded by EBV, alters several functional and oncogenic properties, including transformation, cell death and survival in epithelial cells in NPC. LMP1 may increase protein modification, such as phosphorylation, and initiate aberrant signalling via derailed activation of host adaptor molecules and transcription factors. Here, we summarise the novel features of different domains of LMP1 and several new LMP1-mediated signalling pathways in NPC. When then focus on the potential roles of LMP1 in cancer stem cells, metabolism reprogramming, epigenetic modifications and therapy strategies in NPC.
Collapse
|
20
|
Sun Y, Zhu D, Wang G, Wang D, Zhou H, Liu X, Jiang M, Liao L, Zhou Z, Hu J. Pro-Inflammatory Cytokine IL-1β Up-Regulates CXC Chemokine Receptor 4 via Notch and ERK Signaling Pathways in Tongue Squamous Cell Carcinoma. PLoS One 2015; 10:e0132677. [PMID: 26176534 PMCID: PMC4503771 DOI: 10.1371/journal.pone.0132677] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 06/18/2015] [Indexed: 12/12/2022] Open
Abstract
Chronic inflammation contributes to tumor development through the induction of oncogenic mutations, genomic instability, early tumor promotion, and enhanced angiogenesis. Here, we report that IL-1 receptor 1 (IL-1R1) was expressed in 40 of 41 human tongue squamous cell carcinomas (TSCC). IL-1β up-regulated the expression of CXCR4, a CXC chemokine receptor that mediates cancer growth and metastasis, at both mRNA and protein levels in Tca8113 TSCC cells. IL-1β treatment of Tca8113 cells promoted migration in response to CXCR4 ligand stromal-derived factor α (SDF-1α). The inhibition of IL-1R1 by its antagonist IL-1Ra or RNA interference significantly reversed the up-regulation of CXCR4 induced by IL-1β. IL-1R1 activation also up-regulated the expression of IL-1β itself, suggesting a positive feedback regulation of CXCR4 expression. Furthermore, IL-1β induced the activation of Notch, which was originally considered a stem cell regulator. Pharmacological inhibition of Notch signaling reversed the up-regulation of CXCR4 induced by IL-1β, suggesting that Notch signaling may be involved in the growth and metastasis of cancers via up-regulation of CXCR4. In addition, IL-1β induced the activation of extracellular signal regulated kinase (ERK) and ERK inhibition decreased the up-regulation of CXCR4 induced by IL-1β, suggesting the involvement of ERK signaling in cancer metastasis. Taken together these data suggest that IL-1β and IL-1R1 promote cancer growth and metastasis by up-regulating CXCR4 expression and that CXCR4 may be a link between inflammation and cancer.
Collapse
Affiliation(s)
- Yi Sun
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
- Medical Research Center, Changsha Central Hospital, Changsha, China
| | - Demao Zhu
- Department of Pathology, Changsha Central Hospital, Changsha, China
| | - Guihua Wang
- Department of Oncology, Changsha Central Hospital, Changsha, China
| | - Di Wang
- Department of Oncology, Changsha Central Hospital, Changsha, China
| | - Huashan Zhou
- Department of Pathology, Changsha Central Hospital, Changsha, China
| | - Xueting Liu
- Medical Research Center, Changsha Central Hospital, Changsha, China
| | - Manli Jiang
- Medical Research Center, Changsha Central Hospital, Changsha, China
| | - Lingjuan Liao
- Medical Research Center, Changsha Central Hospital, Changsha, China
| | - Zhiguang Zhou
- Institute of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
- * E-mail: (ZZ); (JH)
| | - Jinyue Hu
- Medical Research Center, Changsha Central Hospital, Changsha, China
- * E-mail: (ZZ); (JH)
| |
Collapse
|
21
|
Lei KF, Huang CH. Paper-based microreactor integrating cell culture and subsequent immunoassay for the investigation of cellular phosphorylation. ACS APPLIED MATERIALS & INTERFACES 2014; 6:22423-22429. [PMID: 25421089 DOI: 10.1021/am506388q] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Investigation of cellular phosphorylation and signaling pathway has recently gained much attention for the study of pathogenesis of cancer. Related conventional bioanalytical operations for this study including cell culture and Western blotting are time-consuming and labor-intensive. In this work, a paper-based microreactor has been developed to integrate cell culture and subsequent immunoassay on a single paper. The paper-based microreactor was a filter paper with an array of circular zones for running multiple cell cultures and subsequent immunoassays. Cancer cells were directly seeded in the circular zones without hydrogel encapsulation and cultured for 1 day. Subsequently, protein expressions including structural, functional, and phosphorylated proteins of the cells could be detected by their specific antibodies, respectively. Study of the activation level of phosphorylated Stat3 of liver cancer cells stimulated by IL-6 cytokine was demonstrated by the paper-based microreactor. This technique can highly reduce tedious bioanalytical operation and sample and reagent consumption. Also, the time required by the entire process can be shortened. This work provides a simple and rapid screening tool for the investigation of cellular phosphorylation and signaling pathway for understanding the pathogenesis of cancer. In addition, the operation of the paper-based microreactor is compatible to the molecular biological training, and therefore, it has the potential to be developed for routine protocol for various research areas in conventional bioanalytical laboratories.
Collapse
Affiliation(s)
- Kin Fong Lei
- Graduate Institute of Medical Mechatronics, Chang Gung University , Kwei-Shan, Tao-Yuan 333, Taiwan
| | | |
Collapse
|
22
|
Targeting Epstein-Barr virus oncoprotein LMP1-mediated glycolysis sensitizes nasopharyngeal carcinoma to radiation therapy. Oncogene 2014; 33:4568-78. [PMID: 24662831 PMCID: PMC4162460 DOI: 10.1038/onc.2014.32] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 11/26/2013] [Accepted: 12/16/2013] [Indexed: 12/13/2022]
Abstract
Our goal in this work was to illustrate the Epstein-Barr virus (EBV)-modulated global biochemical profile and provide a novel metabolism-related target to improve the therapeutic regimen of nasopharyngeal carcinoma (NPC). We used a metabolomics approach to investigate EBV-modulated metabolic changes, and found that the exogenous overexpression of the EBV-encoded latent membrane protein 1 (LMP1) significantly increased glycolysis. The deregulation of several glycolytic genes, including hexokinase 2 (HK2), was determined to be responsible for the reprogramming of LMP1-mediated glucose metabolism in NPC cells. The upregulation of HK2 elevated aerobic glycolysis and facilitated proliferation by blocking apoptosis. More importantly, HK2 was positively correlated with LMP1 in NPC biopsies, and high HK2 levels were significantly associated with poor overall survival of NPC patients following radiation therapy. Knockdown of HK2 effectively enhanced the sensitivity of LMP1-overexpressing NPC cells to irradiation. Finally, c-Myc was demonstrated to be required for LMP1-induced upregulation of HK2. The LMP1-mediated attenuation of the PI3-K/Akt-GSK3beta-FBW7 signaling axis resulted in the stabilization of c-Myc. These findings indicate a close relationship between EBV and glycolysis in NPC. Notably, LMP1 is the key regulator of the reprogramming of EBV-mediated glycolysis in NPC cells. Given the importance of EBV-mediated deregulation of glycolysis, anti-glycolytic therapy might represent a worthwhile avenue of exploration in the treatment of EBV-related cancers.
Collapse
|
23
|
Serum CXCL9 levels are associated with tumor progression and treatment outcome in patients with nasopharyngeal carcinoma. PLoS One 2013; 8:e80052. [PMID: 24278236 PMCID: PMC3836991 DOI: 10.1371/journal.pone.0080052] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 09/29/2013] [Indexed: 01/11/2023] Open
Abstract
Objectives The aim of this cohort study was to examine the role of the chemokine (C-X-C motif) ligand 9 (CXCL9) on nasopharyngeal carcinoma (NPC). Materials & Methods Sera from 205 NPC patients and 231 healthy individuals, and 86 NPC tumor samples were enrolled. CXCL9 expression in tissue samples was analyzed by quantitative real-time PCR and immunohistochemistry. CXCL9 serum concentrations were measured by enzyme-linked immunosorbent assay. Results CXCL9 expression was significantly higher in tumors than in normal epithelium. CXCL9 serum concentrations were also significantly higher in NPC patients compared to those in healthy individuals (516.8±617.6 vs. 170.7±375.0 pg/mL, P<0.0001). Serum CXCL9 levels were significantly higher in NPC patients with higher tumor stages, nodal stages, and overall stages (P<0.001, P = 0.001, and P<0.001, respectively). We found a statistically significant correlation between the concentrations of CXCL9 and EBV DNA load in the NPC patients (Spearman’s correlation analysis; r = 0.473, P<0.001; 95% confidence interval, 0.346–0.582). Moreover, NPC patients with higher CXCL9 levels (>290 pg/mL, median) before treatment had worse prognoses for overall survival and disease-free survival (P = 0.045 and P = 0.008, respectively). Multivariate logistic regression analyses also indicated that higher CXCL9 serum levels were an independent prognostic factor for disease-free survival (P = 0.015). Conclusion Our study demonstrated that CXCL9 is associated with tumor burden and aggressiveness of NPC tumors and the serum level of this ligand may be useful as a prognostic indicator.
Collapse
|
24
|
Lu J, Luo H, Liu X, Peng Y, Zhang B, Wang L, Xu X, Peng X, Li G, Tian W, He ML, Kung H, Li XP. miR-9 targets CXCR4 and functions as a potential tumor suppressor in nasopharyngeal carcinoma. Carcinogenesis 2013; 35:554-63. [PMID: 24170200 DOI: 10.1093/carcin/bgt354] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
MicroRNA deregulation and pathway alterations have been implicated in nasopharyngeal carcinoma (NPC), a highly invasive and metastatic cancer widely prevalent in Southern China. In this study, we report that miR-9 is commonly downregulated in NPC specimens and NPC cell lines with important functional consequences. The reduced expression of miR-9 was inversely correlated with clinical stages and marked the progression from locoregional to metastatic tumors. The CpG island hypermethylation contributed to miR-9 silencing in NPC cell lines and tissues. Ectopic expression of miR-9 dramatically inhibited the proliferative, migratory and invasive capacities of NPC cells in vitro and in vivo. We found that miR-9 strongly reduced the expression of CXCR4 in NPC cells. Luciferase assay demonstrated that miR-9 could directly bind to the 3' untranslated region of CXCR4. Similar to the restoring miR-9 expression, CXCR4 downregulation inhibited cell growth, migration and invasion, whereas CXCR4 overexpression rescued the suppressive effect of miR-9. Mechanistic investigations revealed that CXCR4 functionally mediated the SDF-1-stimulated activation of p38 mitogen-activated protein kinase pathway in NPC cells with miR-9 downregulation or CXCR4 overexpression. In clinical specimens, CXCR4 and phospho-p38 were widely overexpressed, and the levels increased with the progression from locoregional to metastatic tumors in NPC tissues. The levels of CXCR4 were inversely correlated with miR-9 or phospho-p38 expression. Taken together, our results indicate that miR-9 functions as a tumor-suppressive microRNA in NPC, and that its suppressive effects are mediated chiefly by repressing CXCR4 expression.
Collapse
Affiliation(s)
- Juan Lu
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Luo DH, Chen QY, Liu H, Xu LH, Zhang HZ, Zhang L, Tang LQ, Mo HY, Huang PY, Guo X, Mai HQ. The independent, unfavorable prognostic factors endothelin A receptor and chemokine receptor 4 have a close relationship in promoting the motility of nasopharyngeal carcinoma cells via the activation of AKT and MAPK pathways. J Transl Med 2013; 11:203. [PMID: 23987636 PMCID: PMC3765987 DOI: 10.1186/1479-5876-11-203] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2012] [Accepted: 08/23/2013] [Indexed: 01/21/2023] Open
Abstract
Background Recent studies have indicated that the expression of endothelin A receptor (ETAR) and chemokine receptor 4 (CXCR4) could be used as an indicator of the metastatic potential of nasopharyngeal carcinoma (NPC). The aim of this study was to determine the prognostic value of ETAR and CXCR4 in NPC patients and to reveal the interplay of the endothelin-1 (ET-1)/ETAR and stromal-derived factor-1(SDF-1)/CXCR4 pathways in promoting NPC cell motility. Methods Survival analysis was used to analyze the prognostic value of ETAR and CXCR4 expression in 153 cases of NPC. Chemotaxis assays were used to evaluate alterations in the migration ability of non-metastatic 6-10B and metastatic 5-8F NPC cells. Real-time PCR, immunoblotting, and flow cytometric analyses were used to evaluate changes in the expression levels of CXCR4 mRNA and protein induced by ET-1. Results The expression levels of ETAR and CXCR4 were closely related to each other and both correlated with a poor prognosis. A multivariate analysis showed that the expression levels of both ETAR and CXCR4 were independent prognostic factors for overall survival (OS), progression-free survival (PFS), and distant metastasis-free survival (DMFS). The migration of 6-10B and 5-8F cells was elevated by ET-1 in combination with SDF-1α. The knockdown of ETAR protein expression by siRNA reduced CXCR4 protein expression in addition to ETAR protein expression, leading to a decrease in the metastatic potential of the 5-8F cells. ET-1 induced CXCR4 mRNA and protein expression in the 6-10B NPC cells in a time- and concentration-dependent fashion and was inhibited by an ETAR antagonist and PI3K/AKT/mTOR and MAPK/ERK1/2 pathway inhibitors. Conclusions ETAR and CXCR4 expression levels are potential prognostic biomarkers in NPC patients. ETAR activation partially promoted NPC cell migration via a mechanism that enhanced functional CXCR4 expression.
Collapse
Affiliation(s)
- Dong-Hua Luo
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong 510060, P, R, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Song Q, Wang G, Chu Y, Zhou L, Jiang M, He Q, Liu M, Qin J, Hu J. TNF-α up-regulates cellular inhibitor of apoptosis protein 2 (c-IAP2) via c-Jun N-terminal kinase (JNK) pathway in nasopharyngeal carcinoma. Int Immunopharmacol 2013; 16:148-53. [PMID: 23597429 DOI: 10.1016/j.intimp.2013.03.034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 03/27/2013] [Accepted: 03/27/2013] [Indexed: 01/19/2023]
Abstract
Inhibitor of apoptosis proteins (IAPs) contribute to both tumor progression and tumor metastasis. Here, we show that pro-inflammatory cytokine TNF-α induced the up-regulation of c-IAP2 in the potential metastatic nasopharyngeal carcinoma (NPC) cells in a dose- and time-dependent manner. This up-regulation is tolerant, as the pre-treatment of NPC cells with TNF-α reversed the up-regulation of c-IAP2 induced by TNF-α re-stimulation. TNF-α activated MAKP signals, including ERK, JNK and p38, and NF-κB signal, but only inhibition of JNK signal transduction reversed the induction of c-IAP2, suggesting that JNK signaling contributed to the c-IAP2 induction. The results from in vitro scratch wound-healing assays showed that TNF-α promoted cell invasion, which was reversed by the inhibition of JNK signaling. Taken together, these studies suggested that pro-inflammation cytokine TNF-α may be a promoter for NPC metastasis, and the anti-inflammatory therapy may be of benefit to the prevention of NPC metastasis.
Collapse
Affiliation(s)
- Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Xu J, Deng X, Tang M, Li L, Xiao L, Yang L, Zhong J, Bode AM, Dong Z, Tao Y, Cao Y. Tyrosylprotein sulfotransferase-1 and tyrosine sulfation of chemokine receptor 4 are induced by Epstein-Barr virus encoded latent membrane protein 1 and associated with the metastatic potential of human nasopharyngeal carcinoma. PLoS One 2013; 8:e56114. [PMID: 23472069 PMCID: PMC3589389 DOI: 10.1371/journal.pone.0056114] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 01/08/2013] [Indexed: 12/24/2022] Open
Abstract
The latent membrane protein 1 (LMP1), which is encoded by the Epstein-Barr virus (EBV), is an important oncogenic protein that is closely related to carcinogenesis and metastasis of nasopharyngeal carcinoma (NPC), a prevalent cancer in China. We previously reported that the expression of the functional chemokine receptor CXCR4 is associated with human NPC metastasis. In this study, we show that LMP1 induces tyrosine sulfation of CXCR4 through tyrosylprotein sulfotransferase-1 (TPST-1), an enzyme that is responsible for catalysis of tyrosine sulfation in vivo, which is likely to contribute to the highly metastatic character of NPC. LMP1 could induce tyrosine sulfation of CXCR4 and its associated cell motility and invasiveness in a NPC cell culture model. In contrast, the expression of TPST-1 small interfering RNA reversed LMP1-induced tyrosine sulfation of CXCR4. LMP1 conveys signals through the epidermal growth factor receptor (EGFR) pathway, and EGFR-targeted siRNA inhibited the induction of TPST-1 by LMP1. We used a ChIP assay to show that EGFR could bind to the TPST-1 promoter in vivo under the control of LMP1. A reporter gene assay indicated that the activity of the TPST-1 promoter could be suppressed by deleting the binding site between EGFR and TPST-1. Finally, in human NPC tissues, the expression of TPST-1 and LMP1 was directly correlated and clinically, the expression of TPST-1 was associated with metastasis. These results suggest the up-regulation of TPST-1 and tyrosine sulfation of CXCR4 by LMP1 might be a potential mechanism contributing to NPC metastasis.
Collapse
Affiliation(s)
- Juan Xu
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis, Ministry of Heath, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xiyun Deng
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis, Ministry of Heath, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Min Tang
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis, Ministry of Heath, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Lili Li
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Lanbo Xiao
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis, Ministry of Heath, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Lifang Yang
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis, Ministry of Heath, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Molecular Imaging Center, The First Affiliated Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Juanfang Zhong
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis, Ministry of Heath, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ann M. Bode
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| | - Zigang Dong
- The Hormel Institute, University of Minnesota, Austin, Minnesota, United States of America
| | - Yongguang Tao
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis, Ministry of Heath, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- * E-mail: (YT); (YC)
| | - Ya Cao
- Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Carcinogenesis, Ministry of Heath, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Molecular Imaging Center, The First Affiliated Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- * E-mail: (YT); (YC)
| |
Collapse
|
28
|
Lo KW, Chung GTY, To KF. Acquired Genetic and Epigenetic Alterations in Nasopharyngeal Carcinoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013. [DOI: 10.1007/978-1-4614-5947-7_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
29
|
ZHU ZQ, NING ZL, SHAO F, HU B, LIANG W. Expression of CXCR4 in gastric cancer and its relationship with patient prognosis. ACTA ACUST UNITED AC 2012. [DOI: 10.3724/sp.j.1008.2012.00043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
30
|
Wang JP, Hu WM, Wang KS, Luo BH, Wu C, Chen ZH, Luo GQ, Liu YW, Liu QL, Yu J, Li JH, Wen JF. Upregulation of C-X-C chemokine receptor type 1 expression is associated with late-stage gastric adenocarcinoma. Exp Ther Med 2012; 4:55-60. [PMID: 23060922 DOI: 10.3892/etm.2012.568] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 04/30/2012] [Indexed: 12/31/2022] Open
Abstract
Chemokine receptors play multiple roles in the development and progression of various tumor types. The aim of this study was to examine C-X-C chemokine receptor type 1 (CXCR1) protein expression in gastric adenocarcinoma and to investigate the clinical implications of CXCR1 upregulation. Expression of CXCR1 protein in 83 specimens of sporadic gastric adenocarcinoma and their corresponding non-neoplastic mucosa obtained by gastrectomy was assayed using immunohistochemistry. The intensity of immunostaining in tumor tissue was considered strong when tumor tissue staining was more intense than in the corresponding non-neoplastic mucosa; the intensity was null when staining was weaker in the tumor than in the corresponding non-neoplastic mucosa; and the intensity was weak when staining was similar in both tissues. Microvascular density in tumor tissue and its corresponding non-neoplastic mucosa was measured using monoclonal antibody against CD34. A strong correlation was observed between elevated CXCR1 protein expression and tumor stage (P<0.05). T stage, N stage and overall stage positively correlated with CXCR1 protein expression. Microvascular density was higher in tumors with strong CXCR1 protein expression, but the correlation with CXCR1 was not linear (P=0.07). Multiple logistic regression analyses showed that, compared to no or weak expression, overexpression of CXCR1 protein was a significant risk factor for high N stage (N2, N3). These results indicate that CXCR1 may be considered as a new and promising target for gastric adenocarcinoma therapy.
Collapse
Affiliation(s)
- Jun Pu Wang
- Department of Pathology, School of Basic Medicine
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Xiao Z, Yang M, Lv Q, Wang W, Deng M, Liu X, He Q, Chen X, Chen M, Fang L, Xie X, Hu J. P2Y11 impairs cell proliferation by induction of cell cycle arrest and sensitizes endothelial cells to cisplatin-induced cell death. J Cell Biochem 2011; 112:2257-65. [PMID: 21503959 DOI: 10.1002/jcb.23144] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Extracellular ATP mediates a wide range of physiological effects, including cell proliferation, differentiation, maturation, and migration. However, the effect of ATP on cell proliferation has been contradictory, and the mechanism is not fully understood. In the current study, we found that extracellular ATP significantly inhibited the proliferation of human umbilical vein endothelial cells (HUVECs) and human aortic endothelial cells (HAECs). Treatment with ATP did not induce cell apoptosis but instead induced cell cycle arrest in S phase. ATP induced the phosphorylation of ERK1/2, but the ERK inhibitors, U0126 and PD9809, did not regulate the inhibition of cell proliferation induced by ATP. However, ATP-induced inhibition of cell proliferation was blocked by suramin, a nonspecific antagonist of the P2Y receptors, and endothelial cells expressed P2Y11, a P2Y receptor that specifically binds ATP. Moreover, the down-regulation of P2Y11 by RNA interference not only reversed the inhibition of cell proliferation but also ameliorated cell cycle arrest in S phase. In addition, P2Y11 sensitized endothelial cells to cisplatin-induced cell death by down-regulation of the expression of Bcl-2. Taken together, these results suggest that extracellular ATP impairs cell proliferation by triggering signaling to induce cell cycle arrest and sensitizes cell to death via P2Y11 in endothelial cells.
Collapse
Affiliation(s)
- Zhilin Xiao
- Department of Geriatric Cardiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ping YF, Bian XW. Consice review: Contribution of cancer stem cells to neovascularization. Stem Cells 2011; 29:888-94. [PMID: 21557392 DOI: 10.1002/stem.650] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cancer stem cells (CSCs), a special subpopulation of tumor cells, are considered as tumor initiating cells. More recently, these cells have also been identified as initiators of tumor neovascularization. A better understanding of the contribution of CSCs to neovascularization should elucidate the mechanisms of cancer initiation and progression as well as establish new concepts for cancer diagnosis and treatment. In this review, we discuss the evidence for the roles of CSCs in tumor vascularization, including production of proangiogenic factors, transdifferentiation into vascular mural cells such as endothelial and smooth muscle-like cells, and formation of nonendothelium-lined vasculogenic mimicry. In addition, the potential therapeutic significance of targeting CSCs is envisaged.
Collapse
Affiliation(s)
- Yi-Fang Ping
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | |
Collapse
|
33
|
CXCR4 in Cancer and Its Regulation by PPARgamma. PPAR Res 2011; 2008:769413. [PMID: 18779872 PMCID: PMC2528256 DOI: 10.1155/2008/769413] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2008] [Revised: 06/25/2008] [Accepted: 07/10/2008] [Indexed: 12/20/2022] Open
Abstract
Chemokines are peptide mediators involved in normal development,
hematopoietic and immune regulation, wound healing, and
inflammation. Among the chemokines is CXCL12, which binds
principally to its receptor CXCR4 and regulates leukocyte
precursor homing to bone marrow and other sites. This role of
CXCL12/CXCR4 is “commandeered” by cancer cells to facilitate the
spread of CXCR4-bearing tumor cells to tissues with high CXCL12
concentrations. High CXCR4 expression by cancer cells predisposes
to aggressive spread and metastasis and ultimately to poor patient
outcomes. As well as being useful as a marker for disease
progression, CXCR4 is a potential target for anticancer therapies.
It is possible to interfere directly with the CXCL12:CXCR4 axis
using peptide or small-molecular-weight antagonists. A further
opportunity is offered by promoting strategies that downregulate
CXCR4 pathways: CXCR4 expression in the tumor microenvironment is
modulated by factors such as hypoxia, nucleosides, and
eicosanoids. Another promising approach is through targeting PPAR
to suppress CXCR4 expression. Endogenous PPARγ such as 15-deoxy-Δ12,14-PGJ2 and synthetic agonists such as the
thiazolidinediones both cause downregulation of CXCR4 mRNA and
receptor. Adjuvant therapy using PPARγ agonists may, by
stimulating PPARγ-dependent downregulation of CXCR4 on cancer cells, slow the rate of metastasis and impact beneficially on
disease progression.
Collapse
|
34
|
CXCR4 Overexpression Is Associated with Poor Outcome in Females Diagnosed with Stage IV Non-small Cell Lung Cancer. J Thorac Oncol 2011; 6:1169-78. [DOI: 10.1097/jto.0b013e3182199a99] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
35
|
Zhao BC, Wang ZJ, Mao WZ, Ma HC, Han JG, Zhao B, Xu HM. CXCR4/SDF-1 axis is involved in lymph node metastasis of gastric carcinoma. World J Gastroenterol 2011; 17:2389-96. [PMID: 21633638 PMCID: PMC3103791 DOI: 10.3748/wjg.v17.i19.2389] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Revised: 11/13/2010] [Accepted: 11/20/2010] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of CXC chemokine receptor-4 (CXCR4) and stromal cell-derived factor-1 (SDF-1) in lymph node metastasis of gastric carcinoma.
METHODS: In 40 cases of gastric cancer, expression of CXCR4 mRNA in cancer and normal mucous membrane and SDF-1 mRNA in lymph nodes around the stomach was detected using quantitative polymerase chain reaction (PCR) (TaqMan) and immunohistochemistric assay. SGC-7901 and MGC80-3 cancer cells were used to investigate the effect of SDF-1 on cell proliferation and migration.
RESULTS: Quantitative reverse transcription PCR and immunohistochemistry revealed that the expression level of CXCR4 in gastric cancer was significantly higher than that in normal mucous membrane (1.6244 ± 1.3801 vs 1.0715 ± 0.5243, P < 0.05). The expression level of CXCR4 mRNA in gastric cancer with lymph node metastasis was also significantly higher than that without lymph node metastasis (0.823 ± 0.551 vs 0.392 ± 0.338, P < 0.05). CXCR4 expression was significantly related to poorly differentiated, high tumor stage and lymph node metastasis. Significant differences in the expression level of SDF-1 mRNA were found between lymph nodes in metastatic gastric cancer and normal nodes (0.5432 ± 0.4907 vs 0.2640 ± 0.2601, P < 0.05). The positive expression of SDF-1 mRNA in lymph nodes of metastatic gastric cancer was consistent with the positive expression of CXCR4 mRNA in gastric cancer (r = 0.776, P < 0.01). Additionally, human gastric cancer cell lines expressed CXCR4 and showed vigorous proliferation and migratory responses to SDF-1. AMD3100 (a specific CXCR4 antagonist) was also found to effectively reduce the migration of gastric cancer cells.
CONCLUSION: The CXCR4/SDF-1 axis is involved in the lymph node metastasis of gastric cancer. CXCR4 is considered as a potential therapeutic target in the treatment of gastric cancer.
Collapse
|
36
|
Yeh KY, Chang JWC, Li YY, Wang CH, Wang HM. Tumor growth inhibition of metastatic nasopharyngeal carcinoma cell lines by low dose of arsenic trioxide via alteration of cell cycle progression and induction of apoptosis. Head Neck 2010; 33:734-42. [PMID: 20737493 DOI: 10.1002/hed.21535] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2010] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Although arsenic trioxide (ATO) has displayed anticancer activity against primary nasopharyngeal carcinoma (NPC), its efficacy in metastatic NPC deserved further investigation because the biological/therapeutic difference in cancer cells probably exists between primary and distant sites. METHODS Two human metastatic NPC cell lines (NPC-BM1 and NPC-BM2) were investigated. We measured cellular proliferation, cell cycle, and apoptotic extent of BM1 and BM2 cells treated with ATO in vitro. Furthermore, we evaluated the tumor growth after ATO treatment in vivo. RESULTS Low-dose ATO treatment is sufficient to induce an antiproliferative effect, alter the cell cycle, and increase apoptosis in BM1 and BM2 cells. BM1 tumor growth in a xenograft model with low-dose and short-schedule (1 mg/kg/day, intraperitoneal injection for 5 consecutive days) of ATO treatment significantly slowed in vivo. CONCLUSION ATO at low dose seems to be an encouraging schedule for palliative treatment of metastatic NPC.
Collapse
Affiliation(s)
- Kun-Yun Yeh
- Division of Hemato-oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung & Chang Gung University, College of Medicine, Taiwan, Providence of China.
| | | | | | | | | |
Collapse
|
37
|
Nijmeijer S, Leurs R, Smit MJ, Vischer HF. The Epstein-Barr virus-encoded G protein-coupled receptor BILF1 hetero-oligomerizes with human CXCR4, scavenges Gαi proteins, and constitutively impairs CXCR4 functioning. J Biol Chem 2010; 285:29632-41. [PMID: 20622011 DOI: 10.1074/jbc.m110.115618] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cells express distinct G protein-coupled receptor (GPCR) subtypes on their surface, allowing them to react to a corresponding variety of extracellular stimuli. Cross-regulation between different ligand-GPCR pairs is essential to generate appropriate physiological responses. GPCRs can physically affect each other's functioning by forming heteromeric complexes, whereas cross-regulation between activated GPCRs also occurs through integration of shared intracellular signaling networks. Human herpesviruses utilize virally encoded GPCRs to hijack cellular signaling networks for their own benefit. Previously, we demonstrated that the Epstein-Barr virus-encoded GPCR BILF1 forms heterodimeric complexes with human chemokine receptors. Using a combination of bimolecular complementation and bioluminescence resonance energy transfer approaches, we now show the formation of hetero-oligomeric complexes between this viral GPCR and human CXCR4. BILF1 impaired CXCL12 binding to CXCR4 and, consequently, also CXCL12-induced signaling. In contrast, the G protein uncoupled mutant BILF1-K(3.50)A affected CXCL12-induced CXCR4 signaling to a much lesser extent, indicating that BILF1-mediated CXCR4 inhibition is a consequence of its constitutive activity. Co-expression of Gα(i1) with BILF1 and CXCR4 restored CXCL12-induced signaling. Likewise, BILF1 formed heteromers with the human histamine H(4) receptor (H(4)R). BILF1 inhibited histamine-induced Gα(i)-mediated signaling by H(4)R, however, without affecting histamine binding to this receptor. These data indicate that functional cross-regulation of Gα(i)-coupled GPCRs by BILF1 is at the level of G proteins, even though these GPCRs are assembled in hetero-oligomeric complexes.
Collapse
Affiliation(s)
- Saskia Nijmeijer
- Leiden/Amsterdam Center for Drug Research (LACDR), Division of Medicinal Chemistry, Faculty of Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
38
|
Liu Y, Ji R, Li J, Gu Q, Zhao X, Sun T, Wang J, Li J, Du Q, Sun B. Correlation effect of EGFR and CXCR4 and CCR7 chemokine receptors in predicting breast cancer metastasis and prognosis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2010; 29:16. [PMID: 20181250 PMCID: PMC2845107 DOI: 10.1186/1756-9966-29-16] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Accepted: 02/24/2010] [Indexed: 12/16/2022]
Abstract
BACKGROUND The chemokine receptors CXCR4 and CCR7 play an important role in cancer invasion and metastasis. This study investigated the expression of CXCR4, CCR7, CXCL12, CCL21, and EGFR to illustrate the role of these biomarkers in breast cancer metastasis and prognosis. METHODS The CXCR4, CCR7, CXCL12, CCL21, and EGFR biomarkers were analyzed along with ER, PR, and HER-2/neu in breast cancer tissue microarray (TMA) specimens, including 200 primary breast cancer specimens by immunohistochemistry. Corresponding lymph nodes from the same patients were also examined using the same method. RESULTS Together with their CXCL12 and CCL21 ligands, CXCR4 and CCR7 were significantly highly expressed in tumor cells with lymph node (LN) metastasis. Similarly, EGFR was expressed highly in tumors with LN metastasis. The ligands were especially expressed in metastatic tumors than in primary tumors from the same patients. Moreover, the expression of both CXCR4 accompanied by CCR7 and CXCL12 accompanied by CCL21 were up-regulated. Kaplan-Meier survival analysis revealed that patients exhibiting high CXCR4, CCR7, and EGFR expression experienced a shorter survival period compared with those with low expression. CONCLUSIONS The expression of CXCR4, CCR7, and EGFR may be associated with LN metastasis. Moreover, the expression of these receptors can serve as an indicator of undesirable prognosis in patients with breast cancer.
Collapse
Affiliation(s)
- Yixin Liu
- Department of Pathology, Tianjin Medical University, Tianjin, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Deng L, Chen N, Li Y, Zheng H, Lei Q. CXCR6/CXCL16 functions as a regulator in metastasis and progression of cancer. Biochim Biophys Acta Rev Cancer 2010; 1806:42-9. [PMID: 20122997 DOI: 10.1016/j.bbcan.2010.01.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2009] [Revised: 01/12/2010] [Accepted: 01/24/2010] [Indexed: 02/05/2023]
Abstract
Metastasis is considered the obvious mark for most aggressive cancers. However, little is known about the molecular mechanism of the regulation of cancer metastasis. Recent evidence increasingly suggests that the interaction between chemokines and chemokine receptors is pivotal in the process of metastasis. The chemokine receptor CXCR4 and its ligand CXCL12, for example, have been reported to play a vital role in cancer metastasis. Another chemokine and chemokine receptor pair, the CXCL16/CXCR6 axis, has been studied by several independent research groups. Here, we summarize recent advances in our knowledge of the function of CXC chemokine receptor CXCR6 and its ligand CXCL16 in regulating metastasis and invasion of cancer. CXCR6 and CXCL16 are up-regulated in multiple cancer tissue types and cancer cell lines relative to normal tissues and cell lines. In addition, both CXCR6 and CXCL16 levels increase as tumor malignancy increases. Trans-membranous CXCL16 chemokine reduces proliferation while soluble CXCL16 chemokine enhances proliferation and migration. TM-CXCL16 functions as an inducer for lymphocyte build-up around tumor sites. High trans-membranous CXCL16 expression correlates with a good prognosis. Moreover, the Akt/mTOR signal pathway is involved in activating the CXCR6/CXCL16 axis. These findings suggest multiple opportunities for blocking the CXCR6/CXCL16 axis and the Akt/mTOR signal pathway in novel cancer therapies.
Collapse
Affiliation(s)
- Ling Deng
- Department of Radiation Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | | | | | | | | |
Collapse
|
40
|
Characterization of a new small cell lung cancer (SCLC) cell line STP54 derived from a metastatic bioptate of a combined type of SCLC with Non-SCLC component. Folia Histochem Cytobiol 2009; 47:111-5. [PMID: 19419948 DOI: 10.2478/v10042-009-0018-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Small cell lung cancer constitutes 15-20% cases of lung cancers, currently the leading cause of death from malignant diseases. It also causes the demise of >90% of affected individuals in 5 years. We have established a new SCLC cell line STP54 derived from fine needle aspirate of metastatic supraclavicular lymph node of 54 -year-old women for model experiments. The primary tumor was diagnosed by histopathological examination as combined type of small cell lung cancer with a non-small cell component. We cultured the cancer cells in the RPMI 1640 medium. In the long-term culture only the small cell component survived. The cell line was established after 30 passages and then characterized by performing cell morphology, cell growth analysis, tumorigenicity in vitro and flow cytometry analysis of selected markers (like NCAM, cytokeratines, HLA-ABC, Fas, Bcl-2, p53, CXCR4, CD210). The cells were growing in floating aggregates and show features suggesting its invasiveness. We suggest that this new cell line may serve as a valuable tool for further studies on lung tumor biology, molecular pathogenesis and metastatic mechanism.
Collapse
|
41
|
Xiang ZL, Zeng ZC, Tang ZY, Fan J, Zhuang PY, Liang Y, Tan YS, He J. Chemokine receptor CXCR4 expression in hepatocellular carcinoma patients increases the risk of bone metastases and poor survival. BMC Cancer 2009; 9:176. [PMID: 19508713 PMCID: PMC2704220 DOI: 10.1186/1471-2407-9-176] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Accepted: 06/09/2009] [Indexed: 12/21/2022] Open
Abstract
Background The chemokine and bone marrow-homing receptor CXCR4 is implicated in metastases of various cancers. This study was conducted to analyze the association of CXCR4 expression with hepatocellular carcinoma (HCC) bone metastasis and patient survival. Methods Tumor tissue from HCC patients with (n = 43) and without (n = 138) bone metastasis was subjected to immunohistochemical staining for CXCR4 using tissue microarrays. Immunoreactivity was evaluated semi-quantitatively. A receiver-operating characteristic-based approach and logistical regression analysis were used to determine the predictive value of clinicopathologic factors, including CXCR4 expression, in bone metastasis. Patient survival was analyzed by Kaplan-Meier curves and log-rank tests. Results CXCR4 overexpression was detected in 34 of 43 (79.1%) patients with bone metastases and in 57 of 138 (41.3%) without bone metastases. CXCR4 expression correlated with (correlation coefficient: 0.551, P < 0.001) and was predictive of HCC bone metastases (AUC: 0.689; 95%CI: 0.601 – 0.776; P < 0.001). CXCR4 staining intensity correlated with the bone metastasis-free survival (correlation coefficient: -0.359; P = 0.018). CXCR4 overexpression in primary tumors (n = 91) decreased overall median survival (18.0 months vs. 36.0 months, P <0.001). Multivariable analysis identified CXCR4 as a strong, independent risk factor for reduced disease-free survival (relative risk [RR]: 5.440; P = 0.023) and overall survival (RR: 7.082; P = 0.001). Conclusion CXCR4 expression in primary HCCs may be an independent risk factor for bone metastasis and may be associated with poor clinical outcome.
Collapse
Affiliation(s)
- Zuo-Lin Xiang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
The CXCR4/SDF-1 chemokine receptor axis: a new target therapeutic for non-small cell lung cancer. J Thorac Oncol 2009; 3:1379-83. [PMID: 19057260 DOI: 10.1097/jto.0b013e31818dda9d] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Chemokines are proinflammatory chemoattractant cytokines that regulate cell trafficking and adhesion. The CXCR4 chemokine receptor and its ligand, stromal cell derived factor (SDF-1), constitute a chemokine/receptor axis that has attracted great interest because of an increasing understanding of its role in cancer, including lung cancer. The CXCR4/SDF-1 complex activates several pathways that mediate chemotaxis, migration and secretion of angiopoietic factors. Neutralization of SDF-1 by anti-SDF-1 or anti-CXCR4 monoclonal antibody in preclinical in vivo studies results in a significant decrease of non-small cell lung cancer metastases. Since anti-SDF-1/CXCR4 strategies have already been developed for use in combating human immunodeficiency virus infections, it is likely that these approaches will be used in clinical trials in non-small cell lung cancer in the very near future.
Collapse
|
43
|
Sirera G, Videla S, Romeu J, Cañadas M, Fernández MT, Balo S, Cirauqui B, Darwich L, Rey-Joly C, Clotet B. Fatal fast-evolution of nasopharyngeal squamous cell carcinoma in an HIV patient with EBV and HPV (-16 AND -33) in blood serum. Open AIDS J 2008; 2:1-2. [PMID: 18923693 PMCID: PMC2556196 DOI: 10.2174/1874613600802010001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Revised: 01/14/2008] [Accepted: 01/22/2008] [Indexed: 11/22/2022] Open
Abstract
Our case illustrates the first report of an HIV-infected patient with a nasopharyngeal squamous cell carcinoma with viremia by one Epstein-Barr virus (EBV) and seropositivity by two high risk oncogenic human papilloma viruses (HPV)-types (HPV-16 and HPV-33), previous to his death. This patient presented a fatal fast-evolution.
Collapse
Affiliation(s)
- Guillem Sirera
- HIV Clinical Unit, Department of Medicine, Lluita Contra La SIDA Foundation, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
Targeting novel pathways associated with tumor angiogenesis, invasion and immunity, may lead to improvement in patient outcomes for renal cell carcinoma. Chemokines potentiate tumor growth, metastasis, angiogenesis and immune evasion through interactions with stromal cells and neoplastic cells. Further understanding of the mechanisms involved in chemokine-mediated angiogenesis and metastasis may lead to improved therapeutic strategies in this disease. Interactions between chemokine expression and signaling, and the VEGF and hypoxia-inducible factor pathways offer important opportunities to intervene in the process of renal cell carcinoma proliferation, angiogenesis and invasion. Modulation of the CXCR3/CXCR3-ligand or the CXCR4/CXCL12 biologic axis may be potential therapeutic targets for the treatment of renal cell carcinoma. Furthermore, combination treatment with agents targeting chemokine signaling with therapies directed at angiogenesis and tumor immunity may lead to improved outcomes in this disease.
Collapse
Affiliation(s)
- Karen L Reckamp
- Assistant Professor of Medicine, Divisions of Medical Oncology and Therapeutics Research & Hematology and Hematopoeitic Transplantaion, City of Hope and Beckman Research Institute, 1500 E Duarte Road, MOB 1001, Duarte, CA 91010, USA, Tel.: +1 626 256 4673; +1 626 256 3155, Fax: +1 626 930 5461,
| | - Robert M Strieter
- Henry B Mulholland Professor of Internal Medicine, Chairman, Department of Internal Medicine, University of Virginia School of Medicine, PO Box 800466, Charlottesville, VA 22908–0466, USA, Tel.: +1 434 982 6999, Fax: +1 434 243 0399,
| | - Robert A Figlin
- Arthur and Rosalie Kaplan Professor of Medical Oncology, Chair, Division of Medical Oncology & Therapeutics Research, City of Hope and Beckman Research Institute, Associate Director for Clinical Research, City of Hope Comprehensive Cancer Center, 1500 E Duarte Road, Duarte, CA 91010–3000, Tel.: +1 626 471 9290, Fax: +1 626 930 5461,
| |
Collapse
|
45
|
Huang J, Chen K, Gong W, Dunlop NM, Wang JM. G-protein coupled chemoattractant receptors and cancer. FRONT BIOSCI-LANDMRK 2008; 13:3352-63. [PMID: 18508437 DOI: 10.2741/2930] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chemoattractant receptors are a group of seven transmembrane, G protein coupled receptors (GPCRs). They were initially identified mainly on leukocytes to mediate cell migration in response to pathogen or host-derived chemotactic factors. During the past decade, chemoattractant GPCRs have been discovered not only to mediate leukocyte chemotaxis thus promoting innate and adaptive host immune responses, but also to play essential roles in development, homeostasis, HIV infection, angiogenesis and wound healing. A growing body of evidence further indicates that chemoattractant GPCRs contribute to tumor growth, invasion, angiogenesis/angiostasis and metastasis. The diverse properties of GPCRs in the progression of malignant tumors have attracted intense interest in their potential as novel anti-tumor pharmacological targets.
Collapse
Affiliation(s)
- Jian Huang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD 21702, USA
| | | | | | | | | |
Collapse
|
46
|
Endothelial cells enhance tumor cell invasion through a crosstalk mediated by CXC chemokine signaling. Neoplasia 2008; 10:131-9. [PMID: 18283335 DOI: 10.1593/neo.07815] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2007] [Revised: 11/21/2007] [Accepted: 11/26/2007] [Indexed: 12/30/2022] Open
Abstract
Field cancerization involves the lateral spread of premalignant or malignant disease and contributes to the recurrence of head and neck tumors. The overall hypothesis underlying this work is that endothelial cells actively participate in tumor cell invasion by secreting chemokines and creating a chemotactic gradient for tumor cells. Here we demonstrate that conditioned medium from head and neck tumor cells enhance Bcl-2 expression in neovascular endothelial cells. Oral squamous cell carcinoma-3 (OSCC3) and Kaposi's sarcoma (SLK) show enhanced invasiveness when cocultured with pools of human dermal microvascular endothelial cells stably expressing Bcl-2 (HDMEC-Bcl-2), compared to cocultures with empty vector controls (HDMEC-LXSN). Xenografted OSCC3 tumors vascularized with HDMEC-Bcl-2 presented higher local invasion than OSCC3 tumors vascularized with control HDMEC-LXSN. CXCL1 and CXCL8 were upregulated in primary endothelial cells exposed to vascular endothelial growth factor (VEGF), as well as in HDMEC-Bcl-2. Notably, blockade of CXCR2 signaling, but not CXCR1, inhibited OSCC3 and SLK invasion toward endothelial cells. These data demonstrate that CXC chemokines secreted by endothelial cells induce tumor cell invasion and suggest that the process of lateral spread of tumor cells observed in field cancerization is guided by chemotactic signals that originated from endothelial cells.
Collapse
|
47
|
Abstract
Chemokines are a large group of low molecular weight cytokines that are known to selectively attract and activate different cell types. Although the primary function of chemokines is well recognized as leukocyte attractants, recent evidences indicate that they also play a role in number of tumor-related processes, such as growth, angiogenesis and metastasis. Chemokines activate cells through cell surface seven trans-membranes, G-protein-coupled receptors (GPCR). The role played by chemokines and their receptors in tumor pathophysiology is complex as some chemokines favor tumor growth and metastasis, while others may enhance anti-tumor immunity. These diverse functions of chemokines establish them as key mediators between the tumor cells and their microenvironment and play critical role in tumor progression and metastasis. In this review, we present some of the recent advances in chemokine research with special emphasis on its role in tumor angiogenesis and metastasis.
Collapse
|
48
|
Wen DS, Zhu XL, Guan SM, Wu YM, Yu LL, Wu JZ. Silencing of CXCR4 inhibits the proliferation, adhesion, chemotaxis and invasion of salivary gland mucoepidermoid carcinoma Mc3 cells in vitro. Oral Oncol 2007; 44:545-54. [PMID: 17936060 DOI: 10.1016/j.oraloncology.2007.06.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Revised: 06/28/2007] [Accepted: 06/30/2007] [Indexed: 10/22/2022]
Abstract
Mucoepidermoid carcinoma is the most common malignant tumor in salivary glands and high-grade mucoepidermoid carcinoma is often accompanied with poor prognosis. Many recent research works demonstrated that stromal cell-derived factor-1 (SDF-1) and its receptor CXC chemokine receptor-4 (CXCR4) interaction was critical for metastasis of various cancers. In this study, the immunoexpression of CXCR4 in human salivary gland mucoepidermoid carcinoma in different grades was detected by immunohistochemical analysis and the expression of CXCR4 and its ligand SDF-1 in mucoepidermoid carcinoma MEC-1 cell line and its highly metastatic clone Mc3 was examined by RT-PCR, flow cytometry and immunocytochemical analysis. It was found that CXCR4 was over-expressed in Mc3 cell line and SDF-1 was expressed in both cell lines at a nearly equal level. We further constructed CXCR4-shRNA expression vector to stably transfect Mc3 cells. We found that silencing of endogenous CXCR4 gene expression in Mc3 cells resulted in inhibition of the proliferation, adhesion, chemotaxis and invasion of Mc3 cells in vitro. This study implies that CXCR4 molecule is a potential factor controlling the proliferation and metastasis of Mc3 cells.
Collapse
Affiliation(s)
- De-Sheng Wen
- Department of Oral Biology, School of Stomatology, Fourth Military Medical University, 145 Chang Le Xi Road, Xi'an, PR China
| | | | | | | | | | | |
Collapse
|
49
|
Uchida D, Onoue T, Tomizuka Y, Begum NM, Miwa Y, Yoshida H, Sato M. Involvement of an Autocrine Stromal Cell–Derived Factor-1/CXCR4 System on the Distant Metastasis of Human Oral Squamous Cell Carcinoma. Mol Cancer Res 2007; 5:685-94. [PMID: 17634424 DOI: 10.1158/1541-7786.mcr-06-0368] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We have previously shown that a stromal cell-derived factor-1 (SDF-1; CXCL12)/CXCR4 system is involved in the establishment of lymph node metastasis, but not in that of distant metastasis, in oral squamous cell carcinoma (SCC). In this study, we investigated the role of the autocrine SDF-1/CXCR4 system, with a focus on distant metastasis in oral SCC cells. The immunohistochemical staining of SDF-1 and CXCR4 using primary oral SCCs and metastatic lymph nodes showed a significantly higher number of SDF-1-positive cases among the metastatic lymph nodes than among the primary oral SCCs, which was associated with a poor survival rate among those of the former group. The forced expression of SDF-1 in B88 cells, which exhibit functional CXCR4 and lymph node metastatic potential (i.e., the autocrine SDF-1/CXCR4 system), conferred enhanced cell motility and anchorage-independent growth potential onto the cells. Orthotopic inoculation of the transfectant into nude mice was associated with an increase in the number of metastatic lymph nodes and more aggressive metastatic foci in the lymph nodes. Furthermore, the SDF-1 transfectant (i.e., the autocrine SDF-1/CXCR4 system) exhibited dramatic metastasis to the lung after i.v. inoculation, whereas the mock transfectant (i.e., the paracrine SDF-1/CXCR4 system) did not. Under the present conditions, AMD3100, a CXCR4 antagonist, significantly inhibited the lung metastasis of the SDF-1 transfectant, ameliorated body weight loss, and improved the survival rate of tumor-bearing nude mice. These results suggested that, in cases of oral SCC, the paracrine SDF-1/CXCR4 system potentiates lymph node metastasis, but distant metastasis might require the autocrine SDF-1/CXCR4 system.
Collapse
Affiliation(s)
- Daisuke Uchida
- Second Department of Oral and Maxillofacial Surgery, Tokushima University School of Dentistry, 3-18-15 Kuramoto, Tokushima 770-8504, Japan.
| | | | | | | | | | | | | |
Collapse
|
50
|
Ruffini PA, Morandi P, Cabioglu N, Altundag K, Cristofanilli M. Manipulating the chemokine-chemokine receptor network to treat cancer. Cancer 2007; 109:2392-404. [PMID: 17503430 DOI: 10.1002/cncr.22706] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Chemokines are chemoattractant cytokines that regulate the trafficking and activation of leukocytes and other cell types under a variety of inflammatory and noninflammatory conditions. Over the past few years, studies have increasingly shown that chemokines play an important role in several aspects of tumor progression. Tumor cells express functional chemokine receptors, which can sustain proliferation, angiogenesis, and survival and promote organ-specific localization of distant metastases. Chemokine expression in human malignancies is associated with a leukocyte infiltration favoring the establishment of immune escape mechanisms. A literature review of relevant publications on preclinical testing of cancer therapies based on interference with the cancer chemokine network was performed. The feasibility, potential advantages, and limitations of the clinical translation of the results of such studies in treatment of different tumor types and settings are discussed. The chemokine network is a key player in the establishment of metastases. In the preclinical setting, blocking agents and antibodies directed against CXCR4 prevent metastasis of different cancers. In mouse models, overexpression of selected chemokines causes tumor infiltration by distinct leukocyte subsets, resulting in tumor regression and tumor-specific immunity generation. Researchers have also successfully used chemokines as carriers and/or adjuvants for cancer vaccines. The cancer chemokine network is a multifaceted therapeutic target.
Collapse
Affiliation(s)
- Pier A Ruffini
- Divisione Oncologia Medica Falck, Ospedale Niguarda Ca' Granda, Milano, Italy.
| | | | | | | | | |
Collapse
|