1
|
Tojo S, Nakashiro K, Kuribayashi N, Uchida D. Serum CXCL13 as a Novel Biomarker in Oral Squamous Cell Carcinoma. Cancer Med 2024; 13:e70263. [PMID: 39344390 PMCID: PMC11440027 DOI: 10.1002/cam4.70263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Despite its low sensitivity (approximately 30%), squamous cell carcinoma (SCC) antigen is commonly utilized as a serum tumor marker for oral SCC (OSCC) in clinical settings. The objective of this research was to identify novel biomarkers for OSCC. METHODS Initially, we performed microarray analysis to evaluate the gene expression signatures of primary OSCC and normal oral mucosal tissues. Our findings showed the C-X-C motif chemokine ligand 13 (CXCL13) to be a promising novel biomarker as it was consistently overexpressed in primary OSCC tissues, a conclusion corroborated by polymerase chain reaction results. Subsequently, we measured serum CXCL13 levels in 125 patients with OSCC using a sandwich enzyme-linked immunosorbent assay and compared the results with those of 29 healthy individuals. RESULTS Remarkably, the levels of serum CXCL13 were consistently elevated in patients with OSCC, and the high expression of serum CXCL13 was notably associated with tumor size and neck lymph node metastasis. Patients with advanced OSCC with high-serum CXCL13 levels exhibited poor prognosis regarding both overall and disease-free survival. Finally, spatial transcriptome analysis revealed CXCL13 and CD8 expressions within tumor area clusters but not in adjacent normal areas, suggesting specific overexpression of CXCL13 in primary OSCC tissues. CONCLUSION These findings imply that serum CXCL13 holds diagnostic and prognostic value, showing promise as a novel biomarker for OSCC.
Collapse
Affiliation(s)
- Shin Tojo
- Department of Oral and Maxillofacial SurgeryEhime University Graduate School of MedicineToonJapan
| | - Koh‐ichi Nakashiro
- Department of Oral and Maxillofacial SurgeryEhime University Graduate School of MedicineToonJapan
| | - Nobuyuki Kuribayashi
- Department of Oral and Maxillofacial SurgeryEhime University Graduate School of MedicineToonJapan
| | - Daisuke Uchida
- Department of Oral and Maxillofacial SurgeryEhime University Graduate School of MedicineToonJapan
| |
Collapse
|
2
|
Rajendran P. Unveiling the power of flavonoids: A dynamic exploration of their impact on cancer through matrix metalloproteinases regulation. Biomedicine (Taipei) 2024; 14:12-28. [PMID: 38939095 PMCID: PMC11204124 DOI: 10.37796/2211-8039.1447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/15/2023] [Accepted: 12/22/2023] [Indexed: 06/29/2024] Open
Abstract
Cancer stands as a significant contributor to global mortality rates, primarily driven by its progression and widespread dissemination. Despite notable strides in cancer therapy, the efficacy of current treatment strategies is compromised due to their inherent toxicity and the emergence of chemoresistance. Consequently, there is a critical need to evaluate alternative therapeutic approaches, with natural compounds emerging as promising candidates, showcasing demonstrated anticancer capabilities in various research models. This review manuscript presents a comprehensive examination of the regulatory mechanisms governing the expression of matrix metalloproteinases (MMPs) and delves into the potential therapeutic role of flavonoids as agents exhibiting specific anticancer activity against MMPs. The primary aim of this study is to elucidate the diverse functions associated with MMP production in cancer and to investigate the potential of flavonoids in modulating MMP expression to inhibit metastasis.
Collapse
Affiliation(s)
- Peramaiyan Rajendran
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa, 31982, Saudi Arabia
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, Tamil Nadu, India
| |
Collapse
|
3
|
Ding Y, Chu L, Cao Q, Lei H, Li X, Zhuang Q. A meta-validated immune infiltration-related gene model predicts prognosis and immunotherapy sensitivity in HNSCC. BMC Cancer 2023; 23:45. [PMID: 36639648 PMCID: PMC9837972 DOI: 10.1186/s12885-023-10532-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 01/09/2023] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Tumor microenvironment (TME) is of great importance to regulate the initiation and advance of cancer. The immune infiltration patterns of TME have been considered to impact the prognosis and immunotherapy sensitivity in Head and Neck squamous cell carcinoma (HNSCC). Whereas, specific molecular targets and cell components involved in the HNSCC tumor microenvironment remain a twilight zone. METHODS Immune scores of TCGA-HNSCC patients were calculated via ESTIMATE algorithm, followed by weighted gene co-expression network analysis (WGCNA) to filter immune infiltration-related gene modules. Univariate, the least absolute shrinkage and selection operator (LASSO), and multivariate cox regression were applied to construct the prognostic model. The predictive capacity was validated by meta-analysis including external dataset GSE65858, GSE41613 and GSE686. Model candidate genes were verified at mRNA and protein levels using public database and independent specimens of immunohistochemistry. Immunotherapy-treated cohort GSE159067, TIDE and CIBERSORT were used to evaluate the features of immunotherapy responsiveness and immune infiltration in HNSCC. RESULTS Immune microenvironment was significantly associated with the prognosis of HNSCC patients. Total 277 immune infiltration-related genes were filtered by WGCNA and involved in various immune processes. Cox regression identified nine prognostic immune infiltration-related genes (MORF4L2, CTSL1, TBC1D2, C5orf15, LIPA, WIPF1, CXCL13, TMEM173, ISG20) to build a risk score. Most candidate genes were highly expressed in HNSCC tissues at mRNA and protein levels. Survival meta-analysis illustrated high prognostic accuracy of the model in the discovery cohort and validation cohort. Higher proportion of progression-free outcomes, lower TIDE scores and higher expression levels of immune checkpoint genes indicated enhanced immunotherapy responsiveness in low-risk patients. Decreased memory B cells, CD8+ T cells, follicular helper T cells, regulatory T cells, and increased activated dendritic cells and activated mast cells were identified as crucial immune cells in the TME of high-risk patients. CONCLUSIONS The immune infiltration-related gene model was well-qualified and provided novel biomarkers for the prognosis of HNSCC.
Collapse
Affiliation(s)
- Yinghe Ding
- grid.216417.70000 0001 0379 7164Transplantation Center, The 3rd Xiangya Hospital, Central South University, Changsha, 410013 Hunan China
| | - Ling Chu
- grid.216417.70000 0001 0379 7164Department of Pathology, The 3rd Xiangya Hospital, Central South University, Changsha, 410013 Hunan China
| | - Qingtai Cao
- grid.216417.70000 0001 0379 7164Transplantation Center, The 3rd Xiangya Hospital, Central South University, Changsha, 410013 Hunan China
| | - Hanyu Lei
- grid.216417.70000 0001 0379 7164Transplantation Center, The 3rd Xiangya Hospital, Central South University, Changsha, 410013 Hunan China
| | - Xinyu Li
- grid.216417.70000 0001 0379 7164Transplantation Center, The 3rd Xiangya Hospital, Central South University, Changsha, 410013 Hunan China
| | - Quan Zhuang
- grid.216417.70000 0001 0379 7164Transplantation Center, The 3rd Xiangya Hospital, Central South University, Changsha, 410013 Hunan China ,Research Center of National Health Ministry on Transplantation Medicine, Changsha, 410013 Hunan China
| |
Collapse
|
4
|
Beider K, Voevoda-Dimenshtein V, Zoabi A, Rosenberg E, Magen H, Ostrovsky O, Shimoni A, Weiss L, Abraham M, Peled A, Nagler A. CXCL13 chemokine is a novel player in multiple myeloma osteolytic microenvironment, M2 macrophage polarization, and tumor progression. J Hematol Oncol 2022; 15:144. [PMID: 36217194 PMCID: PMC9549634 DOI: 10.1186/s13045-022-01366-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/02/2022] [Indexed: 12/04/2022] Open
Abstract
Background We assessed the mechanism by which multiple myeloma (MM) shapes the bone marrow (BM) microenvironment and affects MΦ polarization. Methods In vivo xenograft model of BM-disseminated human myeloma, as well as analysis of MM cell lines, stromal components, and primary samples from patients with MM, was utilized. Results Analysis of the BM from MM-bearing mice inoculated with human CXCR4-expressing RPMI8226 cells revealed a significant increase in M2 MΦ cell numbers (p < 0.01). CXCL13 was one of the most profoundly increased factors upon MM growth with increased levels in the blood of MM-bearing animals. Myeloid cells were the main source of the increased murine CXCL13 detected in MM-infiltrated BM. MM cell lines induced CXCL13 and concurrent expression of M2 markers (MERTK, CD206, CD163) in co-cultured human MΦ in vitro. Interaction with MΦ reciprocally induced CXCL13 expression in MM cell lines. Mechanistically, TGFβ signaling was involved in CXCL13 induction in MM cells, while BTK signaling was implicated in MM-stimulated increase of CXCL13 in MΦ. Recombinant CXCL13 increased RANKL expression and induced TRAP+ osteoclast (OC) formation in vitro, while CXCL13 neutralization blocked these activities. Moreover, mice inoculated with CXCL13-silenced MM cells developed significantly lower BM disease. Reduced tumor load correlated with decreased numbers of M2 MΦ in BM, decreased bone disease, and lower expression of OC-associated genes. Finally, higher levels of CXCL13 were detected in the blood and BM samples of MM patients in comparison with healthy individuals. Conclusions Altogether, our findings suggest that bidirectional interactions of MΦ with MM tumor cells result in M2 MΦ polarization, CXCL13 induction, and subsequent OC activation, enhancing their ability to support bone resorption and MM progression. CXCL13 may thus serve as a potential novel target in MM. Supplementary Information The online version contains supplementary material available at 10.1186/s13045-022-01366-5.
Collapse
Affiliation(s)
- Katia Beider
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | | | - Ali Zoabi
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Evgenia Rosenberg
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Hila Magen
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Olga Ostrovsky
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Avichai Shimoni
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel
| | - Lola Weiss
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, Jerusalem, Israel
| | - Michal Abraham
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, Jerusalem, Israel
| | - Amnon Peled
- Goldyne Savad Institute of Gene Therapy, Hebrew University Hospital, Jerusalem, Israel
| | - Arnon Nagler
- Division of Hematology and CBB, Chaim Sheba Medical Center, Tel Aviv University, Tel-Hashomer, Israel.
| |
Collapse
|
5
|
Wang B, Wang M, Ao D, Wei X. CXCL13-CXCR5 axis: Regulation in inflammatory diseases and cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188799. [PMID: 36103908 DOI: 10.1016/j.bbcan.2022.188799] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 01/10/2023]
Abstract
Chemokine C-X-C motif ligand 13 (CXCL13), originally identified as a B-cell chemokine, plays an important role in the immune system. The interaction between CXCL13 and its receptor, the G-protein coupled receptor (GPCR) CXCR5, builds a signaling network that regulates not only normal organisms but also the development of many diseases. However, the precise action mechanism remains unclear. In this review, we discussed the functional mechanisms of the CXCL13-CXCR5 axis under normal conditions, with special focus on its association with diseases. For certain refractory diseases, we emphasize the diagnostic and therapeutic role of CXCL13-CXCR5 axis.
Collapse
Affiliation(s)
- Binhan Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Danyi Ao
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
6
|
Potential Role of CXCL13/CXCR5 Signaling in Immune Checkpoint Inhibitor Treatment in Cancer. Cancers (Basel) 2022; 14:cancers14020294. [PMID: 35053457 PMCID: PMC8774093 DOI: 10.3390/cancers14020294] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/30/2021] [Accepted: 01/04/2022] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Immunotherapy is currently the backbone of new drug treatments for many cancer patients. CXC chemokine ligand 13 (CXCL13) is an important factor involved in recruiting immune cells that express CXC chemokine receptor type 5 (CXCR5) in the tumor microenvironment and serves as a key molecular determinant of tertiary lymphoid structure (TLS) formation. An increasing number of studies have identified the influence of CXCL13 on prognosis in patients with cancer, regardless of the use of immunotherapy treatment. However, no comprehensive reviews of the role of CXCL13 in cancer immunotherapy have been published to date. This review aims to provide an overview of the CXCL13/CXCR5 signaling axis to summarize its mechanisms of action in cancer cells and lymphocytes, in addition to effects on immunity and cancer pathobiology, and its potential as a biomarker for the response to cancer immunotherapy. Abstract Immune checkpoint inhibitors (ICIs), including antibodies that target programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), or cytotoxic T lymphocyte antigen 4 (CTLA4), represent some of the most important breakthroughs in new drug development for oncology therapy from the past decade. CXC chemokine ligand 13 (CXCL13) exclusively binds CXC chemokine receptor type 5 (CXCR5), which plays a critical role in immune cell recruitment and activation and the regulation of the adaptive immune response. CXCL13 is a key molecular determinant of the formation of tertiary lymphoid structures (TLSs), which are organized aggregates of T, B, and dendritic cells that participate in the adaptive antitumor immune response. CXCL13 may also serve as a prognostic and predictive factor, and the role played by CXCL13 in some ICI-responsive tumor types has gained intense interest. This review discusses how CXCL13/CXCR5 signaling modulates cancer and immune cells to promote lymphocyte infiltration, activation by tumor antigens, and differentiation to increase the antitumor immune response. We also summarize recent preclinical and clinical evidence regarding the ICI-therapeutic implications of targeting the CXCL13/CXCR5 axis and discuss the potential role of this signaling pathway in cancer immunotherapy.
Collapse
|
7
|
Gao X, Jiang M, Chu Y, Han Y, Jin Y, Zhang W, Wang W, Yang S, Li W, Fan A, Cao J, Wang J, Liu H, Fu X, Chen D, Nie Y, Fan D. ETV4 promotes pancreatic ductal adenocarcinoma metastasis through activation of the CXCL13/CXCR5 signaling axis. Cancer Lett 2022; 524:42-56. [PMID: 34582976 DOI: 10.1016/j.canlet.2021.09.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/05/2021] [Accepted: 09/14/2021] [Indexed: 01/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has the highest fatality rate of any solid tumor, with a five-year survival rate of only 10% in the USA. PDAC is characterized by early metastasis. More than 50% of patients present with distant metastases at the time of diagnosis, and the majority of patients will develop metastasis within 4 years after tumor resection. Despite extensive studies, the molecular mechanisms underlying PDAC metastasis remain unclear. The polyoma enhancer activator protein (PEA3) subfamily was reported to play a vital role in the initiation and progression of multiple tumors. Herein, we found that ETS variant 4 (ETV4) was highly expressed in PDAC tissues and associated with poor survival. Univariate and multivariate analyses revealed that ETV4 expression was an independent prognostic factor for patient survival. Further experiments showed that ETV4 overexpression promoted PDAC invasion and metastasis both in vitro and in vivo. For the first time, we demonstrated that, mechanistically, ETV4 increased CXCR5 expression by directly binding to the CXCR5 promoter region. Knockdown of CXCR5 significantly reversed ETV4-mediated PDAC migration and invasion, while CXCR5 overexpression exerted the opposite effects. Intriguingly, we found that CXCL13, a specific ligand of CXCR5, increased ETV4 expression and promoted PDAC invasion and metastasis by activating the ERK1/2 pathway. ETV4 knockdown significantly abrogated the enhanced migratory and invasive abilities induced by the CXCL13/CXCR5 axis. In addition, a CXCR5 neutralizing antibody disrupted the CXCL13/ETV4/CXCR5 positive feedback loop and inhibited cell migration and invasion. Overall, in this study, we demonstrated that ETV4 plays a vital role in PDAC metastasis and defined a novel CXCL13/ETV4/CXCR5 positive feedback loop. Targeting this pathway has implications for potential therapeutic strategies for PDAC treatment.
Collapse
Affiliation(s)
- Xiaoliang Gao
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Mingzuo Jiang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Yi Chu
- Department of Gastroenterology, The Second Medical Center of PLA General Hospital, Beijing, 100853, China
| | - Yuying Han
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Yirong Jin
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Wenyao Zhang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Weijie Wang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Suzhen Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Wenjiao Li
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Ahui Fan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Jiayi Cao
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Jiayao Wang
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Hao Liu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Xin Fu
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Di Chen
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
| | - Yongzhan Nie
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China.
| | - Daiming Fan
- State Key Laboratory of Cancer Biology and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
8
|
CXCL13 in Cancer and Other Diseases: Biological Functions, Clinical Significance, and Therapeutic Opportunities. Life (Basel) 2021; 11:life11121282. [PMID: 34947813 PMCID: PMC8708574 DOI: 10.3390/life11121282] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/31/2021] [Accepted: 11/12/2021] [Indexed: 12/11/2022] Open
Abstract
The development of cancer is a multistep and complex process involving interactions between tumor cells and the tumor microenvironment (TME). C-X-C chemokine ligand 13 (CXCL13) and its receptor, CXCR5, make crucial contributions to this process by triggering intracellular signaling cascades in malignant cells and modulating the sophisticated TME in an autocrine or paracrine fashion. The CXCL13/CXCR5 axis has a dominant role in B cell recruitment and tertiary lymphoid structure formation, which activate immune responses against some tumors. In most cancer types, the CXCL13/CXCR5 axis mediates pro-neoplastic immune reactions by recruiting suppressive immune cells into tumor tissues. Tobacco smoke and haze (smohaze) and the carcinogen benzo(a)pyrene induce the secretion of CXCL13 by lung epithelial cells, which contributes to environmental lung carcinogenesis. Interestingly, the knockout of CXCL13 inhibits benzo(a)pyrene-induced lung cancer and azoxymethane/dextran sodium sulfate-induced colorectal cancer in mice. Thus, a better understanding of the context-dependent functions of the CXCL13/CXCR5 axis in tumor tissue and the TME is required to design an efficient immune-based therapy. In this review, we summarize the molecular events and TME alterations caused by CXCL13/CXCR5 and briefly discuss the potentials of agents targeting this axis in different malignant tumors.
Collapse
|
9
|
Zheng H, Liu H, Lu Y, Li H. Identification of a Novel Signature Predicting Overall Survival in Head and Neck Squamous Cell Carcinoma. Front Surg 2021; 8:717084. [PMID: 34631779 PMCID: PMC8498039 DOI: 10.3389/fsurg.2021.717084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 08/27/2021] [Indexed: 12/21/2022] Open
Abstract
Background: Head and neck squamous cell carcinoma (HNSCC) is a highly heterogeneous tumor with a high incidence and poor prognosis. Therefore, effective predictive models are needed to evaluate patient outcomes and optimize treatment. Methods: Robust Rank Aggregation (RRA) method was used to identify highly robust differentially-expressed genes (DEGs) between HNSCC and normal tissue in 9 GEO and TCGA datasets. Univariate Cox regression analysis and Lasso Cox regression analysis were performed to identify DEGs related to the Overall survival (OS) and to construct a prognostic gene signature (HNSCCSig). External validation was performed using GSE65858 dataset. Moreover, comprehensive bioinformatics analyses were used to identify the association between HNSCCSig and tumor immune environment. Results: A total of 257 reliable DEGs were identified by differentially analysis result of TCGA and GSE65858 datasets. The HNSCCSig including 7 mRNAs (SLURP1, SCARA5, CLDN10, MYH11, CXCL13, HLF, and ITGA3) were developed and validated to identify high-risk group who had a worse OS than low-risk group in TCGA and GSE65858 datasets. Cox regression analysis showed that the HNSCCSig could independently predict OS in both the TCGA and the GSE65858 datasets. Further research demonstrated that the infiltration bundance of CD8 + T cells, B cells, neutrophils, and NK cells were significantly lower in the high-risk group. A nomogram was also constructed by combining the HNSCCSig and clinical characters. Conclusion: We established and validated the HNSCCSig consisting of SLURP1, SCARA5, CLDN10, MYH11, CXCL13, HLF, and ITGA3. A nomogram combining HNSCCSig and some clinical parameters was constructed to identify high-risk HNSCC-patients with poor prognosis.
Collapse
Affiliation(s)
- Haige Zheng
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Huixian Liu
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yumin Lu
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Hengguo Li
- Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
10
|
CXCL13 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1302:71-90. [PMID: 34286442 DOI: 10.1007/978-3-030-62658-7_6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Chemokines have emerged as important players in tumorigenic process. An extensive body of literature generated over the last two or three decades strongly implicate abnormally activated or functionally disrupted chemokine signaling in liaising most-if not all-hallmark processes of cancer. It is well-known that chemokine signaling networks within the tumor microenvironment are highly versatile and context-dependent: exert both pro-tumoral and antitumoral activities. The C-X-C motif chemokine ligand 13 (CXCL13), and its cognate receptor CXCR5, represents an emerging example of chemokine signaling axes, which express the ability to modulate tumor growth and progression in either way. Collateral evidence indicate that CXCL13-CXCR5 axis may directly modulate tumor growth by inducing proliferation of cancer cells, as well as promoting invasive phenotypes and preventing their apoptosis. In addition, CXCL13-CXCR5 axis may also indirectly modulate tumor growth by regulating noncancerous cells, particularly the immune cells, within the tumor microenvironment. Here, we review the role of CXCL13, together with CXCR5, in the human tumor microenvironment. We first elaborate their patterns of expression, regulation, and biological functions in normal physiology. We then consider how their aberrant activity, as a result of differential overexpression or co-expression, may directly or indirectly modulate the growth of tumors through effects on both cancerous and noncancerous cells.
Collapse
|
11
|
Zhang X, Yang M, Liu Y, Liu H, Yang J, Luo J, Zhou H. A novel 4-gene signature model simultaneously predicting malignant risk of oral potentially malignant disorders and oral squamous cell carcinoma prognosis. Arch Oral Biol 2021; 129:105203. [PMID: 34252587 DOI: 10.1016/j.archoralbio.2021.105203] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 06/10/2021] [Accepted: 06/26/2021] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Oral squamous cell carcinoma (OSCC) is often diagnosed at late stage with a poor prognosis. The study hereunder aimed to construct a multi-gene model to simultaneously promote early diagnosis of OSCC by evaluating malignant risk of oral potentially malignant disorders (OPMDs) and predict prognosis. MATERIALS AND METHODS 3 GEO datasets including OPMDs and OSCC samples were obtained for overlapping differentially expressed genes (DEGs) being screened. The predictive model was built with optimal DEGs by SVM algorithm, estimated by receiver operator characteristic curves and validated for double prediction via oral cancer-free survival (for malignant risk of OPMDs) and overall survival time (for OSCC) analysis respectively compared to other models. The protein expression of biomarkers in the model was validated in human samples by immunohistochemistry. RESULTS A novel predictive model of 4-gene signature was built based on 12 common DEGs revealed from 3 GEO datasets. It could well distinguish OSCC from OPMDs and normal tissues. Both oral cancer-free survival and overall survival time analysis were significantly poorer in high-risk patients than in low-risk ones in Kaplan Meier survival curve respectively. The protein expression of biomarkers in OSCC was with significant difference compared to normal and OPMDs. CONCLUSIONS The novel 4-gene signature model presents strong ability in simultaneous prediction of the malignant risk of OPMDs and OSCC progression, potentially benefiting both the early diagnosis and therapeutic outcomes of OSCC.
Collapse
Affiliation(s)
- Xinyue Zhang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China; Department of Stomatology, Chengdu Fifth People's Hospital/The Second Clinical Medical College, Chengdu University of TCM, Chengdu, Sichuan, People's Republic of China
| | - Miao Yang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yangfan Liu
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Hailong Liu
- Freelance Computer Engineer, Chengdu, Sichuan, People's Republic of China
| | - Jin Yang
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | - Jingjing Luo
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| | - Hongmei Zhou
- State Key Laboratory of Oral Diseases, National Center of Stomatology, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, People's Republic of China
| |
Collapse
|
12
|
Jin K, Cao Y, Gu Y, Fang H, Fei Y, Wang J, Liu X, Lv K, He X, Lin C, Liu H, Li H, He H, Li R, Zhang H, Xu J. Poor clinical outcomes and immunoevasive contexture in CXCL13+CD8+ T cells enriched gastric cancer patients. Oncoimmunology 2021; 10:1915560. [PMID: 33996266 PMCID: PMC8081038 DOI: 10.1080/2162402x.2021.1915560] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
As an adverse survival prognosticator, chemokine (C-X-C motif) ligand 13 (CXCL13) has been studied in several types of malignancies. The secretion and physiological roles of CXCL13 in follicular helper T cells (TFH) cells have been well described, while the clinical significance of CD8+ tumor-infiltrating lymphocytes (TILs)-associated CXCL13 remains unknown. This study aims to investigate the clinical significance of CXCL13+CD8+ T cells in survival and chemotherapeutic responsiveness prediction in gastric cancer. In this study, 440 patients enrolled from Zhongshan Hospital with tumor microarray (TMA) specimens were randomly divided into testing set (n = 220) and validation set (n = 220) for analysis. CXCL13+CD8+ T cells were detected by multicolor immunohistochemistry. Fresh tumor tissue samples from another 60 gastric cancer patients were collected to detect CXCL13+CD8+ T cells functional status by flow cytometry (FCM). We found that high intratumoral CXCL13+CD8+ T cells infiltration predicted poor overall survival and inferior chemotherapeutic responsiveness in gastric cancer. CXCL13+CD8+T cells were associated with immunoevasive contexture with increased regulatory T (Treg) cells and dysfunctional cytotoxic T lymphocytes (CTLs). Moreover, the combinational analysis of CXCL13+CD8+ T cells and CD8+ T cells infiltration stratified patients into distinct risk groups with different clinical outcomes and chemotherapeutic responsiveness. Conclusively, intratumoral CXCL13+CD8+ T cells infiltration could be an independent prognostic and predictive marker for gastric cancer patients. CXCL13+CD8+ T cells represented an exhausted CD8+ T cell subset, and might be a potential immunotherapeutic target in gastric cancer.
Collapse
Affiliation(s)
- Kaifeng Jin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yifan Cao
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Gu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hanji Fang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuchao Fei
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jieti Wang
- Department of Gastric Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Xin Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kunpeng Lv
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xudong He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Chao Lin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - He Li
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hongyong He
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ruochen Li
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Heng Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Transcriptomic analysis identifies differences in gene expression in actinic keratoses after treatment with imiquimod and between responders and non responders. Sci Rep 2021; 11:8775. [PMID: 33888854 PMCID: PMC8062619 DOI: 10.1038/s41598-021-88424-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/08/2021] [Indexed: 02/06/2023] Open
Abstract
The presence of actinic keratoses (AKs) increases a patient’s risk of developing squamous cell carcinoma by greater than six-fold. We evaluated the effect of topical treatment with imiquimod on the tumor microenvironment by measuring transcriptomic differences in AKs before and after treatment with imiquimod 3.75%. Biopsies were collected prospectively from 21 patients and examined histologically. RNA was extracted and transcriptomic analyses of 788 genes were performed using the nanoString assay. Imiquimod decreased number of AKs by study endpoint at week 14 (p < 0.0001). Post-imiquimod therapy, levels of CDK1, CXCL13, IL1B, GADPH, TTK, ILF3, EWSR1, BIRC5, PLAUR, ISG20, and C1QBP were significantly lower (adjusted p < 0.05). Complete responders (CR) exhibited a distinct pattern of inflammatory gene expression pre-treatment relative to incomplete responders (IR), with alterations in 15 inflammatory pathways (p < 0.05) reflecting differential expression of 103 genes (p < 0.05). Presence of adverse effects was associated with improved treatment response. Differences in gene expression were found between pre-treatment samples in CR versus IR, suggesting that higher levels of inflammation pre-treament may play a part in regression of AKs. Further characterization of the immune micro-environment in AKs may help develop biomarkers predictive of response to topical immune modulators and may guide therapy.
Collapse
|
14
|
Liu W, Li CJ, Li LJ. [Advances in molecular mechanisms of bone invasion by oral cancer]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2021; 39:221-226. [PMID: 33834679 DOI: 10.7518/hxkq.2021.02.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Bone invasion by oral cancer is a common clinical problem, which affects the choice of treatment and predicts a poor prognosis. Unfortunately, the molecular mechanism of this phenomenon has not been fully elucidated. Current studies have revealed that oral cancer cells modulate the formation and function of osteoclasts through the expression of a series of signal molecules. Many signal pathways are involved in this process, of which receptor activator of nuclear factor-κB ligand/receptor activator of nuclear factor-κB/osteoprotegerin signaling pathway attracted much attention. In this review, we introduce recent progress in molecular mechanisms of bone invasion by oral cancer.
Collapse
Affiliation(s)
- Wei Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chun-Jie Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Long-Jiang Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
15
|
Son SH, Park J, Jung MJ, Lee SK, Kim H, Kim KR, Park KK, Chung WY. Transforming growth factor-β-regulated fractalkine as a marker of erosive bone invasion in oral squamous cell carcinoma. Eur J Oral Sci 2021; 129:e12750. [PMID: 33503283 DOI: 10.1111/eos.12750] [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: 07/13/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022]
Abstract
Patients with oral squamous cell carcinoma (OSCC) bone invasion are surgically treated with bone resection, which results in severe physical and psychological damage. Here, we investigated the potential of fractalkine (CX3CL1), which is regulated by transforming growth factor (TGF-β), as a novel biomarker for correct prediction and early detection of OSCC-associated bone invasion. TGF-β knockdown and treatment with a TGF-β-neutralizing antibody decreased the level of fractalkine in the culture media of HSC-2 and YD10B OSCC cells. Treatment with a fractalkine-neutralizing antibody reduced TGF-β-stimulated invasion by HSC-2 and YD10B cells. Fractalkine treatment increased the viability, invasion, and uPA secretion of both OSCC cell lines. Furthermore, OSCC cell bone invasion was assessed following subcutaneous inoculation of wild-type or TGF-β knockdown OSCC cells in mouse calvaria. TGF-β knockdown prevented erosive bone invasion, reduced the number of osteoclasts at the tumor-bone interface, and downregulated fractalkine expression in mouse tumor tissues. Our results indicate that the production of fractalkine is stimulated by TGF-β and mediates TGF-β-induced cell invasion in several OSCC cell lines showing an erosive pattern of bone invasion. Fractalkine may be a useful predictive marker and therapeutic target for OSCC-induced bone destruction.
Collapse
Affiliation(s)
- Seung Hwa Son
- Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea
| | - Junhee Park
- Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea
| | - Min Ju Jung
- Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea.,Department of Applied Life Science, The Graduate School, Yonsei University, Seoul, Korea
| | - Sun Kyoung Lee
- Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea
| | - Hyungkeun Kim
- Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea.,Department of Applied Life Science, The Graduate School, Yonsei University, Seoul, Korea.,Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Korea
| | - Ki Rim Kim
- Department of Dental Hygiene, College of Science and Engineering, Kyungpook National University, Sangju, Korea
| | - Kwang-Kyun Park
- Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea
| | - Won-Yoon Chung
- Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea.,Department of Applied Life Science, The Graduate School, Yonsei University, Seoul, Korea.,Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Korea
| |
Collapse
|
16
|
Li Y, Wu T, Gong S, Zhou H, Yu L, Liang M, Shi R, Wu Z, Zhang J, Li S. Analysis of the Prognosis and Therapeutic Value of the CXC Chemokine Family in Head and Neck Squamous Cell Carcinoma. Front Oncol 2021; 10:570736. [PMID: 33489879 PMCID: PMC7820708 DOI: 10.3389/fonc.2020.570736] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 10/29/2020] [Indexed: 12/24/2022] Open
Abstract
The CXC chemokines belong to a family which includes 17 different CXC members. Accumulating evidence suggests that CXC chemokines regulate tumor cell proliferation, invasion, and metastasis in various types of cancers by influencing the tumor microenvironment. The different expression profiles and specific function of each CXC chemokine in head and neck squamous cell carcinoma (HNSCC) are not yet clarified. In our work, we analyzed the altered expression, interaction network, and clinical data of CXC chemokines in patients with HNSCC by using the following: the Oncomine dataset, cBioPortal, Metascape, String analysis, GEPIA, and the Kaplan–Meier plotter. The transcriptional level analysis suggested that the mRNA levels of CXCL1, CXCL2, CXCL3, CXCL5, CXCL6, CXCL8, CXCL9, CXCL10, CXCL11, and CXCL13 increased in HNSCC tissue samples when compared to the control tissue samples. The expression levels of CXCL9, CXCL10, CXCL11, CXCL12, and CXCL14 were associated with various tumor stages in HNSCC. Clinical data analysis showed that high transcription levels of CXCL2, CXCL3, and CXCL12, were linked with low relapse-free survival (RFS) in HNSCC patients. On the other hand, high CXCL14 levels predicted high RFS outcomes in HNSCC patients. Meanwhile, increased gene transcription levels of CXCL9, CXCL10, CXCL13, CXCL14, and CXCL17 were associated with a higher overall survival (OS) advantage in HNSCC patients, while high levels of CXCL1, and CXCL8 were associated with poor OS in all HNSCC patients. This study implied that CXCL1, CXCL2, CXCL3, CXCL8, and CXCL12 could be used as prognosis markers to identify low survival rate subgroups of patients with HNSCC as well as be potential suitable therapeutic targets for HNSCC patients. Additionally, CXCL9, CXCL10, CXCL13, CXCL14, and CXCL17 could be used as functional prognosis biomarkers to identify better survival rate subgroups of patients with HNSCC.
Collapse
Affiliation(s)
- Yongchao Li
- Key Laboratory of Protection & Utilization of Biological Resources in Tarim Basin, College of Life Sciences, Tarim University, Alar, China
| | - Tinghui Wu
- Key Laboratory of Protection & Utilization of Biological Resources in Tarim Basin, College of Life Sciences, Tarim University, Alar, China
| | - Shujuan Gong
- Key Laboratory of Protection & Utilization of Biological Resources in Tarim Basin, College of Life Sciences, Tarim University, Alar, China
| | - Hangzheng Zhou
- Key Laboratory of Protection & Utilization of Biological Resources in Tarim Basin, College of Life Sciences, Tarim University, Alar, China
| | - Lufei Yu
- Key Laboratory of Protection & Utilization of Biological Resources in Tarim Basin, College of Life Sciences, Tarim University, Alar, China
| | - Meiyan Liang
- Key Laboratory of Protection & Utilization of Biological Resources in Tarim Basin, College of Life Sciences, Tarim University, Alar, China
| | - Ruijun Shi
- Key Laboratory of Protection & Utilization of Biological Resources in Tarim Basin, College of Life Sciences, Tarim University, Alar, China
| | - Zhenhui Wu
- Key Laboratory of Protection & Utilization of Biological Resources in Tarim Basin, College of Life Sciences, Tarim University, Alar, China
| | - Jinpei Zhang
- Key Laboratory of Protection & Utilization of Biological Resources in Tarim Basin, College of Life Sciences, Tarim University, Alar, China
| | - Shuwei Li
- Key Laboratory of Protection & Utilization of Biological Resources in Tarim Basin, College of Life Sciences, Tarim University, Alar, China
| |
Collapse
|
17
|
Long J, Zhang S, Zeng X, Ouyang Y, Wang Y, Hu Z, Ye Y, Wu W, Jin F, Zhou S, Zeng Z. Development of an Immunogenomic Landscape-Based Prognostic Index of Head and Neck Squamous Cell Carcinoma. Front Mol Biosci 2020; 7:586344. [PMID: 33330624 PMCID: PMC7732611 DOI: 10.3389/fmolb.2020.586344] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the eighth leading cancer by incidence worldwide, with approximately 700,000 new cases in 2018 (accounting for 11% of all cancers). The occurrence and development of tumors are closely related to the immunological function of the body and sensitivity to treatment schemes as well as prognosis. It is urgent for clinicians to systematically study patients’ immune gene maps to help select a treatment plan and analyze the potential to cure HNSCC. Here, the transcriptomic data of HNSCC samples were downloaded from The Cancer Genome Atlas (TCGA), and 4,793 genes differentially expressed in normal and cancer tissues of HNSCC were identified, including 1,182 downregulated and 3,611 upregulated genes. From these genes, 400 differentially expressed immune-related genes (IRGs) were extracted, including 95 downregulated genes and 305 upregulated genes. The prognostic values of IRGs were evaluated by univariate Cox analysis, and 236 genes that were significantly related to the overall survival (OS) of patients were identified. The signaling pathways that play roles in the prognosis of IRGs were investigated by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, and the expression profiles of IRGs and OS in 499 HNSCC patients based on TCGA dataset were integrated. Potential molecular mechanisms and characteristics of these HNSCC-specific IRGs were further explored with the help of a new prognostic index based on IRGs developed by least absolute shrinkage and selection operator (LASSO) Cox analysis. A total of 64 hub genes (IRGs associated with prognosis) were markedly associated with the clinical outcome of HNSCC patients. KEGG functional enrichment analysis revealed that these genes were actively involved in several pathways, e.g., cytokine–cytokine receptor interaction, T-cell receptor signaling, and natural killer cell-mediated cytotoxicity. IRG-based prognostic signatures performed moderately in prognostic predictions. Interestingly, the prognostic index based on IRGs reflected infiltration by several types of immune cells. These data screened several IRGs of clinical significance and revealed drivers of the immune repertoire, demonstrating the importance of a personalized IRG-based immune signature in the recognition, surveillance, and prognosis of HNSCC.
Collapse
Affiliation(s)
- Jinhua Long
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,School of Biology and Engineering, Guizhou Medical University, Guiyang, China.,Key Laboratory of Infectious Immune and Antibody Engineering in Guizhou Province, Guizhou Medical University, Guiyang, China.,Department of Head and Neck Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shichao Zhang
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,School of Biology and Engineering, Guizhou Medical University, Guiyang, China.,Key Laboratory of Infectious Immune and Antibody Engineering in Guizhou Province, Guizhou Medical University, Guiyang, China.,Key Laboratory of Biology and Medical Engineering, Immune Cells and Antibody Engineering Research Center of Guizhou Province, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Xianlin Zeng
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,School of Biology and Engineering, Guizhou Medical University, Guiyang, China.,Key Laboratory of Infectious Immune and Antibody Engineering in Guizhou Province, Guizhou Medical University, Guiyang, China.,Key Laboratory of Biology and Medical Engineering, Immune Cells and Antibody Engineering Research Center of Guizhou Province, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Yan Ouyang
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Yun Wang
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,School of Biology and Engineering, Guizhou Medical University, Guiyang, China.,Key Laboratory of Infectious Immune and Antibody Engineering in Guizhou Province, Guizhou Medical University, Guiyang, China.,Key Laboratory of Biology and Medical Engineering, Immune Cells and Antibody Engineering Research Center of Guizhou Province, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Zuquan Hu
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,School of Biology and Engineering, Guizhou Medical University, Guiyang, China.,Key Laboratory of Infectious Immune and Antibody Engineering in Guizhou Province, Guizhou Medical University, Guiyang, China.,Key Laboratory of Biology and Medical Engineering, Immune Cells and Antibody Engineering Research Center of Guizhou Province, School of Biology and Engineering, Guizhou Medical University, Guiyang, China
| | - Yuannong Ye
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,School of Biology and Engineering, Guizhou Medical University, Guiyang, China.,Key Laboratory of Infectious Immune and Antibody Engineering in Guizhou Province, Guizhou Medical University, Guiyang, China
| | - Weili Wu
- Department of Head and Neck Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Feng Jin
- Department of Head and Neck Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shi Zhou
- Department of Intervention, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhu Zeng
- School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,School of Biology and Engineering, Guizhou Medical University, Guiyang, China.,Key Laboratory of Infectious Immune and Antibody Engineering in Guizhou Province, Guizhou Medical University, Guiyang, China.,Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| |
Collapse
|
18
|
Chen J, Meng X, Zhou Q, Feng J, Zheng W, Wang Z, Wang J, Wang Y. Effect of CXCR5-Positive Cell Infiltration on the Immune Contexture and Patient Prognosis in Head and Neck Squamous Cell Carcinoma. Onco Targets Ther 2020; 13:5869-5877. [PMID: 32606797 PMCID: PMC7319516 DOI: 10.2147/ott.s248958] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 04/22/2020] [Indexed: 11/23/2022] Open
Abstract
Purpose CXCR5-positive (CXCR5+) tumor cell infiltration has different prognostic values in different types of cancer. The objective was to evaluate the effect of CXCR5+ cell infiltration in head and neck squamous cell carcinoma (HNSCC). Patients and Methods The study included two patient cohorts: The Cancer Genome Atlas cohort (TCGA, n = 472) and the Renji Hospital cohort (RJHC, n = 201). The TCGA and RJHC cohorts were analyzed for CXCR5-related mRNAs and CXCR5+ cell infiltration, respectively. We then evaluated the correlation between CXCR5 mRNA and CXCR5+ cell infiltration in terms of overall survival and the immune contexture. Results The 5-year overall survival rate was significantly correlated with high CXCR5 mRNA expression and CXCR5+ cell infiltration in the TCGA and RJHC cohorts, respectively (p < 0.01), even after adjusting for confounders. Moreover, high CXCR5 mRNA expression was associated with more CD4+ T cells, CD8+ T cells, plasma cells, and less dendritic cells. A high CXCR5 mRNA expression was also correlated with increased expression of cytotoxic IFNG, TNFSF11 (RANKL), GZMA, GZMB, GZMK, GZMM, and PRF1 and increased expression of the immunosuppressive gene PDCD1 (PD-1), CD274 (PD-L1), CTLA4, LAG3, HAVCR2 (TIM-3), BTLA, and TIGIT. Conclusion HNSCC patients with a high intratumoral CXCR5 expression had a better prognosis than those with low intratumoral CXCR5 expression. Moreover, CXCR5+ cell infiltration could be used as an independent prognostic biomarker or as a potential therapeutic target. The presence of CXCR5+ cells affects the infiltration of immunocytes in head and neck cancer, differently from what was reported in other cancer types. Further randomized controlled trials or studies with more patients are needed to validate our results.
Collapse
Affiliation(s)
- Jun Chen
- Head and Neck Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Xiangchao Meng
- Bone and Joint Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Qinyi Zhou
- Head and Neck Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jialin Feng
- Head and Neck Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Wenjie Zheng
- Head and Neck Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Zhuoying Wang
- Head and Neck Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Jiadong Wang
- Head and Neck Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - You Wang
- Bone and Joint Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
19
|
Wu LF, Wang WY, Zhu DC, He P, Zhu K, Gui GP, Gao HQ, Mo XB, Lu X, Deng FY, Lei SF. Protein array test detected three osteoporosis related plasma inflammatory cytokines in Chinese postmenopausal women. Cytokine 2020; 133:155166. [PMID: 32570159 DOI: 10.1016/j.cyto.2020.155166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 01/12/2023]
Abstract
Inflammatory cytokines were involved in pathological conditions of osteoporosis (OP). However, the specific OP-associated inflammatory cytokines are still awaiting to be detected by using a systemic method. Herein, we adopted an extreme sampling scheme and examined inflammatory cytokines between subjects with low and high bone mineral density (BMD) through protein microarray. First, 8 candidate cytokines including B lymphocyte chemoattractant (BLC), osteopontin (OPN) and insulin-like growth factor-binding protein 4 (IGFBP4) were identified in the discovery extreme sampling subgroup. Then, the different expressions for BLC, OPN and IGFBP4 were validated and replicated in two independent extreme sampling subgroups. Further functional experiments showed that the cytokine BLC was involved in bone metabolism by inhibiting bone formation and promoting bone resorption. Together, this study further revealed that inflammatory cytokines were closely related with OP, and that they highlighted critical roles of BLC in the pathogenesis of OP.
Collapse
Affiliation(s)
- Long-Fei Wu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Wen-Yu Wang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China; Suzhou Center for Disease Prevention and Control, Suzhou, Jiangsu, China
| | - Dong-Cheng Zhu
- Department of Orthopedics, Sihong People's Hospital, Suqian, Jiangsu 223900, China
| | - Pei He
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Kan Zhu
- Loujiang Community Health Service Center, Suzhou Gusu District, Suzhou, Jiangsu, China
| | - Guo-Ping Gui
- Disease Prevention and Control Center of Suzhou High Tech Zone, Suzhou, Jiangsu, China
| | - Hong-Qin Gao
- Shishan Community Health Service Center, Suzhou High Tech Zone, Suzhou, Jiangsu, China
| | - Xing-Bo Mo
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xin Lu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Fei-Yan Deng
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shu-Feng Lei
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
20
|
Park J, Zhang X, Lee SK, Song NY, Son SH, Kim KR, Shim JH, Park KK, Chung WY. CCL28-induced RARβ expression inhibits oral squamous cell carcinoma bone invasion. J Clin Invest 2020; 129:5381-5399. [PMID: 31487270 DOI: 10.1172/jci125336] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 09/03/2019] [Indexed: 12/30/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) frequently invades the maxillary or mandibular bone, and this bone invasion is closely associated with poor prognosis and survival. Here, we show that CCL28 functions as a negative regulator of OSCC bone invasion. CCL28 inhibited invasion and epithelial-mesenchymal transition (EMT), and its inhibition of EMT was characterized by induced E-cadherin expression and reduced nuclear localization of β-catenin in OSCC cells with detectable RUNX3 expression levels. CCL28 signaling via CCR10 increased retinoic acid receptor-β (RARβ) expression by reducing the interaction between RARα and HDAC1. In addition, CCL28 reduced RANKL production in OSCC and osteoblastic cells and blocked RANKL-induced osteoclastogenesis in osteoclast precursors. Intraperitoneally administered CCL28 inhibited tumor growth and osteolysis in mouse calvaria and tibia inoculated with OSCC cells. RARβ expression was also increased in tumor tissues. In patients with OSCC, low CCL28, CCR10, and RARβ expression levels were highly correlated with bone invasion. Patients with OSCC who had higher expression of CCL28, CCR10, or RARβ had significantly better overall survival. These findings suggest that CCL28, CCR10, and RARβ are useful markers for the prediction and treatment of OSCC bone invasion. Furthermore, CCL28 upregulation in OSCC cells or CCL28 treatment can be a therapeutic strategy for OSCC bone invasion.
Collapse
Affiliation(s)
- Junhee Park
- Department of Dentistry and.,Department of Applied Life Science, Graduate School, Yonsei University, Seoul, Korea.,Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea
| | - Xianglan Zhang
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Korea.,Department of Pathology, Yanbian University Hospital, Yanji city, China
| | - Sun Kyoung Lee
- Department of Applied Life Science, Graduate School, Yonsei University, Seoul, Korea.,Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea
| | - Na-Young Song
- Department of Dentistry and.,Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea
| | - Seung Hwa Son
- Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Korea
| | - Ki Rim Kim
- Department of Dental Hygiene, College of Science and Engineering, Kyungpook National University, Sangju, Korea
| | - Jae Hoon Shim
- Department of Applied Life Science, Graduate School, Yonsei University, Seoul, Korea
| | - Kwang-Kyun Park
- Department of Dentistry and.,Department of Applied Life Science, Graduate School, Yonsei University, Seoul, Korea.,Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea
| | - Won-Yoon Chung
- Department of Dentistry and.,Department of Applied Life Science, Graduate School, Yonsei University, Seoul, Korea.,Department of Oral Biology and BK21 PLUS project, Yonsei University College of Dentistry, Seoul, Korea.,Oral Cancer Research Institute, Yonsei University College of Dentistry, Seoul, Korea
| |
Collapse
|
21
|
Chemokines and their receptors promoting the recruitment of myeloid-derived suppressor cells into the tumor. Mol Immunol 2019; 117:201-215. [PMID: 31835202 DOI: 10.1016/j.molimm.2019.11.014] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 11/27/2019] [Accepted: 11/30/2019] [Indexed: 02/07/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) expand in tumor-bearing host. They suppress anti-tumor immune response and promote tumor growth. Chemokines play a vital role in recruiting MDSCs into tumor tissue. They can also induce the generation of MDSCs in the bone marrow, maintain their suppressive activity, and promote their proliferation and differentiation. Here, we review CCL2/CCL12-CCR2, CCL3/4/5-CCR5, CCL15-CCR1, CX3CL1/CCL26-CX3CR1, CXCL5/2/1-CXCR2, CXCL8-CXCR1/2, CCL21-CCR7, CXCL13-CXCR5 signaling pathways, their role in MDSCs recruitment to tumor tissue, and their correlation with tumor development, metastasis and prognosis. Targeting chemokines and their receptors may serve as a promising strategy in immunotherapy, especially combined with other strategies such as chemotherapy, cyclin-dependent kinase or immune checkpoints inhibitors.
Collapse
|
22
|
Zhao W, Cui Y, Liu L, Qi X, Liu J, Ma S, Hu X, Zhang Z, Wang Y, Li H, Wang Z, Liu Z, Wu J. Splicing factor derived circular RNA circUHRF1 accelerates oral squamous cell carcinoma tumorigenesis via feedback loop. Cell Death Differ 2019; 27:919-933. [PMID: 31570856 DOI: 10.1038/s41418-019-0423-5] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/16/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023] Open
Abstract
Emerging evidences have suggested the vital roles of circular RNA (circRNA) in the human cancers. However, the underlying biological functions and biogenesis of circRNA in the oral squamous cell carcinoma (OSCC) is still ambiguous. Here, we investigate the oncogenic roles and biogenesis of the novel identified circRNA, circUHRF1 (hsa_circ_0002185), in the OSCC tumorigenesis. Results showed that circUHRF1 was markedly upregulated in the OSCC cells and tissue, besides, the overexpression was closely correlated with the poor prognosis of OSCC patients. Functionally, circUHRF1 promoted the proliferation, migration, invasion, and epithelial mesenchymal transformation (EMT) in vitro and the tumor growth in vivo. Mechanically, circUHRF1 acted as the sponge of miR-526b-5p, thereby positively regulating c-Myc. Transcription factor c-Myc could accelerate the transcription of TGF-β1 and ESRP1. Moreover, splicing factor ESRP1 promoted the circularization and biogenesis of circUHRF1 by targeting the flanking introns, forming the circUHRF1/miR-526b-5p/c-Myc/TGF-β1/ESRP1 feedback loop. In conclusion, our research identified the oncogenic roles of circUHRF1 in the OSCC tumorigenesis and EMT via circUHRF1/miR-526b-5p/c-Myc/TGF-β1/ESRP1 feedback loop, shedding light on the pathogenic mechanism of circUHRF1 for OSCC and providing the potential therapeutic target.
Collapse
Affiliation(s)
- Wei Zhao
- The School and Hospital of Stomatology, Tianjin Medical University, 300070, Tianjin, PR China
| | - Yameng Cui
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, PR China
| | - Lina Liu
- Department of Prosthodontics, Tianjin Stomatological Hospital, Hospital of Stomatology, NanKai University, 300041, Tianjin, PR China
| | - Xiaoqian Qi
- The School and Hospital of Stomatology, Tianjin Medical University, 300070, Tianjin, PR China
| | - Jingwen Liu
- The School and Hospital of Stomatology, Tianjin Medical University, 300070, Tianjin, PR China
| | - Shiqing Ma
- The School and Hospital of Stomatology, Tianjin Medical University, 300070, Tianjin, PR China
| | - Xin Hu
- The School and Hospital of Stomatology, Tianjin Medical University, 300070, Tianjin, PR China
| | - Zheng Zhang
- Department of Periodontics, Tianjin Stomatological Hospital, Hospital of Stomatology, NanKai University, 300041, Tianjin, PR China
| | - Yue Wang
- The School and Hospital of Stomatology, Tianjin Medical University, 300070, Tianjin, PR China
| | - Hongfa Li
- The School and Hospital of Stomatology, Tianjin Medical University, 300070, Tianjin, PR China
| | - Zuomin Wang
- Beijing Chaoyang Hospital, Capital Medical University, Department of Stomatology, 100022, Beijing, PR China.
| | - Zihao Liu
- The School and Hospital of Stomatology, Tianjin Medical University, 300070, Tianjin, PR China.
| | - Jie Wu
- The School and Hospital of Stomatology, Tianjin Medical University, 300070, Tianjin, PR China.
| |
Collapse
|
23
|
Ethiraj P, Sambandam Y, Hathaway-Schrader JD, Haque A, Novince CM, Reddy SV. RANKL triggers resistance to TRAIL-induced cell death in oral squamous cell carcinoma. J Cell Physiol 2019; 235:1663-1673. [PMID: 31309556 DOI: 10.1002/jcp.29086] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/21/2019] [Indexed: 12/29/2022]
Abstract
Oral squamous cell carcinoma (OSCC) occurs as a malignancy of the oral cavity. RANK ligand (RANKL) is essential for osteoclast formation/bone resorption. Recently, we showed autoregulation of receptor activator of nuclear factor-κB ligand (RANKL) stimulates OSCC cell proliferation. OSCC cells show resistance to tumor necrosis factor related apoptosis inducing ligand (TRAIL) treatment. Therefore, we hypothesize that RANKL promotes resistance for TRAIL induction of OSCC apoptotic cell death. In this study, SCC14A and SCC74A cells cultured with TRAIL revealed high-level expression of RANKL which increased resistance to TRAIL inhibition of tumor cell proliferation. RANKL stimulation inhibited terminal deoxynucleotidyl transferase dUTP nick end labeling positive staining in TRAIL-treated cells. CRISPR/Cas-9 knockout of RANKL (RANKL-KO) increased caspase-9, caspase-3 activity and cytochrome c release in OSCC cells. RANKL inhibited proapoptotic proteins BAD and BAX expression. TRAIL treatment suppressed the SQSTM1/p62 and RANKL restored the expression. Interestingly, RANKL alone significantly increased proteasome activity. RANKL-KO in OSCC cells inhibited autophagic activity as evidenced by decreased light chain 3B-II and beclin-1 expression. Thus, RANKL stimulation of OSCC tumor cells triggered resistance for TRAIL-induced OSCC cell death. Taken together, blockade of RANKL may inhibit OSCC tumor progression and enhance the potential of TRAIL induced OSCC tumor cell apoptosis.
Collapse
Affiliation(s)
- Purushoth Ethiraj
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Yuvaraj Sambandam
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| | - Jessica D Hathaway-Schrader
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, South Carolina.,Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | - Chad M Novince
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Sakamuri V Reddy
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
24
|
Kazanietz MG, Durando M, Cooke M. CXCL13 and Its Receptor CXCR5 in Cancer: Inflammation, Immune Response, and Beyond. Front Endocrinol (Lausanne) 2019; 10:471. [PMID: 31354634 PMCID: PMC6639976 DOI: 10.3389/fendo.2019.00471] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 06/28/2019] [Indexed: 12/25/2022] Open
Abstract
It is well-established that the chemokine C-X-C motif ligand 13 (CXCL13) and its receptor, the G-protein coupled receptor (GPCR) CXCR5, play fundamental roles in inflammatory, infectious and immune responses. Originally identified as a B-cell chemoattractant, CXCL13 exerts important functions in lymphoid neogenesis, and has been widely implicated in the pathogenesis of a number of autoimmune diseases and inflammatory conditions, as well as in lymphoproliferative disorders. Current evidence also indicates that the CXCL13:CXCR5 axis orchestrates cell-cell interactions that regulate lymphocyte infiltration within the tumor microenvironment, thereby determining responsiveness to cytotoxic and immune-targeted therapies. In this review, we provide a comprehensive perspective of the involvement of CXCL13 and its receptor in cancer progression. Studies in recent years postulated novel roles for this chemokine in controlling the cancer cell phenotype, and suggest important functions in the growth and metastatic dissemination of solid tumors. Carcinogens have been found to induce CXCL13 production, and production of this chemokine within the tumor milieu has been shown to impact the proliferation, migration, and invasive properties of cancer cells. Thus, the complex networks of cellular interactions involving tumoral CXCL13 and CXCR5 integrate to promote cancer cell autonomous and non-autonomous responses, highlighting the relevance of autocrine and paracrine interactions in dictating the cancer phenotype. Dissecting the molecular and signaling events regulated by CXCL13 and how this chemokine dynamically controls the interaction between the cancer cell and the tumor microenvironment is key to identify novel effectors and therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
| | | | - Mariana Cooke
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
25
|
Hai L, Szwarc MM, Lonard DM, Rajapakshe K, Perera D, Coarfa C, Ittmann M, Fernandez-Valdivia R, Lydon JP. Short-term RANKL exposure initiates a neoplastic transcriptional program in the basal epithelium of the murine salivary gland. Cytokine 2019; 123:154745. [PMID: 31226438 DOI: 10.1016/j.cyto.2019.154745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 06/05/2019] [Indexed: 12/14/2022]
Abstract
Although salivary gland cancers comprise only ∼3-6% of head and neck cancers, treatment options for patients with advanced-stage disease are limited. Because of their rarity, salivary gland malignancies are understudied compared to other exocrine tissue cancers. The comparative lack of progress in this cancer field is particularly evident when it comes to our incomplete understanding of the key molecular signals that are causal for the development and/or progression of salivary gland cancers. Using a novel conditional transgenic mouse (K5:RANKL), we demonstrate that Receptor Activator of NFkB Ligand (RANKL) targeted to cytokeratin 5-positive basal epithelial cells of the salivary gland causes aggressive tumorigenesis within a short period of RANKL exposure. Genome-wide transcriptomic analysis reveals that RANKL markedly increases the expression levels of numerous gene families involved in cellular proliferation, migration, and intra- and extra-tumoral communication. Importantly, cross-species comparison of the K5:RANKL transcriptomic dataset with The Cancer Genome Atlas cancer signatures reveals the strongest molecular similarity with cancer subtypes of the human head and neck squamous cell carcinoma. These studies not only provide a much needed transcriptomic resource to mine for novel molecular targets for therapy and/or diagnosis but validates the K5:RANKL transgenic as a preclinical model to further investigate the in vivo oncogenic role of RANKL signaling in salivary gland tumorigenesis.
Collapse
Affiliation(s)
- Lan Hai
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; Reproductive Medicine Center of Henan Provincial People's Hospital, Zhengzhou, Henan Province, PR China
| | - Maria M Szwarc
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Dimuthu Perera
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Michael Ittmann
- Department of Pathology, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | | | - John P Lydon
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
26
|
Zhu L, Zhang G, Wang P, Zhang L, Ji J, Liu X, Zhou Z, Zhao J, Wang X. The effect of C-X-C motif chemokine ligand 13 in cutaneous squamous cell carcinoma treated with aminolevulinic acid-photodynamic therapy. Photodiagnosis Photodyn Ther 2019; 26:389-394. [DOI: 10.1016/j.pdpdt.2019.04.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/20/2019] [Accepted: 04/19/2019] [Indexed: 11/26/2022]
|
27
|
Hussain M, Adah D, Tariq M, Lu Y, Zhang J, Liu J. CXCL13/CXCR5 signaling axis in cancer. Life Sci 2019; 227:175-186. [PMID: 31026453 DOI: 10.1016/j.lfs.2019.04.053] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/22/2019] [Accepted: 04/22/2019] [Indexed: 02/07/2023]
Abstract
The tumor microenvironment comprises stromal and tumor cells which interact with each other through complex cross-talks that are mediated by a variety of growth factors, cytokines, and chemokines. The chemokine ligand 13 (CXCL13) and its chemokine receptor 5 (CXCR5) are among the key chemotactic factors which play crucial roles in deriving cancer cell biology. CXCL13/CXCR5 signaling axis makes pivotal contributions to the development and progression of several human cancers. In this review, we discuss how CXCL13/CXCR5 signaling modulates cancer cell ability to grow, proliferate, invade, and metastasize. Furthermore, we also discuss the preliminary evidence on context-dependent functioning of this axis within the tumor-immune microenvironment, thus, highlighting its potential dichotomy with respect to anticancer immunity and cancer immune-evasion mechanisms. At the end, we briefly shed light on the therapeutic potential or implications of targeting CXCL13/CXCR5 axis within the tumor microenvironment.
Collapse
Affiliation(s)
- Muzammal Hussain
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Science Park, Guangzhou 510530, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Dickson Adah
- University of Chinese Academy of Sciences, Beijing 100049, PR China; State Key Laboratory of Respiratory Disease, Center for Infection and Immunity, Guangzhou Institutes of Biomedicine and Heath, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Science Park, Guangzhou 510530, PR China
| | - Muqddas Tariq
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Science Park, Guangzhou 510530, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Yongzhi Lu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Science Park, Guangzhou 510530, PR China
| | - Jiancun Zhang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Science Park, Guangzhou 510530, PR China.
| | - Jinsong Liu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Science Park, Guangzhou 510530, PR China.
| |
Collapse
|
28
|
Sambandam Y, Ethiraj P, Hathaway-Schrader JD, Novince CM, Panneerselvam E, Sundaram K, Reddy SV. Autoregulation of RANK ligand in oral squamous cell carcinoma tumor cells. J Cell Physiol 2018; 233:6125-6134. [PMID: 29323724 DOI: 10.1002/jcp.26456] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/05/2018] [Indexed: 12/16/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is the most common malignancy among oral cancers and shows potent activity for local bone invasion. Receptor activator of nuclear factor κB (RANK) ligand (RANKL) is critical for bone-resorbing osteoclast formation. We previously demonstrated that OSCC tumor cells express high levels of RANKL. In this study, confocal microscopy demonstrated RANKL specific receptor, RANK expression in OSCC tumor cell lines (SCC1, SCC12, and SCC14a). We also confirmed the expression of RANK and RANKL in primary human OSCC tumor specimens. However, regulatory mechanisms of RANKL expression and a functional role in OSCC tumor progression are unclear. Interestingly, we identified that RANKL expression is autoregulated in OSCC tumor cells. The RANKL specific inhibitor osteoprotegerin (OPG) treatment to OSCC cells inhibits autoregulation of RANKL expression. Further, we showed conditioned media from RANKL CRISPR-Cas9 knockout OSCC cells significantly decreased osteoclast formation and bone resorption activity. In addition, RANKL increases OSCC tumor cell proliferation. RANKL treatment to OSCC cells demonstrated a dose-dependent increase in RANK intracellular adaptor protein, TRAF6 expression, and activation of IKK and IκB signaling molecules. We further identified that transcription factor NFATc2 mediates autoregulation of RANKL expression in OSCC cells. Thus, our results implicate RANKL autoregulation as a novel mechanism that facilitates OSCC tumor cell growth and osteoclast differentiation/bone destruction.
Collapse
Affiliation(s)
- Yuvaraj Sambandam
- Department of Pediatrics/Endocrinology, Darby Children's Research Institute, Medical University of South Carolina, Charleston, South Carolina
| | - Purushoth Ethiraj
- Department of Pediatrics/Endocrinology, Darby Children's Research Institute, Medical University of South Carolina, Charleston, South Carolina
| | - Jessica D Hathaway-Schrader
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Chad M Novince
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Ezhil Panneerselvam
- Department of Pediatrics/Endocrinology, Darby Children's Research Institute, Medical University of South Carolina, Charleston, South Carolina
| | | | - Sakamuri V Reddy
- Department of Pediatrics/Endocrinology, Darby Children's Research Institute, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
29
|
Wei Y, Lin C, Li H, Xu Z, Wang J, Li R, Liu H, Zhang H, He H, Xu J. CXCL13 expression is prognostic and predictive for postoperative adjuvant chemotherapy benefit in patients with gastric cancer. Cancer Immunol Immunother 2018; 67:261-269. [PMID: 29085997 PMCID: PMC11028286 DOI: 10.1007/s00262-017-2083-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 10/18/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Chemokine (C-X-C motif) ligand 13 (CXCL13/BLC/BCA-1) is a cytokine from C-X-C chemokine family, which is selectively chemotactic for B cells. Previous research has demonstrated that high CXCL13 expression is correlated to poor prognosis in various cancers. However, the association between CXCL13 expression and gastric cancer is still unclear. METHODS Intratumoral CXCL13 expression was evaluated by immunohistochemistry using a semi-quantitative method (modified H-score) in a testing set of 214 and a validation set of 227 randomly selected gastric cancer patients resected in 2008 in one institution. The median value was used as the cut-off point. We performed correlative analysis of CXCL-13 expression with clinicopathological variables, Kaplan-Meier analysis for association with overall survival (OS), and multivariate modeling. RESULTS High CXCL13 expression was associated with larger tumor diameter and shorter OS. By multivariate analysis, CXCL13 expression was associated with OS independently from clinicopathological factors. Within the T2-4 stage patients group, low CXCL13 expression was associated with longer survival, especially in the subgroup of patients (57.6%) who received adjuvant chemotherapy. CONCLUSIONS Intratumoral CXCL13 expression appears as an independent prognostic marker for patients after gastric cancer resection. In addition, CXCL13 expression may serve as a predictive biomarker of response to postoperative adjuvant chemotherapy in these patients.
Collapse
Affiliation(s)
- Yichou Wei
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Room 358, Building West 7, 130 Dongan Rd, Shanghai, 200032, China
| | - Chao Lin
- Department of General Surgery, Zhongshan Hospital, Fudan University, Room 1004, Building No.1, 180 Fenglin Rd, Shanghai, 200032, China
| | - He Li
- Department of General Surgery, Zhongshan Hospital, Fudan University, Room 1004, Building No.1, 180 Fenglin Rd, Shanghai, 200032, China
| | - Zhiying Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Room 358, Building West 7, 130 Dongan Rd, Shanghai, 200032, China
| | - Jieti Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Room 358, Building West 7, 130 Dongan Rd, Shanghai, 200032, China
| | - Ruochen Li
- Department of General Surgery, Zhongshan Hospital, Fudan University, Room 1004, Building No.1, 180 Fenglin Rd, Shanghai, 200032, China
| | - Hao Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Room 1004, Building No.1, 180 Fenglin Rd, Shanghai, 200032, China
| | - Heng Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Room 1004, Building No.1, 180 Fenglin Rd, Shanghai, 200032, China
| | - Hongyong He
- Department of General Surgery, Zhongshan Hospital, Fudan University, Room 1004, Building No.1, 180 Fenglin Rd, Shanghai, 200032, China.
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Room 358, Building West 7, 130 Dongan Rd, Shanghai, 200032, China.
| |
Collapse
|
30
|
B Cell-Attracting Chemokine-1 and Progranulin in Bronchoalveolar Lavage Fluid of Patients with Advanced Non-small Cell Lung Cancer: New Prognostic Factors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1150:11-16. [PMID: 30357709 DOI: 10.1007/5584_2018_285] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Progranulin is a growth and survival factor implicated in tumorigenesis and drug resistance. Several studies showed that progranulin is expressed in breast cancer tissue and inversely correlates with survival. B lymphocyte chemoattractant, also known as B cell-attracting chemokine 1 (BCA-1), is a member of the CXC subtype of the chemokine superfamily. BCA-1 is critical for secondary lymphoid tissue development and navigation of lymphocytes within the microcompartments of the tissue. There are no data on the content of progranulin and BCA-1 in bronchoalveolar lavage fluid (BALF) of non-small cell lung cancer (NSCLC) patients. To study this issue, we measured BALF content of progranulin and BCA-1 in 46 NSCLC patients before chemotherapy and 15 healthy subjects. Both markers were elevated in cancer patients compared to healthy subjects (progranulin: 61.4 (1.6-384.0) vs. 6.5 (0.6-12.9) ng/ml, p = 0.001 and BCA-1: 30.8 (24.3-70.8) vs. 15.4 (13.3-19.5) pg/ml, p = 0.0001). The cut-off BALF level concerning NSCLC vs. controls, investigated using the receiver-operating characteristic (ROC) curve, yielded 6.5 ng/ml for progranulin and 15.4 pg/ml for BCA-1. We failed to find any association between the BALF content of progranulin or BCA-1 and the stage of tumor or prospectively assessed treatment response. However, BALF progranulin associated with time to tumor progression (r = 0.61; p = 0.04). In addition, a higher BALF content of BCA-1 in NSCLC patients associated with shorter overall survival. We conclude that progranulin and BCA-1 in BALF of NSCLC patients before chemotherapy may be prognostic factors of cancer progression.
Collapse
|
31
|
Protein Kinase C Epsilon Cooperates with PTEN Loss for Prostate Tumorigenesis through the CXCL13-CXCR5 Pathway. Cell Rep 2017; 19:375-388. [PMID: 28402859 DOI: 10.1016/j.celrep.2017.03.042] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 02/01/2017] [Accepted: 03/13/2017] [Indexed: 11/22/2022] Open
Abstract
PKCε, an oncogenic member of the PKC family, is aberrantly overexpressed in epithelial cancers. To date, little is known about functional interactions of PKCε with other genetic alterations, as well as the effectors contributing to its tumorigenic and metastatic phenotype. Here, we demonstrate that PKCε cooperates with the loss of the tumor suppressor Pten for the development of prostate cancer in a mouse model. Mechanistic analysis revealed that PKCε overexpression and Pten loss individually and synergistically upregulate the production of the chemokine CXCL13, which involves the transcriptional activation of the CXCL13 gene via the non-canonical nuclear factor κB (NF-κB) pathway. Notably, targeted disruption of CXCL13 or its receptor, CXCR5, in prostate cancer cells impaired their migratory and tumorigenic properties. In addition to providing evidence for an autonomous vicious cycle driven by PKCε, our studies identified a compelling rationale for targeting the CXCL13-CXCR5 axis for prostate cancer treatment.
Collapse
|
32
|
Irshad S, Flores-Borja F, Lawler K, Monypenny J, Evans R, Male V, Gordon P, Cheung A, Gazinska P, Noor F, Wong F, Grigoriadis A, Fruhwirth GO, Barber PR, Woodman N, Patel D, Rodriguez-Justo M, Owen J, Martin SG, Pinder SE, Gillett CE, Poland SP, Ameer-Beg S, McCaughan F, Carlin LM, Hasan U, Withers DR, Lane P, Vojnovic B, Quezada SA, Ellis P, Tutt ANJ, Ng T. RORγt + Innate Lymphoid Cells Promote Lymph Node Metastasis of Breast Cancers. Cancer Res 2017; 77:1083-1096. [PMID: 28082403 DOI: 10.1158/0008-5472.can-16-0598] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Revised: 12/09/2016] [Accepted: 12/10/2016] [Indexed: 11/16/2022]
Abstract
Cancer cells tend to metastasize first to tumor-draining lymph nodes, but the mechanisms mediating cancer cell invasion into the lymphatic vasculature remain little understood. Here, we show that in the human breast tumor microenvironment (TME), the presence of increased numbers of RORγt+ group 3 innate lymphoid cells (ILC3) correlates with an increased likelihood of lymph node metastasis. In a preclinical mouse model of breast cancer, CCL21-mediated recruitment of ILC3 to tumors stimulated the production of the CXCL13 by TME stromal cells, which in turn promoted ILC3-stromal interactions and production of the cancer cell motile factor RANKL. Depleting ILC3 or neutralizing CCL21, CXCL13, or RANKL was sufficient to decrease lymph node metastasis. Our findings establish a role for RORγt+ILC3 in promoting lymphatic metastasis by modulating the local chemokine milieu of cancer cells in the TME. Cancer Res; 77(5); 1083-96. ©2017 AACR.
Collapse
Affiliation(s)
- Sheeba Irshad
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
| | - Fabian Flores-Borja
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Katherine Lawler
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
- Institute for Mathematical and Molecular Biomedicine, King's College London, London, United Kingdom
| | - James Monypenny
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Rachel Evans
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Victoria Male
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
| | - Peter Gordon
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Anthony Cheung
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Patrycja Gazinska
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
| | - Farzana Noor
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
| | - Felix Wong
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Anita Grigoriadis
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
| | - Gilbert O Fruhwirth
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
- Leukocyte Dynamics Group, Beatson Advanced Imaging Resource, CRUK Beatson Institute, Glasgow, United Kingdom
| | - Paul R Barber
- Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, United Kingdom
| | - Natalie Woodman
- King's Health Partners Cancer Biobank, King's College London, London, United Kingdom
| | - Dominic Patel
- International Center for Infectiology Research, University of Lyon, Lyon, France
| | | | - Julie Owen
- King's Health Partners Cancer Biobank, King's College London, London, United Kingdom
| | - Stewart G Martin
- Division of Cancer and Stem Cells, Department of Clinical Oncology, School of Medicine, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
| | - Sarah E Pinder
- King's Health Partners Cancer Biobank, King's College London, London, United Kingdom
- Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, London, United Kingdom
| | - Cheryl E Gillett
- King's Health Partners Cancer Biobank, King's College London, London, United Kingdom
- Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, London, United Kingdom
| | - Simon P Poland
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Simon Ameer-Beg
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
| | - Frank McCaughan
- Department of Asthma, Allergy, and Lung Biology, King's College London, London, United Kingdom
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Leo M Carlin
- Leukocyte Dynamics Group, Beatson Advanced Imaging Resource, CRUK Beatson Institute, Glasgow, United Kingdom
| | - Uzma Hasan
- International Center for Infectiology Research, University of Lyon, Lyon, France
| | - David R Withers
- MRC Centre for Immune Regulation, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Peter Lane
- MRC Centre for Immune Regulation, Institute for Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Borivoj Vojnovic
- Gray Institute for Radiation Oncology & Biology, University of Oxford, Oxford, United Kingdom
| | - Sergio A Quezada
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London, United Kingdom
| | - Paul Ellis
- Department of Medical Oncology, Guy's and St Thomas Foundation Trust, London, United Kingdom
| | - Andrew N J Tutt
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom
- ICR, BCN Research Unit, Toby Robins Research Centre, London, United Kingdom
| | - Tony Ng
- Breast Cancer Now (BCN) Research Unit, King's College London, London, United Kingdom.
- Richard Dimbleby, Randall Division & Division of Cancer Studies, King's College London, London, United Kingdom
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London, United Kingdom
| |
Collapse
|
33
|
Sambandam Y, Sakamuri S, Balasubramanian S, Haque A. RANK Ligand Modulation of Autophagy in Oral Squamous Cell Carcinoma Tumor Cells. J Cell Biochem 2016; 117:118-25. [PMID: 26095774 DOI: 10.1002/jcb.25255] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 06/08/2015] [Indexed: 01/08/2023]
Abstract
Autophagy is a cellular process to recycle nutrients and has been implicated in cancer treatment. Oral squamous cell carcinoma (OSCC) is the most common oral cancer which ranks 3% of cancers in men and 2% in women. In this study, immunohistochemical staining of OSCC tumor specimens from human subjects and an athymic mouse model demonstrated high levels of autophagy markers LC3-II and ATG5 expression. Further, we identified high levels LC3-II expression in OSCC tumor cell lines (SCC-1, SCC-12, and SCC-14a) compared to normal human epithelial (RWPE-1) cells. OSCC cells express high levels of RANK ligand (RANKL); however, a functional role in autophagy is unknown. Interestingly, RANKL stimulation significantly increased autophagosome-related gene expressions such as LC3, ATG5, BECN1, and PI3KC3 mRNA expression in OSCC cells. Further, Western blot analysis of total cell lysates demonstrated a dose-dependent increase in LC3-II and ATG5 expression in RANKL-stimulated cells. In addition, RANKL increased expression of LC3-I and LC3-II, essential for autophagosome formation. Confocal microscopy analysis of LC3-II and localization with lysosome further confirms autophagosome formation in response to RANKL treatment in OSCC cells. Collectively, our results indicate a novel function of RANKL to induce autophagosome formation, and could be a potential therapeutic target to control OSCC tumor progression.
Collapse
Affiliation(s)
- Yuvaraj Sambandam
- Darby Children's Research Institute, Medical University of South Carolina, Charleston, South Carolina, 29425
| | - Sashank Sakamuri
- Department of Microbiology and Immunology and Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, 29425
| | | | - Azizul Haque
- Department of Microbiology and Immunology and Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina, 29425
| |
Collapse
|
34
|
Varga N, Mózes J, Keegan H, White C, Kelly L, Pilkington L, Benczik M, Zsuzsanna S, Sobel G, Koiss R, Babarczi E, Nyíri M, Kovács L, Attila S, Kaltenecker B, Géresi A, Kocsis A, O'Leary J, Martin CM, Jeney C. The Value of a Novel Panel of Cervical Cancer Biomarkers for Triage of HPV Positive Patients and for Detecting Disease Progression. Pathol Oncol Res 2016; 23:295-305. [PMID: 27497597 DOI: 10.1007/s12253-016-0094-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/14/2016] [Indexed: 01/12/2023]
Abstract
In the era of primary vaccination against HPV and at the beginning of the low prevalence of cervical lesions, introduction of screening methods that can distinguish between low- and high-grade lesions is necessary in order to maintain the positive predictive value of screening. This case-control study included 562 women who attended cervical screening or were referred for colposcopy and 140 disease free controls, confirmed by histology and/or cytology. The cases were stratified by age. Using routine exfoliated liquid based cytological samples RT-PCR measurements of biomarker genes, high-risk HPV testing and liquid based cytology were performed and used to evaluate different testing protocols including sets of genes/tests with different test cut-offs for the diagnostic panels. Three new panels of cellular biomarkers for improved triage of hrHPV positive women (diagnostic panel) and for prognostic assessment of CIN lesions were proposed. The diagnostic panel (PIK3AP1, TP63 and DSG3) has the potential to distinguish cytologically normal hrHPV+ women from hrHPV+ women with CIN2+. The prognostic gene panels (KRT78, MUC5AC, BPIFB1 and CXCL13, TP63, DSG3) have the ability to differentiate hrHPV+ CIN1 and carcinoma cases. The diagnostic triage panel showed good likelihood ratios for all age groups. The panel showed age-unrelated performance and even better diagnostic value under age 30, a unique feature among the established cervical triage tests. The prognostic gene-panels demonstrated good discriminatory power and oncogenic, anti-oncogenic grouping of genes. The study highlights the potential for the gene expression panels to be used for diagnostic triage and lesion prognostics in cervical cancer screening.
Collapse
Affiliation(s)
- Norbert Varga
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Johanna Mózes
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Helen Keegan
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Christine White
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Lynne Kelly
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Loretto Pilkington
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Márta Benczik
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Schaff Zsuzsanna
- 2nd Department of Pathology, Semmelweis University, Üllöi út 93, Budapest, 1091, Hungary
| | - Gábor Sobel
- 2nd Department of Obstetrics and Gynecology, Semmelweis University, Üllői út 78/a, Budapest, 1082, Hungary
| | - Róbert Koiss
- Department of Gynecology-Oncology, United Hospital of St. Stephan and Laszlo, Nagyvárad tér 1, Budapest, 1087, Hungary
| | - Edit Babarczi
- Department of Gynecology-Oncology, United Hospital of St. Stephan and Laszlo, Nagyvárad tér 1, Budapest, 1087, Hungary
| | - Miklos Nyíri
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Laura Kovács
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Sebe Attila
- Institute of Pathophysiology, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary
| | | | - Adrienn Géresi
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - Adrienn Kocsis
- CellCall Ltd, Röppentyű utca 48, Budapest, 1134, Hungary
| | - John O'Leary
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Cara M Martin
- Department of Histopathology, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Molecular Pathology Laboratory, Department of Pathology, Coombe Women and Infants University Hospital, Dublin, 8, Ireland
| | - Csaba Jeney
- Department of Medical Microbiology, Semmelweis University, Nagyvárad tér 4, Budapest, 1089, Hungary.
| |
Collapse
|
35
|
Panda S, Padhiary SK, Routray S. Chemokines accentuating protumoral activities in oral cancer microenvironment possess an imperious stratagem for therapeutic resolutions. Oral Oncol 2016; 60:8-17. [PMID: 27531867 DOI: 10.1016/j.oraloncology.2016.06.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 05/01/2016] [Accepted: 06/10/2016] [Indexed: 12/15/2022]
Abstract
Chemokines, the chemotactic cytokines have established their role in tumorigenesis and tumor progression. Studies, which explored their role in oral cancer for protumoral activity, point towards targeting chemokines for oral squamous cell carcinoma therapy. The need of the hour is to emphasize/divulge in the activities of chemokine ligands and their receptors in the tumor microenvironment for augmentation of such stratagems. This progressing sentience of chemokines and their receptors has inspired this review which is an endeavour to comprehend their role as an aid in accentuating hallmarks of cancer and targeted therapy.
Collapse
Affiliation(s)
- Swagatika Panda
- Department of Oral Pathology and Microbiology, Institute of Dental Sciences, Siksha 'O' Anusandhan University, Bhubaneswar 751030, India.
| | - Subrat Kumar Padhiary
- Department of Oral and Maxillofacial Surgery, Institute of Dental Sciences, Siksha 'O' Anusandhan University, Bhubaneswar 751030, India.
| | - Samapika Routray
- Department of Oral Pathology and Microbiology, Institute of Dental Sciences, Siksha 'O' Anusandhan University, Bhubaneswar 751030, India.
| |
Collapse
|
36
|
da Silva JM, Soave DF, Moreira dos Santos TP, Batista AC, Russo RC, Teixeira MM, Silva TAD. Significance of chemokine and chemokine receptors in head and neck squamous cell carcinoma: A critical review. Oral Oncol 2016; 56:8-16. [DOI: 10.1016/j.oraloncology.2016.02.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 01/14/2016] [Accepted: 02/29/2016] [Indexed: 12/17/2022]
|
37
|
Samadi AK, Bilsland A, Georgakilas AG, Amedei A, Amin A, Bishayee A, Azmi AS, Lokeshwar BL, Grue B, Panis C, Boosani CS, Poudyal D, Stafforini DM, Bhakta D, Niccolai E, Guha G, Vasantha Rupasinghe HP, Fujii H, Honoki K, Mehta K, Aquilano K, Lowe L, Hofseth LJ, Ricciardiello L, Ciriolo MR, Singh N, Whelan RL, Chaturvedi R, Ashraf SS, Shantha Kumara HMC, Nowsheen S, Mohammed SI, Keith WN, Helferich WG, Yang X. A multi-targeted approach to suppress tumor-promoting inflammation. Semin Cancer Biol 2015; 35 Suppl:S151-S184. [PMID: 25951989 PMCID: PMC4635070 DOI: 10.1016/j.semcancer.2015.03.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 03/13/2015] [Accepted: 03/16/2015] [Indexed: 12/15/2022]
Abstract
Cancers harbor significant genetic heterogeneity and patterns of relapse following many therapies are due to evolved resistance to treatment. While efforts have been made to combine targeted therapies, significant levels of toxicity have stymied efforts to effectively treat cancer with multi-drug combinations using currently approved therapeutics. We discuss the relationship between tumor-promoting inflammation and cancer as part of a larger effort to develop a broad-spectrum therapeutic approach aimed at a wide range of targets to address this heterogeneity. Specifically, macrophage migration inhibitory factor, cyclooxygenase-2, transcription factor nuclear factor-κB, tumor necrosis factor alpha, inducible nitric oxide synthase, protein kinase B, and CXC chemokines are reviewed as important antiinflammatory targets while curcumin, resveratrol, epigallocatechin gallate, genistein, lycopene, and anthocyanins are reviewed as low-cost, low toxicity means by which these targets might all be reached simultaneously. Future translational work will need to assess the resulting synergies of rationally designed antiinflammatory mixtures (employing low-toxicity constituents), and then combine this with similar approaches targeting the most important pathways across the range of cancer hallmark phenotypes.
Collapse
Affiliation(s)
| | - Alan Bilsland
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematics and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Amr Amin
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates; Faculty of Science, Cairo University, Cairo, Egypt
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin Health Sciences Institute, Miami, FL, United States
| | - Asfar S Azmi
- Department of Pathology, Wayne State Univeristy, Karmanos Cancer Center, Detroit, MI, USA
| | - Bal L Lokeshwar
- Department of Urology, University of Miami, Miller School of Medicine, Miami, FL, United States; Miami Veterans Administration Medical Center, Miami, FL, United States
| | - Brendan Grue
- Department of Environmental Science, Dalhousie University, Halifax, Nova Scotia, Canada; Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Carolina Panis
- Laboratory of Inflammatory Mediators, State University of West Paraná, UNIOESTE, Paraná, Brazil
| | - Chandra S Boosani
- Department of BioMedical Sciences, School of Medicine, Creighton University, Omaha, NE, United States
| | - Deepak Poudyal
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Diana M Stafforini
- Huntsman Cancer Institute and Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Dipita Bhakta
- School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | | | - Gunjan Guha
- School of Chemical and Biotechnology, SASTRA University, Thanjavur, Tamil Nadu, India
| | - H P Vasantha Rupasinghe
- Department of Environmental Sciences, Faculty of Agriculture and Department of Pathology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Hiromasa Fujii
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Kanya Honoki
- Department of Orthopedic Surgery, Nara Medical University, Kashihara, Nara, Japan
| | - Kapil Mehta
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Katia Aquilano
- Department of Biology, University of Rome "Tor Vergata", Rome, Italy
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia, Canada.
| | - Lorne J Hofseth
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Luigi Ricciardiello
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advanced Research), King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Richard L Whelan
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Rupesh Chaturvedi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - S Salman Ashraf
- Department of Chemistry, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - H M C Shantha Kumara
- Department of Surgery, St. Luke's Roosevelt Hospital, New York, NY, United States
| | - Somaira Nowsheen
- Medical Scientist Training Program, Mayo Graduate School, Mayo Medical School, Mayo Clinic, Rochester, MN, United States
| | - Sulma I Mohammed
- Department of Comparative Pathobiology, Purdue University Center for Cancer Research, West Lafayette, IN, United States
| | - W Nicol Keith
- Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland, UK
| | | | - Xujuan Yang
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| |
Collapse
|
38
|
Tsai YM, Chong IW, Hung JY, Chang WA, Kuo PL, Tsai MJ, Hsu YL. Syringetin suppresses osteoclastogenesis mediated by osteoblasts in human lung adenocarcinoma. Oncol Rep 2015; 34:617-26. [PMID: 26044862 DOI: 10.3892/or.2015.4028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 05/04/2015] [Indexed: 11/05/2022] Open
Abstract
Bone metastasis in lung cancer results in an unfavorable outcome for patients by not only impairing the quality of life, yet also increasing the cancer-related death rates. In the present study, we discuss a novel treatment strategy that may benefit these patients. Human CD14+ monocytes treated with macrophage-colony stimulating factor (M-CSF)/receptor activator of nuclear factor κB ligand (RANKL) differentiated into osteoclasts, whereas syringetin (SGN), a flavonoid derivative found in both grapes and wine, suppressed the osteoclastogenesis in vitro in a dose-dependent manner. In addition, SGN inhibited osteoclast formation induced by human lung adenocarcinoma A549 and CL1-5 cells. The associated signaling transduction pathway in osteoclastogenesis and SGN inhibition was found to be via the AKT/mammalian target of rapamycin (mTOR) signaling pathway. Blocking AKT and mTOR by respective inhibitors significantly decreased lung adenocarcinoma-mediated osteoclastogenesis. Moreover, SGN regulated the lung adenocarcinoma-mediated interaction between osteoblasts and osteoclasts by suppressing the stimulatory effect of lung adenocarcinoma on M-CSF and RANKL production in osteoblasts, and reversing the inhibitory effect of the lung adenocarcinoma on OPG production in osteoblasts. The present study has two novel findings. It is the first to illustrate lung adenocarcinoma-mediated interaction between osteoblasts and osteoclasts, leading to osteolytic bone metastasis. It also reveals that SGN, a flavonoid derivative, directly inhibits osteoclastogenesis and reverses lung adenocarcinoma-mediated osteoclastogenesis. In conclusion, the present study suggests that SGN, a natural compound, prevents and treats bone metastasis in patients with lung cancer.
Collapse
Affiliation(s)
- Ying-Ming Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Inn-Wen Chong
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Jen-Yu Hung
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Wei-An Chang
- Division of Pulmonary and Critical Care Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Po-Lin Kuo
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Ming-Ju Tsai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| |
Collapse
|
39
|
CXCL13-CXCR5 axis promotes the growth and invasion of colon cancer cells via PI3K/AKT pathway. Mol Cell Biochem 2014; 400:287-95. [PMID: 25476740 DOI: 10.1007/s11010-014-2285-y] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/15/2014] [Indexed: 12/11/2022]
Abstract
CXCL13, an inflammatory factor in the microenvironment, plays a vital role in the progression of inflammatory diseases and tumors. CXCL13 and its receptor CXCR5 have been reported to be associated with poor prognosis of advanced colon cancer. However, the molecular mechanisms of CXCL13-CXCR5 axis in colon cancer remain elusive. The aim of this study was to investigate the role of CXCR5-CXCL13 axis in the growth and invasion of colon cancer cells. Our results showed that CXCL13 promoted the growth, migration, and matrigel invasion of colon cancer cells. Furthermore, CXCL13 increased the expression and secretion of MMP-13, and stimulated the activation of PI3K/AKT pathway. After knockdown of CXCR5 by siRNA, the biological functions of colon cancer cells regulated by CXCL13 were significantly inhibited. In addition, inhibition of PI3K/AKT pathway by specific inhibitor LY294002 suppressed the CXCL13-mediated growth, migration, and invasion of colon cancer cells. Together, our findings suggest that CXCL13-CXCR5 axis promotes the growth, migration, and invasion of colon cancer cells, probably via PI3K/AKT pathway. Thus, CXCL13 may be a useful biomarker for the detection and treatment of colon cancer.
Collapse
|
40
|
Zhou J, Xiang Y, Yoshimura T, Chen K, Gong W, Huang J, Zhou Y, Yao X, Bian X, Wang JM. The role of chemoattractant receptors in shaping the tumor microenvironment. BIOMED RESEARCH INTERNATIONAL 2014; 2014:751392. [PMID: 25110692 PMCID: PMC4119707 DOI: 10.1155/2014/751392] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 06/17/2014] [Indexed: 12/13/2022]
Abstract
Chemoattractant receptors are a family of seven transmembrane G protein coupled receptors (GPCRs) initially found to mediate the chemotaxis and activation of immune cells. During the past decades, the functions of these GPCRs have been discovered to not only regulate leukocyte trafficking and promote immune responses, but also play important roles in homeostasis, development, angiogenesis, and tumor progression. Accumulating evidence indicates that chemoattractant GPCRs and their ligands promote the progression of malignant tumors based on their capacity to orchestrate the infiltration of the tumor microenvironment by immune cells, endothelial cells, fibroblasts, and mesenchymal cells. This facilitates the interaction of tumor cells with host cells, tumor cells with tumor cells, and host cells with host cells to provide a basis for the expansion of established tumors and development of distant metastasis. In addition, many malignant tumors of the nonhematopoietic origin express multiple chemoattractant GPCRs that increase the invasiveness and metastasis of tumor cells. Therefore, GPCRs and their ligands constitute targets for the development of novel antitumor therapeutics.
Collapse
Affiliation(s)
- Jiamin Zhou
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Endoscopic Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yi Xiang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
- Department of Pulmonary Medicine, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Teizo Yoshimura
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Keqiang Chen
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Wanghua Gong
- Basic Research Program, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Jian Huang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ye Zhou
- Department of Gastric Cancer and Soft Tissue Surgery, Fudan University Cancer Center, Shanghai 200032, China
| | - Xiaohong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Ji Ming Wang
- Laboratory of Molecular Immunoregulation, Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
41
|
Xing J, Li X, Sui J, Cao G, Fu C. C-X-C chemokine receptor type 5 gene polymorphism affects gene expression in CD4+ T cells and is associated with increased risk of colorectal cancer. Tumour Biol 2014; 35:7929-34. [DOI: 10.1007/s13277-014-2069-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 05/06/2014] [Indexed: 10/25/2022] Open
|
42
|
Pimenta EM, Barnes BJ. Role of Tertiary Lymphoid Structures (TLS) in Anti-Tumor Immunity: Potential Tumor-Induced Cytokines/Chemokines that Regulate TLS Formation in Epithelial-Derived Cancers. Cancers (Basel) 2014; 6:969-97. [PMID: 24762633 PMCID: PMC4074812 DOI: 10.3390/cancers6020969] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/19/2014] [Accepted: 03/31/2014] [Indexed: 12/12/2022] Open
Abstract
Following the successes of monoclonal antibody immunotherapies (trastuzumab (Herceptin®) and rituximab (Rituxan®)) and the first approved cancer vaccine, Provenge® (sipuleucel-T), investigations into the immune system and how it can be modified by a tumor has become an exciting and promising new field of cancer research. Dozens of clinical trials for new antibodies, cancer and adjuvant vaccines, and autologous T and dendritic cell transfers are ongoing in hopes of identifying ways to re-awaken the immune system and force an anti-tumor response. To date, however, few consistent, reproducible, or clinically-relevant effects have been shown using vaccine or autologous cell transfers due in part to the fact that the immunosuppressive mechanisms of the tumor have not been overcome. Much of the research focus has been on re-activating or priming cytotoxic T cells to recognize tumor, in some cases completely disregarding the potential roles that B cells play in immune surveillance or how a solid tumor should be treated to maximize immunogenicity. Here, we will summarize what is currently known about the induction or evasion of humoral immunity via tumor-induced cytokine/chemokine expression and how formation of tertiary lymphoid structures (TLS) within the tumor microenvironment may be used to enhance immunotherapy response.
Collapse
Affiliation(s)
- Erica M Pimenta
- Rutgers Biomedical and Health Sciences, New Jersey Medical School-Cancer Center, Newark, NJ 07103, USA.
| | - Betsy J Barnes
- Department of Biochemistry and Molecular Biology, Rutgers Biomedical and Health Sciences, New Jersey Medical School-Cancer Center, Newark, NJ 07103, USA.
| |
Collapse
|
43
|
Abstract
Acting through its cognate receptor, receptor activator of nuclear factor-κB (RANK), RANK ligand (RANKL) is an essential mediator of osteoclast function and survival. Preclinical data have now firmly established that blockade of tumor-induced osteoclastogenesis by RANKL inhibition will not only protect against bone destruction but will also inhibit the progression of established bone metastases and delay the formation of de novo bone metastases in cancer models. In patients with bone metastases, skeletal complications are driven by increased osteoclastic activity and may result in pathological fractures, spinal cord compression and the need for radiotherapy to the bone or orthopedic surgery (collectively known as skeletal-related events (SREs)). Denosumab, a fully human monoclonal antibody against RANKL, has been demonstrated to prevent or delay SREs in patients with solid tumors that have metastasized to bone. In addition to its central role in tumor-induced osteolysis, bone destruction and skeletal tumor progression, there is emerging evidence for direct pro-metastatic effects of RANKL, independent of osteoclasts. For example, RANKL also stimulates metastasis via activity on RANK-expressing cancer cells, resulting in increased invasion and migration. Pharmacological inhibition of RANKL may also reduce bone and lung metastasis through blockade of the direct action of RANKL on metastatic cells. This review describes these distinct but potentially overlapping mechanisms by which RANKL may promote metastases.
Collapse
|
44
|
Feigin ME. Harnessing the genome for characterization of G-protein coupled receptors in cancer pathogenesis. FEBS J 2013; 280:4729-38. [PMID: 23927072 PMCID: PMC4283816 DOI: 10.1111/febs.12473] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Revised: 07/31/2013] [Accepted: 08/01/2013] [Indexed: 12/21/2022]
Abstract
G-protein coupled receptors (GPCRs) mediate numerous physiological processes and represent the targets for a vast array of therapeutics for diseases ranging from depression to hypertension to reflux. Despite the recognition that GPCRs can act as oncogenes and tumour suppressors by regulating oncogenic signalling networks, few drugs targeting GPCRs are utilized in cancer therapy. Recent large-scale genome-wide analyses of multiple human tumours have uncovered novel GPCRs altered in cancer. However, work aiming to determine which GPCRs from these lists are the drivers of tumourigenesis, and hence valid therapeutic targets, comprises a formidable challenge. The present review highlights recent studies providing evidence that GPCRs are relevant targets for cancer therapy through their effects on known cancer signalling pathways, tumour progression, invasion and metastasis, and the microenvironment. Furthermore, the review also explores how genomic analysis is beginning to highlight GPCRs as therapeutic targets in the age of personalized medicine.
Collapse
Affiliation(s)
- Michael E. Feigin
- Cold Spring Harbor Laboratory, 1 Bungtown Road, Cold Spring Harbor, NY 11724, 516-367-8385 (phone)
| |
Collapse
|
45
|
Rochefort GY, Benhamou CL. Osteocytes are not only mechanoreceptive cells. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2013; 29:1082-1088. [PMID: 23776074 DOI: 10.1002/cnm.2561] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 03/25/2013] [Accepted: 04/23/2013] [Indexed: 06/02/2023]
Abstract
Osteocytes represent 95% of all bone cells. These cells are old osteoblasts occupying the lacunar space surrounded by the bone matrix. They possess cytoplasmic dendrites that form a canalicular network for communication between osteocytes and the bone surface. They have a mechanosensing role that is dependent upon the frequency, the intensity, and the duration of strain. The mechanical information transmitted into the cytoplasm also triggers a biological cascade, starting with nitric oxide and prostaglandin E 2 and followed by Wnt/ β-catenin signaling. This information is transmitted to the bone surface through the canalicular network, particularly to the lining cells, and is able to trigger bone remodeling by directing the osteoblast activity and the osteoclastic resorption. Furthermore, the osteocyte death seems to play an important role. The outcome of microcracks in the vicinity of osteocytes may interrupt the canalicular network and trigger cell apoptosis in the immediate surrounding environment thus transmitting a message to the bone surface and activate remodeling. This network also plays a recognized endocrine role, particularly concerning phosphate regulation and vitamin D metabolism.
Collapse
|
46
|
Kuo CH, Chen KF, Chou SH, Huang YF, Wu CY, Cheng DE, Chen YW, Yang CJ, Hung JY, Huang MS. Lung tumor-associated dendritic cell-derived resistin promoted cancer progression by increasing Wolf-Hirschhorn syndrome candidate 1/Twist pathway. Carcinogenesis 2013; 34:2600-9. [PMID: 23955539 DOI: 10.1093/carcin/bgt281] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The interaction between tumors and their microenvironments leads to a vicious cycle, which strengthens both immune suppression and cancer progression. The present study demonstrates for the first time that tumor-associated dendritic cells (TADCs) are a source of resistin, which is responsible for increasing lung cancer epithelial-to-mesenchymal transition. In addition, large amounts of resistin in the condition medium (CM) of TADCs increase cell migration and invasion, as well as the osteolytic bone metastatic properties of lung cancer cells. Neutralization of resistin from TADC-CM prevents the advanced malignancy-inducing features of TADC-CM. Significantly elevated levels of resistin have been observed in mice transplanted with lung cancer cells, tumor-infiltrating CD11c(+) DCs in human lung cancer samples and lung cancer patients' sera. Induction of lung cancer progression by TADC-derived resistin is associated with increased expression of Wolf-Hirschhorn syndrome candidate 1 (WHSC1), a histone methyltransferase. Resistin-induced WHSC1 increases the dimethylation of histone 3 at lysine 36 and decreases the trimethylation of histone 3 at lysine 27 on the promoter of Twist, resulting in an enhancement of the expression of Twist. Knockdown of WHSC1 by small interfering RNA transfection significantly decreases resistin-mediated cancer progression by decreasing the upregulation of Twist, suggesting that WHSC1 plays a critical role in the regulation of Twist by epigenetic modification. Furthermore, mice that received antiresistin antibodies showed a decreased incidence of cancer development and metastasis. These findings suggest that TADC-derived resistin may be a novel candidate in promoting the development of lung cancer.
Collapse
Affiliation(s)
- Chih-Hsin Kuo
- The Affiliated Senior High School of National Kaohsiung Normal University, Kaohsiung 802, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Yedida GR, Nagini S, Mishra R. The importance of oncogenic transcription factors for oral cancer pathogenesis and treatment. Oral Surg Oral Med Oral Pathol Oral Radiol 2013; 116:179-88. [PMID: 23619350 DOI: 10.1016/j.oooo.2013.02.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 02/05/2013] [Accepted: 02/15/2013] [Indexed: 01/03/2023]
Abstract
Oral squamous cell carcinoma is a major cause of morbidity and mortality worldwide. Current experimental evidence shows that most important risk factors for oral cancer include tobacco use and excessive alcohol consumption and less well-defined risks include viral infection and a diet deficient in antioxidants. The positive correlation between various risk/etiologic factors of oral cancer and the activation of various transcription factors (TFs) has been reported in the literature. Although initially, TFs were considered to be very difficult targets for use in clinical treatment, recent technological advances have provided the ability to control these factors of cancer progression. This review focuses on the role of oncogenic transcription factors in oral cancer, their modes of activation through various biological pathways, the promises and pitfalls in viewing them as potent oncotargets, the way they can be controlled based on the current understanding, and the future research to be done in this area.
Collapse
Affiliation(s)
- Govinda Raju Yedida
- Centre for Life Sciences, School of Natural Sciences, Central University of Jharkhand, Ranchi, Jharkhand, India
| | | | | |
Collapse
|
48
|
Sundaram K, Rao DS, Ries WL, Reddy SV. CXCL5 stimulation of RANK ligand expression in Paget's disease of bone. J Transl Med 2013; 93:472-9. [PMID: 23439434 DOI: 10.1038/labinvest.2013.5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Paget's disease of bone (PDB) is a chronic focal skeletal disorder that affects 2-3% of the population over 55 years of age. PDB is marked by highly localized areas of bone turnover with increased osteoclast activity. Evidence suggests a functional role for measles virus nucleocapsid protein (MVNP) in the pathogenesis of PDB. In the present study, we identified elevated levels (≈ 180-fold) of CXCL5 mRNA expression in bone marrow cells from patients with PDB compared with that in normal subjects. In addition, CXCL5 levels are increased (five-fold) in serum samples from patients with PDB. Furthermore, MVNP transduction in human bone marrow monocytes significantly increased CXCL5 mRNA expression. Real-time PCR analysis showed that CXCL5 stimulation increased (6.8-fold) RANKL mRNA expression in normal human bone marrow-derived stromal (SAKA-T) cells. Moreover, CXCL5 increased (5.2-fold) CXCR1 receptor expression in these cells. We further showed that CXCL5 treatment elevated the expression levels of phospho-ERK1/2 and phospho-p38. CXCL5 also significantly increased phosphorylation of CREB (cAMP response element-binding) in bone marrow stromal/preosteoblast cells. Chromatin immuneprecipitation (ChIP) assay confirmed phospho-CREB binding to RANKL gene promoter region. Further, the suppression of p-CREB expression by the inhibitors of ERK1/2, p38 and PKA significantly decreased CXCL5 stimulation of hRANKL gene promoter activity. Thus, our results suggest that CREB is a downstream effector of CXCL5 signaling and that increased levels of CXCL5 contribute to enhanced levels of RANKL expression in PDB.
Collapse
Affiliation(s)
- Kumaran Sundaram
- Charles P Darby Children's Research Institute, Charleston, SC 29425, USA
| | | | | | | |
Collapse
|
49
|
Zhu Q, Han X, Peng J, Qin H, Wang Y. The role of CXC chemokines and their receptors in the progression and treatment of tumors. J Mol Histol 2012; 43:699-713. [PMID: 22752457 DOI: 10.1007/s10735-012-9435-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 06/21/2012] [Indexed: 12/25/2022]
Abstract
Chemokines are a class of functional chemotactic peptides that contribute to a number of tumor-related processes. They are functionally defined as soluble factors that are able to control the directional migration of leukocytes, in particular, during infection and inflammation. It appears, however, that the biological effects mediated by chemokines are far more complex, and virtually all cells, including many tumor cell types, can express chemokines and chemokine receptors. A growing body of evidence indicates that they also contribute to a number of tumor-related processes, such as tumor cell growth, angiogenesis/angiostasis, local invasion, and mediate organ-specific metastases of cancer. The CXC chemokine class is a subfamily of a large family of chemokines. During the occurrence and development of tumor cells, this chemokine class is often accompanied by a series of molecular and biological changes. The CXC chemokine subfamily is closely related to the body's immune response to tumors and biological behaviors of tumors. In this paper, CXC chemokines and their role in the progression and treatment of tumors will be reviewed.
Collapse
Affiliation(s)
- Qingchao Zhu
- Department of Surgery, The Sixth People's Hospital Affiliated to Shanghai Jiao Tong University, 600 Yishan Road, Shanghai, 200233, People's Republic of China.
| | | | | | | | | |
Collapse
|