1
|
Hu X, Wu H, Hu K, Kang Y, Hua G, Cheng M, Yan W, Huang W. Establishing patient-derived tumor organoids of bone metastasis from lung adenocarcinoma reveals the transcriptomic changes underlying denosumab treatment. Clin Exp Metastasis 2024; 42:8. [PMID: 39739069 DOI: 10.1007/s10585-024-10321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 11/17/2024] [Indexed: 01/02/2025]
Abstract
Patient-derived tumor organoids (PDTOs) models have been widely used to investigate the response of primary cancer tissues to anti-cancer agents. Nonetheless, only few case study tried to establish PDTOs and test treatment response based on bone metastasis (BoM) tissues. Fresh BoM tissues were obtained from lung cancer (LC) patients who underwent spinal metastatic tumor surgery for PDTOs culture. Morphology of LC-BoM-PDTOs were characterized during the process: they were high-efficient in self-assembly and regeneration, forming mature 3D-multicellular structures in 2-3 weeks. To be more specific, organoids of BoM derived from patients with EGFR mutation tended to be follicular conglomeration and resembled "a bunch of grapes", while organoids of BoM derived from patients without driver gene mutation were featured with full sphere and "a ripe sunflower". PDTOs of BoM retained good consistencies of HE morphology and immunohistochemical markers expression with their parental BoM tissues. Down-regulation of receptor activator of nuclear factor kappa-B ligand (RANKL) expression in LC-BoM-PDTOs after in vitro DMAb intervention was associated with earlier clinical ossification efficacy of DMAb on BoM (median time: 5 vs. 8 months, P = 0.049). Accordingly, BoM-PDTOs can be expected to be a preferred model for predicting treatment response of bone metastatic tumors, considering its high-efficient expansion and good biological consistency with parental bone tumor tissues.
Collapse
Affiliation(s)
- Xianglin Hu
- Department of Musculoskeletal Oncology, Spine Tumor Center, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Huajian Wu
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center Xiamen Hospital, Xiamen, China
| | - Kewen Hu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yani Kang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Guoqiang Hua
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Research and Early Development (RED) of D1 Medical Technology Company, Shanghai, China
| | - Mo Cheng
- Department of Musculoskeletal Oncology, Spine Tumor Center, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Wangjun Yan
- Department of Musculoskeletal Oncology, Spine Tumor Center, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Wending Huang
- Department of Musculoskeletal Oncology, Spine Tumor Center, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Wu D, Liu Y, Liu J, Ma L, Tong X. Myeloid cell differentiation-related gene signature for predicting clinical outcome, immune microenvironment, and treatment response in lung adenocarcinoma. Sci Rep 2024; 14:17460. [PMID: 39075165 PMCID: PMC11286868 DOI: 10.1038/s41598-024-68111-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/19/2024] [Indexed: 07/31/2024] Open
Abstract
Considering the key role of myeloid cell differentiation-related genes in the tumor microenvironment (TME), we aimed to build a prognostic risk model using these genes for Lung adenocarcinoma (LUAD). The mRNA gene expression profiles of LUAD patients from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were downloaded as the training and validation sets. Then, "edgeR" R package was applied to screen out the differentially expressed genes (DEGs) and univariate cox regression, backward stepwise selection analyses were performed to construct a prognostic model for LUAD. ESTIMATE, TIMER, XCELL, CIBERSORT abs, QUANTISEQ, MCPCOUNTER, EPIC, and CIBERSORT algorithms were conducted to access the association of risk levels with the stromal and immune cell infiltration levels in LUAD. Six genes (F2RL1, PRKDC, TNFSF11, INHA, PLA2G3 and TUBB1) were utilized to construct the prognostic model. The risk model showed excellent prognostic performance for LUAD in both TCGA and GEO datasets. Also, compared to the low-risk patients, the high-risk patients had higher expression of immune checkpoint molecules and showed a lower IC50 value to the chemotherapy agents. Our findings provided a myeloid cell differentiation-related gene signature that could effectively predict prognosis and guide treatment strategies for LUAD patients.
Collapse
Affiliation(s)
- Di Wu
- Experimental Research Center, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201700, China
| | - Yibing Liu
- Experimental Research Center, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201700, China
| | - Jian Liu
- Department of Otolaryngology-Head and Neck Surgery, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Li Ma
- Experimental Research Center, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201700, China
| | - Xiaoxia Tong
- Experimental Research Center, Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, 201700, China.
| |
Collapse
|
3
|
Li Z, Lu W, Yin F, Zeng P, Li H, Huang A. Overexpression of TNFSF11 reduces GPX4 levels and increases sensitivity to ferroptosis inducers in lung adenocarcinoma. J Transl Med 2024; 22:340. [PMID: 38594779 PMCID: PMC11005202 DOI: 10.1186/s12967-024-05112-y] [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: 09/30/2023] [Accepted: 03/20/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD), the most common and lethal subtype of lung cancer, continues to be a major health concern worldwide. Despite advances in targeted and immune therapies, only a minority of patients derive substantial benefits. As a result, the urgent need for novel therapeutic strategies to improve lung cancer treatment outcomes remains undiminished. METHODS In our study, we employed the TIMER database to scrutinize TNFSF11 expression across various cancer types. We further examined the differential expression of TNFSF11 in normal and tumor tissues utilizing the TCGA-LUAD dataset and tissue microarray, and probed the associations between TNFSF11 expression and clinicopathological parameters within the TCGA-LUAD dataset. We used the GSE31210 dataset for external validation. To identify genes strongly linked to TNFSF11, we engaged LinkedOmics and conducted a KEGG pathway enrichment analysis using the WEB-based Gene SeT AnaLysis Toolkit. Moreover, we investigated the function of TNFSF11 through gene knockdown or overexpression approaches and explore its function in tumor cells. The therapeutic impact of ferroptosis inducers in tumors overexpressing TNFSF11 were also investigated through in vivo and in vitro experiments. Through these extensive analyses, we shed light on the potential role of TNFSF11 in lung adenocarcinoma, underscoring potential therapeutic targets for this malignancy. RESULTS This research uncovers the overexpression of TNFSF11 in LUAD patients and its inverse correlation with peroxisome-related enzymes. By utilizing gene knockdown or overexpression assays, we found that TNFSF11 was negatively associated with GPX4. Furthermore, cells with TNFSF11 overexpression were relatively more sensitive to the ferroptosis inducers. CONCLUSIONS Our research has provided valuable insights into the role of TNFSF11, revealing its negative regulation of GPX4, which could be influential in crafting therapeutic strategies. These findings set the stage for further exploration into the mechanisms underpinning the relationship between TNFSF11 and GPX4, potentially opening up new avenues for precision medicine in the treatment of LUAD.
Collapse
Affiliation(s)
- Zizhen Li
- Department of Medical Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510000, China
| | - Wenhua Lu
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Guangzhou, 510000, China
| | - Feng Yin
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Guangzhou, 510000, China
| | - Peiting Zeng
- Department of Hematology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Heping Li
- Department of Medical Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510000, China.
| | - Amin Huang
- Department of Medical Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510000, China.
| |
Collapse
|
4
|
Fotia G, Stellato M, Guadalupi V, Sepe P, Claps M, Giannatempo P, Bottiglieri A, Rametta A, Taglialatela I, Vela C, Procopio G, Verzoni E. Current Status of Predictive Biomarker Development in Metastatic Renal Cell Carcinoma. Curr Oncol Rep 2023; 25:671-677. [PMID: 37000341 DOI: 10.1007/s11912-023-01395-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2023] [Indexed: 04/01/2023]
Abstract
PURPOSE OF REVIEW In this review, we analyze the current state of research in development of new biomarkers that may be useful in managing metastatic renal cell carcinoma (mRCC) setting. RECENT FINDINGS Combining tumor-based biomarkers (gene expression profile) and blood-based biomarkers (ctDNA, cytokines) would be helpful in acquiring information regarding RCC and might be significant in the decision-making process. Renal cell carcinoma (RCC) is the sixth most frequently diagnosed neoplasm in men and tithe in women, making it responsible for 5% and 3% of all diagnosed cancers respectively. Metastatic stage represents a non-negligible percentage at diagnosis and is characterized by poor prognosis. Despite clinical features and prognostic score could guide clinicians in therapeutic approach of this disease, biomarkers predictive of response to treatment remain an unmet need.
Collapse
Affiliation(s)
- Giuseppe Fotia
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Marco Stellato
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy.
| | - Valentina Guadalupi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Pierangela Sepe
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Melanie Claps
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Patrizia Giannatempo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Achille Bottiglieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Alessandro Rametta
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Ida Taglialatela
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Chiara Vela
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Giuseppe Procopio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| | - Elena Verzoni
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, Milan, Italy
| |
Collapse
|
5
|
Eichberger J, Weber F, Spanier G, Gerken M, Schreml S, Schulz D, Fiedler M, Ludwig N, Bauer RJ, Reichert TE, Ettl T. Loss of MMP-27 Predicts Mandibular Bone Invasion in Oral Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14164044. [PMID: 36011038 PMCID: PMC9406335 DOI: 10.3390/cancers14164044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/13/2022] [Accepted: 08/17/2022] [Indexed: 12/24/2022] Open
Abstract
Simple Summary The growth of oral squamous cell carcinoma into the mandible poses significant challenges to head and neck surgery. The resulting need for extensive procedures has a decisive influence on subsequent esthetics and function and therefore also on the patient’s quality of life. The molecular mechanism behind this remains obscure to date. Hence, we investigated the influence of MMP-27, Osteoprotegerin and RANKL, three proteins with importance in bone remodeling. The results showed that tumors exhibited less bone-invasive behavior in the presence of MMP-27. This may be an incentive for further studies to elucidate the molecular mechanisms of mandibular bone invasion in OSCC. Abstract Invasion of the mandibular bone is frequent in oral squamous cell carcinoma (OSCC), which often results in extensive ablative and reconstructive procedures for the patient. The purpose of this single-center, retrospective study was to identify and evaluate potential biomarkers and risk factors for bone invasion in OSCC. Initially, in silico gene expression analysis was performed for different HNSCC tumor T-stages to find factors associated with invasive (T4a) tumor growth. Afterwards, the protein expression of bone-metabolizing MMP-27, TNFRSF11B (Osteoprotegerin, OPG), and TNFSF11 (RANKL) was investigated via Tissue Microarrays (TMAs) for their impact on mandibular bone invasion. TMAs were assembled from the bone–tumor interface of primary OSCCs of the floor of the mouth and gingiva from 119 patients. Sixty-four carcinomas with patho-histological jaw invasion (pT4a) were compared to 55 carcinomas growing along the mandible without invasion (pT2, pT3). Tissue samples were additionally evaluated for patterns of invasion using the WPOI grading system. Statistical analysis of in silico data revealed decreased MMP-27 mRNA expression to be strongly associated with the pT4a-stage in OSCC, indicating invasive tumor growth with infiltration of adjacent anatomical structures. Our own clinico-pathological data on OSCCs presented a significant decrease of MMP-27 in tumors invading the nearby mandible (pT4a), compared to pT2 and pT3 tumors without bone invasion. Loss of MMP27 evolved as the strongest predictor of mandibular bone invasion in binary logistic regression analysis. To our knowledge, this is the first study investigating the role of MMP-27 expression in OSCC and demonstrating the importance of the loss of MMP-27 in mandibular bone invasion.
Collapse
Affiliation(s)
- Jonas Eichberger
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
- Department of Oral and Maxillofacial Surgery and Center for Medical Biotechnology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Florian Weber
- Institute of Pathology, University of Regensburg, 93053 Regensburg, Germany
| | - Gerrit Spanier
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Michael Gerken
- Tumor Center Regensburg, Institute for Quality Assurance and Health Services Research, University of Regensburg, 93053 Regensburg, Germany
| | - Stephan Schreml
- Department of Dermatology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Daniela Schulz
- Department of Oral and Maxillofacial Surgery and Center for Medical Biotechnology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Mathias Fiedler
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
- Department of Oral and Maxillofacial Surgery and Center for Medical Biotechnology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Nils Ludwig
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
- Department of Oral and Maxillofacial Surgery and Center for Medical Biotechnology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Richard Josef Bauer
- Department of Oral and Maxillofacial Surgery and Center for Medical Biotechnology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Torsten Eugen Reichert
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Tobias Ettl
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
- Correspondence:
| |
Collapse
|
6
|
RANKL regulates testicular cancer growth and Denosumab treatment has suppressive effects on GCNIS and advanced seminoma. Br J Cancer 2022; 127:408-421. [PMID: 35418213 PMCID: PMC9345904 DOI: 10.1038/s41416-022-01810-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 03/10/2022] [Accepted: 03/29/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Testicular germ cell tumours (TGCTs) have a high sensitivity to chemotherapy and a high cure rate, although with serious adverse effects. In the search for tumour suppressive drugs, the RANKL inhibitor Denosumab, used to treat osteoporosis, came up as a candidate since RANKL signalling was recently identified in the testis. METHODS Expression of RANKL, RANK and OPG, and the effects of RANKL inhibition were investigated in human TGCTs, TGCT-derived cell-lines, and TGCT-xenograft models. Serum RANKL was measured in TGCT-patients. RESULTS RANKL, RANK, and OPG were expressed in germ cell neoplasia in situ (GCNIS), TGCTs, and TGCT-derived cell lines. RANKL-inhibition reduced proliferation of seminoma-derived TCam-2 cells, but had no effect on embryonal carcinoma-derived NTera2 cells. Pretreatment with Denosumab did not augment the effect of cisplatin in vitro. However, inhibition of RANKL in vivo reduced tumour growth exclusively in the TCam-2-xenograft model and Denosumab-treatment decreased proliferation in human GCNIS cultures. In TGCT-patients serum RANKL had no prognostic value. CONCLUSIONS This study shows that the RANKL signalling system is expressed in GCNIS and seminoma where RANKL inhibition suppresses tumour growth in vitro and in vivo. Future studies are needed to determine whether RANKL is important for the malignant transformation or transition from GCNIS to invasive tumours.
Collapse
|
7
|
Feleke M, Feng W, Song D, Li H, Rothzerg E, Wei Q, Kõks S, Wood D, Liu Y, Xu J. Single-cell RNA sequencing reveals differential expression of EGFL7 and VEGF in giant-cell tumor of bone and osteosarcoma. Exp Biol Med (Maywood) 2022; 247:1214-1227. [PMID: 35695550 PMCID: PMC9379604 DOI: 10.1177/15353702221088238] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Dysregulation of angiogenesis is associated with tumor development and is accompanied by altered expression of pro-angiogenic factors. EGFL7 is a newly identified antigenic factor that plays a role in various cancers such as breast cancer, lung cancer, and acute myeloid leukemia. We have recently found that EGFL7 is expressed in the bone microenvironment, but its role in giant-cell tumor of bone (GCTB) and osteosarcoma (OS) is unknown. The aims of this study are to examine the gene expression profile of EGFL7 in GCTB and OS and compare with that of VEGF-A-D and TNFSF11 using single-cell RNA sequencing data. In-depth differential expression analyses were employed to characterize their expression in the constituent cell types of GCTB and OS. Notably, EGFL7 in GCTB was expressed at highest levels in the endothelial cell (EC) cluster followed by osteoblasts, myeloid cells, and chondrocytes, respectively. In OS, EGFL7 exhibited highest expression in EC cell cluster followed by osteoblastic OS cells, myeloid cells 1, and carcinoma associated fibroblasts (CAFs), respectively. In comparison, VEGF-A is expressed at highest levels in myeloid cells followed by OCs in GCTB, and in myeloid cells, and OCs in OS. VEGF-B is expressed at highest levels in chondrocytes in GCTB and in OCs in OS. VEGF-C is strongly enriched in ECs and VEGF-D is expressed at weak levels in all cell types in both GCTB and OS. TNFSF11 (or RANKL) shows high expression in CAFs and osteoblastic OS cells in OS, and osteoblasts in GCTB. This study investigates pro-angiogenic genes in GCTB and OS and suggests that these genes and their expression patterns are cell-type specific and could provide potential prognostic biomarkers and cell type target treatment for GCTB and OS.
Collapse
Affiliation(s)
- Mesalie Feleke
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Wenyu Feng
- Department of Orthopaedics, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Dezhi Song
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
- Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning 530021, China
| | - Hengyuan Li
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
- Department of Orthopedics, Centre for Orthopedic Research, Second Affiliated Hospital, School of Medicine, Orthopedics Research Institute, Zhejiang University, Hangzhou, China
| | - Emel Rothzerg
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
- Perron Institute for Neurological and Translational Science, QEII Medical Centre, Nedlands, WA 6009, Australia
| | - Qingjun Wei
- Department of Orthopaedics, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Sulev Kõks
- Perron Institute for Neurological and Translational Science, QEII Medical Centre, Nedlands, WA 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA 6150, Australia
| | - David Wood
- Medical School, University of Western Australia, Perth, WA 6009, Australia
| | - Yun Liu
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
- Department of Orthopaedics, First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
- Jiake Xu.
| |
Collapse
|
8
|
A Tailored Approach for Appendicular Impending and Pathologic Fractures in Solid Cancer Metastases. Cancers (Basel) 2022; 14:cancers14040893. [PMID: 35205641 PMCID: PMC8870648 DOI: 10.3390/cancers14040893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/05/2022] [Accepted: 02/10/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Patients with bone metastases often suffer with complications, such as bone fractures, which have a substantial negative impact on clinical outcomes. To optimize clinical results, a tailored approach should be defined for managing impending or pathologic fractures in each individual case. The ability to control systemic disease, the extent, location and nature of bone metastases, and the biology of the underlying tumor, are the main factors that will define the strategy to follow. Abstract Advances in medical and surgical treatment have played a major role in increasing the survival rates of cancer patients with metastatic bone disease. The clinical course of patients with bone metastases is often impaired by bone complications, such as bone fractures, which have a substantial negative impact on clinical outcomes. To optimize clinical results and prevent a detrimental impact on patients’ health, a tailored approach should be defined for any given patient. The optimal management of impending or pathologic fractures is unknown and relies on a multidisciplinary approach to tailor clinical decisions to each individual patient. The ability to control systemic disease, the extent, location and nature of bone metastases, and the biology of the underlying tumor, are the main factors that will define the strategy to follow. The present review covers the most recent data regarding impending and pathologic fractures in patients with bone metastases, and discusses the medical and surgical management of patients presenting with metastatic bone disease in different clinical settings.
Collapse
|
9
|
Madel MB, Elefteriou F. Mechanisms Supporting the Use of Beta-Blockers for the Management of Breast Cancer Bone Metastasis. Cancers (Basel) 2021; 13:cancers13122887. [PMID: 34207620 PMCID: PMC8228198 DOI: 10.3390/cancers13122887] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Bone represents the most common site of metastasis for breast cancer and the establishment and growth of metastatic cancer cells within the skeleton significantly reduces the quality of life of patients and their survival. The interplay between sympathetic nerves and bone cells, and its influence on the process of breast cancer bone metastasis is increasingly being recognized. Several mechanisms, all dependent on β-adrenergic receptor signaling in stromal bone cells, were shown to promote the establishment of disseminated cancer cells into the skeleton. This review provides a summary of these mechanisms in support of the therapeutic potential of β-blockers for the early management of breast cancer metastasis. Abstract The skeleton is heavily innervated by sympathetic nerves and represents a common site for breast cancer metastases, the latter being the main cause of morbidity and mortality in breast cancer patients. Progression and recurrence of breast cancer, as well as decreased overall survival in breast cancer patients, are associated with chronic stress, a condition known to stimulate sympathetic nerve outflow. Preclinical studies have demonstrated that sympathetic stimulation of β-adrenergic receptors in osteoblasts increases bone vascular density, adhesion of metastatic cancer cells to blood vessels, and their colonization of the bone microenvironment, whereas β-blockade prevented these events in mice with high endogenous sympathetic activity. These findings in preclinical models, along with clinical data from breast cancer patients receiving β-blockers, support the pathophysiological role of excess sympathetic nervous system activity in the formation of bone metastases, and the potential of commonly used, safe, and low-cost β-blockers as adjuvant therapy to improve the prognosis of bone metastases.
Collapse
Affiliation(s)
| | - Florent Elefteriou
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
10
|
Liao HQ, Han MT, Cheng W, Zhang C, Li H, Li MQ, Zhu R. Decidual-derived RANKL facilitates macrophages accumulation and residence at the maternal-fetal interface in human early pregnancy. Am J Reprod Immunol 2021; 86:e13406. [PMID: 33629434 DOI: 10.1111/aji.13406] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/13/2021] [Accepted: 02/22/2021] [Indexed: 01/18/2023] Open
Abstract
PROBLEM During the first trimester, the accumulation of macrophages, which is the second largest decidual leukocyte population (~20%) at the maternal-fetal interface, is quite vital for a successful pregnancy, including embryo implantation, trophoblast invasion, and vascular remodeling. The mechanism of the enrichment and redistribution of macrophages in the uterine decidua of early pregnancy is largely unclear. METHOD OF STUDY A total of 37 women with normal early pregnancies were included. Primary decidual macrophages (dMφs) (n = 37) and primary decidual stromal cells (DSCs) (n = 37) were isolated, and the adhesion molecules were analyzed by flow cytometry (FCM). Adhesive experiment was carried out to evaluate the adhesion capacity by counting cell numbers of dMφs adhered to DSCs in a co-culture system. RESULTS We found that RANK+ dMφs was the dominating subtype at the maternal-fetal interface. The expression of adhesion molecules (eg, CD29, CD31, CD54, and CD62L) on the surface of RANK+ dMφs was higher than that of RANK- dMφs. After co-culture with DSCs, the expression of adhesion molecules on dMφs was up-regulated in a RANKL-dependent manner. Meanwhile, dMφs promoted the releasing of RANKL on DSCs after co-culture. Consistently, dMφs exhibited the lessoned capacity of adhesion to DSCs when blocking the crosstalk of RANKL-RANK between the DSCs and dMφs in vitro. CONCLUSION These results suggest that the interaction of RANKL-RANK up-regulates the expression of adhesion molecules on the surface of dMφs, contributing to the accumulation and residence of dMφs in human early pregnancy.
Collapse
Affiliation(s)
- Hai-Qiong Liao
- Center for Human Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Mu-Tian Han
- Center for Human Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Wei Cheng
- Center for Human Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Ce Zhang
- Center for Human Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Hong Li
- Center for Human Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Rui Zhu
- Center for Human Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.,State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| |
Collapse
|
11
|
Abousaud AI, Barbee MS, Davis CC, Caulfield SE, Wang Z, Boykin A, Carthon BC, Gogineni K. Safety and efficacy of extended dosing intervals of denosumab in patients with solid cancers and bone metastases: a retrospective study. Ther Adv Med Oncol 2021; 12:1758835920982859. [PMID: 33488782 PMCID: PMC7768832 DOI: 10.1177/1758835920982859] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022] Open
Abstract
Aim More than half of patients with breast, lung, or prostate cancer who have bone metastases have evidence of skeletal-related events (SREs). Denosumab is a fully human monoclonal antibody that binds to and neutralizes receptor activator of nuclear factor kappa-B ligand (RANKL) on osteoblasts and their precursors. The United States Food and Drug Administration (FDA)-approved dose of denosumab is 120 mg every 4 weeks; however, other schedules have been used in practice for patient convenience. Evidence for the safety and efficacy of alternative dosing intervals is lacking. Patient & Methods Adult patients with solid cancers and bone metastases who received at least two doses of denosumab 120 mg were reviewed. Patients were grouped based on an average denosumab dosing interval of <5 weeks (short-interval) versus 5-11 weeks (medium-interval) versus ⩾12 weeks (long-interval). The primary outcome was the time to first SRE while on denosumab between the short- and medium-interval groups. The secondary outcomes were overall survival (OS), efficacy comparisons between the other groups, and safety events. Results There was no significant difference in median time to first SRE between the short- and medium-interval denosumab groups [33.2 versus 28.4 months, hazard ratio (HR): 1.13, 95% confidence interval (CI): 0.66-1.92, p = 0.91] or the medium- and long-interval dosing groups (28.4 versus 32.2 months, HR: 1.15, 95% CI: 0.66-2.01, p = 0.62). Median OS was not found to differ significantly between any of the groups. There were significantly more hospitalizations in the short-interval dosing group than the other groups (55.2% versus 33.8% versus 30.4%, p < 0.001). Conclusion Extending denosumab dosing intervals does not appear to negatively impact time to first SRE and is associated with fewer hospitalizations in real-world patients with solid cancers and bone metastases.
Collapse
Affiliation(s)
- Aseala I Abousaud
- Department of Pharmaceutical Services, Emory Healthcare, 5665 Peachtree Dunwoody Road NE, Atlanta, GA 30342, USA
| | - Meagan S Barbee
- Global Medical Information Specialist, Oncology at Med Communications Inc
| | - Christine C Davis
- Department of Pharmaceutical Services, Emory Healthcare, Atlanta, GA, USA
| | - Sarah E Caulfield
- Department of Pharmaceutical Services, Emory Healthcare, Atlanta, GA, USA
| | - Zeyuan Wang
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health
| | - Alexa Boykin
- Medical Science Liason, Novocure Inc., Atlanta, GA, USA
| | - Bradley C Carthon
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Keerthi Gogineni
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| |
Collapse
|
12
|
Qiu Z, Xie Z, Qin R, Chen M, He H, Zhang Z, Wang Y, Hong M, Tang W, Xi Y, Zhang S. Evaluation of ICAM-1 rs5498 and rs3093030 Polymorphisms in Chinese Patients with Colorectal Cancer. DNA Cell Biol 2020; 40:384-392. [PMID: 33347388 DOI: 10.1089/dna.2020.6089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Colorectal cancer (CRC) is a common cancer threatening human health. Intercellular adhesion molecule-1 (ICAM-1, CD54) displays a key role in carcinogenesis and previous studies have suggested that ICAM-1 single-nucleotide polymorphisms (SNPs) are predicted to increase the risk of CRC. However, the relationship of ICAM-1 SNPs with CRC susceptibility was controversial. We conducted a case-control study to clarify the association of ICAM-1 SNPs (rs5498 and rs3093030) with the CRC risk. A total of 1003 CRC patients and 1303 controls were recruited to determine ICAM-1 SNPs (rs5498 and rs3093030) by SNPscan method. In the case-control study, we found that ICAM-1 rs5498 polymorphism did not influence CRC risk (AG vs. AA: adjusted p = 0.179; GG vs. AA: adjusted p = 0.281, AG+GG vs. AA: adjusted p = 0.398; GG vs. AA+AG: adjusted p = 0.153), and ICAM-1 rs3093030 polymorphism did not influence CRC risk (CT vs. CC: adjusted p = 0.841; TT vs. CC: adjusted p = 0.175, CT+TT vs. CC: adjusted p = 0.574 and TT vs. CC+TT: adjusted p = 0.180). In a subgroup of age >61, ICAM-1 rs5498 decreased the risk of CRC (p = 0.047). Multivariate analysis revealed that smoking (p = 0.002; odds ratio [OR]: 1.76, 95% confidence interval [CI]: 1.18-2.63), alcohol intake (p < 0.001; OR: 1.99, 95% CI: 1.31-3.05), and body mass index <24 (p < 0.001; OR: 1.55, 95% CI: 1.06-2.26) increased the risk of CRC. Our findings showed that ICAM-1 rs3093030 was not correlated with the susceptibility of CRC, and ICAM-1 rs5498 increased the risk of CRC in the subgroup of age ≥61. In the future, larger and ethnically homogeneous populations are needed to confirm our results.
Collapse
Affiliation(s)
- Zhiyuan Qiu
- Department of Oncology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhiqiang Xie
- Department of Clinical Laboratory, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Rong Qin
- Department of Oncology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Meifang Chen
- Department of Oncology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Han He
- Department of Oncology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhao Zhang
- Department of Oncology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yan Wang
- Department of Oncology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ming Hong
- Center for Foreign Language Education Research, Zhejiang International Studies University, Hangzhou, Zhejiang, China
| | - Weifeng Tang
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yan Xi
- Department of Geriatrics, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Sheng Zhang
- Department of General Surgery, Changzhou No. 3 People's Hospital, Changzhou, Jiangsu, China
| |
Collapse
|
13
|
Kaur M, Gupta KB, Thakur S, Kaur S, Dhiman M. Parthenium hysterophorus mediated inflammation and hyper-responsiveness via NF-κB pathway in human A549 lung cancer cell line. ENVIRONMENTAL TOXICOLOGY 2020; 35:1241-1250. [PMID: 32686900 DOI: 10.1002/tox.22989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/31/2020] [Accepted: 06/06/2020] [Indexed: 06/11/2023]
Abstract
Being one of the notorious weed P. hysterophorus has invaded almost every part India and is the lead cause of skin allergies and severe dermatitis among farmers and rural population. It is an invasive obnoxious weed capable of surviving extreme environmental conditions and various parts of this plant are reported to cause severe contact allergies in humans due to the presence of high concentrations of toxic sesquiterpene lactones viz. parthenin. It can stimulate numerous cellular and immune responses that may translate into Oxidative stress, allergies, and inflammation. The effect of P. hysterophorus flower extract was evaluated on cell viability, oxidative stress and inflammation in A549 lung cancer cell line by spectrophotometric and reverse transcriptase-polymerase chain reaction methods. Schrodinger software based docking was performed for possible interactions studies. The A549 cells treated with P. hysterophorus flower extract favors increase in cell viability, reactive oxygen species generation. The mRNA expression of proinflammatory cytokines such as IFN-γ, TNF-α, and IL-1β was significantly increased whereas no change in IL-18 expression was observed. Significant increase in protein expression of NF-κB was observed, suggesting the role of NF-κB signalling in allergic responses. The docking studies demonstrated the potential interaction between Parthenin and NF-κB/IL-1β/IL-18 suggesting their activation leading to inflammation. The current study emphasize that P. hysterophorus mediates oxidative stress, and inflammatory process via alterations in expression of proinflammatory cytokines such as IL-1β, IFN-γ through NF-κB activation which was also confirmed in docking studies. Cellular and molecular mechanisms involved in pathogenesis of allergic/chronic inflammation and severe dermatitis need to be further investigated to identify specific binding partners responsible for severe inflammation which can provide some leads in developing effective targets against severe dermatitis and skin allergies.
Collapse
Affiliation(s)
- Mandeep Kaur
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India
| | - Kunj Bihari Gupta
- Department of Microbiology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Shweta Thakur
- Department of Zoology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Sukhchain Kaur
- Department of Microbiology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| | - Monisha Dhiman
- Department of Microbiology, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, India
| |
Collapse
|
14
|
Chu QSC. How Is Receptor Activator of Nuclear Factor-κβ/Receptor Activator of Nuclear Factor-κβ Ligand Targeting Ranked as an Immuno-Oncology Target for NSCLC: a Pathway Unclear as yet? J Thorac Oncol 2020; 15:1561-1564. [PMID: 32981598 DOI: 10.1016/j.jtho.2020.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 10/23/2022]
Affiliation(s)
- Quincy S C Chu
- Cross Cancer Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
15
|
Guo Q, Gao J, Cheng L, Yang X, Li F, Jiang G. The Epstein-Barr virus-encoded G protein-coupled receptor BILF1 upregulates ICAM-1 through a mechanism involving the NF-қB pathway. Biosci Biotechnol Biochem 2020; 84:1810-1819. [PMID: 32567483 DOI: 10.1080/09168451.2020.1777525] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Abstract
Although the Epstein-Barr virus (EBV) infection is usually asymptomatic, a primary encounter with the virus can cause mononucleosis. EBV infection is also strongly associated with lymphoma and epithelial cancers. The structure and infection mechanism of EBV have been well studied, but the EBV-encoded G protein-coupled receptor, BILF1, is not fully understood. Here, it was found that the EBV BILF1 was expressed early in the viral lytic cycle and its ectopic expression strikingly upregulated the ICAM-1 expression in Raji cells. The positive effect of BILF1 on the ICAM-1 promoter was observed and the BILF1 deficiency attenuated ICAM-1 promoter activity. Moreover, NF-κB binding sites were important for the regulation of ICAM-1 promoter by BILF1. Furthermore, BILF1 reduced the constitutive level of the IқB-a protein and increased the amount of nuclear NF-қB in Raji cells. In conclusion, this study determined that BILF1 upregulated ICAM-1 in a mechanism involving NF-қB.
Collapse
Affiliation(s)
- Qingwei Guo
- Department of Hematology, Qilu Children’s Hospital of Shandong University, Jinan, China
| | - Jie Gao
- Department of Central Lab, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lu Cheng
- Department of Respiratory, Qilu Children’s Hospital of Shandong University, Jinan, China
| | - Xiaomei Yang
- Department of Hematology, Qilu Children’s Hospital of Shandong University, Jinan, China
| | - Fu Li
- Department of Hematology, Qilu Children’s Hospital of Shandong University, Jinan, China
| | - Guosheng Jiang
- Department of Immunology, Binzhou Medical University, Yantai, China
| |
Collapse
|
16
|
Ming J, Cronin SJF, Penninger JM. Targeting the RANKL/RANK/OPG Axis for Cancer Therapy. Front Oncol 2020; 10:1283. [PMID: 32850393 PMCID: PMC7426519 DOI: 10.3389/fonc.2020.01283] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
RANKL and RANK are expressed in different cell types and tissues throughout the body. They were originally described for their essential roles in bone remodeling and the immune system but have subsequently been shown to provide essential signals from regulating mammary gland homeostasis during pregnancy to modulating tumorigenesis. The success of RANKL/RANK research serves as a paragon for translational research from the laboratory to the bedside. The case in point has been the development of Denosumab, a RANKL-blocking monoclonal antibody which has already helped millions of patients suffering from post-menopausal osteoporosis and skeletal related events in cancer. Here we will provide an overview of the pathway from its origins to its clinical relevance in disease, with a special focus on emerging evidence demonstrating the therapeutic value of targeting the RANKL/RANK/OPG axis not only in breast cancer but also as an addition to the cancer immunotherapy arsenal.
Collapse
Affiliation(s)
- Jie Ming
- Department of Breast and Thyroid Surgery, Wuhan Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shane J F Cronin
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna Biocenter, Vienna, Austria
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Science, Vienna Biocenter, Vienna, Austria.,Department of Medical Genetics, Life Science Institute, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
17
|
Deligiorgi MV, Trafalis DT. Repurposing denosumab in lung cancer beyond counteracting the skeletal related events: an intriguing perspective. Expert Opin Biol Ther 2020; 20:1331-1346. [PMID: 32658547 DOI: 10.1080/14712598.2020.1790522] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Repurposing denosumab in lung cancer therapeutics capitalizes on its well-established role in preventing the skeletal related events (SREs) and its emerging, yet elusive, bone-independent role, assigned to inhibit the contribution of RANKL to cancer initiation and progression. AREAS COVERED The present review presents the available preclinical and clinical data indicating that denosumab may provide survival benefit to lung cancer patients beyond the counteraction of SREs. EXPERT OPINION Despite the preliminary data heralding the potential of denosumab to increase overall survival in lung cancer, the embracement of this strategy in clinical practice cannot be advocated until large randomized clinical trials consolidate its safety and efficacy. Given the improvement of lung cancer prognosis ascribed to revolutionary targeted treatment agents, the possibility of denosumab-related increased risk of second primary malignancies merits further evaluation. Many challenges in endorsing denosumab as a strategy to treat lung cancer beyond SREs prevention are pending counteraction, including: (i) patient selection guided by validated predictive and prognostic biomarkers; (ii) assessment of long-term outcomes; (iii) evaluation of benefit-risk ratio; (iv) translational research; (v) combination of denosumab with other targeted therapies; (vi) integration of genomic biomarkers, immune-related biomarkers, and biomarkers of active RANKL pathway to guide the decision-making process.
Collapse
Affiliation(s)
- Maria V Deligiorgi
- Department of Pharmacology - Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine , Athens, Greece
| | - Dimitrios T Trafalis
- Department of Pharmacology - Clinical Pharmacology Unit, National and Kapodistrian University of Athens, Faculty of Medicine , Athens, Greece
| |
Collapse
|
18
|
Deligiorgi MV, Panayiotidis MI, Griniatsos J, Trafalis DT. Harnessing the versatile role of OPG in bone oncology: counterbalancing RANKL and TRAIL signaling and beyond. Clin Exp Metastasis 2020; 37:13-30. [PMID: 31578655 DOI: 10.1007/s10585-019-09997-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 09/24/2019] [Indexed: 12/15/2022]
Abstract
More than 2 decades ago, the discovery of osteoprotegerin (OPG) as inhibitor of the receptor of activator of nuclear factor Kb (RANK) ligand (RANKL) revolutionized our understanding of bone biology and oncology. Besides acting as decoy receptor for RANKL, OPG acts as decoy receptor for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). OPG, RANKL, and TRAIL are ubiquitously expressed, stimulating per se pivotal signaling cascades implicated in cancer. In the context of cancer cell-bone cell interactions, cancer cells skew the OPG/RANKL/RANK (RANKL cognate receptor) balance towards bone destruction and tumor growth through favoring the RANKL/RANK interface, circumventing OPG. Numerous preclinical and clinical studies demonstrate the dual role of OPG in cancer: antitumor and tumor-promoting. OPG potentially conveys an antitumor signal through inhibiting the tumor-promoting RANKL signaling-both the osteoclast-dependent and the osteoclast-independent-and the tumor-promoting TRAIL signaling. On the other hand, the presumed tumor-promoting functions of OPG are: (i) abrogation of TRAIL-induced apoptosis of cancer cells; (ii) abrogation of RANKL-induced antitumor immunity; and (iii) stimulation of oncogenic and prometastatic signaling cascades downstream of the interaction of OPG with diverse proteins. The present review dissects the role of OPG in bone oncology. It presents the available preclinical and clinical data sustaining the dual role of OPG in cancer and focuses on the imbalanced RANKL/RANK/OPG interplay in the landmark "vicious cycle" of skeletal metastatic disease, osteosarcoma, and multiple myeloma. Finally, current challenges and future perspectives in exploiting OPG signaling in bone oncology therapeutics are discussed.
Collapse
Affiliation(s)
- Maria V Deligiorgi
- Clinical Pharmacology Unit, Laboratory of Pharmacology, Faculty of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias Str, Goudi, 11527, Athens, Greece.
| | - Mihalis I Panayiotidis
- Department of Applied Sciences, Northumbria University, Ellison Building, Room A516, Newcastle upon Tyne, NE1 8ST, UK
| | - John Griniatsos
- 1st Department of Surgery, Faculty of Medicine, National and Kapodistrian University of Athens, Laikon General Hospital, 17 Agiou Thoma Str, Goudi, 115-27, Athens, Greece
| | - Dimitrios T Trafalis
- Clinical Pharmacology Unit, Laboratory of Pharmacology, Faculty of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias Str, Goudi, 11527, Athens, Greece
| |
Collapse
|
19
|
Dynamic changes of Receptor activator of nuclear factor-κB expression in Circulating Tumor Cells during Denosumab predict treatment effectiveness in Metastatic Breast Cancer. Sci Rep 2020; 10:1288. [PMID: 31992773 PMCID: PMC6987166 DOI: 10.1038/s41598-020-58339-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 01/06/2020] [Indexed: 12/11/2022] Open
Abstract
Receptor-activator of nuclear-factor -κB-ligand (RANKL) and its receptor RANK have been recently identified as key players in breast cancer bone metastases. Since Circulating Tumor Cells (CTCs) are considered a crucial step of metastatic process, we explored RANK expression on CTCs in metastatic breast cancer (MBC), and the predictive value of RANK-positive CTCs in monitoring patients during treatment with denosumab (anti-RANKL antibody). To this purpose, we developed a novel CTC assay to quantify RANK-positive CTCs in forty-two bone MBC patients, candidates to denosumab treatment. Companion algorithms ΔAUC and Slope were developed, and correlated with time to first skeletal-related-events (SRE), time to bone metastasis progression and time to visceral metastasis progression. Twenty-seven patients had at least one CTC at baseline and, among these, nineteen (70%) had one or more RANK-positive CTCs. Notably, the baseline total CTCs, but not the RANK-positive, were associated with Time-to-first-SRE, Time-to-Bone-Metastasis-Progression and Time-to-Visceral-Metastasis-Progression. Conversely, during treatment monitoring, positive ΔAUC value, expression of RANK-positive CTCs persistence, correlated with longer Time-to-first-SRE (p = 0.0002) and Time-to-Bone-Metastasis-Progression (p = 0.0012). Furthermore, the early increase at second day, in RANK-positive CTCs (Positive-Slope) was associated with delay in time-to-first-SRE (p = 0.0038) and Time-to-Bone-Metastasis-Progression (p = 0.0024). We demonstrate, for the first time, the expression of RANK on CTCs in MBC patients and that the persistence of RANK expression determines denosumab effectiveness.
Collapse
|
20
|
Wan X, Song Y, Fang H, Xu L, Che X, Wang S, Zhang X, Zhang L, Li C, Fan Y, Hou K, Li Z, Wang X, Liu Y, Qu X. RANKL/RANK promotes the migration of gastric cancer cells by interacting with EGFR. Clin Transl Med 2020; 9:3. [PMID: 31933009 PMCID: PMC6957613 DOI: 10.1186/s40169-019-0249-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 12/12/2019] [Indexed: 01/05/2023] Open
Abstract
Background The incidence and mortality rates of gastric cancer (GC) rank in top five among all malignant tumors. Chemokines and their receptor-signaling pathways reportedly play key roles in the metastasis of malignant tumor cells. Receptor activator of nuclear factor κB ligand (RANKL) is a member of the tumor necrosis factor family, with strong chemokine-like effects. Some studies have pointed out that the RANKL/RANK pathway is vital for the metastasis of cancer cells, but the specific mechanisms in GC remain poorly understood. Results This study reports original findings in cell culture models and in patients with GC. Flow cytometry and western blotting analyses showed that RANK was expressed in BGC-823 and SGC-7901 cells in particular. Chemotaxis experiments and wound healing assay suggested that RANKL spurred the migration of GC cells. This effect was offset by osteoprotegerin (OPG), a decoy receptor for RANKL. RANKL contributed to the activation of human epidermal growth factor receptor (HER) family pathways. The lipid raft core protein, caveolin 1 (Cav-1), interacted with both RANK and human epidermal growth factor receptor-1(EGFR). Knockdown of Cav-1 blocked the activation of EGFR and cell migration induced by RANKL. Moreover, RANK-positive GC patients who displayed higher levels of EGFR expression had poor overall survival. Conclusions In summary, we confirmed that with the promotion of RANKL, RANK and EGFR can form complexes with the lipid raft core protein Cav-1, which together promote GC cell migration. The formation of the RANK-Cav-1-EGFR complex provides a novel mechanism for the metastasis of GC. These observations warrant confirmation in independent studies, in vitro and in vivo. They also inform future drug target discovery research and innovation in the treatment of GC progression.
Collapse
Affiliation(s)
- Xing Wan
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Yongxi Song
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Honghong Fang
- Jining No.1 People's Hospital, Shandong, 272011, China
| | - Ling Xu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Shuo Wang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaomeng Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Lingyun Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Ce Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Yibo Fan
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Kezuo Hou
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Zhi Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xueqing Wang
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China.
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
21
|
Wang M, Chao CC, Chen PC, Liu PI, Yang YC, Su CM, Huang WC, Tang CH. Thrombospondin enhances RANKL-dependent osteoclastogenesis and facilitates lung cancer bone metastasis. Biochem Pharmacol 2019; 166:23-32. [PMID: 31075265 DOI: 10.1016/j.bcp.2019.05.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/04/2019] [Indexed: 01/09/2023]
Abstract
Lung cancers have a predilection for metastasizing to bone. The matricellular glycoprotein thrombospondin (TSP)-2 regulates multiple biological functions and has a critical role in tumor development and metastasis, although its effects are uncertain in lung cancer bone metastasis. This study demonstrates that TSP-2 expression is highly correlated with lung cancer tumor stage and that the TSP-2 neutralizing antibody reduces osteoclast formation in conditioned medium obtained from lung cancer cells. We also found that TSP-2 promotes osteoclastogenesis through the RANKL-dependent pathway and that TSP-2-mediated osteoclastogenesis involves the transactivation of nuclear factor of activated T-cells cytoplasmic 1 (NFATc1) via the inhibition of miR-486-3p expression. Osteoblasts played a critical role in osteoclast differentiation and incubation of osteoblasts with TSP-2 altered the RANKL:OPG ratio. Furthermore, TSP-2 knockdown inhibited lung cancer osteolytic metastasis in vivo. TSP-2 appears to be worth targeting for the prevention of bone metastasis in lung cancer.
Collapse
Affiliation(s)
- Maofeng Wang
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Chia-Chia Chao
- Department of Respiratory Therapy, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Po-Chun Chen
- Central Laboratory, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Po-I Liu
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan; Department of Thoracic Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Yi-Chen Yang
- Department of Nursing, National Taichung University of Science and Technology, Taichung, Taiwan
| | - Chen-Ming Su
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Wei-Chien Huang
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan; Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, Taiwan; Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan.
| |
Collapse
|
22
|
Xuan NT, Trung DM, Minh NN, Nghia VX, Giang NV, Canh NX, Toan NL, Cam TD, Nga NT, Tien TV, Hoang NH. Regulation of p38MAPK-mediated dendritic cell functions by the deubiquitylase otubain 1. HLA 2019; 93:462-470. [PMID: 30908891 DOI: 10.1111/tan.13534] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 02/13/2019] [Accepted: 03/21/2019] [Indexed: 12/14/2022]
Abstract
Dendritic cells (DCs) are professional antigen presenting cells (APCs) that represent the essential link between innate and acquired immunity. Otubain (OTUB) 1 is shown to deubiquitinate TRAFs to suppress virus-induced inflammatory response. MAPK, a downstream molecule of TRAFs, is involved in regulating LPS-induced immune reactions and its activation is sensitive to the presence of OTUB1. Little is known about contributions of OTUB1 to changes in biological properties of DCs. The present study, therefore, explored whether DC functions are influenced by OTUB1. To this end, DCs were isolated and cultured with GM-CSF to attain bone marrow-derived DCs (BMDCs) and followed by treatment with lipopolysaccharide (LPS) in the presence or absence of OTUB1 siRNA. Expression of markers of cellular maturation and proliferation were analyzed by flow cytometry, and secretion of inflammatory cytokines and ability to stimulate CD4+ T-cells in allogenic mixed leukocyte reaction (allo-MLR) by ELISA, cell migration by a transwell migration assay and phagocytic capacity by FITC-dextran uptake measurement. As a result, treatment of the cells with OTUB1 siRNA prolonged activation of p38MAPK, increased CD54 expression and IL-6 release and reduced FITC-dextran uptake. Moreover, cytokine release produced from CD4+ T-cells in allo-MLR was different. The enhanced level of IFN-γ, but not other cytokine production was observed in the presence of siRNA OTUB1. All the effects were completely abolished when the cells were exposed with p38MAPK inhibitor SB203580. In conclusion, OTUB1 prevents the prolonged activation of p38MAPK, which in turn compromises DC functions.
Collapse
Affiliation(s)
- Nguyen Thi Xuan
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam.,Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Do Minh Trung
- Institute of Biomedicine and Pharmacy, Vietnam Military Medical University, Hanoi, Vietnam
| | - Nghiem Ngoc Minh
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam.,Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Vu Xuan Nghia
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| | - Nguyen Van Giang
- Faculty of Biotechnology, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Nguyen Xuan Canh
- Faculty of Biotechnology, Vietnam National University of Agriculture, Hanoi, Vietnam
| | - Nguyen Linh Toan
- Department of Pathophysiology, Vietnam Military Medical University, Hanoi, Vietnam
| | - Truong Dinh Cam
- Department of Cardiology, 175 Military Medical Hospital, Ho Chi Minh, Vietnam
| | - Nguyen Thanh Nga
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Tran Viet Tien
- Department of Infectious Diseases, 103 Hospital, Vietnam Military Medical University, Hanoi, Vietnam
| | - Nguyen Huy Hoang
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam.,Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| |
Collapse
|
23
|
Chang RQ, Shao J, Meng YH, Wang J, Li DJ, Li MQ. Decidual RANKL/RANK interaction promotes the residence and polarization of TGF-β1-producing regulatory γδ T cells. Cell Death Dis 2019; 10:113. [PMID: 30737372 PMCID: PMC6368618 DOI: 10.1038/s41419-019-1380-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 12/19/2018] [Accepted: 01/15/2019] [Indexed: 12/14/2022]
Abstract
ABSTACT Decidual γδΤ (dγδΤ) cells play an essential role during successful pregnancy; however, the residence and polarization of γδΤ cells in decidua remain unclear. In this study, we observed higher levels of receptor activator for nuclear factor-κ B ligand (RANKL) on decidual stromal cells (DSCs), and its receptor RANK on dγδΤ cells in decidua from normal pregnancy compared with patients with recurrent spontaneous abortion (RSA). RANKL expressed by DSCs can induce the polarization of peripheral blood γδΤ (pγδΤ) and dγδΤ cells to Foxp3 + γδΤ cells, and upregulate the expression of transforming growth factor (TGF)-β1. This process is mediated through activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). In addition, RANKL promotes the adhesion of dγδΤ cells to DSCs in vitro, which is associated with the upregulation of ICAM-1 and VCAM-1 on DSCs and integrins on dγδΤ cells. RANKL knockout leads to the decreased numbers of uterus total γδΤ cells, Foxp3+γδΤ cells and the expression of TGF-β1, and the increased pregnancy loss in mice. These results suggest that RANKL is a pivotal regulator of maternal-fetal tolerance by triggering the polarization and residence of TGF-β1-producing Foxp3+γδΤ cells in early pregnancy. The abnormal low level of RANKL/RANK results in pregnancy loss because of the dialogue disorder between DSCs and dγδΤ cells. This observation provides a scientific basis on which a potential marker can be detected to early warning of pregnancy loss.
Collapse
MESH Headings
- Abortion, Habitual/immunology
- Abortion, Habitual/metabolism
- Abortion, Habitual/pathology
- Adult
- Animals
- Cell Plasticity/physiology
- Coculture Techniques
- Decidua/cytology
- Decidua/immunology
- Decidua/metabolism
- Decidua/pathology
- Female
- Humans
- Male
- Maternal-Fetal Exchange
- Mice
- Mice, Knockout
- Phenotype
- Pregnancy
- RANK Ligand/immunology
- RANK Ligand/metabolism
- Receptor Activator of Nuclear Factor-kappa B/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/biosynthesis
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/pathology
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- T-Lymphocytes, Helper-Inducer/pathology
- Transfection
- Transforming Growth Factor beta1/metabolism
Collapse
Grants
- This study supported by the National Basic Research Program of China (No. 2015CB943300), the National Natural Science Foundation of China (NSFC) (No. 31671200, 81490744, 91542108, 81471513, 81471548, 81571509, 81501275), the National Key Research and Development Program of China (2017YFC1001404), the Shanghai Rising-Star Program (No. 16QA1400800), the Innovation-oriented Science and Technology Grant from NPFPC Key Laboratory of Reproduction Regulation (CX2017-2), the Program for Zhuoxue of Fudan University.
Collapse
Affiliation(s)
- Rui-Qi Chang
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, 200011, Shanghai, People's Republic of China
| | - Jun Shao
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, 200011, Shanghai, People's Republic of China
| | - Yu-Han Meng
- Reproductive Medical Center, Affiliated Hospital of Weifang Medical University, 261030, Weifang, People's Republic of China
| | - Jian Wang
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, 200011, Shanghai, People's Republic of China
| | - Da-Jin Li
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, 200011, Shanghai, People's Republic of China.
| | - Ming-Qing Li
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, 200011, Shanghai, People's Republic of China.
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, 200011, Shanghai, People's Republic of China.
| |
Collapse
|
24
|
Yang YC, Chiou PC, Chen PC, Liu PY, Huang WC, Chao CC, Tang CH. Melatonin reduces lung cancer stemness through inhibiting of PLC, ERK, p38, β-catenin, and Twist pathways. ENVIRONMENTAL TOXICOLOGY 2019; 34:203-209. [PMID: 30421542 DOI: 10.1002/tox.22674] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/16/2018] [Accepted: 10/20/2018] [Indexed: 06/09/2023]
Abstract
Lung cancer is one of the most common cancer in cancer-related deaths worldwide, which is characterized by its strong metastatic potential. The melatonin hormone secreted by the pineal grand has an antioxidant effect and protects cells against carcinogenic substances. However, the effects of melatonin in lung cancer stemness are largely unknown. We found that melatonin reduces CD133 expression by ~50% in lung cancer cell lines, while results of a sphere formation assay showed that melatonin inhibits lung cancer stemness. These effects of melatonin were reversed when the cell lines were incubated with phospholipase C (PLC), ERK/p38, and a β-catenin activator. Transfection with Twist siRNA augmented the inhibitory effects of melatonin, indicating that melatonin suppresses lung cancer stemness by inhibiting the PLC, ERK/p38, β-catenin, and Twist signaling pathways. We also found CD133 expression is positively correlated with Twist expression in lung cancer specimens. Melatonin shows promise in the treatment of lung cancer stemness and deserves further study.
Collapse
Affiliation(s)
- Yi-Chen Yang
- Department of Nursing, National Taichung University of Science and Technology, Taichung, Taiwan
| | - Pei-Chen Chiou
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Po-Chun Chen
- Central Laboratory, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Po-Yi Liu
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
- Department of Thoracic Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Wei-Chien Huang
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
| | - Chia-Chia Chao
- Department of Respiratory Therapy, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Graduate Institute of Biomedical Science, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|
25
|
Wang Y, Song Y, Che X, Zhang L, Wang Q, Zhang X, Qu J, Li Z, Xu L, Zhang Y, Fan Y, Hou K, Liu Y, Qu X. Caveolin‑1 enhances RANKL‑induced gastric cancer cell migration. Oncol Rep 2018; 40:1287-1296. [PMID: 30015970 PMCID: PMC6072394 DOI: 10.3892/or.2018.6550] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 06/21/2018] [Indexed: 12/16/2022] Open
Abstract
The classical pathway involving receptor activator of nuclear factor‑κB (RANK) and its ligand (RANKL) induces the activation of osteoclasts and the migration of a variety of tumor cells, including breast and lung cancer. In our previous study, the expression of RANK was identified on the surface of gastric cancer cells, however, whether the RANKL/RANK pathway is involved in the regulation of gastric cancer cell migration remains to be fully elucidated. Lipid rafts represent a major platform for the regulation of cancer signaling; however, their involvement in RANKL‑induced migration remains to be elucidated. To investigate the potential roles and mechanism of RANKL/RANK in gastric cancer migration and metastasis, the present study examined the expression of RANK by western blot analysis and the expression of caveolin‑1 (Cav‑1) in gastric cancer tissues by immunohistochemistry, in addition to cell migration which is measured by Transwell migration assay. The aggregation of lipid reft was observed by fluorescence microscopy and western blotting was used to measure signaling changes in associated pathways. The results showed that RANKL induced gastric cancer cell migration, accompanied by the activation of Cav‑1 and aggregation of lipid rafts. Nystatin, a lipid raft inhibitor, inhibited the activation of Cav‑1 and markedly reversed RANKL‑induced gastric cancer cell migration. The RANKL‑induced activation of Cav‑1 has been shown to occur with the activation of proto‑oncogene tyrosine‑protein kinase Src (c‑Src). The c‑Src inhibitor, PP2, inhibited the activation of Cav‑1 and lipid raft aggregation, and reversed RANKL‑induced gastric cancer cell migration. Furthermore, it was demonstrated that Cav‑1 was involved in RANKL‑induced cell migration in lung, renal and breast cancer cells. These results suggested that RANKL induced gastric cancer cell migration, likely through mechanisms involving the c‑Src/Cav‑1 pathway and lipid raft aggregation.
Collapse
Affiliation(s)
- Yan Wang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yongxi Song
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Lingyun Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Qian Wang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xiaomeng Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Jinglei Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Zhi Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ling Xu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ye Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yibo Fan
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Kezuo Hou
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
26
|
Faget J, Contat C, Zangger N, Peters S, Meylan E. RANKL Signaling Sustains Primary Tumor Growth in Genetically Engineered Mouse Models of Lung Adenocarcinoma. J Thorac Oncol 2018; 13:387-398. [DOI: 10.1016/j.jtho.2017.11.121] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 10/09/2017] [Accepted: 11/14/2017] [Indexed: 01/23/2023]
|
27
|
Zhang X, Song Y, Song N, Zhang L, Wang Y, Li D, Wang Z, Qu X, Liu Y. Rankl expression predicts poor prognosis in gastric cancer patients: results from a retrospective and single-center analysis. ACTA ACUST UNITED AC 2018; 51:e6265. [PMID: 29340518 PMCID: PMC5769752 DOI: 10.1590/1414-431x20176265] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 10/25/2017] [Indexed: 12/27/2022]
Abstract
The receptor activator of nuclear factor κB ligand (RANKL)/RANK pathway plays an important role in the prognosis of several solid tumor types, but its role in gastric cancer prognosis has been poorly characterized. A total of 116 gastric cancer patients who underwent surgical resection were enrolled in this study. Expressions of RANKL and RANK in gastric cancer tissues were detected using immunohistochemical staining. Thirty-eight patients (33%) showed a high level of RANKL expression and 61 patients (53%) showed a high level of RANK expression. There was a positive correlation between expressions of RANKL and RANK (P=0.014, r=0.221). A high level of RANKL expression indicated shorter overall survival (OS) (P=0.008), and was associated with a higher pathological tumor/lymph node/metastasis (pTNM) stage (P=0.035), while no significant correlation was detected between RANK expression and clinicopathological parameters. RANKL also predicted poor prognosis in patients with high RANK expression (P=0.008) and Bormann's type III/IV (P=0.002). Furthermore, RANKL expression correlated with pTNM stage according to high RANK expression (P=0.009), while no significance was found in patients with low RANK expression (P=1.000). Together, our results revealed that high expression of RANKL could predict worse outcomes in gastric cancer especially combined with RANK detection, and thereby this pathway could be a useful prognostic indicator of gastric cancer.
Collapse
Affiliation(s)
- X Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Y Song
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - N Song
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - L Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Y Wang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - D Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Z Wang
- Department of Surgical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - X Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Y Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
28
|
Rao S, Sigl V, Wimmer RA, Novatchkova M, Jais A, Wagner G, Handschuh S, Uribesalgo I, Hagelkruys A, Kozieradzki I, Tortola L, Nitsch R, Cronin SJ, Orthofer M, Branstetter D, Canon J, Rossi J, D'Arcangelo M, Botling J, Micke P, Fleur LL, Edlund K, Bergqvist M, Ekman S, Lendl T, Popper H, Takayanagi H, Kenner L, Hirsch FR, Dougall W, Penninger JM. RANK rewires energy homeostasis in lung cancer cells and drives primary lung cancer. Genes Dev 2017; 31:2099-2112. [PMID: 29118048 PMCID: PMC5733500 DOI: 10.1101/gad.304162.117] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/13/2017] [Indexed: 12/15/2022]
Abstract
Rao et al. report that RANK, the key regulator of osteoclastogenesis, is frequently expressed in primary lung tumors, and clonal genetic inactivation of RANK in mouse lung epithelial cells markedly impairs the progression of KRasG12D-driven lung cancer. RANK rewires energy homeostasis in human and murine lung cancer cells and promotes expansion of lung cancer stem-like cells. Lung cancer is the leading cause of cancer deaths. Besides smoking, epidemiological studies have linked female sex hormones to lung cancer in women; however, the underlying mechanisms remain unclear. Here we report that the receptor activator of nuclear factor-kB (RANK), the key regulator of osteoclastogenesis, is frequently expressed in primary lung tumors, an active RANK pathway correlates with decreased survival, and pharmacologic RANK inhibition reduces tumor growth in patient-derived lung cancer xenografts. Clonal genetic inactivation of KRasG12D in mouse lung epithelial cells markedly impairs the progression of KRasG12D-driven lung cancer, resulting in a significant survival advantage. Mechanistically, RANK rewires energy homeostasis in human and murine lung cancer cells and promotes expansion of lung cancer stem-like cells, which is blocked by inhibiting mitochondrial respiration. Our data also indicate survival differences in KRasG12D-driven lung cancer between male and female mice, and we show that female sex hormones can promote lung cancer progression via the RANK pathway. These data uncover a direct role for RANK in lung cancer and may explain why female sex hormones accelerate lung cancer development. Inhibition of RANK using the approved drug denosumab may be a therapeutic drug candidate for primary lung cancer.
Collapse
Affiliation(s)
- Shuan Rao
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| | - Verena Sigl
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| | - Reiner Alois Wimmer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| | - Maria Novatchkova
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| | - Alexander Jais
- Department of Laboratory Medicine, Medical University Vienna, Vienna 1090, Austria.,Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne 50931, Germany
| | - Gabriel Wagner
- Department of Laboratory Medicine, Medical University Vienna, Vienna 1090, Austria
| | - Stephan Handschuh
- VetCore Facility for Research, University of Veterinary Medicine, Vienna 1220, Austria
| | - Iris Uribesalgo
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| | - Astrid Hagelkruys
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| | - Ivona Kozieradzki
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| | - Luigi Tortola
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| | - Roberto Nitsch
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| | - Shane J Cronin
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| | - Michael Orthofer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| | | | - Jude Canon
- Department of Oncology Research, Amgen, Inc., Seattle, Washington 98119, USA
| | - John Rossi
- Department of Molecular Sciences, Amgen, Inc., Seattle, Washington 98119, USA
| | | | - Johan Botling
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala 75185, Sweden
| | - Patrick Micke
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala 75185, Sweden
| | - Linnea La Fleur
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala 75185, Sweden
| | - Karolina Edlund
- Leibniz Research Center for Working Environment and Human Factors, Dortmund 44139, Germany
| | | | - Simon Ekman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm 17177, Sweden
| | - Thomas Lendl
- Gregor Mendel Institute of Molecular Plant Biology (GMI), Vienna 1030, Austria
| | - Helmut Popper
- Research Unit Molecular Lung and Pleura Pathology, Institute of Pathology, Medical University Graz, Graz 8036, Austria
| | | | - Lukas Kenner
- Department of Clinical Pathology, Medical University Vienna, Vienna 1090, Austria.,Ludwig Boltzmann Institute for Cancer Research, Vienna 1090, Austria.,Unit of Pathology of Laboratory Animals, University of Veterinary Medicine Vienna, Vienna 1220, Austria
| | - Fred R Hirsch
- University of Colorado Cancer Center, Aurora, Colorado 80045, USA
| | - William Dougall
- Department of Oncology Research, Amgen, Inc., Seattle, Washington 98119, USA
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna 1030, Austria
| |
Collapse
|
29
|
Schmitt BM, Laschke MW, Rössler OG, Huang W, Scheller A, Menger MD, Ampofo E. Nerve/glial antigen (NG) 2 is a crucial regulator of intercellular adhesion molecule (ICAM)-1 expression. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:57-66. [PMID: 28964848 DOI: 10.1016/j.bbamcr.2017.09.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 09/01/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
The proteoglycan nerve/glial antigen (NG) 2 is expressed on multiple cell types and mediates cell proliferation and migration. However, little is known about its function in gene regulation. In this study, we demonstrate that in pericytes and glioblastoma cells intercellular adhesion molecule (ICAM)-1, an essential protein for leukocyte adhesion and transmigration, underlies a NG2-dependent expression. As shown by flow cytometry, Western blot analysis and quantitative real-time polymerase chain reaction (qRT-PCR), silencing of NG2 in human placenta-derived pericytes increased the expression of ICAM-1. Pathway analyses revealed that this is mediated by extracellular-regulated-kinases (ERK) 1/2 signaling. Moreover, leukocyte adhesion to NG2 siRNA-treated pericytes was significantly enhanced when compared to scrambled (scr) siRNA-treated control cells. In vivo, we detected increased ICAM-1 protein levels in the retina of mice lacking NG2 expression. To exclude that this novel mechanism is pericyte-specific, we additionally analyzed the expression of ICAM-1 in dependency of NG2 in two glioblastoma cell lines. We found that A1207 and M059K cells exhibit an inverse expression pattern of NG2 and ICAM-1. Finally, downregulation of NG2 in A1207 cells significantly increased ICAM-1 expression. Taken together, these findings indicate that NG2 may represent a promising target for the modulation of ICAM-1-mediated immune responses.
Collapse
Affiliation(s)
- Beate M Schmitt
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Oliver G Rössler
- Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg/Saar, Germany
| | - Wenhui Huang
- Department of Molecular Physiology, CIPMM (Center for Integrative Physiology and Molecular Medicine), Saarland University, 66421 Homburg/Saar, Germany
| | - Anja Scheller
- Department of Molecular Physiology, CIPMM (Center for Integrative Physiology and Molecular Medicine), Saarland University, 66421 Homburg/Saar, Germany
| | - Michael D Menger
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany
| | - Emmanuel Ampofo
- Institute for Clinical & Experimental Surgery, Saarland University, 66421 Homburg/Saar, Germany.
| |
Collapse
|
30
|
Sisay M, Mengistu G, Edessa D. The RANK/RANKL/OPG system in tumorigenesis and metastasis of cancer stem cell: potential targets for anticancer therapy. Onco Targets Ther 2017; 10:3801-3810. [PMID: 28794644 PMCID: PMC5538694 DOI: 10.2147/ott.s135867] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The molecular triad involving receptor activator of nuclear factor kβ (RANK)/RANK ligand (RANKL)/osteoprotegerin cytokine system has been well implicated in several physiological and pathological processes including bone metabolism, mammary gland development, regulation of the immune function, tumorigenesis and metastasis of cancer stem cell, thermoregulation, and vascular calcification. However, this review aimed to summarize several original and up-to-date articles focusing on the role of this signaling system in cancer cell development and metastasis as well as potential therapeutic agents targeting any of the three tumor necrotic factor super family proteins and/or their downstream signaling pathways. The RANK/RANKL axis has direct effects on tumor cell development. The system is well involved in the development of several primary and secondary tumors including breast cancer, prostate cancer, bone tumors, and leukemia. The signaling of this triad system has also been linked to tumor invasiveness in the advanced stage. Bone is by far the most common site of cancer metastasis. Several therapeutic agents targeting this system have been developed. Among them, a monoclonal antibody, denosumab, was clinically approved for the treatment of osteoporosis and cancer-related diseases.
Collapse
Affiliation(s)
| | | | - Dumessa Edessa
- Department of Clinical Pharmacy, School of Pharmacy, College of Health and Medical Sciences, Haramaya University, Harar, Eastern Ethiopia
| |
Collapse
|
31
|
AQP5 promotes hepatocellular carcinoma metastasis via NF-κB-regulated epithelial-mesenchymal transition. Biochem Biophys Res Commun 2017; 490:343-348. [PMID: 28619511 DOI: 10.1016/j.bbrc.2017.06.046] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 06/11/2017] [Indexed: 12/15/2022]
Abstract
Aquaporin 5 (AQP5), a transmembrane protein, is known for its involvement in the progress of many diseases such as chronic kidney disease and systemic disease. Recently, AQP5 has been reported to play an important role in cancer progression. However, little is known about its precise functions in hepatocellular carcinoma (HCC). This study aimed to investigate the specific role of AQP5 in HCC. The results showed that AQP5 was highly expressed in HCC cell lines and its down-regulation inhibited HCC cell invasion and tumor metastasis in vitro and in vivo. In addition, down-regulation of AQP5 suppressed the epithelial-mesenchymal transition (EMT) process in HCC cells by modulating EMT-related molecules such as E-cadherin, α-catenin, N-cadherin and Vimentin. Further studies on corresponding mechanisms indicated that AQP5 down-regulation inhibited HCC metastasis and EMT partly via inactivation of the NF-κB signaling pathway. Taken together, these findings suggest that AQP5 may be a potential therapeutic target for HCC.
Collapse
|
32
|
Amphiregulin enhances intercellular adhesion molecule-1 expression and promotes tumor metastasis in human osteosarcoma. Oncotarget 2016; 6:40880-95. [PMID: 26503469 PMCID: PMC4747375 DOI: 10.18632/oncotarget.5679] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 09/13/2015] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma is a common, high malignant, and metastatic bone cancer. Amphiregulin (AREG) has been associated with cancer cellular activities. However, the effect of AREG on metastasis activity in human osteosarcoma cells has yet to be determined. We determined that AREG increases the expression of intercellular adhesion molecule-1 (ICAM-1) through PI3K/Akt signaling pathway via its interaction with the epidermal growth factor receptor, thus resulting in the enhanced cell migration of osteosarcoma. Furthermore, AREG stimulation increased the association of NF-κB to ICAM-1 promoter which then up-regulated ICAM-1 expression. Finally, we observed that shRNA silencing of AREG decreased osteosarcoma metastasis in vivo. Our findings revealed a relationship between osteosarcoma metastatic potential and AREG expression and the modulating effect of AREG on ICAM-1 expression.
Collapse
|
33
|
Liu JF, Tsao YT, Hou CH. Fractalkine/CX3CL1 induced intercellular adhesion molecule-1-dependent tumor metastasis through the CX3CR1/PI3K/Akt/NF-κB pathway in human osteosarcoma. Oncotarget 2016; 8:54136-54148. [PMID: 28903329 PMCID: PMC5589568 DOI: 10.18632/oncotarget.11250] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 07/09/2016] [Indexed: 12/22/2022] Open
Abstract
Osteosarcoma is the most common primary bone tumor in children and teens. The exact molecular mechanism underlying osteosarcoma progression still remains unclear. The CX3CL1/fractalkine has been implicated in various tumors but not in osteosarcoma. This study is the first to show that fractalkine promotes osteosarcoma metastasis by promoting cell migration. Fractalkine expression was higher in osteosarcoma cell lines than in normal osteoblasts. Fractalkine induced cell migration by upregulating intercellular adhesion molecule-1 (ICAM-1) expression via CX3CR1/PI3K/Akt/NF-κB pathway in human osteosarcoma cells. Knockdown of fractalkine expression markedly inhibited cell migration and lung metastasis in osteosarcoma. Finally, we showed a clinical correlation between CX3CL1 expression and ICAM-1 expression as well as tumor stage in human osteosarcoma tissues. In conclusion, our results indicate that fractalkine promotes cell migration and metastasis of osteosarcoma by upregulating ICAM-1 expression. Thus, fractalkine could serve a novel therapeutic target for preventing osteosarcoma metastasis.
Collapse
Affiliation(s)
- Ju-Fang Liu
- Central Laboratory, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Ya-Ting Tsao
- Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Han Hou
- Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
34
|
Abstract
Oncogenic events combined with a favourable environment are the two main factors in the oncological process. The tumour microenvironment is composed of a complex, interconnected network of protagonists, including soluble factors such as cytokines, extracellular matrix components, interacting with fibroblasts, endothelial cells, immune cells and various specific cell types depending on the location of the cancer cells (e.g. pulmonary epithelium, osteoblasts). This diversity defines specific "niches" (e.g. vascular, immune, bone niches) involved in tumour growth and the metastatic process. These actors communicate together by direct intercellular communications and/or in an autocrine/paracrine/endocrine manner involving cytokines and growth factors. Among these glycoproteins, RANKL (receptor activator nuclear factor-κB ligand) and its receptor RANK (receptor activator nuclear factor), members of the TNF and TNFR superfamilies, have stimulated the interest of the scientific community. RANK is frequently expressed by cancer cells in contrast with RANKL which is frequently detected in the tumour microenvironment and together they participate in every step in cancer development. Their activities are markedly regulated by osteoprotegerin (OPG, a soluble decoy receptor) and its ligands, and by LGR4, a membrane receptor able to bind RANKL. The aim of the present review is to provide an overview of the functional implication of the RANK/RANKL system in cancer development, and to underline the most recent clinical studies.
Collapse
|
35
|
Zhang L, Teng Y, Fan Y, Wang Y, Li W, Shi J, Ma Y, Li C, Shi X, Qu X, Liu Y. The E3 ubiquitin ligase Cbl-b improves the prognosis of RANK positive breast cancer patients by inhibiting RANKL-induced cell migration and metastasis. Oncotarget 2016; 6:22918-33. [PMID: 26087197 PMCID: PMC4673209 DOI: 10.18632/oncotarget.4382] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Accepted: 05/22/2015] [Indexed: 11/25/2022] Open
Abstract
The receptor activator of nuclear factor κ-B ligand (RANKL)/RANK pathway plays an important role in breast cancer progression. Despite the known role of Casitas B-lineage lymphoma (Cbl)-b as an essential regulator of the RANKL/RANK pathway, its effect on RANK pathway in breast cancer remains unclear. Thus, the present study investigated the effect of Cbl-b on the prognosis of RANK-expressing breast cancer patients, as well as on RANKL/RANK pathway. The results showed that RANK and Cbl-b expression was separately detected in 154 (154/300, 51.3%) and 165 (165/300, 55.0%) breast cancer tissue samples. In RANK-expressing breast cancer patients, Cbl-b expression was correlated with low metastasis rate (p = 0.004), better disease-free survival (DFS) and breast cancer-specific survival (BCSS) (p = 0.004 and p = 0.036, respectively). In addition, multivariate analysis showed that Cbl-b expression was an independent predictor of DFS (p = 0.038). Animal experiment results demonstrated that silencing Cbl-b expression in breast cancer cells increased the incidence of lung metastasis in nude mice. Further mechanism investigation revealed that Cbl-b down-regulated RANK protein expression and inhibited RANKL-induced breast cancer cell migration by negatively regulating the Src-Akt/ERK pathway. Our results suggest that Cbl-b improves the prognosis of RANK-expressing breast cancer patients by inhibiting RANKL-induced breast cancer cell migration and metastasis.
Collapse
Affiliation(s)
- Lingyun Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Yuee Teng
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Yibo Fan
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Yan Wang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Wei Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Jing Shi
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Yanju Ma
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Ce Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Xiaonan Shi
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
36
|
Elefteriou F. Role of sympathetic nerves in the establishment of metastatic breast cancer cells in bone. J Bone Oncol 2016; 5:132-134. [PMID: 27761374 PMCID: PMC5063226 DOI: 10.1016/j.jbo.2016.03.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/22/2016] [Accepted: 03/23/2016] [Indexed: 01/11/2023] Open
Abstract
The bone marrow microenvironment is characterized by its multicellular nature, and perhaps less obviously by the high mobility of multiple transient and stationary cell lineages present in this environment. The trafficking of hematopoietic and mesenchymal cells between the bone marrow and blood compartments is regulated by a number of bone marrow-derived factors. It is suspected that transformed metastatic cells “hijack” these processes to engraft into the skeleton and eventually cause the skeletal complications associated with metastatic disease. In this short review, experimental and association data supporting the contribution of a less recognized cell type of the bone marrow – the nerves of the sympathetic nervous system – to early events of the breast cancer bone metastatic process, are summarized.
Collapse
Affiliation(s)
- Florent Elefteriou
- Baylor College of Medicine, Center for Skeletal Medicine and Biology, Dept. of Molecular and Human Genetics and Orthopedic Surgery, One Baylor Plaza, ABBR-R716, Houston, TX 77030, United States
| |
Collapse
|
37
|
Liao CG, Yao L, Xie W, Liu L, Wu SD, Lu N, Huang JG, Kong LM, Zhang HL. Basigin-2 upregulated by receptor activator of NF-κB ligand enhances lung cancer-induced osteolytic lesions. Cancer Cell Int 2016; 16:28. [PMID: 27042161 PMCID: PMC4818914 DOI: 10.1186/s12935-016-0302-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/24/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Lung cancer bone metastasis causes poor prognosis. Basigin-2, a novel cancer-associated biomarker, is upregulated in lung cancer and has been linked with tumor progression. But little is known about the role of basigin-2 in lung cancer bone metastasis and osteolytic lesion. METHODS Basigin-2 expression was evaluated in biopsy tissue specimens of 20 lung cancer patients with bone metastases via immunohistochemistry. Invasion assay and MTT proliferation assay were performed to test the invasion and proliferation of lung cancer cell after modulated basigin-2 expression. The osteoclastic activity of basigin-2 was detected in tibia cancer model by injected of lung cancer cells. The regulation role of receptor activator of NF-κB ligand (RANKL) on basigin-2 and its downstream molecules were measured by real-time quantitative RT-PCR, gelatin zymography and western blot analysis. RESULTS We found that basigin-2 was highly expressed in lung cancer bone metastases. Then, we demonstrated that basigin-2 could promote lung cancer cells invasion, metastasis and proliferation through upregulating metalloproteinases-2 (MMP-2), MMP-9 and vascular endothelial growth factor (VEGF) expression. The lung cancer cells overexpressing basigin-2 strongly induced the osteolytic lesions in immunodeficient mice, which were reduced by treatment with basigin-2 blocking antibody. Furthermore, we explored the enhanced basigin-2 molecular mechanism in lung cancer bone metastasis. Our results indicated the RANKL, pivotal for the control of bone resorption, could increase basigin-2 and its downstream molecules MMP-2, MMP-9 and VEGF expression in vitro. CONCLUSIONS Basigin-2 upregulated by RANKL induces MMPs and VEGF, which may increase lung cancer cell metastasis ability and support osteoclastic activity. Thus, our data suggest important roles for basigin-2 in lung cancer-induced osteolytic lesion and implicate this protein potential application as a target for lung cancer bone metastasis therapy.
Collapse
Affiliation(s)
- Cheng-Gong Liao
- Department of Oncology, Tangdu Hospital, Cancer Institute, Fourth Military Medical University, Xi'an, 710038 People's Republic of China.,Department of Oncology, Urumqi General Hospital of Lanzhou Military Command of PLA, Urumqi, 830000 People's Republic of China
| | - Li Yao
- Department of Pathology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038 People's Republic of China
| | - Wei Xie
- Department of Oncology, Urumqi General Hospital of Lanzhou Military Command of PLA, Urumqi, 830000 People's Republic of China
| | - Lili Liu
- Department of Oncology, Tangdu Hospital, Cancer Institute, Fourth Military Medical University, Xi'an, 710038 People's Republic of China
| | - Sheng-Da Wu
- Cadet Brigade, Fourth Military Medical University, Xi'an, 710032 People's Republic of China
| | - Ning Lu
- Department of Oncology, Urumqi General Hospital of Lanzhou Military Command of PLA, Urumqi, 830000 People's Republic of China
| | - Jian-Guo Huang
- Department of Oncology, Urumqi General Hospital of Lanzhou Military Command of PLA, Urumqi, 830000 People's Republic of China
| | - Ling-Min Kong
- Cell Engineering Research Center and Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, 710032 People's Republic of China
| | - He-Long Zhang
- Department of Oncology, Tangdu Hospital, Cancer Institute, Fourth Military Medical University, Xi'an, 710038 People's Republic of China
| |
Collapse
|
38
|
Shang WQ, Li H, Liu LB, Chang KK, Yu JJ, Xie F, Li MQ, Yu JJ. RANKL/RANK interaction promotes the growth of cervical cancer cells by strengthening the dialogue between cervical cancer cells and regulation of IL-8 secretion. Oncol Rep 2015; 34:3007-16. [PMID: 26398902 DOI: 10.3892/or.2015.4303] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 08/04/2015] [Indexed: 11/06/2022] Open
Abstract
Receptor activator for nuclear factor κB ligand (RANKL) is a member of the tumor necrosis factor (TNF) family. The interaction between RANKL and its receptor RANK plays an important role in the development and function of diverse tissues. However, the expression and role of RANKL in cervical cancer are still unknown. In the present study, we found that RANKL and RANK were highly co-expressed in cervical cancer. HeLa and SiHa cells secreted soluble RANKL (sRANKL), expressed member RANKL (mRANKL) and RANK. Recombinant human RANKL protein had no effect on the viability of HeLa and SiHa cells. Yet, blocking RANKL with an anti-human RANKL neutralizing antibody (α-RANKL) or recombinant human osteoprotegrin (OPG) protein resulted in the downregulation of Ki-67 and B-cell lymphoma 2 (Bcl-2) expression and an increase in Fas and Fas ligand (FasL) expression, as well as a high level of viability and a low level of apoptosis in the HeLa and SiHa cells. In addition, α-RANKL led to a decrease in IL-8 secretion. Recombinant human IL-8 protein reversed the effect of α-RANKL on the expression of proliferation- and apoptosis‑related molecules, and proliferation and apoptosis in the HeLa and SiHa cells. The present study suggests that a high level of mRANKL/RANK expression in cervical cancer lesions plays an important role in the rapid growth of cervical cancer cells possibly through strengthening the dialogue between cervical cancer cells and regulation of IL-8 secretion, which may be a possible target for cervical cancer therapy.
Collapse
Affiliation(s)
- Wen-Qing Shang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Hui Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, P.R. China
| | - Li-Bing Liu
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, P.R. China
| | - Kai-Kai Chang
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, P.R. China
| | - Jia-Jun Yu
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, P.R. China
| | - Feng Xie
- Medical Center of Diagnosis and Treatment for Cervical Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, P.R. China
| | - Ming-Qing Li
- Laboratory for Reproductive Immunology, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, P.R. China
| | - Jin-Jin Yu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| |
Collapse
|
39
|
De Castro J, García R, Garrido P, Isla D, Massuti B, Blanca B, Vázquez J. Therapeutic Potential of Denosumab in Patients With Lung Cancer: Beyond Prevention of Skeletal Complications. Clin Lung Cancer 2015; 16:431-46. [PMID: 26264596 DOI: 10.1016/j.cllc.2015.06.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 06/09/2015] [Accepted: 06/16/2015] [Indexed: 12/29/2022]
Abstract
Approximately up to 40% of patients with lung cancer develop bone metastasis, with 22% to 59% of them experiencing skeletal-related events (SREs), which result in an important quality of life deterioration and economic burden. Denosumab, a fully human antibody that targets the receptor activator of nuclear factor-κB (RANK) ligand (RANKL), is indicated for prevention of SREs in patients with solid tumors and has demonstrated superiority in breast and prostate cancer, and in other solid tumors, in reducing the risk of first SRE by 17% versus zoledronic acid. In the subset of patients with non-small-cell lung carcinoma (NSCLC), denosumab has also shown a positive trend to SRE risk reduction. Denosumab might have direct or indirect antitumor effects. Cancer cells produce factors that stimulate increased bone resorption by osteoclasts, which in turn release tumor growth factors into the bone microenvironment, initiating a tumor/bone vicious cycle. An increasing body of evidence suggests RANK/RANKL signaling plays a role in this tumorigenesis. Both proteins are overexpressed in different tumor types including lung cancer cells. RANK/RANKL signaling activates nuclear factor-κB pathways related to lung carcinogenesis and increases intercellular adhesion molecule 1 expression and MEK/extracellular signal-regulated kinase phosphorylation, which in turn enhances tumor cell migration. In animal NSCLC models, denosumab delayed bone metastases and reduced skeletal tumor growth. In patients with lung cancer (post hoc analysis), denosumab prolonged overall survival by 1.2 months versus zoledronic acid (P = .01). This hypothesis-generating outcome warrants further investigation and 2 studies in lung cancer are ongoing to elucidate the therapeutic potential of denosumab beyond SRE prevention.
Collapse
Affiliation(s)
| | - Rosario García
- Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | | | - Dolores Isla
- Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
| | | | | | | |
Collapse
|
40
|
Hu Z, Brooks SA, Dormoy V, Hsu CW, Hsu HY, Lin LT, Massfelder T, Rathmell WK, Xia M, Al-Mulla F, Al-Temaimi R, Amedei A, Brown DG, Prudhomme KR, Colacci A, Hamid RA, Mondello C, Raju J, Ryan EP, Woodrick J, Scovassi AI, Singh N, Vaccari M, Roy R, Forte S, Memeo L, Salem HK, Lowe L, Jensen L, Bisson WH, Kleinstreuer N. Assessing the carcinogenic potential of low-dose exposures to chemical mixtures in the environment: focus on the cancer hallmark of tumor angiogenesis. Carcinogenesis 2015; 36 Suppl 1:S184-S202. [PMID: 26106137 PMCID: PMC4492067 DOI: 10.1093/carcin/bgv036] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 12/12/2014] [Accepted: 12/15/2014] [Indexed: 01/09/2023] Open
Abstract
One of the important 'hallmarks' of cancer is angiogenesis, which is the process of formation of new blood vessels that are necessary for tumor expansion, invasion and metastasis. Under normal physiological conditions, angiogenesis is well balanced and controlled by endogenous proangiogenic factors and antiangiogenic factors. However, factors produced by cancer cells, cancer stem cells and other cell types in the tumor stroma can disrupt the balance so that the tumor microenvironment favors tumor angiogenesis. These factors include vascular endothelial growth factor, endothelial tissue factor and other membrane bound receptors that mediate multiple intracellular signaling pathways that contribute to tumor angiogenesis. Though environmental exposures to certain chemicals have been found to initiate and promote tumor development, the role of these exposures (particularly to low doses of multiple substances), is largely unknown in relation to tumor angiogenesis. This review summarizes the evidence of the role of environmental chemical bioactivity and exposure in tumor angiogenesis and carcinogenesis. We identify a number of ubiquitous (prototypical) chemicals with disruptive potential that may warrant further investigation given their selectivity for high-throughput screening assay targets associated with proangiogenic pathways. We also consider the cross-hallmark relationships of a number of important angiogenic pathway targets with other cancer hallmarks and we make recommendations for future research. Understanding of the role of low-dose exposure of chemicals with disruptive potential could help us refine our approach to cancer risk assessment, and may ultimately aid in preventing cancer by reducing or eliminating exposures to synergistic mixtures of chemicals with carcinogenic potential.
Collapse
Affiliation(s)
- Zhiwei Hu
- To whom correspondence should be addressed. Tel: +1 614 685 4606; Fax: +1-614-247-7205;
| | - Samira A. Brooks
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Valérian Dormoy
- INSERM U1113, team 3 “Cell Signalling and Communication in Kidney and Prostate Cancer”, University of Strasbourg, Facultée de Médecine, 67085 Strasbourg, France
- Department of Cell and Developmental Biology, University of California, Irvine, CA 92697, USA
| | - Chia-Wen Hsu
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Hsue-Yin Hsu
- Department of Life Sciences, Tzu-Chi University, Taiwan, Republic of China
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, Taipei Medical University, Taiwan, Republic of China
| | - Thierry Massfelder
- INSERM U1113, team 3 “Cell Signalling and Communication in Kidney and Prostate Cancer”, University of Strasbourg, Facultée de Médecine, 67085 Strasbourg, France
| | - W. Kimryn Rathmell
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD 20892-3375, USA
| | - Fahd Al-Mulla
- Department of Life Sciences, Tzu-Chi University, Taiwan, Republic of China
| | | | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Florence 50134, Italy
| | - Dustin G. Brown
- Department of Environmental and Radiological Health Sciences
, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523, USA
| | - Kalan R. Prudhomme
- Environmental and Molecular Toxicology, Environmental Health Science Center, Oregon State University, Corvallis, OR 97331, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna, Italy
| | - Roslida A. Hamid
- Faculty of Medicine and Health Sciences, University Putra, Serdang, Selangor, Malaysia
| | - Chiara Mondello
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Jayadev Raju
- Regulatory Toxicology Research Division, Bureau of Chemical Safety, Food Directorate
, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Elizabeth P. Ryan
- Department of Environmental and Radiological Health Sciences
, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523, USA
| | - Jordan Woodrick
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, WashingtonDC 20057, USA
| | - A. Ivana Scovassi
- Institute of Molecular Genetics, National Research Council, Pavia 27100, Italy
| | - Neetu Singh
- Advanced Molecular Science Research Centre (Centre for Advance Research), King George’s Medical University, Lucknow, Uttar Pradesh 226003, India
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna, Italy
| | - Rabindra Roy
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, WashingtonDC 20057, USA
| | - Stefano Forte
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Hosni K. Salem
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Leroy Lowe
- Getting to Know Cancer, Truro, Nova Scotia B2N 1X5, Canada
| | - Lasse Jensen
- Division of Cardiovascular Medicine, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden and
| | - William H. Bisson
- Environmental and Molecular Toxicology, Environmental Health Science Center, Oregon State University, Corvallis, OR 97331, USA
| | - Nicole Kleinstreuer
- Integrated Laboratory Systems, Inc., in support of the National Toxicology Program Interagency Center for the Evaluation of Alternative Toxicological Methods, NIEHS, MD K2-16, RTP, NC 27709, USA
| |
Collapse
|
41
|
Elefteriou F. Chronic stress, sympathetic activation and skeletal metastasis of breast cancer cells. BONEKEY REPORTS 2015; 4:693. [PMID: 25987989 PMCID: PMC4432778 DOI: 10.1038/bonekey.2015.61] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 03/20/2015] [Indexed: 02/07/2023]
Abstract
Improved detection programs and new therapies significantly improved the 5-year survival rate of women with breast cancer. However, some women still relapse and succumb to cancer because of metastatic disease. In particular, chronically depressed patients do not seem to benefit from newly developed treatments and present with shorter survival. The reason for this association is unclear, but recent cues from preclinical studies point to the possible contribution of neuroendocrine factors generated in response to chronic stress and depression. Retrospective clinical studies also suggest a beneficial effect of sympathetic blockade in terms of less advanced disease at diagnosis, lower cancer-specific mortality, longer disease-free survival and reduced metastasis development and tumor recurrence, especially in patients who have taken propranolol before diagnosis. Therefore, β-blockers or therapies normalizing sympathetic tone might be beneficial as early adjuvant therapies to limit skeletal metastases and growth and eventually to improve prognosis in patients with breast cancers.
Collapse
Affiliation(s)
- Florent Elefteriou
- Vanderbilt University School of Medicine, Pharmacology and Cancer Biology, Nashville, TN, USA
| |
Collapse
|
42
|
Wrobel JK, Choi JJ, Xiao R, Eum SY, Kwiatkowski S, Wolff G, Spangler L, Power RF, Toborek M. Selenoglycoproteins attenuate adhesion of tumor cells to the brain microvascular endothelium via a process involving NF-κB activation. J Nutr Biochem 2014; 26:120-9. [PMID: 25465156 DOI: 10.1016/j.jnutbio.2014.09.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 09/09/2014] [Accepted: 09/16/2014] [Indexed: 01/05/2023]
Abstract
Selenium-containing compounds and selenized yeast have anticancer properties. In order to address possible mechanisms involved in these effects, selenoglycoproteins (SGPs) were extracted from selenium-enriched yeast at pH 4.0 and 6.5 (the fractions are called SGP40 and SGP65, respectively), followed by evaluation of their impact on the interactions of lung and breast tumor cells with human brain microvascular endothelial cells (HBMECs). Extracted SGPs, especially SGP40, significantly inhibited adhesion of tumor cells to HBMECs and their transendothelial migration. Because the active components of SGPs are unknown, small selenium-containing compounds [leucyl-valyl-selenomethionyl-arginine (LVSe-MR) and methylselenoadenosine (M-Se-A)], which are normally present in selenized yeast, were introduced as additional treatment groups. Treatment of HBMECs with SGP40, LVSe-MR and M-Se-A induced changes in gene signatures, which suggested a central involvement of nuclear factor (NF)-κB-dependent pathway. These observations were confirmed in the subsequent analysis of NF-κB DNA binding activity, quantitative measurements of the expression of selected genes and proteins, and tumor cell adhesion assay with a specific NF-κB inhibitor as the additional treatment factor. These findings indicate that specific organic selenium-containing compounds have the ability to inhibit tumor cell adhesion to brain endothelial cells via down-regulation of NF-κB. SGPs appear to be more effective than small selenium-containing compounds, suggesting the role of not only selenium but also the glycoprotein component in the observed protective impact.
Collapse
Affiliation(s)
- Jagoda K Wrobel
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jeong June Choi
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Rijin Xiao
- Nutrigenomics Research Center, Alltech, Nicholasville, KY 40356, USA
| | - Sung Yong Eum
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | - Gretchen Wolff
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Leya Spangler
- Nutrigenomics Research Center, Alltech, Nicholasville, KY 40356, USA
| | - Ronan F Power
- Nutrigenomics Research Center, Alltech, Nicholasville, KY 40356, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Jerzy Kukuczka Academy of Physical Education, Katowice 40-065, Poland.
| |
Collapse
|
43
|
RANKL inhibition blocks osteolytic lesions and reduces skeletal tumor burden in models of non-small-cell lung cancer bone metastases. J Thorac Oncol 2014; 9:345-54. [PMID: 24496001 DOI: 10.1097/jto.0000000000000070] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Bone metastasis is a serious complication in patients with lung cancer, occurring in up to 40% of patients. Tumor cell-mediated osteolysis occurs ultimately through induction of RANK ligand (RANKL) within the bone stroma although this hypothesis has not been tested extensively in the setting of non-small-cell lung cancer (NSCLC). By using two novel NSCLC bone metastasis mouse models, we examined the effects of RANKL inhibition on osteolysis and tumor progression. METHODS We treated mice bearing skeletal NSCLC tumors with osteoprotegerin-Fc (OPG-Fc) to assess whether osteoclast inhibition through RANKL inhibition would affect bone metastases at early or late stages of bone colonization. Progression of skeletal tumor was determined by radiography, longitudinal bioluminescent imaging, and histological analyses. RESULTS OPG-Fc reduced development and progression of radiographically evident osteolytic lesions and also significantly reduced skeletal tumor progression in both NSCLC bone metastasis models. In the H1299 human NSCLC bone metastasis model, OPG-Fc plus docetaxel in combination resulted in significantly greater inhibition of skeletal tumor growth compared with either single agent alone. The observed ability of RANKL inhibition to reduce NSCLC osteolytic bone destruction or skeletal tumor burden was associated with decreases in tumor-associated osteoclasts. CONCLUSIONS These results demonstrate that RANKL is required for the development of tumor-induced osteolytic bone destruction caused by NSCLC cells in vivo. RANKL inhibition also reduced skeletal tumor burden, presumably through the indirect mechanism of blocking tumor-induced osteoclastogenesis and resultant production of growth factors and calcium from the bone microenvironment. RANKL inhibition also provided an additive benefit to docetaxel treatment by augmenting the reduction of tumor burden.
Collapse
|
44
|
Song FN, Duan M, Liu LZ, Wang ZC, Shi JY, Yang LX, Zhou J, Fan J, Gao Q, Wang XY. RANKL promotes migration and invasion of hepatocellular carcinoma cells via NF-κB-mediated epithelial-mesenchymal transition. PLoS One 2014; 9:e108507. [PMID: 25268581 PMCID: PMC4182493 DOI: 10.1371/journal.pone.0108507] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 08/21/2014] [Indexed: 12/19/2022] Open
Abstract
Background Metastasis accounts for the most deaths in patients with hepatocellular carcinoma (HCC). Receptor activator of nuclear factor kappa B ligand (RANKL) is associated with cancer metastasis, while its role in HCC remains largely unknown. Methods Immunohistochemistry was performed to determine the expression of RANK in HCC tissue (n = 398). Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot were used to examine the expression of RANK, E-cadherin, N-cadherin, vimentin, Snail, Slug, Twist and MMPs in HCC cells. Wound healing and Transwell assays were used to evaluate cell migration and invasion ability. Results We found that expression of RANK, the receptor of RANKL, was significantly higher in HCC tumor tissues than in peritumor liver tissues (p<0.001). Constitutive expression of RANK was detected in HCC cell lines, which can be up-regulated when HCC cells were stimulated with RANKL. Notably, in vitro experiments showed that activation of RANKL-RANK axis significantly promoted migration and invasion ability of HCC cells. In addition, RANKL stimulation increased the expression levels of N-cadherin, Snail, and Twist, while decreased the expression of E-cadherin, with concomitant activation of NF-κB signaling pathway. Moreover, administration of the NF-κB inhibitor attenuated RANKL-induced migration, invasion and epithelial-mesenchymal transition of HCC cells. Conclusions RANKL could potentiate migration and invasion ability of RANK-positive HCC cells through NF-κB pathway-mediated epithelial-mesenchymal transition, which means that RANKL-RANK axis could be a potential target for HCC therapy.
Collapse
MESH Headings
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antineoplastic Agents, Phytogenic/pharmacology
- Cadherins/agonists
- Cadherins/antagonists & inhibitors
- Cadherins/genetics
- Cadherins/metabolism
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/surgery
- Cell Line, Tumor
- Cell Movement/drug effects
- Collagenases/genetics
- Collagenases/metabolism
- Diffusion Chambers, Culture
- Epithelial-Mesenchymal Transition/drug effects
- Gene Expression Regulation, Neoplastic
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/surgery
- NF-kappa B/agonists
- NF-kappa B/antagonists & inhibitors
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Neoplasm Invasiveness
- Nuclear Proteins/agonists
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- RANK Ligand/genetics
- RANK Ligand/metabolism
- RANK Ligand/pharmacology
- Receptor Activator of Nuclear Factor-kappa B/genetics
- Receptor Activator of Nuclear Factor-kappa B/metabolism
- Sesquiterpenes/pharmacology
- Sesquiterpenes, Guaiane
- Signal Transduction
- Snail Family Transcription Factors
- Transcription Factors/agonists
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Twist-Related Protein 1/agonists
- Twist-Related Protein 1/genetics
- Twist-Related Protein 1/metabolism
- Vimentin/genetics
- Vimentin/metabolism
Collapse
Affiliation(s)
- Fang-Nan Song
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China
| | - Meng Duan
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China
| | - Long-Zi Liu
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China
| | - Zhi-Chao Wang
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China
| | - Jie-Yi Shi
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China
| | - Liu-Xiao Yang
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China
| | - Jian Zhou
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China
- Institute of Biomedical Sciences, Fudan University, Shanghai, P. R. China
| | - Jia Fan
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China
- Institute of Biomedical Sciences, Fudan University, Shanghai, P. R. China
| | - Qiang Gao
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China
- * E-mail: (QG); (XYW)
| | - Xiao-Ying Wang
- Liver Cancer Institute, Zhongshan Hospital, and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Fudan University, Shanghai, P. R. China
- * E-mail: (QG); (XYW)
| |
Collapse
|
45
|
Hirsh V. Targeted treatments of bone metastases in patients with lung cancer. Front Oncol 2014; 4:146. [PMID: 24982847 PMCID: PMC4058774 DOI: 10.3389/fonc.2014.00146] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 05/28/2014] [Indexed: 11/17/2022] Open
Abstract
Until now ~30–40% of patients with advanced lung cancer develop bone metastases, but as the newer therapies are extending survival, the chance of developing bone metastases increases. Bone metastases cause skeletal-related events (SREs) such as pathologic fractures, spinal cord compression, radiation therapy or surgery to bone, or hypercalcemia, which can have debilitating consequences affecting patients’ health-related quality of life (HR-QOL) and performance status (PS). Poor PS then prevents the patients to receive further lines of treatments, which are available today. SREs are associated with increased economic costs. In one clinical trial, the median time to first SRE was only 5 months. Early detection of bone metastases can prevent SREs and avoid inappropriate implementation of major surgery or chemoradiation therapy. With the new generation bisphosphonate zoledronic acid (ZA) or denosumab (anti-RANKL activity), one can reduce the number of patients who experience SREs, decrease the annual incidence of SREs and delay the median time to first SRE. These agents are effective even after the onset of SREs. They are well tolerated, with manageable side effects. The biochemical markers of bone metabolism especially N-telopeptide of type I collagen and bone specific alkaline phosphatase (BALP) can be both prognostic and predictive markers for the patients with bone metastases from non-small cell lung cancer (NSCLC). Anticancer activity of ZA and denosumab further supports their use as soon as bone metastases are diagnosed in patients with NSCLC. Further trials will inform us about the efficacy of these agents for prevention of bone metastases and even about possible effects on visceral metastases.
Collapse
Affiliation(s)
- Vera Hirsh
- McGill University Health Centre, Royal Victoria Hospital , Montreal, QC , Canada
| |
Collapse
|
46
|
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
|
47
|
Abstract
Differential gene expression profiling studies have lead to the identification of several disease biomarkers. However, the oncogenic alterations in coding regions can modify the gene functions without affecting their own expression profiles. Moreover, post-translational modifications can modify the activity of the coded protein without altering the expression levels of the coding gene, but eliciting variations to the expression levels of the regulated genes. These considerations motivate the study of the rewiring of networks co-expressed genes as a consequence of the aforementioned alterations in order to complement the informative content of differential expression. We analyzed 339 mRNAomes of five distinct cancer types to find single genes that presented co-expression patterns strongly differentiated between normal and tumor phenotypes. Our analysis of differentially connected genes indicates the loss of connectivity as a common topological trait of cancer networks, and unveils novel candidate cancer genes. Moreover, our integrated approach that combines the differential expression together with the differential connectivity improves the classic enrichment pathway analysis providing novel insights on putative cancer gene biosystems not still fully investigated.
Collapse
|
48
|
Cheng ML, Fong L. Effects of RANKL-Targeted Therapy in Immunity and Cancer. Front Oncol 2014; 3:329. [PMID: 24432249 PMCID: PMC3882875 DOI: 10.3389/fonc.2013.00329] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 12/23/2013] [Indexed: 01/24/2023] Open
Abstract
The role of the receptor activator of nuclear factor-κB ligand (RANKL)/RANK system is well characterized within bone, where RANKL/RANK signaling mediates osteoclastogenesis and bone resorption. However, this system has also been shown to influence biologic processes beyond the skeletal system, including in the immune system and in cancer. RANKL/RANK signaling is important in lymph-node development, lymphocyte differentiation, dendritic cell survival, T-cell activation, and tolerance induction. The RANKL/RANK axis may also have direct, osteoclast-independent effects on tumor cells. Indeed, activity of the RANKL/RANK pathway in cancer cells has been correlated with tumor progression and advanced disease. Denosumab, a fully human monoclonal antibody against RANKL, inhibits osteoclastogenesis and is widely used not just for the treatment of osteoporosis, but for the prevention of skeletal-related events from bone metastases in solid malignancies such as breast and prostate cancer. The potential effects of denosumab on the immune system have been largely ignored. Nevertheless, with the emergence of immunotherapies for cancer, denosumab may impact the effectiveness of these therapies, especially if they are given in combination. In this article, we review the role of RANKL/RANK in bone, immunity, and cancer. Examining the potential effects of routine treatment with denosumab beyond the bone represents an important area of investigation.
Collapse
Affiliation(s)
- Michael L Cheng
- Division of Hematology and Oncology, Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco , San Francisco, CA , USA
| | - Lawrence Fong
- Division of Hematology and Oncology, Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California San Francisco , San Francisco, CA , USA
| |
Collapse
|
49
|
Syk/JNK/AP-1 signaling pathway mediates interleukin-6-promoted cell migration in oral squamous cell carcinoma. Int J Mol Sci 2014; 15:545-59. [PMID: 24398980 PMCID: PMC3907824 DOI: 10.3390/ijms15010545] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Revised: 12/20/2013] [Accepted: 12/23/2013] [Indexed: 01/15/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) typically migrates and metastasizes. Interleukin-6 (IL-6) is a multifunctional cytokine associated with disease status and cancer outcomes. The effect of IL-6 on human OSCC cells, however, is unknown. Here, we showed that IL-6 increased cell migration and Intercellular adhesion molecule-1 (ICAM-1) expression in OSCC cells. Pretreatment of OSCC cells with IL-6R monoclonal antibody (mAb) significantly abolished IL-6-induced cell migration and ICAM-1 expression. By contrast, IL-6-mediated cell motility and ICAM-1 upregulation were attenuated by the Syk and c-Jun N-terminal kinase (JNK) inhibitors. Stimulation of OSCC cells with IL-6 promoted Syk and JNK phosphorylation. Furthermore, IL-6 enhanced AP-1 activity, and the IL-6R mAb, Syk inhibitor, or JNK inhibitor all reduced IL-6-mediated c-Jun phosphorylation, c-Jun binding to the ICAM-1 promoter, and c-Jun translocation into the nucleus. Our results indicate that IL-6 enhances the migration of OSCC cells by increasing ICAM-1 expression through the IL-6R receptor and the Syk, JNK, and AP-1 signal transduction pathways.
Collapse
|
50
|
Apoptosis signal-regulating kinase 1 is involved in WISP-1-promoted cell motility in human oral squamous cell carcinoma cells. PLoS One 2013; 8:e78022. [PMID: 24205072 PMCID: PMC3804520 DOI: 10.1371/journal.pone.0078022] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 09/12/2013] [Indexed: 11/21/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) has a tendency to migrate and metastasize. WNT1-inducible signaling pathway protein 1 (WISP-1) is a cysteine-rich protein that belongs to the Cyr61, CTGF, Nov (CCN) family of matrix cellular proteins. The effect of WISP-1 on human OSCC cells, however, is unknown. Here, we showed that WISP-1 increased cell migration and intercellular adhesion molecule-1 (ICAM-1) expression in OSCC cells. Pretreatment of cells with integrin αvβ3 monoclonal antibody (mAb) significantly abolished WISP-1–induced cell migration and ICAM-1 expression. On the other hand, WISP-1–mediated cell motility and ICAM-1 upregulation were attenuated by ASK1, JNK, and p38 inhibitor. Furthermore, WISP-1 also enhanced activator protein 1 (AP-1) activation, and the integrin αvβ3 mAb, and ASK1, JNK, and p38 inhibitors reduced WISP-1–mediated AP-1 activation. Moreover, WISP-1 and ICAM-1 expression correlated with the tumor stage of patients with OSCC. Our results indicate that WISP-1 enhances the migration of OSCC cells by increasing ICAM-1 expression through the αvβ3 integrin receptor and the ASK1, JNK/p38, and AP-1 signal transduction pathways.
Collapse
|