1
|
Abedi S, Behmanesh A, Mazhar FN, Bagherifard A, Sami SH, Heidari N, Hossein-Khannazer N, Namazifard S, Kazem Arki M, Shams R, Zarrabi A, Vosough M. Machine learning and experimental analyses identified miRNA expression models associated with metastatic osteosarcoma. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167357. [PMID: 39033966 DOI: 10.1016/j.bbadis.2024.167357] [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: 04/11/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
Osteosarcoma (OS), as the most common primary bone cancer, has a high invasiveness and metastatic potential, therefore, it has a poor prognosis. This study identified early diagnostic biomarkers using miRNA expression profiles associated with osteosarcoma metastasis. In the first step, we used RNA-seq and online microarray data from osteosarcoma tissues and cell lines to identify differentially expressed miRNAs. Then, using seven feature selection algorithms for ranking, the first-ranked miRNAs were selected as input for five machine learning systems. Using network analysis and machine learning algorithms, we developed new diagnostic models that successfully differentiated metastatic osteosarcoma from non-metastatic samples based on newly discovered miRNA signatures. The results showed that miR-34c-3p and miR-154-3p act as the most promising models in the diagnosis of metastatic osteosarcoma. Validation for this model by RT-qPCR in benign tissue and osteosarcoma biopsies confirmed the lower expression of miR-34c-3p and miR-154-3p in OS samples. In addition, a direct correlation between miR-34c-3p expression, miR-154-3p expression and tumor grade was discovered. The combined values of miR-34c-3p and miR-154-3p showed 90 % diagnostic power (AUC = 0.90) for osteosarcoma samples and 85 % (AUC = 0.85) for metastatic osteosarcoma. Adhesion junction and focal adhesion pathways, as well as epithelial-to-mesenchymal transition (EMT) GO terms, were identified as the most significant KEGG and GO terms for the top miRNAs. The findings of this study highlight the potential use of novel miRNA expression signatures for early detection of metastatic osteosarcoma. These findings may help in determining therapeutic approaches with a quantitative and faster method of metastasis detection and also be used in the development of targeted molecular therapy for this aggressive cancer. Further research is needed to confirm the clinical utility of miR-34c-3p and miR-154-3p as diagnostic biomarkers for metastatic osteosarcoma.
Collapse
Affiliation(s)
- Samira Abedi
- Department of Cellular and Molecular Biology, Faculty of Sciences and Advanced Technology in Biology, University of Science and Culture, Tehran, Iran; Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ali Behmanesh
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farid Najd Mazhar
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Bagherifard
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sam Hajialiloo Sami
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Negar Heidari
- Department of Cellular and Molecular Biology, Faculty of Sciences and Advanced Technology in Biology, University of Science and Culture, Tehran, Iran; Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saina Namazifard
- University of Texas at Arlington, Department of Mechanical and Aerospace Engineering, USA
| | - Mandana Kazem Arki
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roshanak Shams
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan; Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
2
|
Sergi CM. Pediatric cancer-pathology and microenvironment influence: a perspective into osteosarcoma and non-osteogenic mesenchymal malignant neoplasms. Discov Oncol 2024; 15:358. [PMID: 39154307 PMCID: PMC11330953 DOI: 10.1007/s12672-024-01240-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024] Open
Abstract
Pediatric cancer remains the leading cause of disease-related death among children aged 1-14 years. A few risk factors have been conclusively identified, including exposure to pesticides, high-dose radiation, and specific genetic syndromes, but the etiology underlying most events remains unknown. The tumor microenvironment (TME) includes stromal cells, vasculature, fibroblasts, adipocytes, and different subsets of immunological cells. TME plays a crucial role in carcinogenesis, cancer formation, progression, dissemination, and resistance to therapy. Moreover, autophagy seems to be a vital regulator of the TME and controls tumor immunity. Autophagy is an evolutionarily conserved intracellular process. It enables the degradation and recycling of long-lived large molecules or damaged organelles using the lysosomal-mediated pathway. The multifaceted role of autophagy in the complicated neoplastic TME may depend on a specific context. Autophagy may function as a tumor-suppressive mechanism during early tumorigenesis by eliminating unhealthy intracellular components and proteins, regulating antigen presentation to and by immune cells, and supporting anti-cancer immune response. On the other hand, dysregulation of autophagy may contribute to tumor progression by promoting genome damage and instability. This perspective provides an assortment of regulatory substances that influence the features of the TME and the metastasis process. Mesenchymal cells in bone and soft-tissue sarcomas and their signaling pathways play a more critical role than epithelial cells in childhood and youth. The investigation of the TME in pediatric malignancies remains uncharted primarily, and this unique collection may help to include novel advances in this setting.
Collapse
Affiliation(s)
- Consolato M Sergi
- Division of Anatomic Pathology, Department of Laboratory Medicine, Children's Hospital of Eastern Ontario (CHEO), University of Ottawa, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada.
- Department of Laboratory Medicine, Stollery Children's Hospital, University of Alberta, Edmonton, AB, Canada.
- University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
3
|
Wang T, Huang J, Chen G, Fu J, Li T, Zou X, Yi H. miR-1293 suppresses osteosarcoma progression by modulating drug sensitivity in response to cisplatin treatment. Int Immunopharmacol 2024; 130:111702. [PMID: 38367464 DOI: 10.1016/j.intimp.2024.111702] [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: 01/17/2024] [Revised: 02/13/2024] [Accepted: 02/13/2024] [Indexed: 02/19/2024]
Abstract
Chemotherapy is considered the primary treatment for osteosarcoma. however, its effectiveness is limited due to drug resistance and toxicity. Thus, identifying novel therapeutic targets to enhance the efficacy of chemotherapy is urgently needed. Here, we identified a novel cisplatin-sensitivity enhancing mechanism via up-regulation of the tumour suppressor gene, miR-1293. Meanwhile, higher levels of miR-1293 observed in prechemotherapy patients were associated with a more favorable prognosis. The mechanism underlying cisplatin upregulated miR-1293 expression involves hypomethylation of the miR-1293 promoter, which blocks the binding of the transcription repressor TFAP2A to the promoter. Furthermore, miR-1293 inhibits osteosarcoma progression by targeting TIMP1 to inactivate the Notch1/Hes1 and TGFBR1/Smad2/3 pathways, thereby promoting tumour cell death. The findings presented herein unveil a novel mechanism for enhancing cisplatin sensitivity and proposed a potential therapeutic strategy for osteosarcoma through pre-chemotherapy supplementation of miR-1293.
Collapse
Affiliation(s)
- Tingxuan Wang
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510005, China
| | - Jincheng Huang
- Department of Orthopedics, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou 450000, China.
| | - Gang Chen
- Department of Orthopedics, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441000, China
| | - Jiahui Fu
- Department of Fetal Medicine and Prenatal Diagnosis, Zhujiang Hospital, Southern Medical University, Guangzhou 510005, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 10032, China.
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510005, China.
| | - Hualin Yi
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510005, China; Guangzhou National Laboratory, Guangzhou 510005, China.
| |
Collapse
|
4
|
Gelman IH. Metastasis suppressor genes in clinical practice: are they druggable? Cancer Metastasis Rev 2023; 42:1169-1188. [PMID: 37749308 DOI: 10.1007/s10555-023-10135-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/01/2023] [Indexed: 09/27/2023]
Abstract
Since the identification of NM23 (now called NME1) as the first metastasis suppressor gene (MSG), a small number of other gene products and non-coding RNAs have been identified that suppress specific parameters of the metastatic cascade, yet which have little or no ability to regulate primary tumor initiation or maintenance. MSG can regulate various pathways or cell biological functions such as those controlling mitogen-activated protein kinase pathway mediators, cell-cell and cell-extracellular matrix protein adhesion, cytoskeletal architecture, G-protein-coupled receptors, apoptosis, and transcriptional complexes. One defining facet of this gene class is that their expression is typically downregulated, not mutated, in metastasis, such that any effective therapeutic intervention would involve their re-expression. This review will address the therapeutic targeting of MSG, once thought to be a daunting task only facilitated by ectopically re-expressing MSG in metastatic cells in vivo. Examples will be cited of attempts to identify actionable oncogenic pathways that might suppress the formation or progression of metastases through the re-expression of specific metastasis suppressors.
Collapse
Affiliation(s)
- Irwin H Gelman
- Department of Cancer Genetics & Genomics, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| |
Collapse
|
5
|
Quartieri M, Puspitasari A, Vitacchio T, Durante M, Tinganelli W. The role of hypoxia and radiation in developing a CTCs-like phenotype in murine osteosarcoma cells. Front Cell Dev Biol 2023; 11:1222809. [PMID: 38033871 PMCID: PMC10687637 DOI: 10.3389/fcell.2023.1222809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/11/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction: Cancer treatment has evolved significantly, yet concerns about tumor recurrence and metastasis persist. Within the dynamic tumor microenvironment, a subpopulation of mesenchymal tumor cells, known as Circulating Cancer Stem Cells (CCSCs), express markers like CD133, TrkB, and CD47, making them radioresistant and pivotal to metastasis. Hypoxia intensifies their stemness, complicating their identification in the bloodstream. This study investigates the interplay of acute and chronic hypoxia and radiation exposure in selecting and characterizing cells with a CCSC-like phenotype. Methods: LM8 murine osteosarcoma cells were cultured and subjected to normoxic (21% O2) and hypoxic (1% O2) conditions. We employed Sphere Formation and Migration Assays, Western Blot analysis, CD133 Cell Sorting, and CD133+ Fluorescent Activated Cell Sorting (FACS) analysis with a focus on TrkB antibody to assess the effects of acute and chronic hypoxia, along with radiation exposure. Results: Our findings demonstrate that the combination of radiation and acute hypoxia enhances stemness, while chronic hypoxia imparts a cancer stem-like phenotype in murine osteosarcoma cells, marked by increased migration and upregulation of CCSC markers, particularly TrkB and CD47. These insights offer a comprehensive understanding of the interactions between radiation, hypoxia, and cellular responses in the context of cancer treatment. Discussion: This study elucidates the complex interplay among radiation, hypoxia, and cellular responses, offering valuable insights into the intricacies and potential advancements in cancer treatment.
Collapse
Affiliation(s)
- Martina Quartieri
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
| | - Anggraeini Puspitasari
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
- Biology Division, Gunma University Heavy Ion Medical Center, Maebashi, Japan
| | - Tamara Vitacchio
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
| | - Marco Durante
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
- Institut für Festkörperphysik, Technische Universität Darmstadt, Darmstadt, Germany
| | - Walter Tinganelli
- Biophysics Department, GSI Helmholtzzentrum für Schwerionenforschung GmbH, Darmstadt, Germany
| |
Collapse
|
6
|
Cui M, Xiaoyu Chen, Luo X, Zhou Z, Chen Z, Zhou Z, Zhou X, Zou H, Xu T, Wang S, Yang M. Dually stimulative single-chain polymeric nano lock with dynamic ligands for sensitive detection of circulating tumor cells. Biosens Bioelectron 2022; 217:114692. [PMID: 36150325 DOI: 10.1016/j.bios.2022.114692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 08/12/2022] [Accepted: 09/02/2022] [Indexed: 12/24/2022]
Abstract
Circulating tumor cells (CTCs) are important markers for cancer diagnosis and monitoring. However, CTCs detection remains challenging due to their scarcity, where most of the detection methods are compromised by the loss of CTCs in pre-enrichment, and by the lack of universal antibodies for capturing different kinds of cancer cells. Herein, we report a single-chain based nano lock (SCNL) polymer incorporating dually stimulative dynamic ligands that can bind with a broad spectrum of cancer cells and CTCs overexpressing sialic acid (SA) with high sensitivity and selectivity. The high sensitivity is realized by the polymeric single chain structure and the multi-valent functional moieties, which improve the accessibility and binding stability between the target cells and the SCNL. The highly selective targeting of cancer cells is achieved by the dynamic and dually stimulative nano lock structures, which can be unlocked and functionalized upon simultaneous exposure to overexpressed SA and acidic microenvironment. We applied the SCNL to detecting cancer cells and CTCs in clinical samples, where the detection threshold of SCNL reached 4 cells/mL. Besides CTCs enumeration, the SCNL approach could also be extended to metastasis assessment through monitoring the expressing level of surface SA on cancer cells.
Collapse
Affiliation(s)
- Miao Cui
- Shenzhen Bay Laboratory, Shenzhen, 518132, China; Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China.
| | - Xiaoyu Chen
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Xu Luo
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Zhihang Zhou
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China; Department of Gastroenterology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhiji Chen
- Department of Gastroenterology, Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhengdong Zhou
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China
| | - Xiaoyu Zhou
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Heng Zou
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China; Cellomics (Shenzhen) Limited, Shenzhen, China
| | - Tao Xu
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China; Cellomics (Shenzhen) Limited, Shenzhen, China
| | - Shubin Wang
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Mengsu Yang
- Department of Biomedical Sciences, and Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong, China; Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China.
| |
Collapse
|
7
|
Chen X, Xiang H, Yu S, Lu Y, Wu T. Research progress in the role and mechanism of Cadherin-11 in different diseases. J Cancer 2021; 12:1190-1199. [PMID: 33442417 PMCID: PMC7797656 DOI: 10.7150/jca.52720] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/29/2020] [Indexed: 12/16/2022] Open
Abstract
Cadherin is an important cell-cell adhesion molecule, which mediates intercellular adhesion through calcium dependent affinity interaction. Cadherin-11 (CDH11, OB-cadherin) is a member of cadherin family, and its gene is situated on chromosome 16q22.1. Increasing lines of researches have proved that CDH11 plays important roles in the occurrence and development of a lot of diseases, such as tumors, arthritis and so on. CDH11 often leads to promoter methylation inactivation, which can induce cancer cell apoptosis, suppress cell motility and invasion, and can inhibit cancer through Wnt/β-catenin, AKT/Rho A and NF-κB signaling pathways. This review focused on the current knowledge of CDH11, including its function and mechanism in different diseases. In this article, we aimed to have a more comprehensive and in-depth understanding of CDH11 and to provide new ideas for the treatment of some diseases.
Collapse
Affiliation(s)
- Xinyi Chen
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hongjiao Xiang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shiyu Yu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yifei Lu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
8
|
Yang Z, Yan C, Yu Z, He C, Li J, Li C, Yan M, Liu B, Wu Y, Zhu Z. Downregulation of CDH11 Promotes Metastasis and Resistance to Paclitaxel in Gastric Cancer Cells. J Cancer 2021; 12:65-75. [PMID: 33391403 PMCID: PMC7738820 DOI: 10.7150/jca.48193] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 10/23/2020] [Indexed: 11/28/2022] Open
Abstract
Background: Gastric cancer (GC) with peritoneal metastasis has an extremely poor prognosis. Paclitaxel (PTX) intraperitoneal infusion provides an effective treatment for these patients. However, GC patients with peritoneal metastasis who receiving PTX treatments tend to occur PTX-resistance accompany with more aggressive ascites and metastasis. How does this happen is still unknown. Here, we aimed to explore the mechanisms that mediate PTX-resistance and metastasis in GC with peritoneal metastasis. Methods: Ascites samples were collected before PTX infusion and after the relapse in 3 GC patients. To determine the expression of significantly changed proteins, we performed tandem mass tag (TMT) quantitative proteomics. Immunohistochemistry (IHC) staining and western blot were performed to confirm the expression of CDH11 in the PTX-resistant tissues and MKN45P-PR cells. Invasion and migration of GC cells were examined by in vitro transwell and wound healing assays and in vivo dissemination experiments. Results: CDH11 expression was downregulated in the relapsed PTX-resistant ascites, tissues and the PTX-resistant cell line MKN45P-PR. Inhibition of CDH11 expression promoted the invasion, migration and PTX resistance of MKN45P cells, while overexpression of CDH11 repressed these biological functions. Moreover, tumors disseminated in the mice peritoneal cavity induced by MKN45P-PR cells and shCDH11 cells displayed higher metastatic ability and resistance to PTX treatment. Conclusions: Our results reveal that CDH11 is inhibited in the relapsed PTX-resistant patients and the downregulated CDH11 expression promotes GC cell invasion, migration and PTX resistance. CDH11 may have the potential to serve as a predictable marker for the occurrence of PTX resistance in GC patients with peritoneal metastasis.
Collapse
Affiliation(s)
- Zhongyin Yang
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chao Yan
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhenjia Yu
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Changyu He
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jianfang Li
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chen Li
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Min Yan
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bingya Liu
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yingli Wu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital / Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhenggang Zhu
- Department of General Surgery, Gastrointestinal Surgery, Shanghai Key laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
9
|
Oleksiewicz U, Machnik M. Causes, effects, and clinical implications of perturbed patterns within the cancer epigenome. Semin Cancer Biol 2020; 83:15-35. [PMID: 33359485 DOI: 10.1016/j.semcancer.2020.12.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/18/2020] [Accepted: 12/18/2020] [Indexed: 02/06/2023]
Abstract
Somatic mutations accumulating over a patient's lifetime are well-defined causative factors that fuel carcinogenesis. It is now clear, however, that epigenomic signature is also largely perturbed in many malignancies. These alterations support the transcriptional program crucial for the acquisition and maintenance of cancer hallmarks. Epigenetic instability may arise due to the genetic mutations or transcriptional deregulation of the proteins implicated in epigenetic signaling. Moreover, external stimulation and physiological aging may also participate in this phenomenon. The epigenomic signature is frequently associated with a cell of origin, as well as with tumor stage and differentiation, which all reflect its high heterogeneity across and within various tumors. Here, we will overview the current understanding of the causes and effects of the altered and heterogeneous epigenomic landscape in cancer. We will focus mainly on DNA methylation and post-translational histone modifications as the key regulatory epigenetic signaling marks. In addition, we will describe how this knowledge is translated into the clinic. We will particularly concentrate on the applicability of epigenetic alterations as biomarkers for improved diagnosis, prognosis, and prediction. Finally, we will also review current developments regarding epi-drug usage in clinical and experimental settings.
Collapse
Affiliation(s)
- Urszula Oleksiewicz
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznan, Poland; Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Centre, Poznan, Poland.
| | - Marta Machnik
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznan, Poland; Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Centre, Poznan, Poland
| |
Collapse
|
10
|
Transformable peptide nanoparticles inhibit the migration of N-cadherin overexpressed cancer cells. CHINESE CHEM LETT 2020. [DOI: 10.1016/j.cclet.2020.03.065] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
11
|
Serial Xenotransplantation in NSG Mice Promotes a Hybrid Epithelial/Mesenchymal Gene Expression Signature and Stemness in Rhabdomyosarcoma Cells. Cancers (Basel) 2020; 12:cancers12010196. [PMID: 31941033 PMCID: PMC7016569 DOI: 10.3390/cancers12010196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/13/2019] [Accepted: 01/09/2020] [Indexed: 02/07/2023] Open
Abstract
Serial xenotransplantation of sorted cancer cells in immunodeficient mice remains the most complex test of cancer stem cell (CSC) phenotype. However, we have demonstrated in various sarcomas that putative CSC surface markers fail to identify CSCs, thereby impeding the isolation of CSCs for subsequent analyses. Here, we utilized serial xenotransplantation of unsorted rhabdomyosarcoma cells in NOD/SCID gamma (NSG) mice as a proof-of-principle platform to investigate the molecular signature of CSCs. Indeed, serial xenotransplantation steadily enriched for rhabdomyosarcoma stem-like cells characterized by enhanced aldehyde dehydrogenase activity and increased colony and sphere formation capacity in vitro. Although the expression of core pluripotency factors (SOX2, OCT4, NANOG) and common CSC markers (CD133, ABCG2, nestin) was maintained over the passages in mice, gene expression profiling revealed gradual changes in several stemness regulators and genes linked with undifferentiated myogenic precursors, e.g., SOX4, PAX3, MIR145, and CDH15. Moreover, we identified the induction of a hybrid epithelial/mesenchymal gene expression signature that was associated with the increase in CSC number. In total, 60 genes related to epithelial or mesenchymal traits were significantly altered upon serial xenotransplantation. In silico survival analysis based on the identified potential stemness-associated genes demonstrated that serial xenotransplantation of unsorted rhabdomyosarcoma cells in NSG mice might be a useful tool for the unbiased enrichment of CSCs and the identification of novel CSC-specific targets. Using this approach, we provide evidence for a recently proposed link between the hybrid epithelial/mesenchymal phenotype and cancer stemness.
Collapse
|
12
|
Mesenchymal Stem Cells in the Adult Human Liver: Hype or Hope? Cells 2019; 8:cells8101127. [PMID: 31546729 PMCID: PMC6830330 DOI: 10.3390/cells8101127] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/18/2019] [Accepted: 09/21/2019] [Indexed: 02/06/2023] Open
Abstract
Chronic liver diseases constitute a significant economic, social, and biomedical burden. Among commonly adopted approaches, only organ transplantation can radically help patients with end-stage liver pathologies. Cell therapy with hepatocytes as a treatment for chronic liver disease has demonstrated promising results. However, quality human hepatocytes are in short supply. Stem/progenitor cells capable of differentiating into functionally active hepatocytes provide an attractive alternative approach to cell therapy for liver diseases, as well as to liver-tissue engineering, drug screening, and basic research. The application of methods generally used to isolate mesenchymal stem cells (MSCs) and maintain them in culture to human liver tissue provides cells, designated here as liver MSCs. They have much in common with MSCs from other tissues, but differ in two aspects-expression of a range of hepatocyte-specific genes and, possibly, inherent commitment to hepatogenic differentiation. The aim of this review is to analyze data regarding liver MSCs, probably another type of liver stem/progenitor cells different from hepatic stellate cells or so-called hepatic progenitor cells. The review presents an analysis of the phenotypic characteristics of liver MSCs, their differentiation and therapeutic potential, methods for isolating these cells from human liver, and discusses issues of their origin and heterogeneity. Human liver MSCs are a fascinating object of fundamental research with a potential for important practical applications.
Collapse
|
13
|
The Tumor Microenvironment of Pediatric Sarcoma: Mesenchymal Mechanisms Regulating Cell Migration and Metastasis. Curr Oncol Rep 2019; 21:90. [PMID: 31418125 PMCID: PMC6695368 DOI: 10.1007/s11912-019-0839-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW This review presents a selection of regulatory molecules of tumor microenvironmental properties and metastasis. Signaling pathways controlling mesenchymal biology in bone and soft-tissue sarcomas found in children and adolescents are prioritized. RECENT FINDINGS The tumor microenvironment of pediatric tumors is still relatively unexplored. Highlighted findings are mainly on deregulated genes associated with cell adhesion, migration, and tumor cell dissemination. How these processes are involved in a mesenchymal phenotype and metastasis is further discussed in relation to the epithelial to mesenchymal transition (EMT) in epithelial tumors. Cell plasticity is emerging as a concept with impact on tumor behavior. Sarcomas belong to a heterogeneous group of tumors where local recurrence and tumor spread pose major challenges despite intense multimodal treatments. Molecular pathways involved in the metastatic process are currently being characterized, and tumor-regulatory properties of structural components, and infiltrating, non-malignant cell types should be further investigated.
Collapse
|
14
|
Nguyen T, Duchesne L, Sankara Narayana GHN, Boggetto N, Fernig DD, Uttamrao Murade C, Ladoux B, Mège RM. Enhanced cell-cell contact stability and decreased N-cadherin-mediated migration upon fibroblast growth factor receptor-N-cadherin cross talk. Oncogene 2019; 38:6283-6300. [PMID: 31312021 DOI: 10.1038/s41388-019-0875-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 06/17/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022]
Abstract
N-cadherin adhesion has been reported to enhance cancer and neuronal cell migration either by mediating actomyosin-based force transduction or initiating fibroblast growth factor receptor (FGFR)-dependent biochemical signalling. Here we show that FGFR1 reduces N-cadherin-mediated cell migration. Both proteins are co-stabilised at cell-cell contacts through direct interaction. As a consequence, cell adhesion is strengthened, limiting the migration of cells on N-cadherin. Both the inhibition of migration and the stabilisation of cell adhesions require the FGFR activity stimulated by N-cadherin engagement. FGFR1 stabilises N-cadherin at the cell membrane through a pathway involving Src and p120. Moreover, FGFR1 stimulates the anchoring of N-cadherin to actin. We found that the migratory behaviour of cells depends on an optimum balance between FGFR-regulated N-cadherin adhesion and actin dynamics. Based on these findings we propose a positive feed-back loop between N-cadherin and FGFR at adhesion sites limiting N-cadherin-based single-cell migration.
Collapse
Affiliation(s)
- Thao Nguyen
- Institut Jacques Monod, CNRS, Université Paris Diderot, 15 Rue Hélène Brion, 75205, Paris Cedex 13, France
| | - Laurence Duchesne
- Univ Rennes, CNRS, IGDR (Institute of Genetics and Development of Rennes) - UMR 6290, F-35000, Rennes, France
| | | | - Nicole Boggetto
- Institut Jacques Monod, CNRS, Université Paris Diderot, 15 Rue Hélène Brion, 75205, Paris Cedex 13, France
| | - David D Fernig
- Department of Biochemistry, Institute of Integrated Biology, University of Liverpool, Liverpool, L69 7ZB, UK
| | | | - Benoit Ladoux
- Institut Jacques Monod, CNRS, Université Paris Diderot, 15 Rue Hélène Brion, 75205, Paris Cedex 13, France
| | - René-Marc Mège
- Institut Jacques Monod, CNRS, Université Paris Diderot, 15 Rue Hélène Brion, 75205, Paris Cedex 13, France.
| |
Collapse
|
15
|
Zhao G, Shi J, Xia J. Analysis of the association between CDH2 gene polymorphism and osteoarthritis risk. Med Sci (Paris) 2018; 34 Focus issue F1:105-112. [PMID: 30403184 DOI: 10.1051/medsci/201834f118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE to define the cadherin 2 (CDH2) gene polymorphism in Chinese osteoarthritis and control populations and to explore the correlation between CDH2 gene polymorphism and the risk of osteoarthritis. METHOD a total of 476 patients with osteoarthritis were collected and 380 control subjects were included in the study. Clinical data such as gender, age and functional score were collected. The blood and tissue samples were collected and genotyped by PCR. Data analysis was performed using SPSS 19.0, Hapioview 4.2 and SNPstats softwares. RESULTS the association of rs11083271 and osteoarthritis was initially validated in this study population (P = 0.016, OR = 1.43 (1.07- 1.93)]. The risk of OA was significantly higher in heterozygous T/C than in homozygous T/T and C/C in rs11083271. By adjusting the age, according to gender stratification analysis, the heterozygous T/C genotype in rs11083271 significantly increased the risk of OA incidence in males [p = 0.011, 3.40 (1.55-7.43)]. The remaining rs sites were not significantly associated with OA. Notably, the association of rs11564299 with OA, regardless of genotyping, gene frequency and RNA expression levels in the study population, was not confirmed. CONCLUSION in this study, we have analyzed the association between CDH2 gene polymorphism and OA in Chinese population. We found that rs11083271 heterozygous T/C genotype significantly increases the risk of OA and the severity of the disease. By contrast, the rs11564299 locus and OA have no significant correlation in the Chinese population. The role of rs11083271 in the regulation of CDH2 expression levels and the mechanisms by which it impacts OA remain to be further studied.
Collapse
Affiliation(s)
- Guanglei Zhao
- Division of orthopaedic surgery, Huashan Hospital, Fudan University, Shanghai, China, 400040 Shanghai, China
| | - Jingsheng Shi
- Division of orthopaedic surgery, Huashan Hospital, Fudan University, Shanghai, China, 400040 Shanghai, China
| | - Jun Xia
- Division of orthopaedic surgery, Huashan Hospital, Fudan University, Shanghai, China, 400040 Shanghai, China
| |
Collapse
|
16
|
Qin Y, Ye J, Zhao F, Hu S, Wang S. TRIM2 regulates the development and metastasis of tumorous cells of osteosarcoma. Int J Oncol 2018; 53:1643-1656. [PMID: 30066883 DOI: 10.3892/ijo.2018.4494] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 06/14/2018] [Indexed: 11/05/2022] Open
Abstract
The present study aimed to investigate candidate genes involved in the development and metastasis of osteosarcoma. Candidate genes were screened preliminarily from the Gene Expression Omnibus database and then validated using actual tumor tissues collected from patients with osteosarcoma. The cells were prepared and transfected with specific gene-targeted small interfering RNA followed by an MTS assay for cell viability detection and Transwell assays for cell migration and invasion capacity detection. The cell apoptosis was determined by flow cytometry and the protein level of the genes was detected by western blot analysis. An in vivo nude model was used and injected with cells to detect the functions of the genes. Transcriptome sequencing was performed to verify the regulation network, followed by reverse transcription-quantitative polymerase chain reaction and western blot analyses for validation. Increased tripartite motif-containing protein 2 (TRIM2) was detected in the osteosarcoma tumor tissues compared with normal tissues. The inhibition of TRIM2 induced lower cell viability and cell invasion capacity, and increased the rate of cell apoptosis. Decreased TRIM2 also inhibited the development and metastasis of osteosarcoma in the nude mouse models. The transcriptome sequencing revealed that the regulation of TRIM2 may be correlated with genes, Sirtuin 4, DNA damage inducible transcript 3, cAMP responsive element binding protein 5, G protein-coupled receptor 65 (GPR65) and ADP-ribosyltransferase 5. Western blot analysis indicated that TRIM2 regulated the development and metastasis of osteosarcoma via the phosphoinositide 3-kinase/protein kinase B signaling pathway. Therefore, TRIM2 performs important functions in regulating the development and metastasis of osteosarcoma.
Collapse
Affiliation(s)
- Yi Qin
- Department of Orthopedics, Zhuhai Hospital, Jinan University, Zhuhai People's Hospital, Zhuhai, Guangdong 519000, P.R. China
| | - Jichao Ye
- Department of Orthopedics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, P.R. China
| | - Fulan Zhao
- Department of Orthopedics, Zhuhai Hospital, Jinan University, Zhuhai People's Hospital, Zhuhai, Guangdong 519000, P.R. China
| | - Shaoyu Hu
- Department of Orthopedics, Zhuhai Hospital, Jinan University, Zhuhai People's Hospital, Zhuhai, Guangdong 519000, P.R. China
| | - Suwei Wang
- Department of Orthopedics, Zhuhai Hospital, Jinan University, Zhuhai People's Hospital, Zhuhai, Guangdong 519000, P.R. China
| |
Collapse
|
17
|
Tang Q, Lu J, Zou C, Shao Y, Chen Y, Narala S, Fang H, Xu H, Wang J, Shen J, Khokha R. CDH4 is a novel determinant of osteosarcoma tumorigenesis and metastasis. Oncogene 2018; 37:3617-3630. [PMID: 29610525 DOI: 10.1038/s41388-018-0231-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 11/26/2017] [Accepted: 12/29/2017] [Indexed: 01/02/2023]
Abstract
The era of cancer genomics now provides an opportunity to discover novel determinants of osteosarcoma (OS), the most common primary bone cancer in children and adolescents known for its poor prognosis due to lung metastasis. Here, we identify CDH4 amplification in 43.6% of human osteosarcoma using array CGH and demonstrate its critical role in osteosarcoma development and progression. Gain or loss-of-function of CDH4, which encodes R-cadherin, causally impacts multiple features of human OS cells including cell migration and invasion, osteogenic differentiation, and stemness. CDH4 overexpression activates c-Jun via the JNK pathway, while CDH4 knockdown suppresses both tumor xenograft growth and lung colonization. In OS patient specimens, high CDH4 expression associates with lung metastases and poor prognosis. Collectively, our bioinformatics, functional, molecular, and clinical analyses uncover an oncogenic function of CDH4 in osteosarcoma and its relationship with patient outcome.
Collapse
Affiliation(s)
- Qinglian Tang
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, M5G 0A3, Canada.,Department of Orthopedic Oncology, First Affiliated Hospital, Sun Yat-Sen University, 510080, Guangzhou, China
| | - Jinchang Lu
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, M5G 0A3, Canada.,Department of Orthopedic Oncology, First Affiliated Hospital, Sun Yat-Sen University, 510080, Guangzhou, China
| | - Changye Zou
- Department of Orthopedic Oncology, First Affiliated Hospital, Sun Yat-Sen University, 510080, Guangzhou, China
| | - Yang Shao
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, M5G 0A3, Canada
| | - Yan Chen
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, M5G 0A3, Canada
| | - Swami Narala
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, M5G 0A3, Canada
| | - Hui Fang
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, M5G 0A3, Canada
| | - Huaiyuan Xu
- Department of Orthopedic Oncology, First Affiliated Hospital, Sun Yat-Sen University, 510080, Guangzhou, China
| | - Jin Wang
- Department of Orthopedic Oncology, First Affiliated Hospital, Sun Yat-Sen University, 510080, Guangzhou, China
| | - Jingnan Shen
- Department of Orthopedic Oncology, First Affiliated Hospital, Sun Yat-Sen University, 510080, Guangzhou, China.
| | - Rama Khokha
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, M5G 0A3, Canada.
| |
Collapse
|
18
|
Kim BJ, Lee YS, Lee SY, Baek WY, Choi YJ, Moon SA, Lee SH, Kim JE, Chang EJ, Kim EY, Yoon J, Kim SW, Ryu SH, Lee SK, Lorenzo JA, Ahn SH, Kim H, Lee KU, Kim GS, Koh JM. Osteoclast-secreted SLIT3 coordinates bone resorption and formation. J Clin Invest 2018; 128:1429-1441. [PMID: 29504949 PMCID: PMC5873876 DOI: 10.1172/jci91086] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 12/28/2017] [Indexed: 01/15/2023] Open
Abstract
Coupling is the process that links bone resorption to bone formation in a temporally and spatially coordinated manner within the remodeling cycle. Several lines of evidence point to the critical roles of osteoclast-derived coupling factors in the regulation of osteoblast performance. Here, we used a fractionated secretomic approach and identified the axon-guidance molecule SLIT3 as a clastokine that stimulated osteoblast migration and proliferation by activating β-catenin. SLIT3 also inhibited bone resorption by suppressing osteoclast differentiation in an autocrine manner. Mice deficient in Slit3 or its receptor, Robo1, exhibited osteopenic phenotypes due to a decrease in bone formation and increase in bone resorption. Mice lacking Slit3 specifically in osteoclasts had low bone mass, whereas mice with either neuron-specific Slit3 deletion or osteoblast-specific Slit3 deletion had normal bone mass, thereby indicating the importance of SLIT3 as a local determinant of bone metabolism. In postmenopausal women, higher circulating SLIT3 levels were associated with increased bone mass. Notably, injection of a truncated recombinant SLIT3 markedly rescued bone loss after an ovariectomy. Thus, these results indicate that SLIT3 plays an osteoprotective role by synchronously stimulating bone formation and inhibiting bone resorption, making it a potential therapeutic target for metabolic bone diseases.
Collapse
Affiliation(s)
- Beom-Jun Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Young-Sun Lee
- Asan Institute for Life Sciences, Seoul, South Korea
| | - Sun-Young Lee
- Asan Institute for Life Sciences, Seoul, South Korea
| | | | | | - Sung Ah Moon
- Asan Institute for Life Sciences, Seoul, South Korea
| | - Seung Hun Lee
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jung-Eun Kim
- Department of Molecular Medicine, Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu, South Korea
| | | | | | - Jin Yoon
- Department of Pharmacology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Seung-Whan Kim
- Department of Pharmacology, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sung Ho Ryu
- Department of Life Science, Pohang University of Science and Technology, Pohang, Kyungbook, South Korea
| | | | - Joseph A. Lorenzo
- Departments of Medicine and Orthopaedics, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Seong Hee Ahn
- Department of Internal Medicine, Inha University School of Medicine, Incheon, South Korea
| | - Hyeonmok Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ki-Up Lee
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ghi Su Kim
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jung-Min Koh
- Division of Endocrinology and Metabolism, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
19
|
Khan I, Steeg PS. Metastasis suppressors: functional pathways. J Transl Med 2018; 98:198-210. [PMID: 28967874 PMCID: PMC6545599 DOI: 10.1038/labinvest.2017.104] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Revised: 06/29/2017] [Accepted: 06/30/2017] [Indexed: 12/13/2022] Open
Abstract
Metastasis is a complex process and a major contributor of death in cancer patients. Metastasis suppressor genes are identified by their ability to inhibit metastasis at a secondary site without affecting the growth of primary tumor. In this review, we have conducted a survey of the metastasis suppressor literature to identify common downstream pathways. The metastasis suppressor genes mechanistically target MAPK, G-protein-coupled receptor, cell adhesion, cytoskeletal, transcriptional regulatory, and metastasis susceptibility pathways. The majority of the metastasis suppressor genes are functionally multifactorial, inhibiting metastasis at multiple points in the cascade, and many operate in a context-dependent fashion. A greater understanding of common pathways/molecules targeted by metastasis suppressor could improve metastasis treatment strategies.
Collapse
|
20
|
Fan M, Liu Y, Xia F, Wang Z, Huang Y, Li J, Wang Z, Li X. Increased expression of EphA2 and E-N cadherin switch in primary hepatocellular carcinoma. TUMORI JOURNAL 2018; 99:689-96. [DOI: 10.1177/030089161309900608] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Aim To investigate the expression and clinical significance of ephrin type-A receptor 2 and epithelial-mesenchymal transition-related proteins in primary hepatocellular carcinoma. Methods Tissues from 52 primary hepatocellular carcinomas and 12 human normal liver tissues were detected for expression of ephrin type-A receptor 2, E-cadherin, and N-cadherin by immunochemistry. Cinicopathological features of hepatocellular carcinoma and tumor recurrence after operation were studied for the association with these molecular expressions and E-N cadherin switch. Results Increased expressions of ephrin type-A receptor 2 and N-cadherin and reduced expression of E-cadherin were significantly detected in hepatocellular carcinoma compared with normal liver tissues. Univariate analysis showed that there were close associations between unfavorable clinicopathological features and expressions of ephrin type-A receptor 2, E-cadherin, N-cadherin, and E-N cadherin switch. Ephrin type-A receptor 2 and E-cadherin expressions were confirmed as independent prognostic factors when corrected with age, gender, AFP, HBsAg, liver cirrhosis, tumor size, nodules, capsule, portal vein invasion, cell differentiation, and TNM stage. Conclusions The overexpression of ephrin type-A receptor 2 protein is correlated with the number of tumors, capsular integrity, portal vein cancer thrombus and clinical stages. Epithelial-mesenchymal transition regulated by ephrin type-A receptor 2 is involved in the aggressive clinicopathological features and prognosis, suggesting that the receptor may play an important role in the progression and metastasis of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Min Fan
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha
- Department of Geriatrics, Xiangya Second Hospital, Central South University, Changsha
| | - Yu Liu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha
- Department of General Surgery, Yueyang First People's Hospital, Yueyang, China
| | - Fada Xia
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha
| | - Zhuolu Wang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha
| | - Yun Huang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha
| | - Jingdong Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha
| | - Zhiming Wang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha
| | - Xinying Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha
| |
Collapse
|
21
|
Zhang ZC, Tang C, Dong Y, Zhang J, Yuan T, Li XL. Targeting LncRNA-MALAT1 suppresses the progression of osteosarcoma by altering the expression and localization of β-catenin. J Cancer 2018; 9:71-80. [PMID: 29290771 PMCID: PMC5743713 DOI: 10.7150/jca.22113] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 10/24/2017] [Indexed: 12/29/2022] Open
Abstract
Osteosarcoma (OS), which affects adolescents especially during a growth spurt, has the highest incidence of any primary malignant bone tumour, and a high rate of early metastasis leading to a very poor prognosis. In recent years, non-coding RNAs, especially long non-coding RNAs (lncRNAs) have attracted more and more attention as novel epigenetic regulators in a variety of tumours, including OS. Most recently, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) was found to play an important role in OS progression by modulating the enhancers of zeste homolog 2 (EZH2). Furthermore, MALAT1 could inhibit the expression of E-cadherin and promote the expression of β-catenin, and this phenomenon might be the outcome of MALAT1-induced EZH2 activation. In this study, we investigated the vital function of MALAT1 in the progression of OS and its potential leading mechanism, altering the expression and localization of β-catenin via epigenetic transcriptional regulation by interacting with EZH2. With the help of MALAT1 silencing using small interfering RNAs (siRNAs), the loss of E-cadherin of MNNG/HOS cells was rescued, and the abnormal expression and localization of β-catenin were corrected at the same time. Overall, our research showed promising potential for new treatment strategies based on epigenetic regulation targeting MALAT1, which will not only coordinate with the patient's immune system, but also eliminate OS in conjunction with chemotherapy.
Collapse
Affiliation(s)
- Zhi-Chang Zhang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Chun Tang
- Department of Nursing, Guangming Traditional Chinese Medicine Hospital, Pudong New Area, Shanghai 201300, China
| | - Yang Dong
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Jing Zhang
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Ting Yuan
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| | - Xiao-Lin Li
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Road, Shanghai 200233, China
| |
Collapse
|
22
|
Chosa N, Ishisaki A. Two novel mechanisms for maintenance of stemness in mesenchymal stem cells: SCRG1/BST1 axis and cell-cell adhesion through N-cadherin. JAPANESE DENTAL SCIENCE REVIEW 2017; 54:37-44. [PMID: 29629000 PMCID: PMC5884250 DOI: 10.1016/j.jdsr.2017.10.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 09/12/2017] [Accepted: 10/18/2017] [Indexed: 01/01/2023] Open
Abstract
Mesenchymal stem cells (MSCs) retain the ability to self-renew and differentiate into mesenchymal cells. Therefore, human MSCs are suitable candidates for use in regenerative medicine and cell therapies. Upon activation by tissue damage, MSCs contribute to tissue repair through a multitude of processes such as self-renewal, migration, and differentiation. However, loss of self-renewal and multi-lineage differentiation potential occurs at a high rate during cell doubling. Effective MSC therapies require the establishment of new techniques that preserve MSC multipotency after lengthy cell expansions. Here, two novel mechanisms are described for maintenance of stemness in MSCs via scrapie responsive gene 1 (SCRG1)/bone marrow stromal cell antigen-1 (BST1) ligand–receptor combination and cell–cell adhesion through N-cadherin. These two mechanisms findings provide a valuable tool for regenerative medicine and cell therapeutic methods that require the ex vivo expansion of human MSCs while maintaining native stem cell potential.
Collapse
Affiliation(s)
- Naoyuki Chosa
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028-3694, Japan
| | - Akira Ishisaki
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028-3694, Japan
| |
Collapse
|
23
|
Bulla SC, Badial PR, Silva RC, Lunsford K, Bulla C. Platelets Inhibit Migration of Canine Osteosarcoma Cells. J Comp Pathol 2016; 156:3-13. [PMID: 27890405 DOI: 10.1016/j.jcpa.2016.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 09/23/2016] [Accepted: 10/05/2016] [Indexed: 10/20/2022]
Abstract
The interaction between platelets and tumour cells is important for tumour growth and metastasis. Thrombocytopenia or antiplatelet treatment negatively impact on cancer metastasis, demonstrating potentially important roles for platelets in tumour progression. To our knowledge, there is no information regarding the role of platelets in cancer progression in dogs. This study was designed to test whether canine platelets affected the migratory behaviour of three canine osteosarcoma cell lines and to give insights of molecular mechanisms. Intact platelets, platelet lysate and platelet releasate inhibited the migration of canine osteosarcoma cell lines. Addition of blood leucocytes to the platelet samples did not alter the inhibitory effect on migration. Platelet treatment also significantly downregulated the transcriptional levels of SNAI2 and TWIST1 genes. The interaction between canine platelets or molecules released during platelet activation and these tumour cell lines inhibits their migration, which suggests that canine platelets might antagonize metastasis of canine osteosarcoma. This effect is probably due to, at least in part, downregulation of genes related to epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- S C Bulla
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, USA.
| | - P R Badial
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, USA
| | - R C Silva
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, USA
| | - K Lunsford
- Department of Clinical Sciences and Animal Health Center, College of Veterinary Medicine, Mississippi State University, MS, USA
| | - C Bulla
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, USA
| |
Collapse
|
24
|
Bai Y, Wang J, Han J, Xie XL, Ji CG, Yin J, Chen L, Wang CK, Jiang XY, Qi W, Jiang HQ. BCL2L10 inhibits growth and metastasis of hepatocellular carcinoma both in vitro and in vivo. Mol Carcinog 2016; 56:1137-1149. [PMID: 27770580 DOI: 10.1002/mc.22580] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 10/10/2016] [Accepted: 10/13/2016] [Indexed: 01/13/2023]
Affiliation(s)
- Yun Bai
- Hebei Key Laboratory of Gastroenterology; Department of Gastroenterology; Hebei Institute of Gastroenterology; The Second Hospital of Hebei Medical University; Shijiazhuang Hebei China
- Department of Gastroenterology; Hebei General Hospital; Shijiazhuang Hebei China
| | - Jia Wang
- Hebei Key Laboratory of Gastroenterology; Department of Gastroenterology; Hebei Institute of Gastroenterology; The Second Hospital of Hebei Medical University; Shijiazhuang Hebei China
- Ronghe Biotechnology Co., Ltd.; Shijiazhuang Hebei China
| | - Jing Han
- Hebei Key Laboratory of Gastroenterology; Department of Gastroenterology; Hebei Institute of Gastroenterology; The Second Hospital of Hebei Medical University; Shijiazhuang Hebei China
| | - Xiao-Li Xie
- Hebei Key Laboratory of Gastroenterology; Department of Gastroenterology; Hebei Institute of Gastroenterology; The Second Hospital of Hebei Medical University; Shijiazhuang Hebei China
| | - Cheng-Guang Ji
- Hebei Key Laboratory of Gastroenterology; Department of Gastroenterology; Hebei Institute of Gastroenterology; The Second Hospital of Hebei Medical University; Shijiazhuang Hebei China
- Department of Gastroenterology; The Third Hospital of Hebei Medical University; Shijiazhuang Hebei China
| | - Jie Yin
- Hebei Key Laboratory of Gastroenterology; Department of Gastroenterology; Hebei Institute of Gastroenterology; The Second Hospital of Hebei Medical University; Shijiazhuang Hebei China
| | - Lei Chen
- Hebei Key Laboratory of Gastroenterology; Department of Gastroenterology; Hebei Institute of Gastroenterology; The Second Hospital of Hebei Medical University; Shijiazhuang Hebei China
| | - Cun-Kai Wang
- Hebei Key Laboratory of Gastroenterology; Department of Gastroenterology; Hebei Institute of Gastroenterology; The Second Hospital of Hebei Medical University; Shijiazhuang Hebei China
| | - Xiao-Yu Jiang
- Hebei Key Laboratory of Gastroenterology; Department of Gastroenterology; Hebei Institute of Gastroenterology; The Second Hospital of Hebei Medical University; Shijiazhuang Hebei China
| | - Wei Qi
- Hebei Key Laboratory of Gastroenterology; Department of Gastroenterology; Hebei Institute of Gastroenterology; The Second Hospital of Hebei Medical University; Shijiazhuang Hebei China
| | - Hui-Qing Jiang
- Hebei Key Laboratory of Gastroenterology; Department of Gastroenterology; Hebei Institute of Gastroenterology; The Second Hospital of Hebei Medical University; Shijiazhuang Hebei China
| |
Collapse
|
25
|
Wang J, Wang B, Chen LQ, Yang J, Gong ZQ, Zhao XL, Zhang CQ, Du KL. miR-10b promotes invasion by targeting KLF4 in osteosarcoma cells. Biomed Pharmacother 2016; 84:947-953. [PMID: 27764757 DOI: 10.1016/j.biopha.2016.09.108] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 09/29/2016] [Indexed: 11/26/2022] Open
Abstract
OBJECTIVE Osteosarcoma is a common malignancy with high rate of metastasis. miR-10b has been reported to be expressed in many types of tumors abnormally and be associated with cancer carcinogenesis and progression. But the function of miR-10b in osteosarcoma is still unknown. So this study was aimed to investigate the role of miR-10b in osteosarcoma development. METHODS miR-10b expression in osteosarcoma tissues and osteosarcoma cells were detected using real time PCR. The effects of miR-10b on osteosarcoma cells proliferation, apoptosis, migration and invasion were detected using CCK-8 assay, flow cytometry, wound-healing assay and transwell assay, respectively. The relationship between miR-10b and KLF4 was evaluated using dual-luciferase assay, correlation analysis. RESULTS miR-10b was highly expressed in osteosarcoma tissues and osteosarcoma cells. Furthermore, inhibition of miR-10b in osteosarcoma cells depressed the cells proliferation, migration and invasion but promoted cells apoptosis. In addition, KLF4 was down-regulated by miR-10b and miR-10b expression was negatively related to KLF4 expression in osteosarcoma tissue, miR-10b participated in the process of osteosarcoma cells invasion by regulating KLF4 expression. CONCLUSION miR-10b is overexpressed in osteosarcoma and KLF4 is the direct target gene of miR-10b. Furthermore, miR-10b promotes osteosarcoma cells progression by downregulating KLF4 expression. These results suggest that miR-10b functions as an oncomiR and play an important role in osteosarcoma cellular processes at least partially through regulating KLF4; miR-10b may be a therapeutic target for osteosarcoma treatment.
Collapse
Affiliation(s)
- Jing Wang
- Department of Rheumatology, The First People's Hospital of Yunnan Province, Kunming 650032, Yunnan, China
| | - Bing Wang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Ling-Qiang Chen
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China.
| | - Jin Yang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Zhi-Qiang Gong
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Xue-Ling Zhao
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Chun-Qiang Zhang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| | - Kai-Li Du
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan, China
| |
Collapse
|
26
|
Ren L, Mendoza A, Zhu J, Briggs JW, Halsey C, Hong ES, Burkett SS, Morrow J, Lizardo MM, Osborne T, Li SQ, Luu HH, Meltzer P, Khanna C. Characterization of the metastatic phenotype of a panel of established osteosarcoma cells. Oncotarget 2016; 6:29469-81. [PMID: 26320182 PMCID: PMC4745740 DOI: 10.18632/oncotarget.5177] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 09/25/2015] [Indexed: 11/25/2022] Open
Abstract
Osteosarcoma (OS) is the most common bone tumor in pediatric patients. Metastasis is a major cause of mortality and morbidity. The rarity of this disease coupled with the challenges of drug development for metastatic cancers have slowed the delivery of improvements in long-term outcomes for these patients. In this study, we collected 18 OS cell lines, confirmed their expression of bone markers and complex karyotypes, and characterized their in vivo tumorgenicity and metastatic potential. Since prior reports included conflicting descriptions of the metastatic and in vivo phenotypes of these models, there was a need for a comparative assessment of metastatic phenotypes using identical procedures in the hands of a single investigative group. We expect that this single characterization will accelerate the study of this metastatic cancer. Using these models we evaluated the expression of six previously reported metastasis-related OS genes. Ezrin was the only gene consistently differentially expressed in all the pairs of high/low metatstatic OS cells. We then used a subtractive gene expression approach of the high and low human metastatic cells to identify novel genes that may be involved in OS metastasis. PHLDA1 (pleckstrin homology-like domain, family A) was identified as one of the genes more highly expressed in the high metastatic compared to low metastatic cells. Knocking down PHLDA1 with siRNA or shRNA resulted in down regulation of the activities of MAPKs (ERK1/2), c-Jun N-terminal kinases (JNK), and p38 mitogen-activated protein kinases (MAPKs). Reducing the expression of PHLDA1 also delayed OS metastasis progression in mouse xenograft models.
Collapse
Affiliation(s)
- Ling Ren
- Molecular Oncology Section - Metastasis Biology Group, Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Arnulfo Mendoza
- Molecular Oncology Section - Metastasis Biology Group, Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Jack Zhu
- Genetic Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Joseph W Briggs
- Molecular Oncology Section - Metastasis Biology Group, Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Charles Halsey
- Molecular Pathology Unit, Laboratory of Cancer Biology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Ellen S Hong
- Molecular Oncology Section - Metastasis Biology Group, Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Sandra S Burkett
- Comparative Molecular Cytogenetics Core Facility, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - James Morrow
- School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Michael M Lizardo
- Molecular Oncology Section - Metastasis Biology Group, Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Tanasa Osborne
- National Institute of Environmental Health, Research Triangle Park, North Carolina, USA
| | - Samuel Q Li
- School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Hue H Luu
- Department of Orthopedic Surgery & Rehabilitation Medicine, University of Chicago, Medicine & Biological Sciences, Chicago, USA
| | - Paul Meltzer
- Genetic Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Chand Khanna
- Molecular Oncology Section - Metastasis Biology Group, Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
27
|
Weekes D, Kashima TG, Zandueta C, Perurena N, Thomas DP, Sunters A, Vuillier C, Bozec A, El-Emir E, Miletich I, Patiño-Garcia A, Lecanda F, Grigoriadis AE. Regulation of osteosarcoma cell lung metastasis by the c-Fos/AP-1 target FGFR1. Oncogene 2016; 35:2852-61. [PMID: 26387545 PMCID: PMC4688957 DOI: 10.1038/onc.2015.344] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 07/05/2015] [Accepted: 07/24/2015] [Indexed: 12/13/2022]
Abstract
Osteosarcoma is the most common primary malignancy of the skeleton and is prevalent in children and adolescents. Survival rates are poor and have remained stagnant owing to chemoresistance and the high propensity to form lung metastases. In this study, we used in vivo transgenic models of c-fos oncogene-induced osteosarcoma and chondrosarcoma in addition to c-Fos-inducible systems in vitro to investigate downstream signalling pathways that regulate osteosarcoma growth and metastasis. Fgfr1 (fibroblast growth factor receptor 1) was identified as a novel c-Fos/activator protein-1(AP-1)-regulated gene. Induction of c-Fos in vitro in osteoblasts and chondroblasts caused an increase in Fgfr1 RNA and FGFR1 protein expression levels that resulted in increased and sustained activation of mitogen-activated protein kinases (MAPKs), morphological transformation and increased anchorage-independent growth in response to FGF2 ligand treatment. High levels of FGFR1 protein and activated pFRS2α signalling were observed in murine and human osteosarcomas. Pharmacological inhibition of FGFR1 signalling blocked MAPK activation and colony growth of osteosarcoma cells in vitro. Orthotopic injection in vivo of FGFR1-silenced osteosarcoma cells caused a marked twofold to fivefold decrease in spontaneous lung metastases. Similarly, inhibition of FGFR signalling in vivo with the small-molecule inhibitor AZD4547 markedly reduced the number and size of metastatic nodules. Thus deregulated FGFR signalling has an important role in osteoblast transformation and osteosarcoma formation and regulates the development of lung metastases. Our findings support the development of anti-FGFR inhibitors as potential antimetastatic therapy.
Collapse
Affiliation(s)
- Daniel Weekes
- Department of Craniofacial Development and Stem Cell Biology, King’s College London, UK
| | - Takeshi G Kashima
- Department of Craniofacial Development and Stem Cell Biology, King’s College London, UK
| | - Carolina Zandueta
- Division of Oncology, Adhesion and Metastasis Laboratory, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Naiara Perurena
- Division of Oncology, Adhesion and Metastasis Laboratory, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - David P Thomas
- Department of Craniofacial Development and Stem Cell Biology, King’s College London, UK
| | - Andrew Sunters
- Department of Craniofacial Development and Stem Cell Biology, King’s College London, UK
| | - Céline Vuillier
- Department of Craniofacial Development and Stem Cell Biology, King’s College London, UK
| | - Aline Bozec
- Department of Rheumatology and Immunology, Universitätsklinikum Erlangen, Germany
| | - Ethaar El-Emir
- Department of Craniofacial Development and Stem Cell Biology, King’s College London, UK
| | - Isabelle Miletich
- Department of Craniofacial Development and Stem Cell Biology, King’s College London, UK
| | - Ana Patiño-Garcia
- Laboratory of Pediatrics, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Fernando Lecanda
- Division of Oncology, Adhesion and Metastasis Laboratory, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | | |
Collapse
|
28
|
Mitogen-activated protein kinase 7 promotes cell proliferation, migration and invasion in SOSP-M human osteosarcoma cell line. TUMORI JOURNAL 2015; 103:483-488. [PMID: 26350187 DOI: 10.5301/tj.5000399] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2015] [Indexed: 01/20/2023]
Abstract
PURPOSE Osteosarcoma (OS) is the most common primary bone tumor and has low cure rates. Our study aimed to evaluate the roles of mitogen-activated protein kinase 7 (MAPK7) in cell proliferation, migration and invasion using the SOSP-M human OS cell line as an in vitro model. METHODS SOSP-M cells were transfected with PCDNA3.1-MAPK7 and siRNA-MAPK7 plasmids using Lipofectamine 2000. Quantitative real-time polymerase chain reaction (RT-PCR) was performed to determine the relative expression level of MAPK7 and Western blot analysis was carried out to determine the expression level of ERK5 protein. Then MTT, scratch wound healing and Matrigel transwell assays were used to investigate the roles of MAPK7 expression in the proliferation, migration and invasion, respectively, of SOSP-M cells in vitro. RESULTS RT-PCR analysis showed that the expression level of MAPK7 increased significantly after transfection with PCDNA3.1-MAPK7 plasmid compared with the blank group, while it decreased significantly after transfection with siRNA-MAPK7 plasmid. Similar results for ERK5 expression were obtained by Western blot analysis. In addition, the cell proliferation rate, cell migration rate and invasive cell number in the PCDNA3.1-MAPK7 transfection group increased significantly compared with the blank group, while they decreased significantly in the siRNA-MAPK7 transfection group. CONCLUSIONS Our results indicate that overexpression of MAPK7 in human OS cells could promote cell proliferation, migration and invasion, whereas knockdown of MAPK7 expression had the opposite effect. All the results suggest that MAPK7 may serve as a potent target for drug development.
Collapse
|
29
|
Qu L, Liu B. Cyclooxygeanse-2 promotes metastasis in osteosarcoma. Cancer Cell Int 2015; 15:69. [PMID: 26180515 PMCID: PMC4502918 DOI: 10.1186/s12935-015-0220-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 06/23/2015] [Indexed: 12/17/2022] Open
Abstract
Cyclooxygenase-2 (COX-2), an inducible form of the enzyme that catalyzes the first step in the synthesis of prostanoids, is associated with carcinogenesis, which is suspected to promote angiogenesis and tissue invasion of tumors and resistance to apoptosis. COX-2 is also involved in metastasis and poor prognosis of cancer. Osteosarcoma with COX-2 positivity is from 67 to 92 %. COX-2-positive rate in metastatic lesions was greater than that of biopsy and/or resected samples of the primary site in osteosarcoma. And, what role does COX-2 play in osteosarcoma metastasis? Genetic studies support a cause-effect connection between COX-2 and tumorigenesis. COX-2 expression had a poor prognosis with regard to metastasis, and patients with increased COX-2 expression in lung metastases died of the disease. COX-2 expression has also been established as a marker in human osteosarcoma, and COX-2 inhibition has been suggested as a possible way of improving therapeutic outcome. In addition, COX-inhibitors inhibit the tumor initiation, matrix metalloproteinases (MMPs), cell differentiation and T cell proliferation and suppression of the antitumor activity of natural killer cells and macrophages, angiogenic mechanism. Therefore, we can exert the COX-inhibitors to potentialize the effects of chemotherapeutic agents, and reverse the metastasis in osteosarcoma to facilitate the patient who may benefit from addition of COX-inhibitors to standard cytotoxic therapy.
Collapse
Affiliation(s)
- Liyan Qu
- Clinical Laboratory Centre, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, 310009 Zhejiang People's Republic of China ; Clinical Laboratory Centre, Binjiang Hospital of Hangzhou, Hangzhou, Zhejiang China
| | - Bing Liu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, 310009 Zhejiang People's Republic of China
| |
Collapse
|
30
|
Wang B, Su Y, Yang Q, Lv D, Zhang W, Tang K, Wang H, Zhang R, Liu Y. Overexpression of Long Non-Coding RNA HOTAIR Promotes Tumor Growth and Metastasis in Human Osteosarcoma. Mol Cells 2015; 38:432-40. [PMID: 25728753 PMCID: PMC4443285 DOI: 10.14348/molcells.2015.2327] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 01/05/2015] [Accepted: 01/26/2015] [Indexed: 01/13/2023] Open
Abstract
Human osteosarcoma usually presented a high tendency to metastatic spread and caused poor outcomes, however, the underlying mechanism was still largely unknown. In the present study, using a series of in vitro experiments and an animal model, we investigated the roles of HOX antisense intergenic RNA (HOTAIR) during the proliferation and invasion of osteosarcoma. According with our results, HOTAIR was commonly overexpressed in osteosarcoma, which significantly correlated with advanced tumor stage, highly histological grade and poor prognosis. In vitro and in vivo experiments demonstrated that knockdown of HOTAIR could notably suppress cellular proliferation, inhibit invasion and decrease the secretion of MMP2 and MMP9 in osteosarcoma. Collectively, our results suggested that HOTAIR might be a potent therapeutic target for osteosarcoma.
Collapse
Affiliation(s)
- Bo Wang
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| | - Yun Su
- Department of Orthopeadics, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning Province, 116001,
China
| | - Qun Yang
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| | - Decheng Lv
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| | - Weiguo Zhang
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| | - Kai Tang
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| | - Hong Wang
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| | - Rui Zhang
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| | - Yang Liu
- Department of Spine Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116011,
China
| |
Collapse
|
31
|
Wang J, Dong L, Xu L, Chu ES, Chen Y, Shen J, Li X, Wong CC, Sung JJ, Yu J. B cell CLL/lymphoma 6 member B inhibits hepatocellular carcinoma metastases in vitro and in mice. Cancer Lett 2014; 355:192-200. [DOI: 10.1016/j.canlet.2014.08.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/16/2014] [Accepted: 08/18/2014] [Indexed: 02/07/2023]
|
32
|
Alizadeh AM, Shiri S, Farsinejad S. Metastasis review: from bench to bedside. Tumour Biol 2014; 35:8483-523. [PMID: 25104089 DOI: 10.1007/s13277-014-2421-z] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 07/29/2014] [Indexed: 12/19/2022] Open
Abstract
Cancer is the final result of uninhibited cell growth that involves an enormous group of associated diseases. One major aspect of cancer is when cells attack adjacent components of the body and spread to other organs, named metastasis, which is the major cause of cancer-related mortality. In developing this process, metastatic cells must successfully negotiate a series of complex steps, including dissociation, invasion, intravasation, extravasation, and dormancy regulated by various signaling pathways. In this review, we will focus on the recent studies and collect a comprehensive encyclopedia in molecular basis of metastasis, and then we will discuss some new potential therapeutics which target the metastasis pathways. Understanding the new aspects on molecular mechanisms and signaling pathways controlling tumor cell metastasis is critical for the development of therapeutic strategies for cancer patients that would be valuable for researchers in both fields of molecular and clinical oncology.
Collapse
Affiliation(s)
- Ali Mohammad Alizadeh
- Cancer Research Center, Tehran University of Medical Sciences, Tehran, 1419733141, Iran,
| | | | | |
Collapse
|
33
|
Marino N, Collins JW, Shen C, Caplen NJ, Merchant AS, Gökmen-Polar Y, Goswami CP, Hoshino T, Qian Y, Sledge GW, Steeg PS. Identification and validation of genes with expression patterns inverse to multiple metastasis suppressor genes in breast cancer cell lines. Clin Exp Metastasis 2014; 31:771-86. [PMID: 25086928 DOI: 10.1007/s10585-014-9667-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 07/04/2014] [Indexed: 12/30/2022]
Abstract
Metastasis suppressor genes (MSGs) have contributed to an understanding of regulatory pathways unique to the lethal metastatic process. When re-expressed in experimental models, MSGs block cancer spread to, and colonization of distant sites without affecting primary tumor formation. Genes have been identified with expression patterns inverse to a single MSG, and found to encode functional, druggable signaling pathways. We now hypothesize that common signaling pathways mediate the effects of multiple MSGs. By gene expression profiling of human MCF7 breast carcinoma cells expressing a scrambled siRNA, or siRNAs to each of 19 validated MSGs (NME1, BRMS1, CD82, CDH1, CDH2, CDH11, CASP8, MAP2K4, MAP2K6, MAP2K7, MAPK14, GSN, ARHGDIB, AKAP12, DRG1, CD44, PEBP1, RRM1, KISS1), we identified genes whose expression was significantly opposite to at least five MSGs. Five genes were selected for further analysis: PDE5A, UGT1A, IL11RA, DNM3 and OAS1. After stable downregulation of each candidate gene in the aggressive human breast cancer cell line MDA-MB-231T, in vitro motility was significantly inhibited. Two stable clones downregulating PDE5A (phosphodiesterase 5A), an enzyme involved in the regulation of cGMP-specific signaling, exhibited no difference in cell proliferation, but reduced motility by 47 and 66 % compared to the empty vector-expressing cells (p = 0.01 and p = 0.005). In an experimental metastasis assay, two shPDE5A-MDA-MB-231T clones produced 47-62 % fewer lung metastases than shRNA-scramble expressing cells (p = 0.045 and p = 0.009 respectively). This study demonstrates that previously unrecognized genes are inversely related to the expression of multiple MSGs, contribute to aspects of metastasis, and may stand as novel therapeutic targets.
Collapse
Affiliation(s)
- Natascia Marino
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Building 37/Room 1126, 37 Convent Drive, Bethesda, MD, 20892, USA,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Mendes R, Martins G, Cristovão A, Saúde L. N-Cadherin Locks Left-Right Asymmetry by Ending the Leftward Movement of Hensen’s Node Cells. Dev Cell 2014; 30:353-60. [DOI: 10.1016/j.devcel.2014.06.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Revised: 05/17/2014] [Accepted: 06/12/2014] [Indexed: 01/29/2023]
|
35
|
Kidani T, Nakamura A, Kamei S, Norimatsu Y, Miura H, Masuno H. Overexpression of cytoplasmic β-catenin inhibits the metastasis of the murine osteosarcoma cell line LM8. Cancer Cell Int 2014; 14:31. [PMID: 24690154 PMCID: PMC3977682 DOI: 10.1186/1475-2867-14-31] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 03/26/2014] [Indexed: 02/07/2023] Open
Abstract
Background Previously, we found that treatment of LM8 murine osteosarcoma cells with genistein, an isoflavone found in soy, increased the cellular level of β-catenin and decreased its invasive and motile potential. The purpose of this study is to investigate whether the expression of β-catenin in LM8 cells is associated with metastatic potential in nude mice. To this end, we used untreated and genistein-treated LM8 cells. Methods LM8 cells were treated for 3 days with or without 50 μM genistein and harvested by trypsinization. Untreated (the control group) and genistein-treated (the genistein group) cells were subcutaneously inoculated into the backs of male nude mice. After 25 days of inoculation, the tumors, lungs, and livers were excised, fixed in 10% formalin, and embedded in paraffin. The sections of formalin-fixed, paraffin-embedded lungs and livers were stained with hematoxylin-eosin (H&E) to confirm the absence or presence of metastatic tumors. The expression of β-catenin within the primary tumor was immunohistochemically examined. Results All mice in the control group (n = 8) exhibited large primary tumors, while in the genistein group (n = 8), one mouse showed no tumor formation and the remaining seven mice exhibited smaller primary tumors compared with the control group. The tumor mass of the genistein group was 23% of that of the control group. In the control group, multiple metastatic tumors were found in the lung and/or liver and the metastatic incidence was 100% in the lung and 87.5% in the liver. Six of seven tumor-bearing mice in the genistein group developed no metastatic tumors in the lung or liver, and this group was termed the genistein/metastasis(-) subgroup. Positive β-catenin immunostaining was observed in the cytoplasm of tumor cells, and the β-catenin-labeling index was higher in the genistein/metastasis(-) subgroup than in the control group. The intensity of cytoplasmic β-catenin immunostaining was stronger in the genistein/metastasis(-) subgroup compared with the control group, and the β-catenin-labeling score was 1.9-times higher in the former subgroup than in the latter group. Conclusions Overexpression of cytoplasmic β-catenin in LM8 cells causes inhibition of the growth of primary tumors and loss of the metastatic potential to the lung and liver.
Collapse
Affiliation(s)
| | | | | | | | | | - Hiroshi Masuno
- Department of Medical Technology, Faculty of Health Sciences, Ehime Prefectural University of Health Sciences, Takooda, Tobe-cho, Iyo-gun, Ehime 791-2101, Japan.
| |
Collapse
|
36
|
Mu X, Sultankulov B, Agarwal R, Mahjoub A, Schott T, Greco N, Huard J, Weiss K. Chick embryo extract demethylates tumor suppressor genes in osteosarcoma cells. Clin Orthop Relat Res 2014; 472:865-73. [PMID: 23761177 PMCID: PMC3916611 DOI: 10.1007/s11999-013-3104-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Epigenetics is the study of changes in gene expression or cellular phenotype caused by mechanisms other than changes in the underlying DNA sequence. It is widely accepted that cancer has genetic and epigenetic origins. The idea of epigenetic reprogramming of cancer cells by an embryonic microenvironment possesses potential interest from the prospect of both basic science and potential therapeutic strategies. Chick embryo extract (CEE) has been used for the successful expansion of many specific stem cells and has demonstrated the ability to facilitate DNA demethylation. QUESTIONS/PURPOSES The current study was conducted to compare the status of DNA methylation in highly metastatic and less metastatic osteosarcoma cells and to investigate whether CEE may affect the epigenetic regulation of tumor suppressor genes and thus change the metastatic phenotypes of highly metastatic osteosarcoma cells. METHODS K7M2 murine OS cells were treated with CEE to determine its potential effect on DNA methylation, cell apoptosis, and invasion capacity. RESULTS Our current results suggest that the methylation status of tumor suppressor genes (p16, p53, and E-cadherin) is significantly greater in highly metastatic mouse ostoesarcoma K7M2 cells in comparison with less metastatic mouse osteosarcoma K12 cells. CEE treatment of K7M2 cells caused demethylation of p16, p53, and E-cadherin genes, upregulated their expression, and resulted in the reversion of metastatic phenotypes in highly metastatic osteosarcoma cells. CONCLUSIONS CEE may promote the reversion of metastatic phenotypes of osteosarcoma cells and can be a helpful tool to study osteosarcoma tumor reversion by epigenetic reprogramming. CLINICAL RELEVANCE Demethylation of tumor suppressor genes in osteosarcoma may represent a novel strategy to diminish the metastatic potential of this neoplasm. Further studies, both in vitro and in vivo, are warranted to evaluate the clinical feasibility of this approach as an adjuvant to current therapy.
Collapse
Affiliation(s)
- Xiaodong Mu
- Cancer Stem Cell Laboratory, Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Bridgeside Point 2, Suite 206, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Bolat Sultankulov
- Department of Biophysics, Nazarbayev University Research and Innovation System, Astana, Kazakhstan
| | - Riddhima Agarwal
- Cancer Stem Cell Laboratory, Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Bridgeside Point 2, Suite 206, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Adel Mahjoub
- Cancer Stem Cell Laboratory, Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Bridgeside Point 2, Suite 206, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Trevor Schott
- Cancer Stem Cell Laboratory, Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Bridgeside Point 2, Suite 206, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Nicholas Greco
- Cancer Stem Cell Laboratory, Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Bridgeside Point 2, Suite 206, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Johnny Huard
- Cancer Stem Cell Laboratory, Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Bridgeside Point 2, Suite 206, 450 Technology Drive, Pittsburgh, PA 15219 USA
| | - Kurt Weiss
- Cancer Stem Cell Laboratory, Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Bridgeside Point 2, Suite 206, 450 Technology Drive, Pittsburgh, PA 15219 USA
| |
Collapse
|
37
|
Abstract
Loss of cadherin 1 (CDH1; also known as epithelial cadherin (E-cadherin)) is used for the diagnosis and prognosis of epithelial cancers. However, it should not be ignored that the superfamily of transmembrane cadherin proteins encompasses more than 100 members in humans, including other classical cadherins, numerous protocadherins and cadherin-related proteins. Elucidation of their roles in suppression versus initiation or progression of various tumour types is a young but fascinating field of molecular cancer research. These cadherins are very diverse in both structure and function, and their mutual interactions seem to influence biological responses in complex and versatile ways.
Collapse
Affiliation(s)
- Frans van Roy
- Department of Biomedical Molecular Biology, Ghent University, B-9052 Ghent, Belgium.The Inflammation Research Center, VIB, B-9052 Ghent, Belgium
| |
Collapse
|
38
|
Dou C, Yan Y, Dong S. Role of cadherin-11 in synovial joint formation and rheumatoid arthritis pathology. Mod Rheumatol 2014. [DOI: 10.3109/s10165-012-0806-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
39
|
Aomatsu E, Chosa N, Nishihira S, Sugiyama Y, Miura H, Ishisaki A. Cell-cell adhesion through N-cadherin enhances VCAM-1 expression via PDGFRβ in a ligand-independent manner in mesenchymal stem cells. Int J Mol Med 2013; 33:565-72. [PMID: 24378362 DOI: 10.3892/ijmm.2013.1607] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 12/17/2013] [Indexed: 11/06/2022] Open
Abstract
Cell-cell adhesions induce various intracellular signals through hierarchical and synergistic molecular interactions. Recently, we demonstrated that a high cell density induces the expression of vascular cell adhesion molecule-1 (VCAM-1) through the nuclear factor-κB (NF-κB) pathway in human bone marrow-derived mesenchymal stem cells (MSCs). However, the specific molecules that activated the NF-κB pathway were not determined. In the present study, in experiments with receptor tyrosine kinase inhibitors, VCAM-1 expression was completely suppressed by platelet-derived growth factor (PDGF) receptor (PDGFR) inhibitors. In addition, VCAM-1 expression was significantly suppressed by knockdown with PDGFRβ siRNA, but not with PDGFRα siRNA. However, VCAM-1 expression did not increase following treatment with PDGF. The overexpression of N-cadherin, a structural molecule in adherence junctions in MSCs, promoted VCAM-1 expression and induced the marked phosphorylation of the intracellular signaling factor, Src. In addition, VCAM-1 expression and Src phosphorylation were reduced by the overexpression of a dominant negative mutant of N-cadherin. These results suggest that cell-cell adhesion, through N-cadherin, enhances the expression of VCAM-1 via PDGFRβ and the activation of Src in a ligand-independent manner in MSCs.
Collapse
Affiliation(s)
- Emiko Aomatsu
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028-3694, Japan
| | - Naoyuki Chosa
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028-3694, Japan
| | - Soko Nishihira
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028-3694, Japan
| | - Yoshiki Sugiyama
- Division of Oral Surgery, Department of Oral and Maxillofacial Surgery, Iwate Medical University School of Dentistry, Morioka, Iwate 020-8505, Japan
| | - Hiroyuki Miura
- Division of Orthodontics, Department of Developmental Oral Health Science, Iwate Medical University School of Dentistry, Morioka, Iwate 020-8505, Japan
| | - Akira Ishisaki
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Yahaba, Iwate 028-3694, Japan
| |
Collapse
|
40
|
Reduced Latency in the Metastatic Niche Contributes to the More Aggressive Phenotype of LM8 Compared to Dunn Osteosarcoma Cells. Sarcoma 2013; 2013:404962. [PMID: 24369449 PMCID: PMC3867932 DOI: 10.1155/2013/404962] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 10/13/2013] [Accepted: 10/13/2013] [Indexed: 01/04/2023] Open
Abstract
Metastasis is the major cause of death of osteosarcoma patients and its diagnosis remains difficult. In preclinical studies, however, forced expression of reporter genes in osteosarcoma cells has remarkably improved the detection of micrometastases and, consequently, the quality of the studies. We recently showed that Dunn cells equipped with a lacZ reporter gene disseminated from subcutaneous primary tumors as frequently as their highly metastatic subline LM8, but only LM8 cells grew to macrometastases. In the present time-course study, tail-vein-injected Dunn and LM8 cells settled within 24 h at the same frequency in the lung, liver, and kidney of mice. Furthermore, Dunn cells also grew to macrometastases, but, compared to LM8, with a delay of two weeks in lung and one week in liver and kidney tissue, consistent with prolonged survival of the mice. Dunn- and LM8-cell-derived ovary and spine metastases occurred less frequently. In vitro, Dunn cells showed less invasiveness and stronger contact inhibition and intercellular adhesion than LM8 cells and several cancer- and dormancy-related genes were differentially expressed. In conclusion, Dunn cells, compared to LM8, have a similar capability but a longer latency to form macrometastases and provide an interesting new experimental system to study tumor cell dormancy.
Collapse
|
41
|
Ruedel A, Stark K, Kaufmann S, Bauer R, Reinders J, Rovensky J, BlažiČková S, Oefner PJ, Bosserhoff AK. N‐cadherin promoter polymorphisms and risk of osteoarthritis. FASEB J 2013; 28:683-91. [DOI: 10.1096/fj.13-238295] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Anke Ruedel
- Institute of PathologyUniversity of RegensburgRegensburgGermany
| | - Klaus Stark
- Department of Internal Medicine IIUniversity of RegensburgRegensburgGermany
- Department of Genetic EpidemiologyUniversity of RegensburgRegensburgGermany
| | - Simone Kaufmann
- Institute of PathologyUniversity of RegensburgRegensburgGermany
| | - Richard Bauer
- Department of Oral and Maxillofacial SurgeryUniversity Hospital RegensburgRegensburgGermany
| | - Joerg Reinders
- Institute of Functional GenomicsUniversity of RegensburgRegensburgGermany
| | - Jozef Rovensky
- National Institute of Rheumatic DiseasesPiestanySlovakia
| | | | - Peter J. Oefner
- Institute of Functional GenomicsUniversity of RegensburgRegensburgGermany
| | | |
Collapse
|
42
|
FUKUDA KOJI, MIURA YASUSHI, MAEDA TOSHIHISA, TAKAHASHI MASAYASU, HAYASHI SHINYA, KUROSAKA MASAHIRO. Decoy receptor 3 regulates the expression of various genes in rheumatoid arthritis synovial fibroblasts. Int J Mol Med 2013; 32:910-6. [DOI: 10.3892/ijmm.2013.1461] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 07/18/2013] [Indexed: 11/06/2022] Open
|
43
|
Deng Z, Niu G, Cai L, Wei R, Zhao X. The prognostic significance of CD44V6, CDH11, and β-catenin expression in patients with osteosarcoma. BIOMED RESEARCH INTERNATIONAL 2013; 2013:496193. [PMID: 23971040 PMCID: PMC3732612 DOI: 10.1155/2013/496193] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Revised: 07/01/2013] [Accepted: 07/01/2013] [Indexed: 11/28/2022]
Abstract
This study aimed to examine the expression of and the relationship between CD44V6, CDH11, and β-catenin. The expression of these cell adhesion molecules was detected in 90 osteosarcoma and 20 osteochondroma specimens using immunohistochemistry. Associations between these parameters and clinicopathological data were also examined. The expression rates of CD44V6, CDH11, and β-catenin were 25.0% (5/20), 70.0% (14/20), and 20.0% (4/20) in osteochondroma specimens, respectively. Compared to osteochondromas, the proportions of expression of CD44V6 and β-catenin in osteosarcoma specimens increased to 65.6% (59/90) and 60.0% (54/90), respectively. However, the expression rate of CDH11 in osteosarcomas was reduced to 40.0% (36/90). The expression of these markers was significantly associated with metastasis and overall survival (P < 0.05). Survival analysis revealed that patients with increased expression of CD44V6 and β-catenin as well as decreased expression of CDH11 were correlated with a shorter survival time. Multivariate analysis indicated that clinical stage, metastasis status, and the expression of CD44V6, CDH11, and β-catenin were found to be associated with overall survival. Further, the expression of β -catenin and that of CD44V6 were positively correlated with each other. Thus, our results indicated abnormal expression of CD44V6, CDH11, and β-catenin in osteosarcomas and osteochondromas, which may provide important indicators for further research.
Collapse
Affiliation(s)
- Zhouming Deng
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhan, Hubei Province 430071, China
| | - Guangfeng Niu
- Department of Spine, Shandong Provincial Hospital, Jinan 250021, China
| | - Lin Cai
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhan, Hubei Province 430071, China
| | - Renxiong Wei
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhan, Hubei Province 430071, China
| | - Xiaolei Zhao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, No. 169 Donghu Road, Wuhan, Hubei Province 430071, China
| |
Collapse
|
44
|
Sharili AS, Allen S, Smith K, Price J, McGonnell IM. Snail2 promotes osteosarcoma cell motility through remodelling of the actin cytoskeleton and regulates tumor development. Cancer Lett 2013; 333:170-9. [PMID: 23352643 PMCID: PMC3644682 DOI: 10.1016/j.canlet.2013.01.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 01/13/2013] [Accepted: 01/15/2013] [Indexed: 12/01/2022]
Abstract
The function of Snail2 in mesenchymal tumors is, to date unknown. Using knockdown and overexpression studies, we show that Snail2 regulates migration and invasion of osteosarcoma cells. Knockdown resulted in significantly decreased motility, remodelling of the actin cytoskeleton, and loss of cellular protrusions. Over-expression increased motility, formation of actin-rich cellular protrusions, and altered expression of some non-canonical Wnt pathway components whilst decreasing expression of the adhesion molecule OB-cadherin. Unexpectedly, knockdown also resulted in significantly smaller tumors in an in vivo CAM assay. Therefore Snail2 may be a potential therapeutic target for clinical intervention of osteosarcoma.
Collapse
Affiliation(s)
- Amir-Shaya Sharili
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
- Barts and the London School of Medicine and Dentistry, Blizard Institute, 4 Newark Street, London E1 2AR, UK
| | - Steve Allen
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Ken Smith
- Department of Pathology and Pathogen Biology, Royal Veterinary College, London, UK
| | - Joanna Price
- Department of Clinical Veterinary Sciences, University of Bristol, Langford House, Langford, North Somerset, UK
| | - Imelda M. McGonnell
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| |
Collapse
|
45
|
Thériault BL, Dimaras H, Gallie BL, Corson TW. The genomic landscape of retinoblastoma: a review. Clin Exp Ophthalmol 2013; 42:33-52. [PMID: 24433356 DOI: 10.1111/ceo.12132] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 04/07/2013] [Indexed: 12/13/2022]
Abstract
Retinoblastoma is a paediatric ocular tumour that continues to reveal much about the genetic basis of cancer development. Study of genomic aberrations in retinoblastoma tumours has exposed important mechanisms of cancer development and identified oncogenes and tumour suppressors that offer potential points of therapeutic intervention. The recent development of next-generation genomic technologies has allowed further refinement of the genomic landscape of retinoblastoma at high resolution. In a relatively short period of time, a wealth of genetic and epigenetic data has emerged on a small number of tumour samples. These data highlight the inherent molecular complexity of this cancer despite the fact that most retinoblastomas are initiated by the inactivation of a single tumour suppressor gene. This review outlines the current understanding of the genomic, genetic and epigenetic changes in retinoblastoma, highlighting recent genome-wide analyses that have identified exciting candidate genes worthy of further validation as potential prognostic and therapeutic targets.
Collapse
Affiliation(s)
- Brigitte L Thériault
- Campbell Family Cancer Research Institute, Ontario Cancer Institute, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
46
|
Zucchini C, Manara MC, Pinca RS, De Sanctis P, Guerzoni C, Sciandra M, Lollini PL, Cenacchi G, Picci P, Valvassori L, Scotlandi K. CD99 suppresses osteosarcoma cell migration through inhibition of ROCK2 activity. Oncogene 2013; 33:1912-21. [PMID: 23644663 DOI: 10.1038/onc.2013.152] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 02/25/2013] [Accepted: 03/08/2013] [Indexed: 12/21/2022]
Abstract
CD99, a transmembrane protein encoded by MIC2 gene is involved in multiple cellular events including cell adhesion and migration, apoptosis, cell differentiation and regulation of protein trafficking either in physiological or pathological conditions. In osteosarcoma, CD99 is expressed at low levels and functions as a tumour suppressor. The full-length protein (CD99wt) and the short-form harbouring a deletion in the intracytoplasmic domain (CD99sh) have been associated with distinct functional outcomes with respect to tumour malignancy. In this study, we especially evaluated modulation of cell-cell contacts, reorganisation of the actin cytoskeleton and modulation of signalling pathways by comparing osteosarcoma cells characterised by different metastasis capabilities and CD99 expression, to identify molecular mechanisms responsible for metastasis. Our data indicate that forced expression of CD99wt induces recruitment of N-cadherin and β-catenin to adherens junctions. In addition, transfection of CD99wt inhibits the expression of several molecules crucial to the remodelling of the actin cytoskeleton, such as ACTR2, ARPC1A, Rho-associated, coiled-coil containing protein kinase 2 (ROCK2) as well as ezrin, an ezrin/radixin/moesin family member that has been clearly associated with tumour progression and metastatic spread in osteosarcoma. Functional studies point to ROCK2 as a crucial intracellular mediator regulating osteosarcoma migration. By maintaining c-Src in an inactive conformation, CD99wt inhibits ROCK2 signalling and this leads to ezrin decrease at cell membrane while N-cadherin and β-catenin translocate to the plasma membrane and function as main molecular bridges for actin cytoskeleton. Taken together, we propose that the re-expression of CD99wt, which is generally present in osteoblasts but lost in osteosarcoma, through inhibition of c-Src and ROCK2 activity, manages to increase contact strength and reactivate stop-migration signals that counteract the otherwise dominant promigratory action of ezrin in osteosarcoma cells.
Collapse
Affiliation(s)
- C Zucchini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - M C Manara
- 1] CRS Development of Biomolecular Therapies, Bologna, Italy [2] Experimental Oncology Laboratory, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - R S Pinca
- 1] CRS Development of Biomolecular Therapies, Bologna, Italy [2] Experimental Oncology Laboratory, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - P De Sanctis
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - C Guerzoni
- 1] Experimental Oncology Laboratory, Istituto Ortopedico Rizzoli, Bologna, Italy [2] PROMETEO Laboratory, STB, RIT Department, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - M Sciandra
- CRS Development of Biomolecular Therapies, Bologna, Italy
| | - P-L Lollini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - G Cenacchi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - P Picci
- Experimental Oncology Laboratory, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - L Valvassori
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - K Scotlandi
- 1] CRS Development of Biomolecular Therapies, Bologna, Italy [2] Experimental Oncology Laboratory, Istituto Ortopedico Rizzoli, Bologna, Italy [3] PROMETEO Laboratory, STB, RIT Department, Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
47
|
Protein kinase C epsilon and genetic networks in osteosarcoma metastasis. Cancers (Basel) 2013; 5:372-403. [PMID: 24216982 PMCID: PMC3730329 DOI: 10.3390/cancers5020372] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Revised: 03/22/2013] [Accepted: 03/26/2013] [Indexed: 12/20/2022] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant tumor of the bone, and pulmonary metastasis is the most frequent cause of OS mortality. The aim of this study was to discover and characterize genetic networks differentially expressed in metastatic OS. Expression profiling of OS tumors, and subsequent supervised network analysis, was performed to discover genetic networks differentially activated or organized in metastatic OS compared to localized OS. Broad trends among the profiles of metastatic tumors include aberrant activity of intracellular organization and translation networks, as well as disorganization of metabolic networks. The differentially activated PRKCε-RASGRP3-GNB2 network, which interacts with the disorganized DLG2 hub, was also found to be differentially expressed among OS cell lines with differing metastatic capacity in xenograft models. PRKCε transcript was more abundant in some metastatic OS tumors; however the difference was not significant overall. In functional studies, PRKCε was not found to be involved in migration of M132 OS cells, but its protein expression was induced in M112 OS cells following IGF-1 stimulation.
Collapse
|
48
|
PosthumaDeBoer J, van Royen B, Helder M. Mechanisms of therapy resistance in osteosarcoma: a review. ACTA ACUST UNITED AC 2013. [DOI: 10.7243/2052-6199-1-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
49
|
Role of cadherin-11 in synovial joint formation and rheumatoid arthritis pathology. Mod Rheumatol 2012; 23:1037-44. [PMID: 23239231 DOI: 10.1007/s10165-012-0806-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Accepted: 11/26/2012] [Indexed: 10/27/2022]
Abstract
Cadherin-11 is a classic cadherin adhesion molecule that mediates homophilic cell-to-cell adhesion. Cadherin-11 is involved in the function of embryonic development, tissue morphogenesis, tumor invasion and metastasis, and signal transduction. This review summarizes the function of cadherin-11 in synovial joint formation and rheumatoid arthritis (RA), including its relative function with bone and cartilage development and growth plate, synovial, and tendon formation. The role of cadherin-11 in RA is also discussed, both in fibroblasts inflammation and fibroblast-like synoviocyte (FLSs) migration and invasion. The potential of anti-cadherin-11 therapy for RA is introduced in comparison with the other current RA therapies.
Collapse
|
50
|
Yin K, Liao Q, He H, Zhong D. Prognostic value of Twist and E-cadherin in patients with osteosarcoma. Med Oncol 2012; 29:3449-55. [PMID: 22847601 DOI: 10.1007/s12032-012-0317-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 07/17/2012] [Indexed: 12/31/2022]
Abstract
Twist, a basic helix-loop-helix transcription factor, and E-cadherin are both correlated with the metastatic progression of several types of cancer. However, it is currently unknown whether their activations have relevance to the progression of osteosarcoma. The purpose of the present study was to investigate the clinicopathological and prognostic value of Twist and E-cadherin in osteosarcoma. Twist and E-cadherin expressions were determined by immunohistochemistry. Patient survival rates were determined by Kaplan-Meier method and log-rank test. Cox regression was adopted for multivariate analysis of prognostic factors. The positive rates of Twist and E-cadherin in 107 osteosarcoma specimens were 31.8 % (34/107) and 20.6 % (22/107), respectively. Twist expression was significantly correlated with that of E-cadherin (r = -0.209, P = 0.031). The positive expression of Twist and E-cadherin was significantly associated with metastasis in 107 osteosarcoma specimens (both P < 0.05). Patients with positive Twist expression had significantly poorer overall survival (OS; P < 0.05) and disease-free survival (DFS, P < 0.05) when compared with patients with the negative expression of Twist. Patients with positive expression of E-cadherin had significantly poorer OS (P < 0.05) when compared with patients with negative E-cadherin expression, but not a significantly poorer DFS (P = 0.081). On multivariate analysis, Twist expression and age were found to be independent prognostic factors for OS (both P < 0.05) and DFS (both P < 0.05). Our results suggest that Twist was expressed significantly more and E-cadherin significantly less in osteosarcoma with metastasis, and expression of both might be related to the prediction of metastasis potency and poor prognosis for patients with osteosarcoma.
Collapse
Affiliation(s)
- Ke Yin
- Department of Orthopaedics, Xiangya Hospital, Central South University, # 87 Xiangya Road, Changsha, 410008, Hunan, China
| | | | | | | |
Collapse
|