1
|
Liu H, Li Z, Xu B, Li Z, Yang X, Luo J. Curcumin suppresses metastasis, invasion, and proliferation in osteosarcoma cells by regulating the EGFR/Src signaling axis. Int J Immunopathol Pharmacol 2024; 38:3946320241308082. [PMID: 39707861 DOI: 10.1177/03946320241308082] [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] [Indexed: 12/23/2024] Open
Abstract
We explored the biological mechanisms by which curcumin (Cur) confronts osteosarcoma (OS) tumorigenesis and potential drug gene targets based on network pharmacology and in vitro cell experiments. Cur has been recognized for its significant role in combating various types of tumors. However, the intrinsic molecular mechanisms through which it affects OS remain uncharted. In this study, we performed network pharmacology methods including protein-protein interaction (PPI) and core target screening, Functional Enrichment Analysis and Network Construction, Molecular Docking, which obtained the potential target of Cur. Meanwhile, cell experiments (wound healing assay, Transwell assay, Western blots, immunofluorescence, et al.) in vitro were performed to verify the targets, and reveal the biological mechanisms. A total of 18 hub genes were identified through our network pharmacological analysis. In vitro studies show that Cur inhibits the proliferation, migration, invasion capabilities of MG63 and U2OS cells. Western blot reveals a down-regulation of p-PI3K, PI3K, p-Akt, Akt, EGFR, Src, p-Src (Tyr416) and STAT3 expression when treated with Cur. Additionally, Cur upregulated epithelial proteins (E-cadherin and Occludin) while decreasing the expression of the mesenchymal protein (N-cadherin). In addition, Cur treatment decreases the EGFR/Src signaling pathway in the presence of active Src overexpression. Cur inhibits the proliferation, migration, invasion, epithelial-mesenchymal transition (EMT) by down-regulating EGFR/Src signaling axis, also resulting in coordinated weakening of its downstream regulatory genes, including Akt, STAT3, Bcl2, ERK1/2, among others signal axis (PI3K/Akt signaling pathway).
Collapse
Affiliation(s)
- Huiying Liu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, P. R. China
- The Institute of Translational Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, P. R. China
| | - Zhiqiang Li
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, P. R. China
| | - Binwu Xu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, P. R. China
| | - Zhipeng Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi, P. R. China
| | - Xili Yang
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, P. R. China
| | - Jun Luo
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, P. R. China
- The Institute of Translational Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, P. R. China
| |
Collapse
|
2
|
Ji Z, Shen J, Lan Y, Yi Q, Liu H. Targeting signaling pathways in osteosarcoma: Mechanisms and clinical studies. MedComm (Beijing) 2023; 4:e308. [PMID: 37441462 PMCID: PMC10333890 DOI: 10.1002/mco2.308] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 07/15/2023] Open
Abstract
Osteosarcoma (OS) is a highly prevalent bone malignancy among adolescents, accounting for 40% of all primary malignant bone tumors. Neoadjuvant chemotherapy combined with limb-preserving surgery has effectively reduced patient disability and mortality, but pulmonary metastases and OS cells' resistance to chemotherapeutic agents are pressing challenges in the clinical management of OS. There has been an urgent need to identify new biomarkers for OS to develop specific targeted therapies. Recently, the continued advancements in genomic analysis have contributed to the identification of clinically significant molecular biomarkers for diagnosing OS, acting as therapeutic targets, and predicting prognosis. Additionally, the contemporary molecular classifications have revealed that the signaling pathways, including Wnt/β-catenin, PI3K/AKT/mTOR, JAK/STAT3, Hippo, Notch, PD-1/PD-L1, MAPK, and NF-κB, have an integral role in OS onset, progression, metastasis, and treatment response. These molecular classifications and biological markers have created new avenues for more accurate OS diagnosis and relevant treatment. We herein present a review of the recent findings for the modulatory role of signaling pathways as possible biological markers and treatment targets for OS. This review also discusses current OS therapeutic approaches, including signaling pathway-based therapies developed over the past decade. Additionally, the review covers the signaling targets involved in the curative effects of traditional Chinese medicines in the context of expression regulation of relevant genes and proteins through the signaling pathways to inhibit OS cell growth. These findings are expected to provide directions for integrating genomic, molecular, and clinical profiles to enhance OS diagnosis and treatment.
Collapse
Affiliation(s)
- Ziyu Ji
- School of Integrated Traditional Chinese and Western MedicineSouthwest Medical UniversityLuzhouSichuanChina
| | - Jianlin Shen
- Department of OrthopaedicsAffiliated Hospital of Putian UniversityPutianFujianChina
| | - Yujian Lan
- School of Integrated Traditional Chinese and Western MedicineSouthwest Medical UniversityLuzhouSichuanChina
| | - Qian Yi
- Department of PhysiologySchool of Basic Medical ScienceSouthwest Medical UniversityLuzhouSichuanChina
| | - Huan Liu
- Department of OrthopaedicsThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouSichuanChina
| |
Collapse
|
3
|
Pharmacogenetics of the Primary and Metastatic Osteosarcoma: Gene Expression Profile Associated with Outcome. Int J Mol Sci 2023; 24:ijms24065607. [PMID: 36982681 PMCID: PMC10059037 DOI: 10.3390/ijms24065607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/08/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023] Open
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor in children and adolescents. In recent decades, OS treatment has reached a plateau and drug resistance is still a major challenge. Therefore, the present study aimed to analyze the expression of the genes related to pharmacogenetics in OS. The expression of 32 target genes in 80 paired specimens (pre-chemotherapeutic primary tumor, post-chemotherapeutic primary tumor and pulmonary metastasis) obtained from 33 patients diagnosed with OS were analyzed by the real-time PCR methodology. As the calibrators (control), five normal bone specimens were used. The present study identified associations between the OS outcome and the expression of the genes TOP2A, DHFR, MTHFR, BCL2L1, CASP3, FASLG, GSTM3, SOD1, ABCC1, ABCC2, ABCC3, ABCC5, ABCC6, ABCC10, ABCC11, ABCG2, RALBP1, SLC19A1, SLC22A1, ERCC1 and MSH2. In addition, the expression of the ABCC10, GGH, GSTM3 and SLC22A1 genes were associated with the disease event, and the metastasis specimens showed a high expression profile of ABCC1, ABCC3 and ABCC4 genes and a low expression of SLC22A1 and ABCC10 genes, which is possibly an important factor for resistance in OS metastasis. Therefore, our findings may, in the future, contribute to clinical management as prognostic factors as well as possible therapeutic targets.
Collapse
|
4
|
Belluomo R, Khodaei A, Amin Yavari S. Additively manufactured Bi-functionalized bioceramics for reconstruction of bone tumor defects. Acta Biomater 2023; 156:234-249. [PMID: 36028198 DOI: 10.1016/j.actbio.2022.08.042] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 02/08/2023]
Abstract
Bone tissue exhibits critical factors for metastatic cancer cells and represents an extremely pleasant spot for further growth of tumors. The number of metastatic bone lesions and primary tumors that arise directly from cells comprised in the bone milieu is constantly increasing. Bioceramics have recently received significant attention in bone tissue engineering and local drug delivery applications. Additionally, additive manufacturing of bioceramics offers unprecedented advantages including the possibilities to fill irregular voids after the resection and fabricate patient-specific implants. Herein, we investigated the recent advances in additively manufactured bioceramics and ceramic-based composites that were used in the local bone tumor treatment and reconstruction of bone tumor defects. Furthermore, it has been extensively explained how to bi-functionalize ceramics-based biomaterials and what current limitations impede their clinical application. We have also discussed the importance of further development into ceramic-based biomaterials and molecular biology of bone tumors to: (1) discover new potential therapeutic targets to enhance conventional therapies, (2) local delivering of bio-molecular agents in a customized and "smart" way, and (3) accomplish a complete elimination of tumor cells in order to prevent tumor recurrence formation. We emphasized that by developing the research focus on the introduction of novel 3D-printed bioceramics with unique properties such as stimuli responsiveness, it will be possible to fabricate smart bioceramics that promote bone regeneration while minimizing the side-effects and effectively eradicate bone tumors while promoting bone regeneration. In fact, by combining all these therapeutic strategies and additive manufacturing, it is likely to provide personalized tumor-targeting therapies for cancer patients in the foreseeable future. STATEMENT OF SIGNIFICANCE: To increase the survival rates of cancer patients, different strategies such as surgery, reconstruction, chemotherapy, radiotherapy, etc have proven to be essential. Nonetheless, these therapeutic protocols have reached a plateau in their effectiveness due to limitations including drug resistance, tumor recurrence after surgery, toxic side-effects, and impaired bone regeneration following tumor resection. Hence, novel approaches to specifically and locally attack cancer cells, while also regenerating the damaged bony tissue, have being developed in the past years. This review sheds light to the novel approaches that enhance local bone tumor therapy and reconstruction procedures by combining additive manufacturing of ceramic biomaterials and other polymers, bioactive molecules, nanoparticles to affect bone tumor functions, metabolism, and microenvironment.
Collapse
Affiliation(s)
- Ruggero Belluomo
- Department of Orthopedics, University Medical Center Utrecht, Utrecht 3508GA, the Netherlands
| | - Azin Khodaei
- Department of Orthopedics, University Medical Center Utrecht, Utrecht 3508GA, the Netherlands
| | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, Utrecht 3508GA, the Netherlands; Regenerative Medicine Utrecht, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
5
|
Hu Z, Wen S, Huo Z, Wang Q, Zhao J, Wang Z, Chen Y, Zhang L, Zhou F, Guo Z, Liu H, Zhou S. Current Status and Prospects of Targeted Therapy for Osteosarcoma. Cells 2022; 11:3507. [PMID: 36359903 PMCID: PMC9653755 DOI: 10.3390/cells11213507] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 09/26/2023] Open
Abstract
Osteosarcoma (OS) is a highly malignant tumor occurring in bone tissue with a high propensity to metastasize, and its underlying mechanisms remain largely elusive. The OS prognosis is poor, and improving the survival of OS patients remains a challenge. Current treatment methods such as surgical approaches, chemotherapeutic drugs, and immunotherapeutic drugs remain ineffective. As research progresses, targeted therapy is gradually becoming irreplaceable. In this review, several treatment modalities for osteosarcoma, such as surgery, chemotherapy, and immunotherapy, are briefly described, followed by a discussion of targeted therapy, the important targets, and new technologies for osteosarcoma treatment.
Collapse
Affiliation(s)
- Zunguo Hu
- Department of Joint Surgery, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang 261061, China
| | - Shuang Wen
- Department of Joint Surgery, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang 261061, China
| | - Zijun Huo
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Qing Wang
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Jiantao Zhao
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Zihao Wang
- Department of Joint Surgery, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang 261061, China
| | - Yanchun Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Lingyun Zhang
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Fenghua Zhou
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Zhangyu Guo
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Huancai Liu
- Department of Joint Surgery, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang 261061, China
| | - Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
Martinez T, Sarda S, Dupret-Bories A, Charvillat C, Projetti F, Drouet C. Toward a doxorubicin-loaded bioinspired bone cement for the localized treatment of osteosarcoma. Future Oncol 2021; 17:3511-3528. [PMID: 34213375 DOI: 10.2217/fon-2021-0128] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aims: Osteosarcoma represents the second most common cause of death in children and young adults. No biomaterial allowing local drug delivery has been specifically developed. However, a biocompatible bioactive implantable material could prevent some amputations, and the local release of an antitumor agent could limit risks of relapse and metastasis. Methods: We propose a proof of concept of a self-setting paste combining amorphous calcium phosphate and doxorubicin-loaded particles of bone-like carbonated nanocrystalline apatite, as a means of local release. Results: The cement formulation and doping, first with folic acid and then with doxorubicin, was successful. Its physicochemistry was scrutinized. Preliminary in vivo data on an invasive osteosarcoma rat model suggest a limiting effect on metastatic events in the lungs without signs of toxicity.
Collapse
Affiliation(s)
- Thomas Martinez
- CIRIMAT, Université de Toulouse, CNRS, Université Toulouse 3 Paul Sabatier, ENSIACET, Toulouse, 31030, France
| | - Stéphanie Sarda
- CIRIMAT, Université de Toulouse, CNRS, Université Toulouse 3 Paul Sabatier, ENSIACET, Toulouse, 31030, France
| | | | - Cédric Charvillat
- CIRIMAT, Université de Toulouse, CNRS, Université Toulouse 3 Paul Sabatier, ENSIACET, Toulouse, 31030, France
| | | | - Christophe Drouet
- CIRIMAT, Université de Toulouse, CNRS, Université Toulouse 3 Paul Sabatier, ENSIACET, Toulouse, 31030, France
| |
Collapse
|
7
|
Prasad SR, Kumar TSS, Jayakrishnan A. Nanocarrier-based drug delivery systems for bone cancer therapy: a review. Biomed Mater 2021; 16. [PMID: 33853043 DOI: 10.1088/1748-605x/abf7d5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/14/2021] [Indexed: 02/07/2023]
Abstract
Bone cancer is a malignant tumor that originates in the bone and destroys the healthy bone tissues. Of the various types of bone tumors, osteosarcoma is the most commonly diagnosed primary bone malignancy. The standard treatment for primary malignant bone tumors comprises surgery, chemotherapy and radiotherapy. Owing to the lack of proven treatments, different forms of alternative therapeutic approaches have been examined in recent decades. Among the new therapeutic methodologies, nanotechnology-based anticancer therapy has paved the way for new targeted strategies for bone cancer treatment and bone regeneration. They include approaches such as the co-delivery of multiple drug cargoes, the enhancement of their biodistribution and transport properties, normalizing accumulation and the optimization of drug release profiles to overcome shortcomings of the existing therapy. This review examines the standard treatments for osteosarcoma, their lacunae, and the evolving therapeutic strategies based on nanocarrier-mediated combinational drug delivery systems, and future perspectives for osteosarcoma therapy.
Collapse
Affiliation(s)
- S Ram Prasad
- Rajiv Gandhi Centre for Biotechnology, Jagathy, Trivandrum 695 014 Kerala, India
| | - T S Sampath Kumar
- Medical Materials Laboratory, Department of Metallurgical and Materials Engineering, Indian Institute of Technology Madras, Chennai 600 036 Tamil Nadu, India
| | - A Jayakrishnan
- Rajiv Gandhi Centre for Biotechnology, Jagathy, Trivandrum 695 014 Kerala, India
| |
Collapse
|
8
|
Gao X, Zhang C, Wang Y, Zhang P, Zhang J, Hong T. Berberine and Cisplatin Exhibit Synergistic Anticancer Effects on Osteosarcoma MG-63 Cells by Inhibiting the MAPK Pathway. Molecules 2021; 26:molecules26061666. [PMID: 33802664 PMCID: PMC8002572 DOI: 10.3390/molecules26061666] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 11/27/2022] Open
Abstract
Berberine (BBR) has been reported to have potent anticancer activity and can increase the anticancer effects of chemotherapy drugs. The present study aims to investigate whether BBR and cisplatin (DDP) exert synergistic effects on the osteosarcoma (OS) MG-63 cell line. In the present study, MG-63 cells were treated with BBR and DDP alone or in combination. The effects of these therapeutics on cell viability, colony formation, migration, invasion, nuclear morphology, apoptosis, and the cell cycle, as well as their role in regulating the expression of proteins related to apoptosis, the cell cycle, and the mitogen-activated protein kinase (MAPK) pathway, were determined. The results demonstrated that BBR or DDP significantly inhibited the proliferation of MG-63 cells in a dose- and time-dependent manner. The combination treatment of BBR and DDP exerted a prominent inhibitory effect on proliferation and colony formation. Furthermore, the results showed that the combination treatment of BBR and DDP enhanced the inhibition of cell migration and invasion and reversed the changes in nuclear morphology. The results showed that the combination treatment of BBR and DDP induced apoptosis and cell cycle arrest in the G0/G1 phase. Mechanistically, the combination treatment of BBR and DDP inhibited the expression of MMP-2/9, Bcl-2, CyclinD1, and CDK4, enhanced the expression of Bax and regulated the activity of the MAPK pathway. Collectively, our data suggest that the combination therapy of BBR and DDP markedly enhanced OS cell death.
Collapse
Affiliation(s)
| | | | | | | | | | - Tie Hong
- Correspondence: ; Tel.: +86-431-8561-9705
| |
Collapse
|
9
|
Zhang L, Yang C, Huang Y, Huang H, Yuan X, Zhang P, Ye C, Wei M, Wang Y, Luo X, Luo J. Cardamonin inhibits the growth of human osteosarcoma cells through activating P38 and JNK signaling pathway. Biomed Pharmacother 2021; 134:111155. [PMID: 33370628 DOI: 10.1016/j.biopha.2020.111155] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023] Open
Abstract
Osteosarcoma (OS) is the most common type of bone malignant tumors. Clinical commonly used therapeutic drugs of OS treatment are prone to toxic and side effects, so it is very urgent to develop new drugs with low toxicity and low side effects. As a Chinese herbal medicine, Cardamonin (CAR) (C16H14O4) has inhibitory effects in various tumors. In the present study, we investigated the effects of CAR on OS cells in vitro and in vivo. We found that CAR inhibited cell proliferation, reduced migration, decreased invasion, and induced G2 / M arrest of OS cells. Notably, we demonstrated that CAR had no obvious effect on proliferation and apoptosis of normal cells. Besides, CAR repressed tumor growth of OS cells in xenograft mouse model. Mechanically, we found that CAR increased the phosphorylation level of P38 and JNK. In summary, our research validates that CAR may inhibit the proliferation, migration, and invasion of OS and promote apoptosis possibly by activating P38 and JNK Mitogen-activated protein kinase (MAPK) signaling pathway.
Collapse
Affiliation(s)
- Lulu Zhang
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| | - Chunmei Yang
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| | - Yanran Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Huakun Huang
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| | - Xiaohui Yuan
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| | - Ping Zhang
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| | - Caihong Ye
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| | - Mengqi Wei
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| | - Yuping Wang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Xiaoji Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Jinyong Luo
- School of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
10
|
Fernandes I, Melo-Alvim C, Lopes-Brás R, Esperança-Martins M, Costa L. Osteosarcoma Pathogenesis Leads the Way to New Target Treatments. Int J Mol Sci 2021; 22:E813. [PMID: 33467481 PMCID: PMC7831017 DOI: 10.3390/ijms22020813] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma (OS) is a rare condition with very poor prognosis in a metastatic setting. Basic research has enabled a better understanding of OS pathogenesis and the discovery of new potential therapeutic targets. Phase I and II clinical trials are already ongoing, with some promising results for these patients. This article reviews OS pathogenesis and new potential therapeutic targets.
Collapse
Affiliation(s)
- Isabel Fernandes
- Medical Oncology Department, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1600 Lisbon, Portugal; (C.M.-A.); (R.L.-B.); (M.E.-M.); (L.C.)
- Luís Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1600 Lisbon, Portugal
| | - Cecília Melo-Alvim
- Medical Oncology Department, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1600 Lisbon, Portugal; (C.M.-A.); (R.L.-B.); (M.E.-M.); (L.C.)
| | - Raquel Lopes-Brás
- Medical Oncology Department, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1600 Lisbon, Portugal; (C.M.-A.); (R.L.-B.); (M.E.-M.); (L.C.)
| | - Miguel Esperança-Martins
- Medical Oncology Department, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1600 Lisbon, Portugal; (C.M.-A.); (R.L.-B.); (M.E.-M.); (L.C.)
- Luís Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1600 Lisbon, Portugal
- Sérgio Dias Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1600 Lisbon, Portugal
| | - Luís Costa
- Medical Oncology Department, Hospital Santa Maria, Centro Hospitalar Universitário Lisboa Norte, 1600 Lisbon, Portugal; (C.M.-A.); (R.L.-B.); (M.E.-M.); (L.C.)
- Luís Costa Lab, Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1600 Lisbon, Portugal
| |
Collapse
|
11
|
Li J, Li J, Qin J, Zeng H, Wang K, Wang D, Wang S. Confocal Raman microspectroscopic analysis on the time-dependent impact of DAPT, a γ-secretase inhibitor, to osteosarcoma cells. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2020; 239:118372. [PMID: 32416170 DOI: 10.1016/j.saa.2020.118372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 06/11/2023]
Abstract
Confocal Raman microspectroscopy (CRM) analysis provides subcellular compositional and morphology related information. In this study, we used CRM in conjunction with multivariate statistical analysis to elucidate the time-dependent impact of the γ-secretase inhibitor, DAPT (N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester) of osteosarcoma (OS) cells. The interactions of DAPT (20 μM) with a murine OS cell line K7M2 at 24 and 48 h were monitored. The spectral characteristics of drug action were identified to illustrate the cellular compositional alterations, showing that DAPT induced apoptosis by reducing the protein, lipid and nucleic acid content and structural changes. Multivariate algorithms, principal component analysis (PCA) and linear discriminant analysis (LDA) revealed a clear separation among cells in the untreated control (UT), 24H (DAPT-treated for 24 h), and 48H (DAPT-treated for 48 h) groups, achieving sensitivities of 100%, 96%, 100% and specificities of 98%, 100%, 100%, respectively. After point-scanned spectral imaging, K-means clustering analysis (KCA) was further used to visualize sub-cellular morphological changes and the underlying spectral characteristics in a temporal sequence. Compared with the UT group, Raman imaging results exhibited gradually increased nuclear division of OS cells with DAPT treatment duration extension, along with changes in the physiology of other organelles within the cell. By providing a unique perspective for understanding the temporary cellular responses to DAPT at molecular level, the achieved results form the foundation of strategies for the application of CRM and other Raman-based techniques for studying the therapeutic responses of other anticancer agents in cancer model systems.
Collapse
Affiliation(s)
- Jie Li
- Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi 710069, China
| | - Jing Li
- Department of Orthopedics, The Second Affiliated Hospital of Xi 'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Jie Qin
- Department of Orthopedics, The Second Affiliated Hospital of Xi 'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Haishan Zeng
- Imaging Unit - Integrative Oncology Department, BC Cancer Research Center, Vancouver, BC V5Z1L3, Canada
| | - Kaige Wang
- Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi 710069, China
| | - Difan Wang
- School of Life, Xidian University, Xi'an, Shaanxi 710071, China
| | - Shuang Wang
- Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
12
|
Abstract
The rediscovery and reinterpretation of the Warburg effect in the year 2000 occulted for almost a decade the key functions exerted by mitochondria in cancer cells. Until recent times, the scientific community indeed focused on constitutive glycolysis as a hallmark of cancer cells, which it is not, largely ignoring the contribution of mitochondria to the malignancy of oxidative and glycolytic cancer cells, being Warburgian or merely adapted to hypoxia. In this review, we highlight that mitochondria are not only powerhouses in some cancer cells, but also dynamic regulators of life, death, proliferation, motion and stemness in other types of cancer cells. Similar to the cells that host them, mitochondria are capable to adapt to tumoral conditions, and probably to evolve to ‘oncogenic mitochondria' capable of transferring malignant capacities to recipient cells. In the wider quest of metabolic modulators of cancer, treatments have already been identified targeting mitochondria in cancer cells, but the field is still in infancy.
Collapse
Affiliation(s)
- Debora Grasso
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Luca X Zampieri
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Tânia Capelôa
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Justine A Van de Velde
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
13
|
Wan B, Hu H, Wang R, Liu W, Chen D. Therapeutic Potential of Circular RNAs in Osteosarcoma. Front Oncol 2020; 10:370. [PMID: 32351876 PMCID: PMC7174900 DOI: 10.3389/fonc.2020.00370] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 03/03/2020] [Indexed: 12/14/2022] Open
Abstract
Osteosarcoma is the most common malignant bone tumor in children and adolescents. Multiagent chemotherapy, together with surgical removal of all detectable lesions, has improved the long-term survival rate to 65-70% in patients with localized osteosarcoma and to 25-30% in patients with metastatic osteosarcoma since the 1970s. However, the conventional strategy has not improved in recent decades. With accumulating knowledge of the natural circular RNA (circRNA) pathogenesis of osteosarcoma, the diagnostic and therapeutic potential of some circRNAs has been explored. Meanwhile, artificial circular RNAs have been designed as onco-microRNA inhibitors to exert antitumor functions. Therefore, natural and artificial circular RNAs, like other RNA counterparts, are attractive new classes of therapeutic molecules for the treatment of osteosarcoma. This review summarizes the latest progress in the relationship between circRNAs and the malignant phenotype of osteosarcoma and sheds light on the therapeutic potential of the two types of circular RNA in the clinic.
Collapse
Affiliation(s)
- Ben Wan
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, China
| | - Hao Hu
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
- Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Renxian Wang
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, China
| | - Weifeng Liu
- Department of Orthopaedic Oncology Surgery, Beijing Jishuitan Hospital, Peking University, Beijing, China
| | - Dafu Chen
- Laboratory of Bone Tissue Engineering, Beijing Laboratory of Biomedical Materials, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Beijing, China
| |
Collapse
|
14
|
Palubeckaitė I, Crooks L, Smith DP, Cole LM, Bram H, Le Maitre C, Clench MR, Cross NA. Mass spectrometry imaging of endogenous metabolites in response to doxorubicin in a novel 3D osteosarcoma cell culture model. JOURNAL OF MASS SPECTROMETRY : JMS 2020; 55:e4461. [PMID: 31654532 DOI: 10.1002/jms.4461] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/27/2019] [Accepted: 10/17/2019] [Indexed: 06/10/2023]
Abstract
Three-dimensional (3D) cell culture is a rapidly emerging field, which mimics some of the physiological conditions of human tissues. In cancer biology, it is considered a useful tool in predicting in vivo chemotherapy responses, compared with conventional two-dimensional (2D) cell culture. We have developed a novel 3D cell culture model of osteosarcoma composed of aggregated proliferative tumour spheroids, which shows regions of tumour heterogeneity formed by aggregated spheroids of polyclonal tumour cells. Aggregated spheroids show local necrotic and apoptotic regions and have sizes suitable for the study of spatial distribution of metabolites by mass spectrometry imaging (MSI). We have used this model to perform a proof-of-principle study showing a heterogeneous distribution of endogenous metabolites that colocalise with the necrotic core and apoptotic regions in this model. Cytotoxic chemotherapy (doxorubicin) responses were significantly attenuated in our 3D cell culture model compared with those of standard cell culture, as determined by resazurin assay, despite sufficient doxorubicin diffusion demonstrated by localisation throughout the 3D constructs. Finally, changes to the distribution of endogenous metabolites in response to doxorubicin were readily detected by MSI. Principal component analysis identified 50 metabolites which differed most in their abundance between treatment groups, and of these, 10 were identified by both in-software t test and mixed-effects analysis of variance (ANOVA). Subsequent independent MSIs of identified species were consistent with principle component analysis findings. This proof-of-principle study shows for the first time that chemotherapy-induced changes in metabolite abundance and distribution may be determined in 3D cell culture by MSI, highlighting this method as a potentially useful tool in the elucidation of chemotherapy responses as an alternative to in vivo testing.
Collapse
Affiliation(s)
- Ieva Palubeckaitė
- Department of Pathology, Leiden University Medical Center, PO Box 9600, 2300, RC, Leiden, The Netherlands
| | - Lucy Crooks
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield, S1 1WB, UK
| | - David P Smith
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield, S1 1WB, UK
| | - Laura M Cole
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield, S1 1WB, UK
| | - Heijs Bram
- Center for Proteomics and Metabolomics, Leiden University Medical Center, PO Box 9600, 2300, RC, Leiden, The Netherlands
| | - Christine Le Maitre
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield, S1 1WB, UK
| | - Malcolm R Clench
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield, S1 1WB, UK
| | - Neil A Cross
- Centre for Mass Spectrometry Imaging, Biomolecular Sciences Research Centre, Sheffield Hallam University, Howard Street, Sheffield, S1 1WB, UK
| |
Collapse
|
15
|
Savio S, Deslivia M, Astawa P, Wiratnaya IGE. The outcome comparison of limb salvage surgery versus amputation for high-grade osteosarcoma: A systematic review and meta-analysis of the last 7-year studies. JOURNAL OF ORTHOPEDICS, TRAUMATOLOGY AND REHABILITATION 2020. [DOI: 10.4103/jotr.jotr_52_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
16
|
Zhao S, Su Y, Duan J, Qiu Q, Ge X, Wang A, Yin Y. Radiomics signature extracted from diffusion-weighted magnetic resonance imaging predicts outcomes in osteosarcoma. J Bone Oncol 2019; 19:100263. [PMID: 31667064 PMCID: PMC6812010 DOI: 10.1016/j.jbo.2019.100263] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/28/2019] [Accepted: 10/03/2019] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Osteosarcoma often requires multidisciplinary treatment including surgery, chemotherapy and radiotherapy. However, tumor behavior can vary widely among patients and selection of appropriate therapies in any individual patient remains a critical challenge. Radiomics seeks to quantify complex aspects of tumor images under the assumption that this information is related to tumor biology. This study tested the hypothesis that a radiomic signature extracted from Diffusion-weighted magnetic resonance images (DWI-MRI) can improve prediction of overall survival (OS) compared with clinical factors alone in localised osteosarcoma. MATERIALS/METHODS Pre-treatment DWI-MRI were collected from 112 patients (9-67 years of age) with histological-proven osteosarcoma that were treated with curative intent. The entire dataset was divided in two subsets: the training and validation cohorts containing 76 and 24% of the data respectively. Clinical data were extracted from our medical record. Two experienced radiotherapists evaluated DWI-MRIs for quality and segmented the tumor. A total of 103 radiomic features were calculated for each image. Least absolute shrinkage and selection operator (LASSO) regression was applied to select features. Association between the radiomics signature and OS was explored. Further validation of the radiomics signature as an independent biomarker was performed by using multivariate Cox regression. The Cox proportional-hazard regression model was also used to analyze the correlation between the prognostic factor and the survival for the clinical (C) model after the univariate analysis. Radiomics (R) model identified radiomics signature, which is the best predictor from the radiomic variable classes based on LASSO regression. Harrell's C-index was used to demonstrate the incremental value of the radiomics signature to the traditional clinical risk factors for the individualized prediction performance. RESULTS Cox proportional-hazard regression model shows that: Tumor size, alkaline phosphatase (ALP) status before treatment and number of courses of chemotherapy were proven as the dependent clinical prognostic factors of osteosarcoma's overall survival time. The radiomics signature was significantly associated with OS, independent of clinical risk factors (radiomics signature: HR: 5.11, 95% CI: 2.85, 9.18, P < 0.001). Incorporating the radiomics signature into the coalition (C+R) model resulted in better performance (P < .001) for the estimation of OS (C-index: 0.813; 95% CI: 0.75, 0.89) than with the clinical (C) model (C-index: 0.764; 95% CI: 0.69, 0.85), or the single radiomics (R) model (C-index: 0.712; 95% CI: 0.65, 0.78). CONCLUSION This study shows that the radiomics signature extracted from pre-treatment DWI-MRI improve prediction of OS over clinical features alone. Combination of the radiomics signature and the traditional clinical risk factors performed better for individualized OS estimation in patients with osteosarcoma, which might enable a step forward precise medicine. This method may help better select patients most likely to benefit from intensified multimodality diagnosis and therapies. Future studies will focus on multi-center validation of an optimized model.
Collapse
Affiliation(s)
- Shuliang Zhao
- School of Medicine, Shandong University, Ji'nan 250012, China
- Department of Radiotherapy, Yantaishan Hospital of Yantai, Yantai 264001, China
| | - Yi Su
- Department of Radiotherapy, Yuhuangding Hospital of Yantai, Yantai 264001, China
| | - Jinghao Duan
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji'nan 250117, China
| | - Qingtao Qiu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji'nan 250117, China
| | - Xingping Ge
- Department of Radiotherapy, Yantaishan Hospital of Yantai, Yantai 264001, China
| | - Aijie Wang
- Department of CT/MR, Yantaishan Hospital, Yantai 264001, China
| | - Yong Yin
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Ji'nan 250117, China
| |
Collapse
|
17
|
Li W, Tao C, Wang J, Le Y, Zhang J. MMP-responsive in situ forming hydrogel loaded with doxorubicin-encapsulated biodegradable micelles for local chemotherapy of oral squamous cell carcinoma. RSC Adv 2019; 9:31264-31273. [PMID: 35527962 PMCID: PMC9072589 DOI: 10.1039/c9ra04343h] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/10/2019] [Indexed: 12/24/2022] Open
Abstract
The complex construction within the oral cavity causes incomplete surgical resection of oral squamous cell carcinoma (OSCC) that may enhance the risk of recurrence and metastasis in the treatment. In situ forming injectable hydrogels with minimally invasive procedures, encapsulation stability and stimuli-responsive degradation have emerged as promising carriers for local drug delivery. In this study, doxorubicin (DOX) was first encapsulated in biodegradable poly(d,l-lactide)-poly(ethylene glycol)-poly(d,l-lactide) (PDLLA-PEG-PDLLA) micelles and then loaded into an in situ injectable hyaluronic acid (HA) hydrogel, which was cross-linked by a matrix metalloproteinase-2 (MMP-2)-responsive peptide (GCRDGPQGIWGQDRCG) through a Michael addition reaction. In vitro studies demonstrated that the HA hydrogel had a sensitive MMP-2-responsive drug release profile. Investigations including MTT, live-dead, apoptosis, and wound healing assays illustrated that DOX micelle-loaded HA hydrogels exhibited outstanding cytotoxicity against squamous carcinoma cells (SCC-15). Furthermore, by in vivo studies, we also proved that HA hydrogels degraded faster in the tumor site than in normal tissue, which led to a local sustained release of DOX-loaded micelles and tumor growth inhibition of oral squamous cell carcinoma (OSCC) without any damage to the organs. Therefore, this work provides a remarkable drug delivery platform for local chemotherapy and other applications.
Collapse
Affiliation(s)
- Wei Li
- State Key Laboratory of Organic-Inorganic Composites, College of Chemical Engineering, Beijing University of Chemical Technology Beijing 100029 PR China
| | - Cheng Tao
- State Key Laboratory of Organic-Inorganic Composites, College of Chemical Engineering, Beijing University of Chemical Technology Beijing 100029 PR China
| | - Jiexin Wang
- State Key Laboratory of Organic-Inorganic Composites, College of Chemical Engineering, Beijing University of Chemical Technology Beijing 100029 PR China
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology Beijing 100029 PR China
- Research Center of the Ministry of Education for High Gravity Engineering and Technology, Beijing University of Chemical Technology Beijing 100029 PR China
| | - Yuan Le
- State Key Laboratory of Organic-Inorganic Composites, College of Chemical Engineering, Beijing University of Chemical Technology Beijing 100029 PR China
- Research Center of the Ministry of Education for High Gravity Engineering and Technology, Beijing University of Chemical Technology Beijing 100029 PR China
| | - Jianjun Zhang
- State Key Laboratory of Organic-Inorganic Composites, College of Chemical Engineering, Beijing University of Chemical Technology Beijing 100029 PR China
| |
Collapse
|
18
|
Zhong B, Shi D, Wu F, Wang S, Hu H, Cheng C, Qing X, Huang X, Luo X, Zhang Z, Shao Z. Dynasore suppresses cell proliferation, migration, and invasion and enhances the antitumor capacity of cisplatin via STAT3 pathway in osteosarcoma. Cell Death Dis 2019; 10:687. [PMID: 31534119 PMCID: PMC6751204 DOI: 10.1038/s41419-019-1917-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 12/17/2022]
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor. The prognosis of metastatic and recurrent OS patients still remains unsatisfactory. Cisplatin reveals undeniable anti-tumor effect while induces severe side effects that threatening patients’ health. Dynasore, a cell-permeable small molecule that inhibits dynamin activity, has been widely studied in endocytosis and phagocytosis. However, the anti-tumor effect of dynasore on OS has not yet been ascertained. In the present study, we suggested that dynasore inhibited cell proliferation, migration, invasion, and induced G0/G1 arrest of OS cells. Besides, dynasore repressed tumorigenesis of OS in xenograft mouse model. In addition, we demonstrated that dynasore improved the anti-tumor effect of cisplatin in vitro and in vivo without inducing nephrotoxicity and hepatotoxicity. Mechanistically, dynasore repressed the expression of CCND1, CDK4, p-Rb, and MMP-2. Furthermore, we found that dynasore exerts anti-tumor effects in OS partially via inhibiting STAT3 signaling pathway but not ERK-MAPK, PI3K-Akt or SAPK/JNK pathways. P38 MAPK pathway served as a negative regulatory mechanism in dynasore induced anti-OS effects. Taken together, our study indicated that dynasore does suppress cell proliferation, migration, and invasion via STAT3 signaling pathway, and enhances the antitumor capacity of cisplatin in OS. Our results suggest that dynasore is a novel candidate drug to inhibit the tumor growth of OS and enhance the anti-tumor effects of cisplatin.
Collapse
Affiliation(s)
- Binlong Zhong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Deyao Shi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Fashuai Wu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Shangyu Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Hongzhi Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Cheng Cheng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Xiangcheng Qing
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Xin Huang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China
| | - Xueying Luo
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan Mental Health Centre, Wuhan Hospital for Psychotherapy, Wuhan, China
| | - Zhicai Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 JieFang Avenue, Wuhan, 430022, China.
| |
Collapse
|
19
|
Cao K, Wang H, Fang Y, Wang Y, Wei L, Chen X, Jiang Z, Wei X, Hu Y. Histone Deacetylase 4 Promotes Osteosarcoma Cell Proliferation and Invasion by Regulating Expression of Proliferating Cell Nuclear Antigen. Front Oncol 2019; 9:870. [PMID: 31552187 PMCID: PMC6743440 DOI: 10.3389/fonc.2019.00870] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 08/21/2019] [Indexed: 01/01/2023] Open
Abstract
Background/Aims: Osteosarcoma (OS) is commonly characterized by lower survival rates and high incidences of local recurrence due to its highly aggressive nature and metastatic tendencies. Studies have shown that histone deacetylase 4 (HDAC4) and proliferating cell nuclear antigen (PCNA) are highly expressed in cancers. Nevertheless, the roles of HDAC4 and PCNA in osteosarcoma (OS) remain unclear. This research aimed to study the expression of HDAC4 and PCNA and their relation to cell proliferation and invasion in human OS. Methods: The levels of HDAC4 and PCNA mRNA and protein were tested in human OS and osteochondroma (OC) tissues. The overexpression and knockdown of HDAC4 in OS cell lines were used to determine the effect of HDAC4 on the expression and degradation of PCNA. The effect of HDAC4 on cell proliferation, invasion and apoptosis was also detected. Additionally, we explored the interaction between HDAC4 and PCNA. Results: The results showed that both HDAC4 and PCNA were increased in human OS tissues. Overexpression of the HDAC4 protein increased the protein level of PCNA, had no effect on the PCNA mRNA level, and decreased the level of ubiquitinated PCNA. We found that overexpression of HDAC4 promoted cell proliferation and invasion and inhibited apoptosis. The opposite effects were observed when HDAC4 was knocked down. The results also showed that HDAC4 could bind to PCNA directly. Conclusions: Our findings suggest that HDAC4 could promote OS cell proliferation and invasion by regulating the expression of PCNA. Thus, our research indicates that HDAC4 may be a potential target for therapy in OS.
Collapse
Affiliation(s)
- Kun Cao
- Department of Orthopaedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hao Wang
- Department of Orthopaedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yueyang Fang
- Department of Orthopaedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuan Wang
- Department of Orthopaedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lei Wei
- Department of Orthopaedics and Department of Surgery, Warren Alpert Medical School of Brown University/Rhode Island Hospital (RIH), Providence, RI, United States
| | - Xi Chen
- Department of Orthopaedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zheng Jiang
- Department of Orthopaedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaochun Wei
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yong Hu
- Department of Orthopaedics, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
20
|
Liu Y, Luo G, He D. Clinical importance of S100A9 in osteosarcoma development and as a diagnostic marker and therapeutic target. Bioengineered 2019; 10:133-141. [PMID: 31055998 PMCID: PMC6527076 DOI: 10.1080/21655979.2019.1607709] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Objective: S100A9 is a calcium- and zinc-binding molecule of S100 family. The aim of the study was to evaluate the role of S100A9 in osteosarcoma (OS) and its value as a diagnostic and therapeutic target in OS. Methods: Quantitative real-time polymerase chain reaction (qRT-PCR), immunohistochemistry and microdissection-based mRNA analysis were used to detect S100A9 mRNA and protein expression in OS and normal bone tissues and its potential as a diagnostic marker in OS. In vitro experiments with RNA interference were performed to evaluate the functional role of S100A9 and its potential as a therapeutic target in OS. Results: S100A9 mRNA levels were significantly higher in OS tissues than that of in normal bone tissues. Receiver operating characteristic curves showed that S100A9 could be a useful diagnostic marker in OS. In vitro data showed that inhibition of S100A9 decreased the proliferation and invasiveness of OS cells, and these findings were supported by microarray data. Conclusions: Assessment of S100A9 mRNA expression is a promising tool for the diagnosis of OS, and S100A9 may be a promising therapeutic target in OS.
Collapse
Affiliation(s)
- Yongliang Liu
- a Department of Trauma Orthopedics , Linyi Central Hospital , Yishui , Shandong , China
| | - Gongzeng Luo
- a Department of Trauma Orthopedics , Linyi Central Hospital , Yishui , Shandong , China
| | - Dongyong He
- b Department of Internal Medcine , The Affiliated Hospital of Qingdao University , Qingdao , Shangdong , China
| |
Collapse
|
21
|
Marques MPM, Batista de Carvalho ALM, Mamede AP, Santos IP, García Sakai V, Dopplapudi A, Cinque G, Wolna M, Gardner P, Batista de Carvalho LAE. Chemotherapeutic Targets in Osteosarcoma: Insights from Synchrotron-MicroFTIR and Quasi-Elastic Neutron Scattering. J Phys Chem B 2019; 123:6968-6979. [PMID: 31339317 DOI: 10.1021/acs.jpcb.9b05596] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
This study aimed at the development of improved drugs against human osteosarcoma, which is the most common primary bone tumor in children and teenagers with a low prognosis. New insights into the impact of an unconventional Pd(II) anticancer agent on human osteosarcoma cells were obtained by synchrotron radiation-Fourier transform infrared microspectroscopy and quasi-elastic neutron scattering (QENS) experiments from its effect on the cellular metabolism to its influence on intracellular water, which can be regarded as a potential secondary pharmacological target. Specific infrared biomarkers of drug action were identified, enabling a molecular-level description of variations in cellular biochemistry upon drug exposure. The main changes were detected in the protein and lipid cellular components, namely, in the ratio of unsaturated-to-saturated fatty acids. QENS revealed reduced water mobility within the cytoplasm for drug-treated cells, coupled to a disruption of the hydration layers of biomolecules. Additionally, the chemical and dynamical profiles of osteosarcoma cells were compared to those of metastatic breast cancer cells, revealing distinct dissimilarities that may influence drug activity.
Collapse
Affiliation(s)
- Maria Paula M Marques
- "Química-Física Molecular", Department of Chemistry , University of Coimbra , 3004-535 Coimbra , Portugal.,Department of Life Sciences , University of Coimbra , 3000-456 Coimbra , Portugal
| | | | - Adriana P Mamede
- "Química-Física Molecular", Department of Chemistry , University of Coimbra , 3004-535 Coimbra , Portugal
| | - Inês P Santos
- "Química-Física Molecular", Department of Chemistry , University of Coimbra , 3004-535 Coimbra , Portugal
| | - Victoria García Sakai
- ISIS Facility , STFC Rutherford Appleton Laboratory , Chilton, Didcot , Oxfordshire OX11 0QX , U.K
| | - Asha Dopplapudi
- ISIS Facility , STFC Rutherford Appleton Laboratory , Chilton, Didcot , Oxfordshire OX11 0QX , U.K
| | - Gianfelice Cinque
- Diamond Light Source , Harwell Science and Innovation Campus , Chilton, Didcot , Oxfordshire OX11 0DE , U.K
| | - Magda Wolna
- Diamond Light Source , Harwell Science and Innovation Campus , Chilton, Didcot , Oxfordshire OX11 0DE , U.K
| | - Peter Gardner
- Manchester Institute of Biotechnology , University of Manchester , Manchester M1 7DN , U.K
| | | |
Collapse
|
22
|
Chen R, Huang LH, Gao YY, Yang JZ, Wang Y. Identification of differentially expressed genes in MG63 osteosarcoma cells with drug‑resistance by microarray analysis. Mol Med Rep 2018; 19:1571-1580. [PMID: 30569145 PMCID: PMC6390052 DOI: 10.3892/mmr.2018.9774] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 11/09/2018] [Indexed: 11/24/2022] Open
Abstract
Osteosarcoma is the most common type of primary malignant bone tumor, with extremely poor prognosis in patients with metastatic disease and resistance to therapy, such as multidrug regimens. The mechanisms of drug resistance are quite complex and have not been fully elucidated; thus, novel therapeutic targets should be identified to alleviate drug resistance in osteosarcoma. In the present study, the transcriptomes of the human osteosarcoma cell line MG63 and vincristine (VCR)-resistant MG63 cells were compared by microarray analysis. A total of 1,300 genes (602 upregulated and 698 downregulated) were reported to be differentially expressed in MG63/VCR compared with MG63 cells. Bioinformatics analysis predicted that the differentially expressed genes were mainly enriched in the B cell receptor, UVA-induced mitogen-activated protein kinases and receptor tyrosine kinase 2/3 signaling pathways. In the present study, 10 of the dysregulated genes, including roundabout homolog 1, death-associated protein kinase 1 and A-kinase anchor protein 12 were further evaluated by reverse transcription-quantitative polymerase chain reaction. These results may aid the validation of candidate biomarkers for the treatment and prognosis of osteosarcoma, and provide novel insight into the molecular mechanisms underlying the drug resistance of osteosarcoma cells.
Collapse
Affiliation(s)
- Rui Chen
- Department of Nuclear Medicine; Changhai Hospital of Shanghai, Shanghai 200433, P.R. China
| | - Li-Hong Huang
- Geriatric Department, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Yi-Yao Gao
- Science Research Center, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 13003, P.R. China
| | - Jian-Zeng Yang
- Henan Medical Key Laboratory of Molecular Imaging, Nuclear Medicine Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yan Wang
- Science Research Center, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 13003, P.R. China
| |
Collapse
|
23
|
Liu D, Liu X. Genetic polymorphisms of ERCC-1 and ERCC-2 are not prognostic markers in osteosarcoma patients with chemotherapy: A meta-analysis in Chinese population. Medicine (Baltimore) 2018; 97:e13358. [PMID: 30544402 PMCID: PMC6310529 DOI: 10.1097/md.0000000000013358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 10/30/2018] [Indexed: 11/26/2022] Open
Abstract
AIM To make an accurate estimation of the association of ERCC1 and ERCC2 polymorphisms with osteosarcoma (OS) prognosis in Chinese population. METHODS Total 7 qualified studies with 1404 osteosarcoma patients were included. Odds ratios (OR) with 95% CIs were pooled for the survival rate in different osteosarcoma patients with ERCC1 and ERCC2 genetic polymorphisms. The heterogeneity was assessed by I test. Potential publication bias was assessed by Begg funnel plot and Egger linear regression test. RESULTS In rs11615, no significant association was found under dominant [TT+TC vs. CC: OR = 1.252, 95% CI:0.864-1.815, P = .235], recessive [TT vs. TC+CC: OR = 0.850, 95% CI: 0.695-1.030, P = .095] or allelic model [T vs. C Allele: OR = 1.219, 95% CI: 0.922-1.612, P = .165]. In rs13181, no significant association was found under dominant [AA+AC vs. CC: OR = 1.031, 95% CI: 0.800-1.329, P = .801], recessive [AA vs. AC+CC: OR = 1.005, 95% CI: 0.875, 1.154, P = .944] or allelic model [A vs. C Allele: OR = 1.009, 95% CI: 0.903-1.128, P = .870]. In rs1799793, no significant association was found under dominant [GG+GA vs. AA: OR = 1.134, 95% CI: 0.884-1.454, P = .322, recessive [GG vs. AG+AA: OR = 1.025, 95% CI: 0.881-1.192, P = .750], or allelic model [G vs. A Allele: OR = 1.046, 95% CI: 0.930-1.177, P = .450]. CONCLUSION This study did not support rs11615, rs13181 or rs1799793 to be used as surrogate markers for clinical outcome of osteosarcoma with chemotherapy.
Collapse
Affiliation(s)
- Dabiao Liu
- Department of Clinical Laboratory, Zhenjiang No.4 Hospital
| | - Xuesong Liu
- Department of Laboratory Medicine, Jiangsu Vocational College of Medicine, Zhenjiang, Jiangsu, China
| |
Collapse
|
24
|
Glutathione Transferases: Potential Targets to Overcome Chemoresistance in Solid Tumors. Int J Mol Sci 2018; 19:ijms19123785. [PMID: 30487385 PMCID: PMC6321424 DOI: 10.3390/ijms19123785] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 11/23/2018] [Accepted: 11/24/2018] [Indexed: 12/14/2022] Open
Abstract
Multifunctional enzymes glutathione transferases (GSTs) are involved in the development of chemoresistance, thus representing a promising target for a novel approach in cancer treatment. This superfamily of polymorphic enzymes exhibits extraordinary substrate promiscuity responsible for detoxification of numerous conventional chemotherapeutics, at the same time regulating signaling pathways involved in cell proliferation and apoptosis. In addition to upregulated GST expression, different cancer cell types have a unique GST signature, enabling targeted selectivity for isoenzyme specific inhibitors and pro-drugs. As a result of extensive research, certain GST inhibitors are already tested in clinical trials. Catalytic properties of GST isoenzymes are also exploited in bio-activation of specific pro-drugs, enabling their targeted accumulation in cancer cells with upregulated expression of the appropriate GST isoenzyme. Moreover, the latest approach to increase specificity in treatment of solid tumors is development of GST pro-drugs that are derivatives of conventional anti-cancer drugs. A future perspective is based on the design of new drugs, which would selectively target GST overexpressing cancers more prone to developing chemoresistance, while decreasing side effects in off-target cells.
Collapse
|
25
|
Jin W, Zhou L, Yan B, Yan L, Liu F, Tong P, Yu W, Dong X, Xie L, Zhang J, Xu Y, Li C, Yuan Q, Shan L, Efferth T. Theabrownin triggers DNA damage to suppress human osteosarcoma U2OS cells by activating p53 signalling pathway. J Cell Mol Med 2018; 22:4423-4436. [PMID: 29993186 PMCID: PMC6111873 DOI: 10.1111/jcmm.13742] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 05/21/2018] [Indexed: 12/21/2022] Open
Abstract
Osteosarcoma becomes the second leading cause of cancer death in the younger population. Current outcomes of chemotherapy on osteosarcoma were unsatisfactory to date, demanding development of effective therapies. Tea is a commonly used beverage beneficial to human health. As a major component of tea, theabrownin has been reported to possess anti‐cancer activity. To evaluate its anti‐osteosarcoma effect, we established a xenograft model of zebrafish and employed U2OS cells for in vivo and in vitro assays. The animal data showed that TB significantly inhibited the tumour growth with stronger effect than that of chemotherapy. The cellular data confirmed that TB‐triggered DNA damage and induced apoptosis of U2OS cells by regulation of Mki67, PARP, caspase 3 and H2AX, and Western blot assay showed an activation of p53 signalling pathway. When P53 was knocked down by siRNA, the subsequent downstream signalling was blocked, indicating a p53‐dependent mechanism of TB on U2OS cells (p53 wt). Using osteosarcoma cell lines with p53 mutations (HOS, SAOS‐2 and MG63), we found that TB exerted stronger inhibitory effect on U2OS cells than that on p53‐mut cell lines, but it also exerted obvious effect on SAOS‐2 cells (p53 null), suggesting an activation of p53‐independent pathway in the p53‐null cells. Interestingly, theabrownin was found to have no toxicity on normal tissue in vivo and could even increase the viability of p53‐wt normal cells. In sum, theabrownin could trigger DNA damage and induce apoptosis on U2OS cells via a p53‐dependent mechanism, being a promising candidate for osteosarcoma therapy.
Collapse
Affiliation(s)
- Wangdong Jin
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Zhou
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bo Yan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Yan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fucun Liu
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Peijian Tong
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenhua Yu
- Hangzhou First People's Hospital, Hangzhou, China
| | | | - Li Xie
- Analysis Center of Agrobiology and Environmental Science, Zhejiang University, Hangzhou, China
| | | | - Yiqiao Xu
- Hunter Biotechnology, Inc, Hangzhou, China
| | - Chunqi Li
- Hunter Biotechnology, Inc, Hangzhou, China
| | - Qiang Yuan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Letian Shan
- The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
26
|
Zhang Y, He Z, Duan Y, Wang C, Kamar S, Shi X, Yang J, Yang J, Zhao N, Han L, Yang Y, Yang Z. Does intensified chemotherapy increase survival outcomes of osteosarcoma patients? A meta-analysis. J Bone Oncol 2018; 12:54-60. [PMID: 29963367 PMCID: PMC6024259 DOI: 10.1016/j.jbo.2018.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 04/04/2018] [Accepted: 04/10/2018] [Indexed: 12/21/2022] Open
Abstract
Study Design Meta-analysis. Background Although some new insights have been offered for clinical and scientific relevance, minor progress has been made in osteosarcoma treatment after a dramatic survival improvement in the late 1980s with the addition of chemotherapy to surgery. Intensified chemotherapy strategies have been suggested to increase the survival rate of patients with osteosarcoma. We performed this study to access whether intensified chemotherapy strategiesincreased survival outcomes of osteosarcoma patients compared with conventional chemotherapy strategies. Methods MEDLINE/PubMed, EMBASE, BIOSIS Previews, and Cochrane Library were searched from database set up to October2016. Randomized controlled trials (RCTs) and comparative clinical trials (CCTs) on intensified versus conventional chemotherapy strategies for osteosarcoma patients met the inclusion criteria, and the methodological quality standard were retrieved and reviewed. Data on participant characteristics, interventions, follow-up period, and outcomes were extracted from the included studies and analyzed by Review Manager 5.3. Results 12 studies (8 RCTs and 4CCT) involving 4112 patients were selected. There were no significant differences between intensified and conventional chemotherapy strategies group in 3-year event-free survival (OR, 1.01; 95% CI, [0.74–1.37]; P = 0.97), 5-year event-free survival (OR, 1.00; 95% CI, [0.86–1.17]; P = 0.97), and 5-year overall survival (OR, 1.04; 95% CI, [0.87–1.26]; P = 0.64), and good histologic response to preoperative chemotherapy (OR, 1.12; 95% CI, [0.78–1.60]; P = 0.55). Pooled analysis of local recurrence rate showed that local recurrence rate was significantly decreased in the intensified group compared with that in the conventional group (OR, 0.60; 95% CI, [0.42–0.85]; P = 0.004). Conclusions Intensified chemotherapy might not be a preferred treatment for all of the osteosarcoma patients.
Collapse
Affiliation(s)
- Ya Zhang
- Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, PR China
| | - Zewei He
- Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, PR China
| | - Yanping Duan
- The Kunming Medical University, Kunming, Yunnan 650000, PR China
| | - Cao Wang
- Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, PR China
| | - Santoshi Kamar
- The Kunming Medical University, Kunming, Yunnan 650000, PR China
| | - Xiaoqian Shi
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, PR China
| | - Jifei Yang
- Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, PR China
| | - Jingqing Yang
- Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, PR China
| | - Na Zhao
- Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, PR China
| | - Lei Han
- Department of Radiology, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, PR China
| | - Yihao Yang
- Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, PR China
| | - Zuozhang Yang
- Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University, Tumor Hospital of Yunnan Province, Kunming, Yunnan 650118, PR China
| |
Collapse
|
27
|
Ding L, Wang C, Cui Y, Han X, Zhou Y, Bai J, Li R. S-phase kinase-associated protein 2 is involved in epithelial-mesenchymal transition in methotrexate-resistant osteosarcoma cells. Int J Oncol 2018; 52:1841-1852. [PMID: 29620168 PMCID: PMC5919717 DOI: 10.3892/ijo.2018.4345] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/23/2018] [Indexed: 12/18/2022] Open
Abstract
Osteosarcoma (OS), a common worldwide primary aggressive bone malignancy, arises from primitive transformed cells of mesenchymal origin and usually attacks adolescents and young adults. Methotrexate (MTX) is the anti-folate drug used as a pivotal chemotherapeutic agent in the treatment of OS. However, patients with OS often develop drug resistance, leading to poor treatment outcomes. In the present study, in order to explore the underlying mechanisms responsible for MTX resistance, we established MTX-resistant OS cells using the U2OS and MG63 cell lines and examined whether MTX-resistant OS cells underwent epithelial-mesenchymal transition (EMT) by Transwell assay, wound healing assay, MTT assay, RT-PCR and western blot analysis. We found that the viability of the MTX-resistant cells remained relatively unaltered following further treatment with MTX compared to the parental cells. The resistant cells appeared to possess a mesenchymal phenotype, with an elongated and more spindle-like shape, and acquired enhanced invasive, migratory and attachment abilities. The measurement of EMT markers also supported EMT transition in the MTX-resistant OS cells. Our result further demonstrated that the overexpression of S-phase kinase-associated protein 2 (Skp2) was closely involved in the resistance of OS cells to MTX and in the acquirement of EMT properties. Thus, the pharmacological inhibition of Skp2 may prove to be a novel therapeutic strategy with which to overcome drug resistance in OS.
Collapse
Affiliation(s)
- Lu Ding
- Department of Orthopedics, Fifth Affiliated Hospital, Xinjiang Medical , Urumqi, Xinjiang 830011, P.R. China
| | - Chengwei Wang
- Department of Orthopedics, Sixth Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang 830002, P.R. China
| | - Yong Cui
- Department of Orthopedics, Fifth Affiliated Hospital, Xinjiang Medical , Urumqi, Xinjiang 830011, P.R. China
| | - Xiaoping Han
- Department of Orthopedics, Fifth Affiliated Hospital, Xinjiang Medical , Urumqi, Xinjiang 830011, P.R. China
| | - Yang Zhou
- Department of Orthopedics, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Jingping Bai
- Department of Orthopedics, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| | - Rong Li
- Department of Maternal, Child and Adolescent Health, College of Public Health, Xinjiang Medical University, Urumqi, Xinjiang 830011, P.R. China
| |
Collapse
|
28
|
Han K, Zhou Y, Tseng KF, Hu H, Li K, Wang Y, Gan Z, Lin S, Sun Y, Min D. PAK5 overexpression is associated with lung metastasis in osteosarcoma. Oncol Lett 2018; 15:2202-2210. [PMID: 29434926 PMCID: PMC5777019 DOI: 10.3892/ol.2017.7545] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 02/23/2017] [Indexed: 02/07/2023] Open
Abstract
p21-activated kinases (PAKs) are multifunctional effectors of Rho GTPases, which are associated with cytoskeletal organization, cellular morphogenesis, migration and survival. PAKs are overactive in a number of tumor tissues and have attracted attention as a potential target for cancer therapy. In the present study, PAK5 levels were analyzed in primary osteosarcoma (OS) samples (n=65) using reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry (IHC) methods. In the primary OS tissue, increased PAK5 expression (IHC score >2, n=37) was associated with significantly decreased overall survival (P=0.036) compared with decreased PAK5 expression (IHC score ≤2, n=28). PAK5 expression was identified to be significantly associated with metastasis (P=0.010). The lung is the most common metastasis site for OS. In addition, the level of PAK5 in lung metastasis tissue (n=13) was detected using RT-qPCR and IHC methods. PAK5 expression was increased in lung metastasis tissue compared with in primary OS samples. PAK5 was silenced using short hairpin RNA in OS cell lines. Wound healing, migration and nude mice model assay results consistently demonstrated that PAK5 knockdown was able to significantly inhibit OS migration. In PAK5-knockdown cells, the alteration in the expression of a number of metastasis-associated factors, including epithelial cadherin, vimentin, fibronectin and matrix metalloproteinase 2 (MMP2), was analyzed. Only MMP2 expression was decreased significantly (P<0.05). The expression level of MMP2 was analyzed in primary OS tissue and lung metastasis tissue using RT-qPCR and IHC methods. Expression of MMP2 was identified to be associated with expression of PAK5. The results of the present study suggest that PAK5 promotes OS cell migration and that PAK5 expression may be used to predict lung metastasis.
Collapse
Affiliation(s)
- Kun Han
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yan Zhou
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Kuo-Fu Tseng
- Biophysics Department of Oregon State University, Corvallis, OR 97330, USA
| | - Haiyan Hu
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Kunpeng Li
- School of Life Sciences, Sun Yat-Sen University, Guangzhou, Guangdong 510275, P.R. China
| | - Yaling Wang
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Zhihua Gan
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Shuchen Lin
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Yongning Sun
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| | - Daliu Min
- Oncology Department, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, P.R. China
| |
Collapse
|
29
|
Chen JM, Zhang J, Xia YM, Wang XX, Li J. The natural sweetener metabolite steviol inhibits the proliferation of human osteosarcoma U2OS cell line. Oncol Lett 2018; 15:5250-5256. [PMID: 29552164 DOI: 10.3892/ol.2018.7962] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 12/13/2017] [Indexed: 12/18/2022] Open
Abstract
Steviol is the colonic metabolite of the natural sweetener steviol glycosides. It does not diffuse to the blood and the half maximal inhibitory concentration of steviol is longer compared with that of current chemotherapy agents, including 5-fluorouracil and doxorubicin. The present study demonstrated that steviol inhibits the proliferation of the human osteosarcoma U2OS cell line in a dose- and time-dependent manner, and that the inhibition rate is comparative with that of doxorubicin and 5-fluorouracil. The mechanism of this anticancer activity is also investigated. The results indicated that steviol inhibits U2OS cells through inducing G1 phase cell cycle arrest, downregulating the ability of colony formation via a mitochondrial apoptotic pathway, which was indicated by an increase of the Bax/Bcl-2 ratio and activation of cyclin-dependent kinase inhibitor 1, tumor protein 53 and cyclin-dependent kinase; whereas a Survivin and Caspase 3-independent mechanism was involved. Considering that steviol appears minimally in the plasma during metabolism, and possesses a median lethal dose of 100-fold greater compared with that of 5-fluorouracil, it may become a potential chemotherapy agent.
Collapse
Affiliation(s)
- Jun-Ming Chen
- State Key Laboratory of Food Science and Technology, School of Chemical and Materials Engineering, Jiangnan University, Wuxi, Jiangsu 214122, P.R. China
| | - Jue Zhang
- Key Laboratory of Nuclear Medicine of Ministry of Health, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, P.R. China
| | - Yong-Mei Xia
- State Key Laboratory of Food Science and Technology, School of Chemical and Materials Engineering, Jiangnan University, Wuxi, Jiangsu 214122, P.R. China
| | - Xiao-Xia Wang
- State Key Laboratory of Food Science and Technology, School of Chemical and Materials Engineering, Jiangnan University, Wuxi, Jiangsu 214122, P.R. China
| | - Jian Li
- Key Laboratory of Nuclear Medicine of Ministry of Health, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, P.R. China
| |
Collapse
|
30
|
IWR-1, a tankyrase inhibitor, attenuates Wnt/β-catenin signaling in cancer stem-like cells and inhibits in vivo the growth of a subcutaneous human osteosarcoma xenograft. Cancer Lett 2018; 414:1-15. [DOI: 10.1016/j.canlet.2017.11.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/31/2017] [Accepted: 11/03/2017] [Indexed: 01/04/2023]
|
31
|
Su L, Li R, Khan S, Clanton R, Zhang F, Lin YN, Song Y, Wang H, Fan J, Hernandez S, Butters AS, Akabani G, MacLoughlin R, Smolen J, Wooley KL. Chemical Design of Both a Glutathione-Sensitive Dimeric Drug Guest and a Glucose-Derived Nanocarrier Host to Achieve Enhanced Osteosarcoma Lung Metastatic Anticancer Selectivity. J Am Chem Soc 2018; 140:1438-1446. [PMID: 29350522 DOI: 10.1021/jacs.7b11462] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Although nanomedicines have been pursued for nearly 20 years, fundamental chemical strategies that seek to optimize both the drug and drug carrier together in a concerted effort remain uncommon yet may be powerful. In this work, two block polymers and one dimeric prodrug molecule were designed to be coassembled into degradable, functional nanocarriers, where the chemistry of each component was defined to accomplish important tasks. The result is a poly(ethylene glycol) (PEG)-protected redox-responsive dimeric paclitaxel (diPTX)-loaded cationic poly(d-glucose carbonate) micelle (diPTX@CPGC). These nanostructures showed tunable sizes and surface charges and displayed controlled PTX drug release profiles in the presence of reducing agents, such as glutathione (GSH) and dithiothreitol (DTT), thereby resulting in significant selectivity for killing cancer cells over healthy cells. Compared to free PTX and diPTX, diPTX@CPGC exhibited improved tumor penetration and significant inhibition of tumor cell growth toward osteosarcoma (OS) lung metastases with minimal side effects both in vitro and in vivo, indicating the promise of diPTX@CPGC as optimized anticancer therapeutic agents for treatment of OS lung metastases.
Collapse
Affiliation(s)
- Lu Su
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Richen Li
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Sarosh Khan
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Ryan Clanton
- Departments of Nuclear Engineering and Veterinary Integrative Biosciences and Texas A&M Institute for Preclinical Studies, Texas A&M University , College Station, Texas 77842, United States
| | - Fuwu Zhang
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Yen-Nan Lin
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States.,College of Medicine, Texas A&M University , Bryan, Texas 77807, United States
| | - Yue Song
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Hai Wang
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Jingwei Fan
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Soleil Hernandez
- Departments of Nuclear Engineering and Veterinary Integrative Biosciences and Texas A&M Institute for Preclinical Studies, Texas A&M University , College Station, Texas 77842, United States
| | - Andrew S Butters
- Departments of Nuclear Engineering and Veterinary Integrative Biosciences and Texas A&M Institute for Preclinical Studies, Texas A&M University , College Station, Texas 77842, United States
| | - Gamal Akabani
- Departments of Nuclear Engineering and Veterinary Integrative Biosciences and Texas A&M Institute for Preclinical Studies, Texas A&M University , College Station, Texas 77842, United States
| | - Ronan MacLoughlin
- Aerogen , IDA Business Park, Dangan, Galway, Ireland .,School of Pharmacy, Royal College of Surgeons , Dublin, Ireland .,School of Pharmacy and Pharmaceutical Sciences, Trinity College , Dublin, Ireland
| | - Justin Smolen
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Karen L Wooley
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| |
Collapse
|
32
|
|
33
|
Hattinger CM, Fanelli M, Tavanti E, Vella S, Riganti C, Picci P, Serra M. Doxorubicin-resistant osteosarcoma: novel therapeutic approaches in sight? Future Oncol 2017; 13:673-677. [PMID: 28183198 DOI: 10.2217/fon-2016-0519] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Claudia M Hattinger
- Pharmacogenomics & Pharmacogenetics Research Unit, Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Marilù Fanelli
- Pharmacogenomics & Pharmacogenetics Research Unit, Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Elisa Tavanti
- Pharmacogenomics & Pharmacogenetics Research Unit, Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Serena Vella
- Pharmacogenomics & Pharmacogenetics Research Unit, Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Chiara Riganti
- Department of Oncology & Center for Experimental Research & Medical Studies (CeRMS), University of Torino, via Santena, 5/bis, 10126, Torino, Italy
| | - Piero Picci
- Pharmacogenomics & Pharmacogenetics Research Unit, Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, via di Barbiano 1/10, 40136 Bologna, Italy
| | - Massimo Serra
- Pharmacogenomics & Pharmacogenetics Research Unit, Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, via di Barbiano 1/10, 40136 Bologna, Italy
| |
Collapse
|
34
|
Zheng Y, Cheng Y, Chen J, Ding J, Li M, Li C, Wang JC, Chen X. Injectable Hydrogel-Microsphere Construct with Sequential Degradation for Locally Synergistic Chemotherapy. ACS APPLIED MATERIALS & INTERFACES 2017; 9:3487-3496. [PMID: 28067493 DOI: 10.1021/acsami.6b15245] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
In recent years, in situ chemotherapy mediated by biodegradable polymer platforms has attracted increased attention. Herein, an advanced drug delivery system, combretastatin A-4 (CA4) and docetaxel (DTX)-loaded microsphere embedded in injectable thermosensitive polypeptide hydrogel (i.e., hydrogel-microsphere (Gel-MP) construct), was reported for sequential release of drugs with different mechanisms to treat osteosarcoma synergistically. The Gel-MP construct showed sequential biodegradability and excellent biocompatibility. CA4 was preferentially released from hydrogel with faster degradation to disturb the vascular structure of the tumor and reduce the exchange of nutrients between the tumor and surrounding tissues, which created interstitial space in the tissue for DTX penetration to inhibit tumor cell proliferation. The in vivo treatment with Gel/CA4-MP/DTX efficiently suppressed the growth of mouse K7 osteosarcoma compared to other formulations. More importantly, by systematical study of histopathology and immunohistochemistry, the Gel-MP construct can significantly upregulate antiproliferation effect and reduce toxicity of drugs. Therefore, this injectable and locally sequential delivery system has a bright prospect in clinical application of in situ synergistic chemotherapy.
Collapse
Affiliation(s)
- Yuhao Zheng
- Department of Orthopedics, Second Hospital of Jilin University , Changchun 130041, People's Republic of China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, People's Republic of China
| | - Yilong Cheng
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, People's Republic of China
| | - Jinjin Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, People's Republic of China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, People's Republic of China
| | - Mingqiang Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, People's Republic of China
| | - Chen Li
- Department of Orthopedics, Second Hospital of Jilin University , Changchun 130041, People's Republic of China
| | - Jin-Cheng Wang
- Department of Orthopedics, Second Hospital of Jilin University , Changchun 130041, People's Republic of China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences , Changchun 130022, People's Republic of China
| |
Collapse
|
35
|
Vella S, Tavanti E, Hattinger CM, Fanelli M, Versteeg R, Koster J, Picci P, Serra M. Targeting CDKs with Roscovitine Increases Sensitivity to DNA Damaging Drugs of Human Osteosarcoma Cells. PLoS One 2016; 11:e0166233. [PMID: 27898692 PMCID: PMC5127503 DOI: 10.1371/journal.pone.0166233] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/25/2016] [Indexed: 01/10/2023] Open
Abstract
Cyclin-dependent kinase 2 (CDK2) has been reported to be essential for cell proliferation in several human tumours and it has been suggested as an appropriate target to be considered in order to enhance the efficacy of treatment regimens based on the use of DNA damaging drugs. We evaluated the clinical impact of CDK2 overexpression on a series of 21 high-grade osteosarcoma (OS) samples profiled by using cDNA microarrays. We also assessed the in vitro efficacy of the CDKs inhibitor roscovitine in a panel of drug-sensitive and drug-resistant human OS cell lines. OS tumour samples showed an inherent overexpression of CDK2, and high expression levels at diagnosis of this kinase appeared to negatively impact on clinical outcome. CDK2 expression also proved to be relevant for in vitro OS cells growth. These findings indicated CDK2 as a promising candidate therapeutic marker for OS and therefore we assessed the efficacy of the CDKs-inhibitor roscovitine in both drug-sensitive and -resistant OS cell lines. All cell lines resulted to be responsive to roscovitine, which was also able to increase the activity of cisplatin and doxorubicin, the two most active DNA damaging drugs used in OS chemotherapy. Our results indicated that combined treatment with conventional OS chemotherapeutic drugs and roscovitine may represent a new candidate intervention approach, which may be considered to enhance tumour cell sensitivity to DNA damaging drugs.
Collapse
Affiliation(s)
- Serena Vella
- Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Italy
| | - Elisa Tavanti
- Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Italy
| | | | - Marilù Fanelli
- Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Italy
| | - Rogier Versteeg
- Department of Human Genetics, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Jan Koster
- Department of Oncogenomics, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Piero Picci
- Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Italy
| | - Massimo Serra
- Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Italy
| |
Collapse
|
36
|
Hattinger CM, Vella S, Tavanti E, Fanelli M, Picci P, Serra M. Pharmacogenomics of second-line drugs used for treatment of unresponsive or relapsed osteosarcoma patients. Pharmacogenomics 2016; 17:2097-2114. [PMID: 27883291 DOI: 10.2217/pgs-2016-0116] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Second-line treatment of high-grade osteosarcoma (HGOS) patients is based on different approaches and chemotherapy protocols, which are not yet standardized. Although several drugs have been used in HGOS second-line protocols, none of them has provided fully satisfactory results and the role of rescue chemotherapy is not well defined yet. This article focuses on the drugs that have most frequently been used for second-line treatment of HGOS, highlighting the present knowledge on their mechanisms of action and resistance and on gene polymorphisms with possible impact on treatment sensitivity or toxicity. In the near future, validation of the so far identified candidate genetic biomarkers may constitute the basis for tailoring treatment by taking the patients' genetic background into account.
Collapse
Affiliation(s)
- Claudia M Hattinger
- Pharmacogenomics & Pharmacogenetics Research Unit of the Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Via di Barbiano 1/10, I-40136 Bologna, Italy
| | - Serena Vella
- Pharmacogenomics & Pharmacogenetics Research Unit of the Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Via di Barbiano 1/10, I-40136 Bologna, Italy
| | - Elisa Tavanti
- Pharmacogenomics & Pharmacogenetics Research Unit of the Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Via di Barbiano 1/10, I-40136 Bologna, Italy
| | - Marilù Fanelli
- Pharmacogenomics & Pharmacogenetics Research Unit of the Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Via di Barbiano 1/10, I-40136 Bologna, Italy
| | - Piero Picci
- Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Via di Barbiano 1/10, I-40136 Bologna, Italy
| | - Massimo Serra
- Pharmacogenomics & Pharmacogenetics Research Unit of the Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Via di Barbiano 1/10, I-40136 Bologna, Italy
| |
Collapse
|
37
|
Liao YY, Tsai HC, Chou PY, Wang SW, Chen HT, Lin YM, Chiang IP, Chang TM, Hsu SK, Chou MC, Tang CH, Fong YC. CCL3 promotes angiogenesis by dysregulation of miR-374b/ VEGF-A axis in human osteosarcoma cells. Oncotarget 2016; 7:4310-25. [PMID: 26713602 PMCID: PMC4826207 DOI: 10.18632/oncotarget.6708] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 12/05/2015] [Indexed: 11/25/2022] Open
Abstract
Osteosarcoma is the most frequent bone tumor, characterized by a high metastatic potential. However, the crosstalk between chemokine (C-C motif) ligand 3 (CCL3), which facilitates tumor progression and metastasis. Vascular endothelial growth factor-A (VEGF-A), an angiogenesis inducer and a highly specific mitogen for endothelial cells, has not been well explored in human osteosarcoma. Here we demonstrate the correlation of CCL3 and VEGF-A expressions, quantified by immunohistochemistry, with the tumor stage of human osteosarcoma tissues. Furthermore, CCL3 promotes VEGF-A expression in human osteosarcoma cells that subsequently induces human endothelial progenitor cell (EPC) migration and tube formation. Phosphorylation of JNK, ERK, and p38 was found after CCL3 stimulation. In addition, JNK, ERK, and p38 inhibitors also abolished CCL3-induced VEGF-A expression and angiogenesis. We noted that CCL3 reduces the expression of miR-374b and miR-374b mimic by reversing CCL3-promoted VEGF-A expression and angiogenesis in vitro and in vivo. This study shows that CCL3 promotes VEGF-A expression and angiogenesis in human osteosarcoma cells by down-regulating miR-374b expression via JNK, ERK, and p38 signaling pathways. Thus, CCL3 may be a new molecular therapeutic target in osteosarcoma angiogenesis and metastasis.
Collapse
Affiliation(s)
- Yuan-Ya Liao
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Hsiao-Chi Tsai
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Pei-Yu Chou
- Department of Nursing, Hung Kuang University, Taichung, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Hsien-Te Chen
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan.,School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Yu-Min Lin
- Department of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Orthopaedics, Taichung Veterans General Hospital, Taichung, Taiwan
| | - I-Ping Chiang
- Department of Pathology, China Medical University Hospital, Taichung, Taiwan
| | - Tzu-Ming Chang
- Department of Orthopedic Surgery, Tungs' Taichung Metroharbor Hospital, Taichung, Taiwan
| | - Shao-Keh Hsu
- Department of Orthopedic Surgery, Tungs' Taichung Metroharbor Hospital, Taichung, Taiwan
| | - Ming-Chih Chou
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| | - Yi-Chin Fong
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan.,Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
| |
Collapse
|
38
|
Sirichativapee W, Wisanuyotin T, Pattanittum P, Paholpak P, Laupattarakasem P, Srisodaphol W, Tsuchiya H, Laopaiboon M, Kosuwon W, Wiangnon S. Chemotherapy for treating high-grade osteosarcoma in children and young adults. Hippokratia 2016. [DOI: 10.1002/14651858.cd012372] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Winai Sirichativapee
- Faculty of Medicine, Khon Kaen University; Orthopedics; 123 Mithraphap rd, Muang District Thailand 40002
| | - Taweechok Wisanuyotin
- Faculty of Medicine, Khon Kaen University; Orthopedics; 123 Mithraphap rd, Muang District Thailand 40002
| | - Porjai Pattanittum
- Khon Kaen University; Department of Biostatistics and Demography, Faculty of Public Health; Mitraparp Road Mueng District Khon Kaen Khon Kaen Thailand 40002
| | - Permsak Paholpak
- Faculty of Medicine, Khon Kaen University; Orthopedics; 123 Mithraphap rd, Muang District Thailand 40002
| | - Pat Laupattarakasem
- Faculty of Medicine, Khon Kaen University; Orthopedics; 123 Mithraphap rd, Muang District Thailand 40002
| | | | - Hiroyuki Tsuchiya
- Graduate School of Medicine, Kanazawa University; Orthopedic Surgery; 13-1 Takara-machi Kanazawa Japan 920-8641
| | - Malinee Laopaiboon
- Khon Kaen University; Department of Epidemiology and Biostatistics, Faculty of Public Health; 123 Mitraparb Road Amphur Muang Khon Kaen Thailand 40002
| | - Weerachai Kosuwon
- Faculty of Medicine, Khon Kaen University; Orthopedics; 123 Mithraphap rd, Muang District Thailand 40002
| | - Surapon Wiangnon
- Faculty of Medicine, Khon Kaen University; Department of Pediatrics; 123 Mithraphap rd, Muang district Thailand 40002
| |
Collapse
|
39
|
Li KT, Chen Q, Wang DW, Duan QQ, Tian S, He JW, Ou YS, Bai DQ. Mitochondrial pathway and endoplasmic reticulum stress participate in the photosensitizing effectiveness of AE-PDT in MG63 cells. Cancer Med 2016; 5:3186-3193. [PMID: 27700017 PMCID: PMC5119974 DOI: 10.1002/cam4.895] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 07/18/2016] [Accepted: 08/08/2016] [Indexed: 01/01/2023] Open
Abstract
Photodynamic therapy (PDT) is a promising treatment in cancer therapy, with a photosensitizer activated by visible light. Aloe‐emodin (AE) is a promising photosensitive agent. In this study, the photosensitizing effects and possible mechanisms of AE‐PDT in MG63 cells were evaluated. The efficiency of AE‐PDT was analyzed by MTT assay. The mode of cell death was investigated by Hoechst 33,342 staining and flow cytometer. The intracellular distribution of AE was detected with confocal microscopy. The formation of reactive oxygen species (ROS) was detected by DCFH‐DA. The mitochondrial membrane potential (MMP) was measured by Rhodamine 123. The expression of proteins including cytochrome c, caspase‐3, ‐9, and ‐12, CHOP and GRP78 was detected by western blot. Apoptosis is the primary mode of cell death in our study, which occurs in a manner of depending on AE concentration and irradiation dose. Confocal microscopy showed that AE was primarily localized on the mitochondria and endoplasmic reticulum (ER) of MG63 cells. AE‐PDT resulted in rapid increases of intracellular ROS production, which reached a peak at 2 h, followed by declining of mitochondrial membrane potential, releasing of cytochrome c from mitochondria into the cytoplasm, and up‐regulation of caspase‐3, ‐9, and ‐12, CHOP and GRP78. These results suggest that death of MG63 cells induced by AE‐PDT is triggered by ROS. Meanwhile, Mitochondria and ER serve as the subcellular targets, which are responsible for AE‐PDT‐induced death of MG63 cells.
Collapse
Affiliation(s)
- Kai-Ting Li
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Chen
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Da-Wu Wang
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qin-Qin Duan
- Department of Gastroenterology, Chinese Medicine Hospital of Longquan, Chengdu, China
| | - Si Tian
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Juan-Wen He
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yun-Sheng Ou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ding-Qun Bai
- Department of Rehabilitation, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
40
|
Gao Y, Liao Y, Shen JK, Feng Y, Choy E, Cote G, Harmon D, Mankin HJ, Hornicek FJ, Duan Z. Evaluation of P-glycoprotein (Pgp) expression in human osteosarcoma by high-throughput tissue microarray. J Orthop Res 2016; 34:1606-12. [PMID: 26790551 DOI: 10.1002/jor.23173] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 01/15/2016] [Indexed: 02/04/2023]
Abstract
Survival of osteosarcoma patients is currently limited by the development of metastases and multidrug resistance (MDR). A well-established cause of MDR involves overexpression of P-glycoprotein (Pgp) in tumor cells. However, some discrepancies still exist as to the clinical significance of Pgp in osteosarcoma. We sought to elucidate further whether the Pgp expression correlated with clinical behavior in a series of patients with osteosarcoma via high-throughput tissue microarray (TMA) analysis. Immunohistochemical analysis of Pgp expression in a TMA of 114 specimens with a retrospective review of 70 osteosarcoma patients admitted to the Massachusetts General Hospital (MGH) was performed. High Pgp expression was correlated with metastasis development and poor response to pre-operative chemotherapy in osteosarcoma patients. Eighteen of the fifty-seven patients initially admitted with primary osteosarcoma showed high Pgp expression. Among these 18 patients with high Pgp expression, 13 of 18 (72%) patients eventually developed metastases. There was no significant clinical relevance between Pgp expression and osteosarcoma survival. These results support that high expression of Pgp is important, but cannot be assigned as, an individual predictor in the development of human osteosarcoma. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1606-1612, 2016.
Collapse
Affiliation(s)
- Yan Gao
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, 02114.,Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan Province, China
| | - Yunfei Liao
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, 02114
| | - Jacson K Shen
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, 02114
| | - Yong Feng
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, 02114
| | - Edwin Choy
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, 02114
| | - Gregory Cote
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, 02114
| | - David Harmon
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, 02114
| | - Henry J Mankin
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, 02114
| | - Francis J Hornicek
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, 02114
| | - Zhenfeng Duan
- Sarcoma Biology Laboratory, Center for Sarcoma and Connective Tissue Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, 02114
| |
Collapse
|
41
|
Serra M, Hattinger CM. The pharmacogenomics of osteosarcoma. THE PHARMACOGENOMICS JOURNAL 2016; 17:11-20. [PMID: 27241064 DOI: 10.1038/tpj.2016.45] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 04/15/2016] [Accepted: 05/04/2016] [Indexed: 12/30/2022]
Abstract
Osteosarcoma (OS), the most common malignant tumor of bone, is presently treated with multidrug neoadjuvant chemotherapy protocols, which allow to cure 60-65% of patients but also induce toxicity events that cannot be predicted or efficiently prevented. The identification and validation of pharmacogenomic biomarkers is, therefore, absolutely warranted to provide the bases for planning personalized treatments with the aim to increase the therapeutic benefits and to avoid or limit unnecessary toxicities. As several targeted therapies against molecular and immunological markers in OS are presently under clinical investigation, it may be speculated that some new agents for innovative treatments may emerge in the next years. However, the real improvement of therapeutic perspectives for OS is strictly connected to the identification of pharmacogenomic biomarkers that may stratify patients in responders or non-responders and identify those individuals with higher susceptibility to treatment-associated toxicity. This review provides an overview of the pharmacogenomic biomarkers identified so far in OS, which appear to be promising candidates for a translation to clinical practice, after further investigation and/or prospective validation.
Collapse
Affiliation(s)
- M Serra
- Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Bologna, Italy
| | - C M Hattinger
- Laboratory of Experimental Oncology, Orthopaedic Rizzoli Institute, Bologna, Italy
| |
Collapse
|
42
|
Lamora A, Mullard M, Amiaud J, Brion R, Heymann D, Redini F, Verrecchia F. Anticancer activity of halofuginone in a preclinical model of osteosarcoma: inhibition of tumor growth and lung metastases. Oncotarget 2016; 6:14413-27. [PMID: 26015407 PMCID: PMC4546476 DOI: 10.18632/oncotarget.3891] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 04/24/2015] [Indexed: 12/11/2022] Open
Abstract
Osteosarcoma is the main malignant primary bone tumor in children and adolescents for whom the prognosis remains poor, especially when metastases are present at diagnosis. Because we recently demonstrated that TGF-β/Smad cascade plays a crucial role in osteosarcoma metastatic progression, we investigated the effect of halofuginone, identified as an inhibitor of the TGF-β/Smad3 cascade, on osteosarcoma progression. A preclinical model of osteosarcoma was used to evaluate the impact of halofuginone on tumor growth, tumor microenvironment and metastasis development. In vivo experiments showed that halofuginone reduces primary tumor growth and lung metastases development. In vitro experiments demonstrated that halofuginone decreases cell viability mainly by its ability to induce caspase-3 dependent cell apoptosis. Moreover, halofuginone inhibits the TGF-β/Smad3 cascade and the response of TGF-β key targets involved in the metastases dissemination process such as MMP-2. In addition, halofuginone treatment affects the “vicious cycle” established between tumor and bone cells, and therefore the tumor-associated bone osteolysis. Together, these results demonstrate that halofuginone decreased primary osteosarcoma development and associated lung metastases by targeting both the tumor cells and the tumor microenvironment. Using halofuginone may be a promising therapeutic strategy against tumor progression of osteosarcoma specifically against lung metastases dissemination.
Collapse
Affiliation(s)
- Audrey Lamora
- INSERM, UMR 957, Equipe Labellisée Ligue contre le Cancer 2012, Nantes, France.,Université de Nantes, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France.,CHU Hôtel Dieu, Nantes, France.,Inserm Liliane Bettencourt School, France
| | - Mathilde Mullard
- INSERM, UMR 957, Equipe Labellisée Ligue contre le Cancer 2012, Nantes, France.,Université de Nantes, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France.,CHU Hôtel Dieu, Nantes, France
| | - Jérôme Amiaud
- INSERM, UMR 957, Equipe Labellisée Ligue contre le Cancer 2012, Nantes, France.,Université de Nantes, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France.,CHU Hôtel Dieu, Nantes, France
| | - Régis Brion
- INSERM, UMR 957, Equipe Labellisée Ligue contre le Cancer 2012, Nantes, France.,Université de Nantes, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France.,CHU Hôtel Dieu, Nantes, France
| | - Dominique Heymann
- INSERM, UMR 957, Equipe Labellisée Ligue contre le Cancer 2012, Nantes, France.,Université de Nantes, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France.,CHU Hôtel Dieu, Nantes, France
| | - Françoise Redini
- INSERM, UMR 957, Equipe Labellisée Ligue contre le Cancer 2012, Nantes, France.,Université de Nantes, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France.,CHU Hôtel Dieu, Nantes, France
| | - Franck Verrecchia
- INSERM, UMR 957, Equipe Labellisée Ligue contre le Cancer 2012, Nantes, France.,Université de Nantes, Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Nantes, France.,CHU Hôtel Dieu, Nantes, France
| |
Collapse
|
43
|
Bao ZQ, Zhang CC, Xiao YZ, Zhou JS, Tao YS, Chai DM. Over-expression of Sox4 and β-catenin is associated with a less favorable prognosis of osteosarcoma. ACTA ACUST UNITED AC 2016; 36:193-199. [PMID: 27072961 DOI: 10.1007/s11596-016-1565-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 10/20/2015] [Indexed: 12/17/2022]
Abstract
The purpose of this study was to examine the association of the expression of Sox4 and β-catenin with the prognosis of osteosarcoma. A total of 108 cases of conventional osteosarcoma were involved in this study and 28 cases of osteochondroma served as controls. The expression of Sox4 and β-catenin was detected by using immunohistochemical staining and Western blotting. The results showed that Sox4 and β-catenin were over-expressed in 67 (62.03%) and 62 (57.41%) of 108 osteosarcoma cases, while in only 3 (10.71%) and 5 (17.86%) of 28 controls, respectively (P<0.05 for all). The expression of Sox4 and β-catenin was associated with the distant metastasis, pathological grade and Enneking stage of patients with osteosarcoma (P<0.05 for all). The mean overall survival time and the 5-year-survival rate in osteosarcoma patients with Sox4 and β-catenin over-expressed were significantly reduced as compared with those in Sox4 and β-catenin low-expression group (P<0.05 for all). Cox multifactor regression analysis revealed that the distant metastasis, Enneking stage, and the expression of Sox4 and β-catenin were independent risk factors of patients with osteosarcoma (P<0.05 for all). The findings indicated that overexpression of Sox4 and β-catenin is associated with a poor prognosis of osteosarcoma.
Collapse
Affiliation(s)
- Zheng-Qi Bao
- Department of Orthopaedics, the First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Chang-Chun Zhang
- Department of Orthopaedics, the First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Yu-Zhou Xiao
- Department of Orthopaedics, the First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Jian-Sheng Zhou
- Department of Orthopaedics, the First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Yi-Sheng Tao
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China
| | - Da-Min Chai
- Department of Pathology, the First Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, China.
| |
Collapse
|
44
|
Meazza C, Scanagatta P. Metastatic osteosarcoma: a challenging multidisciplinary treatment. Expert Rev Anticancer Ther 2016; 16:543-56. [PMID: 26999418 DOI: 10.1586/14737140.2016.1168697] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Osteosarcoma is the most common malignant bone tumor, currently treated with pre-and postoperative chemotherapy in association with the surgical removal of the tumor. About 15-20% of patients have evidence of metastases at diagnosis, mostly in the lungs. Patients with metastatic disease still have a very poor prognosis, with approximately 20-30% of long-term survivors, as compared with 65-70% of patients with localized disease. The optimum management of these patients has not been standardized yet due to several patterns of metastatic disease harboring different prognosis. Complete surgical resection of all sites of disease is mandatory and predictive of survival. Patients with multiple sites of disease not amenable to complete surgery removal should be considered for innovative therapeutic approaches because of poor prognosis.
Collapse
Affiliation(s)
- Cristina Meazza
- a Pediatric Oncology Unit , Fondazione IRCCS Istituto Nazionale dei Tumori , Milano , Italy
| | - Paolo Scanagatta
- b Division of Thoracic Surgery , Fondazione IRCCS Istituto Nazionale dei Tumori , Milano , Italy
| |
Collapse
|
45
|
Cheng S, Zhang X, Huang N, Qiu Q, Jin Y, Jiang D. Down-regulation of S100A9 inhibits osteosarcoma cell growth through inactivating MAPK and NF-κB signaling pathways. BMC Cancer 2016; 16:253. [PMID: 27020242 PMCID: PMC4810516 DOI: 10.1186/s12885-016-2294-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 03/23/2016] [Indexed: 02/05/2023] Open
Abstract
Background Osteosarcoma (OS) is well-known for poor prognosis due to its high incidence of proliferation and metastasis. Researches have provided valuable insights into the tumorigenesis of S100A9 in some cancers. We aimed to understand the expression level, functions and mechanisms of S100A9 in human osteosarcoma for the first time. Methods The expression of S100A9 protein was detected in 120 human osteosarcoma tissues and 40 normal human bone tissues using tissue microarrays analysis. The knockdown of S100A9 induced by RNA interference (RNAi) method in three osteosarcoma cell lines (U2OS, 143B, MG63) was applied to analyze the effects of S100A9 on cell proliferation, cell cycle distribution, migration, invasion and xenotransplanted tumors. Moreover, MAPK-ERK1/2, MAPK-p38, NF-κB-p65, NF-κB-p50, p21, p27, CDK2 and CDK4 were tested. Results The expression of S100A9 was increased in human osteosarcoma issues and was positively correlated with clinical classification and survival rate. Down-regulation of S100A9 inhibited OS cellular proliferation, migration, invasion and cell cycle S phase in vitro and suppressed tumor formation in vivo with the reduction on PCNA and Ki67 proliferation index. Our data also demonstrated that knockdown of S100A9 repressed the protein levels of phospho-ERK1/2, phospho-p50, phospho-p65 except phospho-p38, and prompted up-regulation of p21 and p27 leading to inactivation of cyclin dependent kinase 2(CDK2) and cyclin dependent kinase 4(CDK4). Conclusions S100A9 might be a significant role for predicting osteosarcoma prognosis and down-regulation of S100A9 could be used as a potential target for gene therapy. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2294-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Si Cheng
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Xi Zhang
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Ning Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.,Institute of Life Sciences,Chongqing Medical University, Chongqing, PR China
| | - Quanhe Qiu
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Ying Jin
- Department of Orthodontics, State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, WestChina School of Stomatology, Sichuan University, Chongqing, PR China
| | - Dianming Jiang
- Department of Orthopaedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China.
| |
Collapse
|
46
|
Sane S, Abdullah A, Nelson ME, Wang H, Chauhan SC, Newton SS, Rezvani K. Structural studies of UBXN2A and mortalin interaction and the putative role of silenced UBXN2A in preventing response to chemotherapy. Cell Stress Chaperones 2016; 21:313-26. [PMID: 26634371 PMCID: PMC4786526 DOI: 10.1007/s12192-015-0661-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 11/16/2015] [Accepted: 11/18/2015] [Indexed: 12/26/2022] Open
Abstract
Overexpression of the oncoprotein mortalin in cancer cells and its protein partners enables mortalin to promote multiple oncogenic signaling pathways and effectively antagonize chemotherapy-induced cell death. A UBX-domain-containing protein, UBXN2A, acts as a potential mortalin inhibitor. This current study determines whether UBXN2A effectively binds to and occupies mortalin's binding pocket, resulting in a direct improvement in the tumor's sensitivity to chemotherapy. Molecular modeling of human mortalin's binding pocket and its binding to the SEP domain of UBXN2A followed by yeast two-hybrid and His-tag pull-down assays revealed that three amino acids (PRO442, ILE558, and LYS555) within the substrate-binding domain of mortalin are crucial for UBXN2A binding to mortalin. As revealed by chase experiments in the presence of cycloheximide, overexpression of UBXN2A seems to interfere with the mortalin-CHIP E3 ubiquitin ligase and consequently suppresses the C-terminus of the HSC70-interacting protein (CHIP)-mediated destabilization of p53, resulting in its stabilization in the cytoplasm and upregulation in the nucleus. Overexpression of UBXN2A causes a significant inhibition of cell proliferation and the migration of colon cancer cells. We silenced UBXN2A in the human osteosarcoma U2OS cell line, an enriched mortalin cancer cell, followed by a clinical dosage of the chemotherapeutic agent 5-fluorouracil (5-FU). The UBXN2A knockout U2OS cells revealed that UBXNA is essential for the cytotoxic effect achieved by 5-FU. UBXN2A overexpression markedly increased the apoptotic response of U2OS cells to the 5-FU. In addition, silencing of UBXN2A protein suppresses apoptosis enhanced by UBXN2A overexpression in U2OS. The knowledge gained from this study provides insights into the mechanistic role of UBXN2A as a potent mortalin inhibitor and as a potential chemotherapy sensitizer for clinical application.
Collapse
Affiliation(s)
- Sanam Sane
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, 57069, USA
| | - Ammara Abdullah
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, 57069, USA
| | - Morgan E Nelson
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, 57069, USA
| | - Hongmin Wang
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, 57069, USA
| | - Subhash C Chauhan
- Department of Pharmaceutical Sciences and Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Samuel S Newton
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, 57069, USA
| | - Khosrow Rezvani
- Division of Basic Biomedical Sciences, Sanford School of Medicine, The University of South Dakota, 414 E. Clark Street, Lee Medical Building, Vermillion, SD, 57069, USA.
| |
Collapse
|
47
|
Hattinger CM, Serra M. Role of pharmacogenetics of drug-metabolizing enzymes in treating osteosarcoma. Expert Opin Drug Metab Toxicol 2015; 11:1449-63. [PMID: 26095223 DOI: 10.1517/17425255.2015.1060220] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Drug-metabolizing enzymes (DMEs) biotransform several toxins and xenobiotics in both tumor and normal cells, resulting in either their detoxification or their activation. Since DMEs also metabolize several chemotherapeutic drugs, they can significantly influence tumor response to chemotherapy and susceptibility of normal tissues to collateral toxicity of anticancer treatments. AREAS COVERED This review discusses the pharmacogenetics of DMEs involved in the metabolism of drugs which constitute the backbone of osteosarcoma (OS) chemotherapy, highlighting what is presently known for this tumor and their possible impact on the modulation of future treatment approaches. EXPERT OPINION Achieving further insight into pharmacogenetic markers and biological determinants related to treatment response in OS may ultimately lead to individualized treatment regimens, based on a combination of genotype and tumor characteristics of each patient.
Collapse
Affiliation(s)
- Claudia Maria Hattinger
- a Orthopaedic Rizzoli Institute, Laboratory of Experimental Oncology, Pharmacogenomics and Pharmacogenetics Research Unit , Via di Barbiano 1/10, I-40136 Bologna, Italy +390 516 366 762 ; +390 516 366 763 ;
| | | |
Collapse
|
48
|
CD44 is a direct target of miR-199a-3p and contributes to aggressive progression in osteosarcoma. Sci Rep 2015; 5:11365. [PMID: 26079799 PMCID: PMC4468826 DOI: 10.1038/srep11365] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 05/22/2015] [Indexed: 01/07/2023] Open
Abstract
Osteosarcoma is the most common primary bone malignancy in children and adolescents. Herein, we investigated the role of cluster of differentiation 44 (CD44), a cell-surface glycoprotein involved in cell-cell interactions, cell adhesion, and migration in osteosarcoma. We constructed a human osteosarcoma tissue microarray with 114 patient tumor specimens, including tumor tissues from primary, metastatic, and recurrent stages, and determined the expression of CD44 by immunohistochemistry. Results showed that CD44 was overexpressed in metastatic and recurrent osteosarcoma as compared with primary tumors. Higher expression of CD44 was found in both patients with shorter survival and patients who exhibited unfavorable response to chemotherapy before surgical resection. Additionally, the 3′-untranslated region of CD44 mRNA was the direct target of microRNA-199a-3p (miR-199a-3p). Overexpression of miR-199a-3p significantly inhibited CD44 expression in osteosarcoma cells. miR-199a-3p is one of the most dramatically decreased miRs in osteosarcoma cells and tumor tissues as compared with normal osteoblast cells. Transfection of miR-199a-3p significantly increased the drug sensitivity through down-regulation of CD44 in osteosarcoma cells. Taken together, these results suggest that the CD44-miR-199a-3p axis plays an important role in the development of metastasis, recurrence, and drug resistance of osteosarcoma. Developing strategies to target CD44 may improve the clinical outcome of osteosarcoma.
Collapse
|
49
|
Hattinger CM, Fanelli M, Tavanti E, Vella S, Ferrari S, Picci P, Serra M. Advances in emerging drugs for osteosarcoma. Expert Opin Emerg Drugs 2015; 20:495-514. [PMID: 26021401 DOI: 10.1517/14728214.2015.1051965] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Osteosarcoma (OS), the most common primary malignant bone tumor, is currently treated with pre- and postoperative chemotherapy in association with the surgical removal of the tumor. Conventional treatments allow to cure about 60 - 65% of patients with primary tumors and only 20 - 25% of patients with recurrent disease. New treatment approaches and drugs are therefore highly warranted to improve prognosis. AREAS COVERED This review focuses on the therapeutic approaches that are under development or clinical evaluation in OS. Information was obtained from different and continuously updated data bases, as well as from literature searches, in which particular relevance was given to reports and reviews on new targeted therapies under clinical investigation in high-grade OS. EXPERT OPINION OS is a heterogeneous tumor, with a great variability in treatment response between patients. It is therefore unlikely that a single therapeutic tool will be uniformly successful for all OS patients. This claims for the validation of new treatment approaches together with biologic/(pharmaco)genetic markers, which may select the most appropriate subgroup of patients for each treatment approach. Since some promising novel agents and treatment strategies are currently tested in Phase I/II/III clinical trials, we may hope that new therapies with superior efficacy and safety profiles will be identified in the next few years.
Collapse
|
50
|
Tsai HC, Su HL, Huang CY, Fong YC, Hsu CJ, Tang CH. CTGF increases matrix metalloproteinases expression and subsequently promotes tumor metastasis in human osteosarcoma through down-regulating miR-519d. Oncotarget 2015; 5:3800-12. [PMID: 25003330 PMCID: PMC4116521 DOI: 10.18632/oncotarget.1998] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Osteosarcoma, the most common primary malignant bone tumor, shows potent capacity for local invasion and distant metastasis. Connective tissue growth factor (CTGF/CCN2), a secreted protein, binds to integrins, modulates invasive behavior of certain human cancer cells. Effect of CTGF in metastasis of human osteosarcoma is unknown. We found overexpression of CTGF increasing matrix metalloproteinases (MMPs)-2 and MMP-3 expression as well as promoting cell migration. MicroRNA (miRNA) analysis of CTGF-overexpressed osteosarcoma versus control cells probed mechanisms of CTGF-mediated promotion of migration. Among miRNAs regulated by CTGF, miR-519d was most downregulated after CTGF treatment. Co-transfection with miR-519d mimic reversed CTGF-mediated MMPs expression and cell migration. Also, MEK and ERK inhibitors or mutants reduced CTGF-increased cell migration and miR-519d suppression. By contrast, knockdown of CTGF diminished lung metastasis in vivo. Clinical samples indicate CTGF expression as linked with clinical stage and tumor metastasis. Taken together, data show CTGF elevating MMPs expression and subsequently promoting tumor metastasis in human osteosarcoma, down-regulating miR-519d via MEK and ERK pathways, making CTGF a new molecular therapeutic target in osteosarcoma metastasis.
Collapse
Affiliation(s)
- Hsiao-Chi Tsai
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | | | | | | | | | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan; Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| |
Collapse
|