1
|
Chen Y, Wu L, Li Y, Zheng J, Zhong S, Gu S, Chen J. Necrotizing apoptosis-related genes prognosis and treatment effect analysis of osteosarcoma in children. J Gene Med 2024; 26:e3646. [PMID: 38100138 DOI: 10.1002/jgm.3646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/31/2023] [Accepted: 11/13/2023] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Immune cell homeostasis plays a crucial role in cancer research and therapeutic response. While chemotherapy and immunotherapy hold promise in treating osteosarcoma (OS), identifying patients who are likely to respond would significantly improve clinical practices. Necroptosis, a fundamental mechanism mediating chemotherapy and immunotherapy efficacy, offers valuable insights. In this context, subtypes based on necroptosis-related genes have been established to predict the response of OS patients to immunotherapy and chemotherapy. METHODS We conducted a high-throughput screening test to identify necroptosis-associated genes that regulate the development of osteosarcoma. Subsequently, the ConsensusClusterPlus package was employed to classify OS patients into subtypes, enabling comparisons of prognosis and clinical information between these subtypes. Patients from the TARGET-OS and GSE21257 datasets were stratified into high-risk and low-risk groups, and their prognoses were compared. Additionally, we assessed the accuracy of the Risk Scoring Model in predicting prognosis, identified independent prognostic factors and explored potential chemotherapeutic agents and immunotherapy drugs. RESULTS Through the intersection of expression profiles from the TARGET-OS and GSE21257 datasets, we have identified a total of 92 genes associated with necroptosis. Based on differences in the expression of these genes, patients were divided into three subtypes, and we investigated the differences in tumor-infiltrating immune cells, immune-related pathways, and prognosis among these subtypes. Our nomogram effectively differentiated subtypes with distinct responses to chemotherapy and immunotherapy. The established signature demonstrated superior prediction ability compared with single clinical indicators. CONCLUSIONS This pioneering study unveils the prognostic role of necroptosis-related genes in OS patients, providing a promising alternative for prognostic prediction in clinical disease management. Moreover, our findings highlight the significance of immune cell homeostasis in cancer research and therapeutic response, underscoring its relevance in advancing current treatment strategies.
Collapse
Affiliation(s)
| | - Ling Wu
- Ningbo Women and Children's Hospital, Ningbo, China
| | - Yunyan Li
- Ningbo Women and Children's Hospital, Ningbo, China
| | - Jika Zheng
- Ningbo Women and Children's Hospital, Ningbo, China
| | | | - Shirong Gu
- Department of Orthopaedics, LiHuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Jingyang Chen
- Department of Orthopaedics, LiHuili Hospital Affiliated to Ningbo University, Ningbo, China
| |
Collapse
|
2
|
Zare A, Izanloo S, Khaledi S, Maratovich MN, Kaliyev AA, Abenova NA, Rahmanifar F, Mahdipour M, Bakhshalizadeh S, Shirazi R, Tanideh N, Tamadon A. A Bibliometric and In Silico-Based Analysis of Anti-Lung Cancer Compounds from Sea Cucumber. Mar Drugs 2023; 21:md21050283. [PMID: 37233477 DOI: 10.3390/md21050283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/18/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Lung cancer is one of the most lethal malignancies in the world. However, current curative approaches for treating this type of cancer have some weaknesses. Therefore, scientists are attempting to discover new anti-lung cancer agents. Sea cucumber is a marine-derived source for discovering biologically active compounds with anti-lung cancer properties. To explore the anti-lung cancer properties of sea cucumber, we analyzed surveys using VOSviewer software and identified the most frequently used keywords. We then searched the Google Scholar database for compounds with anti-lung cancer properties within that keyword family. Finally, we used AutoDock 4 to identify the compounds with the highest affinity for apoptotic receptors in lung cancer cells. The results showed that triterpene glucosides were the most frequently identified compounds in studies examining the anti-cancer properties of sea cucumbers. Intercedenside C, Scabraside A, and Scabraside B were the three triterpene glycosides with the highest affinity for apoptotic receptors in lung cancer cells. To the best of our knowledge, this is the first time that anti-lung cancer properties of sea cucumber-derived compounds have been examined in in silico conditions. Ultimately, these three components displayed anti-lung cancer properties in in silico conditions and may be used for the manufacture of anti-lung cancer agents in the near future.
Collapse
Affiliation(s)
- Afshin Zare
- The PerciaVista Biotechnology Company, Shiraz 71676-83745, Iran
| | - Safoura Izanloo
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
- School of Nursing, North Khorasan University of Medical Sciences, Bojnurd 94149-74877, Iran
| | - Sajed Khaledi
- Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran 14117-13116, Iran
| | | | | | - Nurgul Abdullayevna Abenova
- Department of Internal Diseases, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan
| | - Farhad Rahmanifar
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz 71348-14336, Iran
| | - Mahdi Mahdipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz 51666-53431, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 51666-53431, Iran
| | - Shabnam Bakhshalizadeh
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Reza Shirazi
- Department of Anatomy, School of Medical Sciences, Biomedical & Health, UNSW Sydney, Sydney, NSW 1466, Australia
| | - Nader Tanideh
- The PerciaVista Biotechnology Company, Shiraz 71676-83745, Iran
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
- Department of Pharmacology, Medical School, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Amin Tamadon
- The PerciaVista Biotechnology Company, Shiraz 71676-83745, Iran
- Department for Scientific Work, West Kazakhstan Marat Ospanov Medical University, Aktobe 030010, Kazakhstan
| |
Collapse
|
3
|
Abstract
Osteosarcoma is the most common primary bone malignancy in adolescents. Its high propensity to metastasize is the leading cause for treatment failure and poor prognosis. Although the research of osteosarcoma has greatly expanded in the past decades, the knowledge and new therapy strategies targeting metastatic progression remain sparse. The prognosis of patients with metastasis is still unsatisfactory. There is resonating urgency for a thorough and deeper understanding of molecular mechanisms underlying osteosarcoma to develop innovative therapies targeting metastasis. Toward the goal of elaborating the characteristics and biological behavior of metastatic osteosarcoma, it is essential to combine the diverse investigations that are performed at molecular, cellular, and animal levels from basic research to clinical translation spanning chemical, physical sciences, and biology. This review focuses on the metastatic process, regulatory networks involving key molecules and signaling pathways, the role of microenvironment, osteoclast, angiogenesis, metabolism, immunity, and noncoding RNAs in osteosarcoma metastasis. The aim of this review is to provide an overview of current research advances, with the hope to discovery druggable targets and promising therapy strategies for osteosarcoma metastasis and thus to overcome this clinical impasse.
Collapse
Affiliation(s)
- Gaohong Sheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Gao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Guo HM, Zhao ZF, Che YQ, Fan JH. Induction of Apoptosis in the Human HepG2 Hepatoma Cell Line Through the Extrinsic Pathway by Delphinium Alkaloid A. INT J PHARMACOL 2021. [DOI: 10.3923/ijp.2021.562.571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
5
|
Wang S, Wei H, Huang Z, Wang X, Shen R, Wu Z, Lin J. Epidermal growth factor receptor promotes tumor progression and contributes to gemcitabine resistance in osteosarcoma. Acta Biochim Biophys Sin (Shanghai) 2021; 53:317-324. [PMID: 33432347 DOI: 10.1093/abbs/gmaa177] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Indexed: 12/18/2022] Open
Abstract
Osteosarcoma (OS) is the most common type of primary malignant tumors that originate in the bone. Resistance to chemotherapy confers a poor prognosis on OS patients. Dysregulation of the epidermal growth factor receptor (EGFR) signaling has been reported in sarcomas. However, the functional contribution of EGFR hyperactivation to the tumor biology and chemoresistance remains largely unexplored in OS. In this study, we aimed to investigate the role of EGFR in OS progression and in the response of OS to gemcitabine treatment. The EGFR expression was found to be upregulated in fibroblastic OS cell lines. EGFR knockdown suppressed OS cell proliferation, migration, and invasion in vitro and tumor formation in vivo. Conversely, EGFR overexpression promoted the growth and motility of OS cells. In terms of mechanism, the levels of phospho-Akt and phospho-ERK were decreased upon EGFR knockdown but increased as a result of EGFR overexpression, implying a possible involvement of PI3K/Akt and ERK pathways in mediating the effects of EGFR on OS cells. Moreover, the level of phospho-EGFR was increased in OS cells when exposed to gemcitabine treatment. A more profound proliferative inhibition and a higher rate of apoptosis were obtained in OS cells via inducing cell cycle arrest at G1 phase upon gemcitabine treatment combined with EGFR knockdown, as compared to gemcitabine alone. On the contrary, EGFR overexpression counteracted the growth-inhibiting and pro-apoptotic effects of gemcitabine in OS cells. The present study suggests that EGFR promotes tumor progression and contributes to gemcitabine resistance in OS.
Collapse
Affiliation(s)
- Shenglin Wang
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Hongxiang Wei
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Zhen Huang
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Xinwen Wang
- Department of Orthopedics, The People’s Hospital of Jiangmen City, Jiangmen 529051, China
| | - Rongkai Shen
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Zhaoyang Wu
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Jianhua Lin
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| |
Collapse
|
6
|
Xu M, Wang Z, Yu XC, Lin JH, Hu YC. Guideline for Limb-Salvage Treatment of Osteosarcoma. Orthop Surg 2020; 12:1021-1029. [PMID: 32633103 PMCID: PMC7454155 DOI: 10.1111/os.12702] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 04/20/2020] [Indexed: 12/26/2022] Open
Abstract
Osteosarcoma is the most common primary malignant bone tumor, occurring mainly in children and adolescents, and the limbs are the main affected sites. At present, limb‐salvage treatment is considered as an effective basic standard treatment for osteosarcoma of the limb. China has a vast territory, but the development of technology is not balanced,which requires sufficient theoretical coverage, strong technical guidance and the application of limb‐salvage treatment guidelines to the treatment of osteosarcoma. Therefore, to standardize and promote the development of limb‐salvage surgery technology and improve the success rate of limb‐salvage treatment, this guide systematically introduces limb‐salvage techniques for the treatment of patients with limb osteosarcoma through definition of limb‐salvage treatment, surgical methods, efficacy evaluation, postoperative treatment and prevention of complications, rehabilitation guidance, and follow‐up advice.
Collapse
Affiliation(s)
- Ming Xu
- Department of Orthopedics, The 960th Hospital of PLA, Jinan, China
| | - Zhen Wang
- Department of Orthopedics, Xi-jing Hospital, Air Force Military Medical University, Xi'an, China
| | - Xiu-Chun Yu
- Department of Orthopedics, The 960th Hospital of PLA, Jinan, China
| | - Jian-Hua Lin
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yong-Cheng Hu
- Department of Bone Oncology, Tianjin hospital, Tianjin, China
| |
Collapse
|
7
|
Kiany S, Harrison D, Gordon N. The Histone Deacetylase Inhibitor Entinostat/Syndax 275 in Osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1257:75-83. [PMID: 32483732 DOI: 10.1007/978-3-030-43032-0_7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The prognosis for metastatic osteosarcoma (OS) is poor and has not changed in several decades. Therapeutic paradigms that target and exploit novel molecular pathways are desperately needed. Recent preclinical data suggests that modulation of the Fas/FasL pathway may offer benefit in the treatment of refractory osteosarcoma. Fas and FasL are complimentary receptor-ligand proteins. Fas is expressed in multiple tissues, whereas FasL is restricted to privilege organs, such as the lung. Fas expression has been shown to inversely correlate with the metastatic potential of OS cells; tumor cells which express high levels of Fas have decreased metastatic potential and the ones that reach the lung undergo cell death upon interaction with constitutive FasL in the lung. Agents such as gemcitabine and the HDAC inhibitor, entinostat/Syndax 275, have been shown to upregulate Fas expression on OS cells, potentially leading to decreased OS pulmonary metastasis and improved outcome. Clinical trials are in development to evaluate this combination as a potential treatment option for patients with refractory OS.
Collapse
Affiliation(s)
- Simin Kiany
- Department of Pediatrics Research, MD Anderson Cancer Center, Houston, TX, USA
| | - Douglas Harrison
- Department of Pediatrics - Patient Care, MD Anderson Cancer Center, Houston, TX, USA
| | - Nancy Gordon
- Department of Pediatrics Research, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
8
|
Aerosolized Chemotherapy for Osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1257:67-73. [DOI: 10.1007/978-3-030-43032-0_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
9
|
The Fas/FasL Signaling Pathway: Its Role in the Metastatic Process and as a Target for Treating Osteosarcoma Lung Metastases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1258:177-187. [PMID: 32767242 DOI: 10.1007/978-3-030-43085-6_12] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Understanding how the tumor microenvironment participates in inhibiting or supporting tumor growth is critical for the development of novel therapies. Osteosarcoma (OS) metastasizes almost exclusively to the lung, an organ where Fas ligand (FasL) is constitutively expressed. This chapter focuses on our studies dedicated to the interaction of OS cells with the lung microenvironment. We will summarize our studies conducted over the past 20 years showing the importance of the Fas/FasL signaling pathway to the establishment and progression of OS metastases in the lung. We demonstrated that the FasL+ lung microenvironment eliminates Fas-positive (Fas+) OS cells that metastasize to the lungs, through apoptosis induced by Fas signaling following interaction of Fas on the tumor cell surface with FasL on the lung epithelial cells. Expression of the Fas receptor on OS cells inversely correlated with the ability of OS cells to form lung metastases. Blocking this pathway interferes with this process, allowing Fas+ cells to grow in the lung. By contrast, upregulation of Fas on Fas- OS cells inhibited their ability to metastasize to the lung. We demonstrated how the FasL+ lung microenvironment can be leveraged for therapeutic intent through the upregulation of Fas expression. To this end, we demonstrated that the histone deacetylase inhibitor entinostat upregulated Fas expression on OS cells, reduced their ability to form lung metastases, and induced regression of established micrometastases. Fas expression in OS cells is regulated epigenetically by the microRNA miR-20a. We showed that expressions of Fas and miR-20a are inversely correlated, and that delivery of anti-miR-20a in vivo to mice with established osteosarcoma lung metastases resulted in upregulation of Fas and tumor regression. Therefore, targeting the Fas signaling pathway may present therapeutic opportunities, which target the lung microenvironment for elimination of OS lung metastases. We have also shown that in addition to being critically involved in the metastatic potential, the Fas signaling pathway may also contribute to the efficacy of chemotherapy. We demonstrated that the chemotherapeutic agent gemcitabine (GCB) increased Fas expression in both human and mouse OS cells in vitro. In vivo, aerosol GCB therapy induced upregulation of Fas expression and the regression of established osteosarcoma lung metastases. The therapeutic efficacy of GCB was contingent upon a FasL+ lung microenvironment as aerosol GCB had no effect in FasL-deficient mice. Manipulation of Fas expression and the Fas pathway should be considered, as this concept may provide additional novel therapeutic approaches for treating patients with OS lung metastases.
Collapse
|
10
|
Gao T, Zhao P, Yu X, Cao S, Zhang B, Dai M. Use of Saikosaponin D and JNK inhibitor SP600125, alone or in combination, inhibits malignant properties of human osteosarcoma U2 cells. Am J Transl Res 2019; 11:2070-2080. [PMID: 31105818 PMCID: PMC6511790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 03/13/2019] [Indexed: 06/09/2023]
Abstract
Saikosaponin D (Ssd) is a major active ingredient derived from the traditional Chinese medicinal herb Bupleurum falcatum, and SP600125 is a specific inhibitor of JNK that competes with adenosine triphosphate. In this study, we co-analyzed cell proliferation, apoptosis, migration, and invasion in U-2OS osteosarcoma cells treated with Ssd and SP600125 alone or in combination. Cell death and signaling were analyzed using western blotting and flow cytometry. We observed dramatic inhibition of cellular proliferation, invasion, and migration in cells treated with Ssd alone or in combination with SP600125. Ssd, alone or in combination with SP600125, enhanced Cytochrome C release, increased the Bax/Bcl-2 ratio, and activated caspase-3, -8 and -9, indicating that cellular apoptosis was induced via both the mitochondrial and death receptor pathways. The effect of SP600125 alone on U2 cells was not significant. Additional evaluation of Mcl-1, Akt, p-Akt, ERK, and p-ERK supported an anti-tumor effect of Ssd, which was enhanced in combination with SP600125. This study provides a theoretical basis for the treatment of osteosarcoma with Ssd alone or in combination with SP600125.
Collapse
Affiliation(s)
- Tian Gao
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang UniversityNanchang 330006, P. R. China
| | - Ping Zhao
- Medical School of Nanchang UniversityNanchang 330006, P. R. China
| | - Xiaolong Yu
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang UniversityNanchang 330006, P. R. China
| | - Suixia Cao
- Medical School of Nanchang UniversityNanchang 330006, P. R. China
| | - Bin Zhang
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang UniversityNanchang 330006, P. R. China
| | - Min Dai
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang UniversityNanchang 330006, P. R. China
| |
Collapse
|
11
|
Gravett AM, Dalgleish AG, Copier J. In vitro culture with gemcitabine augments death receptor and NKG2D ligand expression on tumour cells. Sci Rep 2019; 9:1544. [PMID: 30733494 PMCID: PMC6367314 DOI: 10.1038/s41598-018-38190-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 12/06/2018] [Indexed: 01/11/2023] Open
Abstract
Much effort has been made to try to understand the relationship between chemotherapeutic treatment of cancer and the immune system. Whereas much of that focus has been on the direct effect of chemotherapy drugs on immune cells and the release of antigens and danger signals by malignant cells killed by chemotherapy, the effect of chemotherapy on cells surviving treatment has often been overlooked. In the present study, tumour cell lines: A549 (lung), HCT116 (colon) and MCF-7 (breast), were treated with various concentrations of the chemotherapeutic drugs cyclophosphamide, gemcitabine (GEM) and oxaliplatin (OXP) for 24 hours in vitro. In line with other reports, GEM and OXP upregulated expression of the death receptor CD95 (fas) on live cells even at sub-cytotoxic concentrations. Further investigation revealed that the increase in CD95 in response to GEM sensitised the cells to fas ligand treatment, was associated with increased phosphorylation of stress activated protein kinase/c-Jun N-terminal kinase and that other death receptors and activatory immune receptors were co-ordinately upregulated with CD95 in certain cell lines. The upregulation of death receptors and NKG2D ligands together on cells after chemotherapy suggest that although the cells have survived preliminary treatment with chemotherapy they may now be more susceptible to immune cell-mediated challenge. This re-enforces the idea that chemotherapy-immunotherapy combinations may be useful clinically and has implications for the make-up and scheduling of such treatments.
Collapse
Affiliation(s)
- Andrew M Gravett
- Oncology Group, Institute for Infection and Immunity, St. George's, University of London, London, UK.
| | - Angus G Dalgleish
- Oncology Group, Institute for Infection and Immunity, St. George's, University of London, London, UK
| | - John Copier
- Oncology Group, Institute for Infection and Immunity, St. George's, University of London, London, UK
| |
Collapse
|
12
|
Wang Q, Yu X, Li F, Lv X, Fu X, Gu H, Liu H, Liu J, Dai M, Zhang B. Efficacy of celastrol combined with cisplatin in enhancing the apoptosis of U-2OS osteosarcoma cells via the mitochondrial and endoplasmic reticulum pathways of apoptosis. Oncol Lett 2019; 17:3305-3313. [PMID: 30867764 PMCID: PMC6396172 DOI: 10.3892/ol.2019.10007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 09/04/2017] [Indexed: 11/11/2022] Open
Abstract
Osteosarcoma is a common primary malignant tumor of bone, and the poor prognosis and low 5-year survival rate have not improved for three decades. The present study aimed to study the effect a combination of celastrol and cisplatin on the human osteosarcoma cell line U-2OS, and to investigate the mechanism by which celastrol/cisplatin induces the apoptosis of osteosarcoma cells. MTT and Annexin V-FITC/PI assays were used to evaluate the effects of combined celastrol/cisplatin on growth and apoptosis, respectively, in U-2OS cells. Morphological changes accompanying cell growth inhibition were observed using a fluorescence microscope. Combination index (CI) analysis was used to evaluate the combinatorial effects of celastrol/cisplatin treatment. Western blotting was used to quantify the expression of apoptosis-associated proteins. It was identified that celastrol/cisplatin inhibited the growth of U-2OS cells in a dose-dependent manner. CI analysis revealed that combined celastrol/cisplatin demonstrated a synergistic effect in U-2OS cells, with CIs ranging from 0.80 to 0.97 at effect levels from IC10 to IC70. In addition, it was observed that celastrol/cisplatin upregulated the expression of Bcl-associated X protein, cytochrome c, caspase-3 and C/EBP homologous protein, and downregulated the expression of Bcl-2, poly(ADP-ribose) polymerase, 78 kDa glucose-regulated protein and caspase-9, whereas the expression of caspase-8 remained unchanged. To conclude, celastrol/cisplatin induced apoptosis in U-2OS cells via the mitochondrial and endoplasmic reticulum pathways, particularly in the former. Celastrol/cisplatin therefore exhibits potential as a novel therapeutic combination for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Orthopedics, Fujian Longyan First Hospital, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, Fujian 364000, P.R. China
| | - Xiaolong Yu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Fan Li
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xin Lv
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaoxing Fu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Houyun Gu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hucheng Liu
- Multidisciplinary Therapy Center of Musculoskeletal Tumors, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Jun Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Min Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Bin Zhang
- Artificial Joint Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
13
|
Lee WH, Loo CY, Ghadiri M, Leong CR, Young PM, Traini D. The potential to treat lung cancer via inhalation of repurposed drugs. Adv Drug Deliv Rev 2018; 133:107-130. [PMID: 30189271 DOI: 10.1016/j.addr.2018.08.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 08/27/2018] [Accepted: 08/31/2018] [Indexed: 01/10/2023]
Abstract
Lung cancer is a highly invasive and prevalent disease with ineffective first-line treatment and remains the leading cause of cancer death in men and women. Despite the improvements in diagnosis and therapy, the prognosis and outcome of lung cancer patients is still poor. This could be associated with the lack of effective first-line oncology drugs, formation of resistant tumors and non-optimal administration route. Therefore, the repurposing of existing drugs currently used for different indications and the introduction of a different method of drug administration could be investigated as an alternative to improve lung cancer therapy. This review describes the rationale and development of repositioning of drugs for lung cancer treatment with emphasis on inhalation. The review includes the current progress of repurposing non-cancer drugs, as well as current chemotherapeutics for lung malignancies via inhalation. Several potential non-cancer drugs such as statins, itraconazole and clarithromycin, that have demonstrated preclinical anti-cancer activity, are also presented. Furthermore, the potential challenges and limitations that might hamper the clinical translation of repurposed oncology drugs are described.
Collapse
Affiliation(s)
- Wing-Hin Lee
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur (RCMP UniKL), Ipoh, Perak, Malaysia; Respiratory Technology, Woolcock Institute of Medical Research, and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, NSW 2037, Australia; Centre for Lung Cancer Research, 431 Glebe Point Road, 2037, Australia.
| | - Ching-Yee Loo
- Faculty of Pharmacy and Health Sciences, Royal College of Medicine Perak, Universiti Kuala Lumpur (RCMP UniKL), Ipoh, Perak, Malaysia; Respiratory Technology, Woolcock Institute of Medical Research, and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, NSW 2037, Australia; Centre for Lung Cancer Research, 431 Glebe Point Road, 2037, Australia
| | - Maliheh Ghadiri
- Respiratory Technology, Woolcock Institute of Medical Research, and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, NSW 2037, Australia; Centre for Lung Cancer Research, 431 Glebe Point Road, 2037, Australia
| | - Chean-Ring Leong
- Section of Bioengineering Technology, Universiti Kuala Lumpur (UniKL) MICET, Alor Gajah, Melaka, Malaysia
| | - Paul M Young
- Respiratory Technology, Woolcock Institute of Medical Research, and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, NSW 2037, Australia; Centre for Lung Cancer Research, 431 Glebe Point Road, 2037, Australia
| | - Daniela Traini
- Respiratory Technology, Woolcock Institute of Medical Research, and Discipline of Pharmacology, Faculty of Medicine and Health, The University of Sydney, NSW 2037, Australia; Centre for Lung Cancer Research, 431 Glebe Point Road, 2037, Australia
| |
Collapse
|
14
|
Wang Y, Wang S, Liu J, Lu Y, Li D. Licoricidin enhances gemcitabine-induced cytotoxicity in osteosarcoma cells by suppressing the Akt and NF-κB signal pathways. Chem Biol Interact 2018; 290:44-51. [PMID: 29782821 DOI: 10.1016/j.cbi.2018.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 04/13/2018] [Accepted: 05/17/2018] [Indexed: 10/16/2022]
Abstract
Osteosarcoma (OS) is the most common bone malignancy in children and adolescents. Combined treatments of anti-cancer drugs can remarkably improve chemotherapeutic outcomes. Gemcitabine and licoricidin both have potential anti-tumor activity in several cancers. However, the combined therapeutic efficiency of gemcitabine and licoricidin for OS has not been explored. Here, we found that licoricidin or gemcitabine inhibited OS cell viability in a dose-dependent manner. Cotreatment with licoricidin and gemcitabine enhanced gemcitabine-induced cytotoxicity in OS cells. Licoricidin suppressed activation of the Akt and nuclear factor-kappa B (NF-κB) pathways. Gemcitabine had no effect on Akt signal, but facilitated the activation of NF-κB signal in OS cells. Moreover, combined treatment of licoricidin and gemcitabine markedly curbed the activation of Akt and NF-κB pathways in OS cells. Inhibition of the Akt and NF-κB pathways enhanced gemcitabine-induced cytotoxicity in OS cells. In vivo assay further manifested that licoricidin enhanced gemcitabine-induced cytotoxicity in tumor xenograft models of OS via inactivation of the Akt and NF-κB pathways. In conclusion, licoricidin enhanced gemcitabine-induced cytotoxicity in OS cells by inactivation of the Akt and NF-κB pathways in vitro and in vivo.
Collapse
Affiliation(s)
- Yifei Wang
- Department of Orthopedics, The First Affiliated Hospital of Henan University, Kaifeng 475001, China
| | - Shengli Wang
- Department of Orthopedics, The First Affiliated Hospital of Henan University, Kaifeng 475001, China.
| | - Jianhua Liu
- Department of Orthopedics, The First Affiliated Hospital of Henan University, Kaifeng 475001, China
| | - Yanxiao Lu
- Department of Orthopedics, The First Affiliated Hospital of Henan University, Kaifeng 475001, China
| | - Donghui Li
- Department of Orthopedics, The First Affiliated Hospital of Henan University, Kaifeng 475001, China
| |
Collapse
|
15
|
Aerosol Gemcitabine after Amputation Inhibits Osteosarcoma Lung Metastases but Not Wound Healing. Sarcoma 2018; 2018:3143096. [PMID: 29610563 PMCID: PMC5828535 DOI: 10.1155/2018/3143096] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/25/2017] [Indexed: 01/29/2023] Open
Abstract
Background In newly diagnosed osteosarcoma (OS) patients, the time between surgery and resumption of chemotherapy is 2-7 weeks. Delays > 16 days are associated with increased risk of relapse and decreased overall survival. Identifying an effective therapy that can be used postoperatively may prevent relapse. We investigated whether aerosol gemcitabine (GCB) initiated after tumor resection inhibited the growth of OS lung metastases without affecting the wound-healing process. Methods Mice were injected intratibially with OS cells. Amputation was performed when the tumor reached 1.5 cm. Full-thickness excisional wounds were also made on the dorsal skin and tail. Aerosol GCB or PBS was initiated 48 hours after amputation (3 times/week for 3 weeks). Wound sections were evaluated by immunohistochemistry for Ki-67 (proliferation), CD31 (vessels), VEGF, IL-10, bFGF, mast cells, macrophages, and M1/M2 macrophage ratios. The lungs were analyzed for macro- and micrometastases. Results Aerosol GCB inhibited the growth of the lung metastases but had no effect on the 3 phases of wound healing in the dorsal skin, tail, or bone. Production of cytokines at the wound sites was the same. Conclusion These data indicate that initiating aerosol GCB postoperatively may kill residual lung metastases thereby preventing relapse and improve survival.
Collapse
|
16
|
Mayr L, Pirker C, Lötsch D, Van Schoonhoven S, Windhager R, Englinger B, Berger W, Kubista B. CD44 drives aggressiveness and chemoresistance of a metastatic human osteosarcoma xenograft model. Oncotarget 2017; 8:114095-114108. [PMID: 29371972 PMCID: PMC5768389 DOI: 10.18632/oncotarget.23125] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 11/28/2017] [Indexed: 12/15/2022] Open
Abstract
Background Osteosarcoma is the most common primary malignant bone tumor with a 5 year survival rate of up to 70%. However, patients with metastatic disease have still a very poor prognosis. Osteosarcoma metastasis models are essential to develop novel treatment strategies for advanced disease. Methods Based on a serial transplantation approach, we have established a U-2 OS osteosarcoma xenograft model with increased metastatic potential and compared it to other metastatic osteosarcoma models from international sources. Subclones with differing invasive potential were compared for genomic gains and losses as well as gene expression changes by several bioinformatic approaches. Based on the acquired results, the effects of a shRNA-mediated CD44 mRNA knockdown on migration, invasion and chemosensitivity were evaluated. Results The CD44 gene was part of an amplified region at chromosome 11p found in both U-2 OS subclones with enhanced metastatic potential but not in parental U-2 OS cells, corresponding with distinct CD44 overexpression. Accordingly, shRNA-mediated CD44 knockdown significantly attenuated osteosarcoma cell migration, invasion, and viability especially in the metastatic subclones of U-2 OS and Saos-2 cells. Metastatic subclones generally were hypersensitive against the integrin inhibitor cilengitide paralleled by alterations in integrin expression pattern following CD44 knock-down. Additionally, attenuation of CD44 expression sensitized these cell models against osteosarcoma chemotherapy with doxorubicin but not methotrexate and cisplatin. Conclusions The osteosarcoma xenograft models with increased metastatic potential developed in this study can be useful for identification of mechanisms driving metastasis and resistance towards clinically used and novel therapeutic regimens.
Collapse
Affiliation(s)
- Lisa Mayr
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, 1090 Vienna, Austria
| | - Christine Pirker
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, 1090 Vienna, Austria
| | - Daniela Lötsch
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, 1090 Vienna, Austria
| | - Sushilla Van Schoonhoven
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, 1090 Vienna, Austria
| | - Reinhard Windhager
- Department of Orthopaedics, Medical University Vienna, 1090 Vienna, Austria
| | - Bernhard Englinger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, 1090 Vienna, Austria
| | - Walter Berger
- Institute of Cancer Research and Comprehensive Cancer Center, Department of Medicine I, Medical University Vienna, 1090 Vienna, Austria
| | - Bernd Kubista
- Department of Orthopaedics, Medical University Vienna, 1090 Vienna, Austria
| |
Collapse
|
17
|
Ye C, Yu X, Zeng J, Dai M, Zhang B. Effects of baicalein on proliferation, apoptosis, migration and invasion of Ewing's sarcoma cells. Int J Oncol 2017; 51:1785-1792. [PMID: 29039470 DOI: 10.3892/ijo.2017.4148] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Accepted: 09/25/2017] [Indexed: 11/06/2022] Open
Abstract
Ewing's sarcoma (ES) is a rare tumor that is more frequent in pediatric and adolescent age groups. In the past few decades, long-term survival in affected patients has improved due to the success of multimodal therapy. However, long-term survival is inevitably restricted by the late side-effects of chemotherapy. Besides, early metastasis also contributes to the poor prognosis of ES. Recently, traditional Chinese medicines (TCMs) have increasingly attracted interest due to the promising clinical results and fewer side-effects for the treatment of cancers. Among the various TCMs, the root of Scutellaria baicalensis exerts anti-inflammatory properties as a well-known herb in traditional Chinese medicine. Baicalein (5,6,7-trihydroxyflavone) derived from the root of Scutellaria baicalensis is a bioactive compound, which possesses a powerful pro-apoptotic activity in various cancers such as hepatocellular carcinoma and myeloma. However, the effects of baicalein on ES cells remain still unknown. We anticipated that baicalein also has apoptotic activity in ES. The aim of the present study was to investigate the effects of baicalein on viability, apoptosis, migration and invasion of ES cells, and further to elaborate the molecular mechanism of baicalein-induced ES cell apoptosis. We found that baicalein markedly inhibited ES cells viability in a time- and dose-dependent manner, especially SK-ES-1 cells and could promote the apoptosis of ES cells. Additionally, baicalein was capable of upregulating the expression of the pro-apoptotic proteins Bax and cytochrome c, reducing the expression of the anti-apoptotic protein Bcl-2, elevating the ratio of Bax/Bcl-2, and triggering the mitochondrial apoptotic pathway, which led to caspase-3 and caspase-9 activation and PARP cleavage. Meanwhile, the activation of caspase-8 and the death receptor pathway was also observed. Besides, baicalein could reduce ES migration and invasion in vitro, which showed its potential to inhibit ES metastasis, besides contributing to the decrease in the expression of matrix metalloproteinases (MMP)-2 and MMP-9. In conclusion, baicalein has a potent tumor-suppressor activity by inducing cell apoptosis through the mitochondrial apoptotic pathway and the death receptor pathway in ES cells, thus it may serve as a novel and effective candidate agent for ES treatment.
Collapse
Affiliation(s)
- Conglin Ye
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Xiaolong Yu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Jin Zeng
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Min Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Bin Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
18
|
Chang H, Dong T, Ma X, Zhang T, Chen Z, Yang Z, Zhang Y. Spondin 1 promotes metastatic progression through Fak and Src dependent pathway in human osteosarcoma. Biochem Biophys Res Commun 2015; 464:45-50. [DOI: 10.1016/j.bbrc.2015.05.092] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 05/28/2015] [Indexed: 10/23/2022]
|
19
|
GRM4 gene polymorphism is associated with susceptibility and prognosis of osteosarcoma in a Chinese Han population. Med Oncol 2015; 31:50. [PMID: 24984297 PMCID: PMC4079940 DOI: 10.1007/s12032-014-0050-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Osteosarcoma (OS), the most common primary bone malignancy, occurs primarily in adolescents and young adults. In earlier genome-wide association studies, rs7591996, rs10208273, rs17206779 and rs1906953 were identified as candidate loci for OS in Caucasians but the association of these single-nucleotide polymorphisms (SNPs) with OS in a Chinese Han population remains unknown. We measured the frequency of these four variants in a Chinese Han population to better understand the genetic etiology of OS. Polymerase chain reaction sequencing was used to detect the genotypes of four candidate SNPs in peripheral blood samples collected from 168 OS patients and 216 healthy controls. Logistic regression models were used to estimate the odds ratios and 95 % confidence intervals. We found rs1906953 in the glutamate receptor metabotropic 4 (GRM4) gene was associated significantly with OS in our Chinese Han population; as with the other SNPs, however, no statistically significant difference was detected. Further analysis showed the association between rs1906953 and OS was independent of gender and age. The rs1906953 locus was not associated with Enneking stages or tumor location; however, it was associated significantly with OS metastasis and prognosis. The GRM4 gene polymorphism was associated with the susceptibility and metastasis of OS in a Chinese Han population.
Collapse
|
20
|
Yu X, Zhou X, Fu C, Wang Q, Nie T, Zou F, Guo R, Liu H, Zhang B, Dai M. Celastrol induces apoptosis of human osteosarcoma cells via the mitochondrial apoptotic pathway. Oncol Rep 2015; 34:1129-36. [PMID: 26165547 PMCID: PMC4530898 DOI: 10.3892/or.2015.4124] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 06/15/2015] [Indexed: 12/28/2022] Open
Abstract
Celastrol is an active compound extracted from the root bark of Triptergium wilfordii Hook F., also known as 'Thunder of God Vine'. It is a well-known Chinese medicinal herb that was found to inhibit tumor cell growth and promote apoptosis in several tumor cell lines. However, research into its effects on osteosarcoma cell apoptosis is still extremely limited. The present study was undertaken to determine the effect of celastrol on viability and apoptosis of osteosarcoma cells and furthermore, to illuminate the molecular mechanism of celastrol-induced osteosarcoma cell apoptosis. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) colorimetric assay was used to evaluate the viability of the cells following treatment with celastrol. The effect of celastrol on the apoptotic rate of the cells was evaluated by flow cytometry using Annexin V-PE/7-AAD staining assay. Fluorescence microscopy was used to detect the morphological changes in the human osteosarcoma U-2OS cell lines. The expression of Bcl-2 family proteins, caspase-3, caspase-8, caspase-9, cytochrome c and PARP was measured by western blotting. We found that celastrol significantly inhibited the growth of osteosarcoma cells in a dose-dependent manner, particularly U-2OS cells. Furthermore, we observed that celastrol upregulated the expression of the pro-apoptotic proteins Bax and cytochrome c and altered the ratio of Bax/Bcl-2, and triggered the mitochondrial apoptotic pathway, resulting in caspase-3 and -9 activation and PARP cleavage. To conclude, the results indicate that celastrol inhibits the proliferation of human osteosarcoma cancer cells by inducing apoptosis via the mitochondrial-dependent pathway.
Collapse
Affiliation(s)
- Xiaolong Yu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Xin Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Changlin Fu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Qiang Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Tao Nie
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Fan Zou
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Runsheng Guo
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Hucheng Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Bin Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Min Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
21
|
Abstract
Osteosarcoma is the most common primary malignancy of bone, typically presenting in the first or second decade of life. Unfortunately, clinical outcomes for osteosarcoma patients have not substantially improved in over 30 years. This stagnation in therapeutic advances is perhaps explained by the genetic, epigenetic, and biological complexities of this rare tumor. In this review we provide a general background on the biology of osteosarcoma and the clinical status quo. We go on to enumerate the genetic and epigenetic defects identified in osteosarcoma. Finally, we discuss ongoing large-scale studies in the field and potential new therapies that are currently under investigation.
Collapse
Affiliation(s)
- James J. Morrow
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Chand Khanna
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
22
|
Guan G, Zhang Y, Lu Y, Liu L, Shi D, Wen Y, Yang L, Ma Q, Liu T, Zhu X, Qiu X, Zhou Y. The HIF-1α/CXCR4 pathway supports hypoxia-induced metastasis of human osteosarcoma cells. Cancer Lett 2014; 357:254-264. [PMID: 25444927 DOI: 10.1016/j.canlet.2014.11.034] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 11/13/2014] [Accepted: 11/14/2014] [Indexed: 12/11/2022]
Abstract
HIF-1α mediates hypoxia-induced expression of the chemokine receptor CXCR4 and contributes to metastasis in many different cancers. We have previously shown that hypoxia promotes migration of human osteosarcoma cells by activating the HIF-1α/CXCR4 pathway. Here, immunohistochemical analysis showed that unlike control osteochondroma samples, osteosarcoma specimens were characterized by elevated expression levels of HIF-1α and CXCR4. Moreover, we found that hypoxia-induced invasiveness was more pronounced in high metastatic potential F5M2 osteosarcoma cells than in low metastatic potential F4 cells, and that this induction was sensitive to treatment with the CXCR4 antagonist AMD3100 and the HIF-1α inhibitor KC7F2. Interestingly, hypoxia-induced CXCR4 expression persisted after cultured osteosarcoma cells were returned to normoxic conditions. These observations were confirmed by experiments in a mouse model of osteosarcoma lung metastasis showing that hypoxia stimulation of pulmonary metastasis was greater in F5M2 than in F4 cells, and was sensitive to treatment with AMD3100. Our study provides further evidence of the contributions of hypoxia and the HIF-1α/CXCR4 pathway to the progression of osteosarcoma, and suggests that this axis might be efficiently leveraged in the development of novel osteosarcoma therapeutics.
Collapse
Affiliation(s)
- Guofeng Guan
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Yinglong Zhang
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Yao Lu
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Lijuan Liu
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Doufei Shi
- Department of Geriatrics, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256603, China
| | - Yanhua Wen
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Lianjia Yang
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Qiong Ma
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Tao Liu
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China
| | - Xiaodong Zhu
- Department of Microsurgery, Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong 256603, China.
| | - Xiuchun Qiu
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China.
| | - Yong Zhou
- Orthopaedic Oncology Institute, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710038, China.
| |
Collapse
|
23
|
Wu J, Feng X, Zhang B, Li J, Xu X, Liu J, Wang X, Wang J, Tong X. Blocking the bFGF/STAT3 interaction through specific signaling pathways induces apoptosis in glioblastoma cells. J Neurooncol 2014; 120:33-41. [PMID: 25048528 DOI: 10.1007/s11060-014-1529-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 06/28/2014] [Indexed: 12/17/2022]
Abstract
We have reported that basic fibroblast growth factor (bFGF) demonstrates an intimate connection with signal transducer and activator of transcription 3 (STAT3) in malignant brain tumor cells. However, its mechanisms are still unclear. In this study, we used inhibitors to block specific signaling pathways, including JAK, PI3K/Akt, and Src pathways, to explore how bFGF mediates crosstalk with STAT3 in two glioblastoma(GBM) cell lines: U251 (mutant p53) and U87 (wild-type p53). Furthermore, we explored how the bFGF/STAT3 pathway affects GBM cell apoptosis. Our results suggest that bFGF can induce the activation of STAT3 mainly through the JAK and PI3K/Akt pathways, and that siRNA-mediated knockdown of STAT3 markedly reduces the bFGF levels in U251 cells. Our results also suggest that STAT3 knockdown increases the expression of pro-apoptotic genes and decreases the expression of anti-apoptotic genes, subsequently collapsing the mitochondrial membrane potentials in vitro and impairs tumor growth in vivo.
Collapse
Affiliation(s)
- Jingchao Wu
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, 300060, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Siena L, Pace E, Ferraro M, Di Sano C, Melis M, Profita M, Spatafora M, Gjomarkaj M. Gemcitabine sensitizes lung cancer cells to Fas/FasL system-mediated killing. Immunology 2014; 141:242-55. [PMID: 24128051 DOI: 10.1111/imm.12190] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 10/08/2013] [Accepted: 10/10/2013] [Indexed: 12/01/2022] Open
Abstract
Gemcitabine is a chemotherapy agent commonly used in the treatment of non-small cell lung cancer (NSCLC) that has been demonstrated to induce apoptosis in NSCLC cells by increasing functionally active Fas expression. The aim of this study was to evaluate the Fas/Fas ligand (FasL) system involvement in gemcitabine-induced lung cancer cell killing. NSCLC H292 cells were cultured in the presence or absence of gemcitabine. FasL mRNA and protein were evaluated by real-time PCR, and by Western blot and flow cytometry, respectively. Apoptosis of FasL-expressing cells was evaluated by flow cytometry, and caspase-8 and caspase-3 activation by Western blot and a colorimetric assay. Cytotoxicity of lymphokine-activated killer (LAK) cells and malignant pleural fluid lymphocytes against H292 cells was analysed in the presence or absence of the neutralizing anti-Fas ZB4 antibody, by flow cytometry. Gemcitabine increased FasL mRNA and total protein expression, the percentage of H292 cells bearing membrane-bound FasL (mFasL) and of mFasL-positive apoptotic H292 cells, as well as caspase-8 and caspase-3 cleavage. Moreover, gemcitabine increased CH11-induced caspase-8 and caspase-3 cleavage and proteolytic activity. Cytotoxicity of LAK cells and pleural fluid lymphocytes was increased against gemcitabine-treated H292 cells and was partially inhibited by ZB4 antibody. These results demonstrate that gemcitabine: (i) induces up-regulation of FasL in lung cancer cells triggering cell apoptosis via an autocrine/paracrine loop; (ii) induces a Fas-dependent apoptosis mediated by caspase-8 and caspase-3 activation; (iii) enhances the sensitivity of lung cancer cells to cytotoxic activity of LAK cells and malignant pleural fluid lymphocytes, partially via Fas/FasL pathway. Our data strongly suggest an active involvement of the Fas/FasL system in gemcitabine-induced lung cancer cell killing.
Collapse
Affiliation(s)
- Liboria Siena
- Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Palermo, Italy
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Participation of the Fas/FasL signaling pathway and the lung microenvironment in the development of osteosarcoma lung metastases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 804:203-17. [PMID: 24924176 DOI: 10.1007/978-3-319-04843-7_11] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The lungs are the most common site for the metastatic spread of osteosarcoma. Success in using chemotherapy to improve overall survival has reached a plateau. Understanding the biologic properties that permit osteosarcoma cells to grow in the lungs may allow the identification of novel therapeutic approaches-the goal being to alter the tumor cells' expression of cell surface proteins so that there is no longer compatibility with the metastatic niche. We have demonstrated that the Fas Ligand positive (FasL(+)) lung microenvironment eliminates Fas(+) osteosarcoma cells that metastasize to the lungs. Indeed, osteosarcoma lung metastases from patients are Fas(-), similar to what we found in several different mouse models. The Fas(+) cells are cleared from the lungs through apoptosis induced by the Fas signaling pathway following interaction of Fas on the tumor cell surface with the lung FasL. Blocking the Fas signaling pathway interferes with this process, allowing the Fas(+) cells to grow in the lungs. Our investigations show that Fas expression in osteosarcoma cells is regulated epigenetically by the micro-RNA miR-20a, encoded by the miR-17-92 cluster. Our studies support the feasibility of finding agents that can re-induce Fas expression as a novel therapeutic approach to treat osteosarcoma patients with lung metastases. We have identified two such agents, the histone deacetylase inhibitor entinostat and the chemotherapeutic agent gemcitabine (GCB). Aerosol GCB and oral entinostat induce the upregulation of Fas and the regression of established osteosarcoma lung metastases. Aerosol GCB was not effective in the FasL-deficient gld mouse confirming that the lung microenvironment was central to the success of this therapy. Our studies establish the critical role of the lung microenvironment in the metastatic process of osteosarcoma to the lungs and suggest an alternative focus for therapy, that is, incorporating the lung microenvironment as part of the treatment strategy against established osteosarcoma disease in the lungs.
Collapse
|
26
|
Greco N, Schott T, Mu X, Rothenberg A, Voigt C, McGough RL, Goodman M, Huard J, Weiss KR. ALDH Activity Correlates with Metastatic Potential in Primary Sarcomas of Bone. ACTA ACUST UNITED AC 2014; 5:331-338. [PMID: 25328803 DOI: 10.4236/jct.2014.54040] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Osteosarcoma (OS), chondrosarcoma (CSA), and Ewings sarcoma (ES) are the most common primary malignancies of bone, and are rare diseases. As with all sarcomas, the prognosis of these diseases ultimately depends on the presence of metastatic disease. Survival is therefore closely linked with the biology and metastatic potential of a particular bone tumor's cells. Here we describe a significant correlation of aldehyde dehydrogenase (ALDH) activity and the presence/absence of distant metastases in ten consecutive cases of human bone sarcomas. Additionally, cultured human CSA cells, which are historically chemo- and radio-resistant, may be sensitive to the ALDH inhibitor, disulfiram. While it is premature to draw broad conclusions from such a small series, the importance of ALDH activity and inhibition in the metastatic potential of primary bone sarcomas should be investigated further.
Collapse
Affiliation(s)
- Nicholas Greco
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, USA ; Cancer Stem Cell Laboratory, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Trevor Schott
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, USA ; Cancer Stem Cell Laboratory, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Xiaodong Mu
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, USA ; Cancer Stem Cell Laboratory, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Adam Rothenberg
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, USA ; Cancer Stem Cell Laboratory, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Clifford Voigt
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, USA ; Cancer Stem Cell Laboratory, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Richard L McGough
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, USA ; University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Mark Goodman
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, USA ; University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, USA ; Cancer Stem Cell Laboratory, University of Pittsburgh School of Medicine, Pittsburgh, USA ; University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Kurt R Weiss
- Department of Orthopaedic Surgery, University of Pittsburgh Medical Center, Pittsburgh, USA ; Cancer Stem Cell Laboratory, University of Pittsburgh School of Medicine, Pittsburgh, USA ; University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, USA
| |
Collapse
|
27
|
O'Farrill JS, Gordon N. Autophagy in osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 804:147-60. [PMID: 24924173 DOI: 10.1007/978-3-319-04843-7_8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Osteosarcoma (OS) metastatic disease is resistant to conventional chemotherapy. Tumor resistance to chemotherapy has been one of the major areas of concern to clinicians and the topic of many laboratory investigators. Evaluation of mechanisms implicated in OS lung metastasis resistance to chemotherapy has been the focus of some of our most recent work. We have previously demonstrated the therapeutic efficacy of aerosol gemcitabine (GCB) in OS lung metastases. However, a subset of cells fails to respond to GCB treatment and persists as isolated lung metastases in vivo. Autophagy, a physiological mechanism that supports nutritional deprivation under stressful conditions, has been implicated in tumor resistance to chemotherapy. We demonstrated the induction of autophagy by GCB in LM7 metastatic human OS cells and K7M3 metastatic murine OS cells. Inhibition of autophagy resulted in increased sensitivity to GCB in LM7 cells. By contrast, inhibiting autophagy in K7M3 cells decreased GCB sensitivity. Defining the role autophagy plays in chemotherapy response in different tumor types has become of greater importance in order to identify the best suitable therapeutic approach. In this chapter, we summarize some of the most recent work related to autophagy in OS, identify some of the known mechanisms, and address the different roles autophagy plays in chemotherapy response.
Collapse
Affiliation(s)
- Janice Santiago O'Farrill
- Department of Pediatrics-Research, The Children's Cancer Hospital, University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | | |
Collapse
|
28
|
Study on Apoptosis of Human Promyelocytic Leukemia HL-60 Cells Induced by Fucosterol via Death Receptor Pathway. ACTA ACUST UNITED AC 2013. [DOI: 10.4028/www.scientific.net/amr.790.607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The purpose of this study is to investigate the effect of fucosterol on the induction of apoptosis and the molecular mechanism involved in Human promyelocytic leukemia HL-60 Cells. HL-60 Cells were treated with different concentrations of fucosterol at different time. MTT method was used to study fucosterol anti-tumor activity. Morphology observation was performed to determine the effects of fucosterol on apoptosis of HL-60 cells. Flow cytometry (FCM) was used to detect the cell cycle. Laser scanning confocal microscope (LSCM) was used to analyze the expressions of Fas, FasL, Fadd and Caspase-8. Caspase activity kits were used to determine the activity of Caspase-8 and Caspase-3. The results showed fucosterol could inhibit the growth of HL-60 cells, and the apoptosis morphology for 48 h treatment was obvious, which showed cell protuberance, cytoplasm concentrated and apoptotic body. Fucosterol treatment for 24 h increased the protein expression of Fas, FasL, Fadd and Caspase-8. It also showed that the activity of Caspase-3 and Caspase-8 has increased significantly. In conclusion, Fucosterol could induce HL-60 cells apoptosis via death receptor pathway.
Collapse
|
29
|
MS-275 sensitizes osteosarcoma cells to Fas ligand-induced cell death by increasing the localization of Fas in membrane lipid rafts. Cell Death Dis 2012; 3:e369. [PMID: 22875006 PMCID: PMC3434650 DOI: 10.1038/cddis.2012.101] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Fas expression is inversely correlated with the metastatic potential of osteosarcoma (OS) cells to the lungs. Fas+ cells are rapidly eliminated when they enter the lungs via their interaction with constitutive Fas ligand (FasL) on the lung epithelium, whereas Fas− OS cells escape this FasL-induced apoptosis and survive in the lung microenvironment. Upregulation of Fas expression in established OS lung metastases results in tumor regression. Here, we demonstrate that treatment of Fas− OS cells with the histone deacetylase inhibitor MS-275 results in the upregulation of Fas mRNA and sensitizes these cells to FasL-induced apoptosis. However, flow cytometry analysis revealed that Fas cell surface protein expression was not significantly increased. Rather, we observed increased levels of Fas within the membrane lipid rafts, as demonstrated by an increase in Fas expression in detergent-insoluble lipid raft fractions and colocalization with GM1+ lipid rafts. We had previously shown that MS-275 treatment inhibited expression of the anti-apoptotic cellular FLICE-inhibitory protein (c-FLIP). Here, we demonstrated that transfection of cells with short hairpin RNA to c-FLIP also resulted in the localization of Fas to lipid rafts. Overall, our studies indicate that MS-275 sensitizes OS cells to FasL by upregulating the expression of Fas in membrane lipid rafts, which correlates with the c-FLIP-dependent distribution of Fas to lipid rafts.
Collapse
|
30
|
Wang X, Cao L, Wang Y, Wang X, Liu N, You Y. Regulation of let-7 and its target oncogenes (Review). Oncol Lett 2012; 3:955-960. [PMID: 22783372 DOI: 10.3892/ol.2012.609] [Citation(s) in RCA: 148] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 02/01/2012] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) are highly evolutionarily-conserved non-coding small RNAs, which were first identified in Caenorhabditis elegans. Let-7 miRNA is involved in the regulation of gene expression in cells. Several novel factors and feedback loops involved in the regulation of the synthesis of let-7 have been identified and additional let-7 target genes have been found. Let-7 has also been shown to be significantly correlated with the occurrence and development of cancer and the results of preliminary studies suggest that it is involved in the regulation of oncogenic pathways in numerous types of tumors. Let-7 is, therefore, a potential molecular target for tumor therapy. Thus, this review examined let-7 and the correlation between let-7 and oncogenic pathways in cancer.
Collapse
Affiliation(s)
- Xirui Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | | | | | | | | | | |
Collapse
|
31
|
Identification of novel candidate oncogenes in chromosome region 17p11.2-p12 in human osteosarcoma. PLoS One 2012; 7:e30907. [PMID: 22292074 PMCID: PMC3266911 DOI: 10.1371/journal.pone.0030907] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 12/29/2011] [Indexed: 11/19/2022] Open
Abstract
Osteosarcoma is the most common primary malignancy of bone. The tumours are characterized by high genomic instability, including the occurrence of multiple regions of amplifications and deletions. Chromosome region 17p11.2–p12 is amplified in about 25% of cases. In previous studies, COPS3 and PMP22 have been identified as candidate oncogenes in this region. Considering the complexity and variation of the amplification profiles for this segment, the involvement of additional causative oncogenes is to be expected. The aim of the present investigation is to identify novel candidate oncogenes in 17p11.2–p12. We selected 26 of in total 85 osteosarcoma samples (31%) with amplification events in 17p11.2–p12, using quantitative PCR for 8 marker genes. These were subjected to high-resolution SNP array analysis and subsequent GISTIC analysis to identify the most significantly amplified regions. Two major amplification peaks were found in the 17p11.2–p12 region. Overexpression as a consequence of gene amplification is a major mechanism for oncogene activation in tumours. Therefore, to identify the causative oncogenes, we next determined expression levels of all genes within the two segments using expression array data that could be generated for 20 of the selected samples. We identified 11 genes that were overexpressed through amplification in at least 50% of cases. Nine of these, c17orf39, RICH2, c17orf45, TOP3A, COPS3, SHMT1, PRPSAP2, PMP22, and RASD1, demonstrated a significant association between copy number and expression level. We conclude that these genes, including COPS3 and PMP22, are candidate oncogenes in 17p11.2–p12 of importance in osteosarcoma tumourigenesis.
Collapse
|
32
|
Villa-Morales M, Fernández-Piqueras J. Targeting the Fas/FasL signaling pathway in cancer therapy. Expert Opin Ther Targets 2012; 16:85-101. [PMID: 22239437 DOI: 10.1517/14728222.2011.628937] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The Fas/FasL system plays a significant role in tumorigenesis. Research has shown that its impairment in cancer cells may lead to apoptosis resistance and contribute to tumor progression. Thus, the development of effective therapies targeting the Fas/FasL system may play an important role in the fight against cancer. AREAS COVERED In this review the recent literature on targeting the Fas/FasL system for therapeutic exploitation at different levels is reviewed. Promising pre-clinical approaches and various exceptions are highlighted. The potential of combined therapies is also explored, whereby tumor sensitivity to Fas-mediated apoptosis is restored, before an effective targeted therapy is employed. EXPERT OPINION The success of the Fas/FasL system targeting for therapeutics will require a better understanding of the alterations conferring resistance, in order to use the most appropriate sensitizing chemotherapeutic or radiotherapeutic agents in combination with effective targeted therapies.
Collapse
Affiliation(s)
- María Villa-Morales
- Department of Biology, Universidad Autónoma de Madrid, CIBER de Enfermedades Raras, Madrid, Spain
| | | |
Collapse
|
33
|
Cole HA, Ichikawa J, Colvin DC, O'Rear L, Schoenecker JG. Quantifying intra-osseous growth of osteosarcoma in a murine model with radiographic analysis. J Orthop Res 2011; 29:1957-62. [PMID: 21656849 DOI: 10.1002/jor.21474] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Accepted: 05/18/2011] [Indexed: 02/04/2023]
Abstract
The orthotopic murine osteosarcoma model is an excellent representation of the human condition as mice develop rapid growth of 'primary' tumor with subsequent lung metastasis. Currently, monitoring tumor growth relies on measuring pulmonary metastases occurring four weeks post injection. Studies show that amputation of the tumor-bearing limb is required before pulmonary metastases are detectable due to rapid growth causing morbidity. Thus, a method measuring 'primary' tumor growth independent of metastasis is required. We hypothesized that serial radiography would allow for longitudinal quantification of 'primary' osteosarcoma growth and explored this idea by utilizing the tibial orthotopic model. Tumor growth was monitored weekly by radiography and calipers, and results were compared with µCT and histology. We found that radiographs demonstrate extra and intra-osseous tumor growth by displaying lytic and blastic lesions and the surrounding radio-opaque area enlarged significantly (p < 0.0001) allowing for quantification. Additionally, radiographs proved more precise than indirect caliper measurements (intra-observer error ±6.64%: inter-observer error ±15.84%). Therefore, we determined that radiography provides accurate, longitudinal quantification of 'primary' osteosarcoma tumor that can be performed serially in the same mouse, does not require introduction of bioluminescence to the host or cell, and is more precise than the current caliper method.
Collapse
Affiliation(s)
- Heather A Cole
- Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1211 Medical Center Dr., Nashville, TN 37232, USA
| | | | | | | | | |
Collapse
|
34
|
Ichikawa J, Cole HA, Magnussen RA, Mignemi NA, Butler M, Holt GE, O'Rear L, Yuasa M, Pabla B, Haro H, Cates JMM, Hamm HE, Schwartz HS, Schoenecker JG. Thrombin induces osteosarcoma growth, a function inhibited by low molecular weight heparin in vitro and in vivo. Cancer 2011; 118:2494-506. [DOI: 10.1002/cncr.26518] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 07/08/2011] [Accepted: 08/03/2011] [Indexed: 12/21/2022]
|