1
|
Morel VJ, Rössler J, Bernasconi M. Targeted immunotherapy and nanomedicine for rhabdomyosarcoma: The way of the future. Med Res Rev 2024; 44:2730-2773. [PMID: 38885148 DOI: 10.1002/med.22059] [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: 06/29/2023] [Revised: 04/17/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024]
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of childhood. Histology separates two main subtypes: embryonal RMS (eRMS; 60%-70%) and alveolar RMS (aRMS; 20%-30%). The aggressive aRMS carry one of two characteristic chromosomal translocations that result in the expression of a PAX3::FOXO1 or PAX7::FOXO1 fusion transcription factor; therefore, aRMS are now classified as fusion-positive (FP) RMS. Embryonal RMS have a better prognosis and are clinically indistinguishable from fusion-negative (FN) RMS. Next to histology and molecular characteristics, RMS risk groupings are now available defining low risk tumors with excellent outcomes and advanced stage disease with poor prognosis, with an overall survival of about only 20% despite intensified multimodal treatment. Therefore, development of novel effective targeted strategies to increase survival and to decrease long-term side effects is urgently needed. Recently, immunotherapies and nanomedicine have been emerging for potent and effective tumor treatments with minimal side effects, raising hopes for effective and safe cures for RMS patients. This review aims to describe the most relevant preclinical and clinical studies in immunotherapy and targeted nanomedicine performed so far in RMS and to provide an insight in future developments.
Collapse
Affiliation(s)
- Victoria Judith Morel
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Jochen Rössler
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Michele Bernasconi
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
2
|
Zhang L, He J, Yu X, Zhang D. Prognostic Factors in Pediatric Alveolar Rhabdomyosarcoma: SEER Analysis of 277 Cases. Clin Pediatr (Phila) 2024; 63:1371-1378. [PMID: 38153032 DOI: 10.1177/00099228231220236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Alveolar rhabdomyosarcoma (ARMS) is a rare but highly aggressive cancer predominantly affecting children and adolescents. This study explores prognostic factors for pediatric and adolescent ARMS, using the Surveillance, Epidemiology, and End Results (SEER) database. Leveraging SEER data (2000-2019), we analyzed 277 cases. Employing Kaplan-Meier survival analysis and Cox proportional hazards models, we identified significant prognostic factors. Gender distribution was nearly equal (56.0% boys, 44.0% girls), with the majority (70.8%) from the white ethnic group. Primary tumors were predominantly in extremities (37.2%). Distant metastases significantly increased mortality risk (hazard ratio [HR], 3.13; 95% CI: 2.14-4.58) and regional lymph node involvement raised mortality risk (HR, 1.36; 95% CI: 0.96-1.92). Chemotherapy-only treatment had higher mortality risk than chemoradiotherapy (HR, 1.16; 95% CI: 0.97-2.67). Conclusively, our study identifies distant metastases, regional lymph node involvement, and treatment modality as crucial predictors of overall survival in pediatric ARMS.
Collapse
Affiliation(s)
- Li Zhang
- Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jiali He
- Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xianhai Yu
- Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Deying Zhang
- Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
| |
Collapse
|
3
|
Zou C, Huang R, Lin T, Wang Y, Tu J, Zhang L, Wang B, Huang J, Zhao Z, Xie X, Huang G, Wang K, Yin J, Shen J. Age-dependent molecular variations in osteosarcoma: implications for precision oncology across pediatric, adolescent, and adult patients. Front Oncol 2024; 14:1382276. [PMID: 38841159 PMCID: PMC11150704 DOI: 10.3389/fonc.2024.1382276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/19/2024] [Indexed: 06/07/2024] Open
Abstract
Background Osteosarcoma is a leading subtype of bone tumor affecting adolescents and adults. Comparative molecular characterization among different age groups, especially in pediatric, adolescents and adults, is scarce. Methods We collected samples from 194 osteosarcoma patients, encompassing pediatric, adolescent, and adult cohorts. Genomic analyses were conducted to reveal prevalent mutations and compare molecular features in pediatric, adolescent, and adult patients. Results Samples from 194 osteosarcoma patients across pediatric to adult ages were analyzed, revealing key mutations such as TP53, FLCN, NCOR1, and others. Children and adolescents showed more gene amplifications and HRD mutations, while adults had a greater Tumor Mutational Burden (TMB). Mutations in those over 15 were mainly in cell cycle and PI3K/mTOR pathways, while under 15s had more in cell cycle and angiogenesis with higher VEGFA, CCND3, TFEB mutations. CNV patterns varied with age: VEGFA and XPO5 amplifications more in under 25s, and CDKN2A/B deletions in over 25s. Genetic alterations in genes like MCL1 and MYC were associated with poor prognosis, with VEGFA mutations also indicating worse outcomes. 58% of patients had actionable mutations, suggesting opportunities for targeted therapies. Age-specific patterns were observed, with Multi-TKI mutations more common in younger patients and CDK4/6 inhibitor mutations in adults, highlighting the need for personalized treatment approaches in osteosarcoma. In a small group of patients with VEGFR amplification, postoperative treatment with multi-kinase inhibitors resulted in a PR in 3 of 13 cases, especially in patients under 15. A significant case involved a 13-year-old with a notable tumor size reduction achieving PR, even with other genetic alterations present in some patients with PD. Conclusion This study delineates the molecular differences among pediatric, adolescent, and adult osteosarcoma patients at the genomic level, emphasizing the necessity for precision diagnostics and treatment strategies, and may offer novel prognostic biomarkers for patients with osteosarcoma. These findings provide a significant scientific foundation for the development of individualized treatment approaches tailored to patients of different age groups.
Collapse
Affiliation(s)
- Changye Zou
- Department of Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Renxuan Huang
- Department of Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tiao Lin
- Department of Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | - Jian Tu
- Department of Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | - Bo Wang
- Department of Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | - Zhiqiang Zhao
- Department of Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xianbiao Xie
- Department of Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Gang Huang
- Department of Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | - Junqiang Yin
- Department of Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jingnan Shen
- Department of Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Hassan SA, Shabaan AAA, Ahmed AR, Issa YA, Fadel SH, El-Sabaa BM. Clinicopathological significance of SOX9 and β-catenin expression in pre-neoadjuvant chemotherapy cases of osteosarcoma: molecular and immunohistochemical study. J Histotechnol 2023; 46:127-138. [PMID: 37013797 DOI: 10.1080/01478885.2023.2193526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/15/2023] [Indexed: 04/05/2023]
Abstract
The molecular pathogenesis of osteosarcoma (OS), the most frequent primary malignant bone tumor of all age groups, is still obscure. Since multidrug chemotherapeutic regimens were introduced in the 1970s, survival rates have been stationary. The Wnt-β-catenin signaling cascade and SOX9 have a significant contribution to skeletal growth, development, and tumorigenesis. In the present work, an attempt was made to examine the role and clinicopathological significance of β-catenin and SOX9 in 46 cases of pre-neoadjuvant chemotherapy OS tissues compared to 10 cases of non-neoplastic bone. The mRNA levels of both markers were assessed by qRT-PCR, and protein levels of β-catenin were analyzed by immunohistochemistry. The results were correlated with different clinicopathological parameters. SOX9 mRNA levels were significantly elevated in OS compared to non-neoplastic bone, and higher levels were significantly associated with the occurrence of fluid-fluid levels (indicating blood-containing cystic spaces) and osteolytic radiological pattern. Although β-catenin mRNA and protein levels were higher in OS compared to non-neoplastic bone, only the protein levels reached statistical significance. Higher β-catenin mRNA levels were significantly associated with tumor size, while higher protein levels were significantly associated with the histologic subtype, mitotic count, and radiological pattern. No significant association was noted with any of the other evaluated parameters. OS showing higher SOX9 mRNA expression and lower β-catenin mRNA and protein expression exhibited longer estimated overall survival times approaching statistical significance. To conclude, while high expression of β-catenin and SOX9 suggests their possible involvement in OS development, their prognostic role may need further research.
Collapse
Affiliation(s)
- Sarah Ahmed Hassan
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | | | - Adel Refaat Ahmed
- Department of Orthopedic Surgery and Traumatology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Yasmine Amr Issa
- Department of Medical Biochemistry, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Shady Hassan Fadel
- Department of Clinical Oncology and Nuclear Medicine, Faculty of Medicine, Faculty of Medicine, Alexandria, Egypt
| | | |
Collapse
|
5
|
Zajec Ž, Dernovšek J, Distel M, Gobec M, Tomašič T. Optimisation of pyrazolo[1,5-a]pyrimidin-7(4H)-one derivatives as novel Hsp90 C-terminal domain inhibitors against Ewing sarcoma. Bioorg Chem 2023; 131:106311. [PMID: 36495678 DOI: 10.1016/j.bioorg.2022.106311] [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: 09/15/2022] [Revised: 11/16/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Ewing sarcoma is the second most prevalent paediatric malignant bone tumour. In most cases, it is driven by the fusion oncoprotein EWS::FLI1, which acts as an aberrant transcription factor and dysregulates gene expression. EWS::FLI1 and a large number of downstream dysregulated proteins are Hsp90 client proteins, making Hsp90 an attractive target for the treatment of Ewing sarcoma. In this article, we report a new structural class of allosteric Hsp90 C-terminal domain (CTD) inhibitors based on the virtual screening hit TVS24, which showed antiproliferative activity in the SK-N-MC Ewing sarcoma cell line with an IC50 value of 15.9 ± 0.7 µM. The optimised compounds showed enhanced anticancer activity in the SK-N-MC cell line. Exposure of Ewing sarcoma cells to the most potent analogue 11c resulted in depletion of critical Hsp90 client proteins involved in cancer pathways such as EWS::FLI1, CDK4, RAF-1 and IGF1R, without inducing a heat shock response. The results of this study highlight Hsp90 CTD inhibitors as promising new agents for the treatment of Ewing sarcoma.
Collapse
Affiliation(s)
- Živa Zajec
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Jaka Dernovšek
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Martin Distel
- St. Anna Children's Cancer Research Institute, Zimmermannplatz 10, 1090 Vienna, Austria
| | - Martina Gobec
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia
| | - Tihomir Tomašič
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia.
| |
Collapse
|
6
|
Bodea J, Caldwell KJ, Federico SM. Bevacizumab, With Sorafenib and Cyclophosphamide Provides Clinical Benefit for Recurrent or Refractory Osseous Sarcomas in Children and Young Adults. Front Oncol 2022; 12:864790. [PMID: 35692751 PMCID: PMC9174993 DOI: 10.3389/fonc.2022.864790] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/11/2022] [Indexed: 11/23/2022] Open
Abstract
Objective Children and adolescents with recurrent and metastatic solid tumors have a poor outcome. A previous phase 1 study (ANGIO1) targeting angiogenesis with bevacizumab, sorafenib, and cyclophosphamide, demonstrated a signal of activity in a subset of patients. Here we report the results of a cohort of pediatric and young adult patients treated at the recommended phase 2 doses. Methods Electronic medical records of patients with refractory or recurrent solid tumors who received ANGIO1 therapy were reviewed. Treatment cycles lasted 21 days and included bevacizumab, sorafenib, and cyclophosphamide. Toxicities were assessed using Common Terminology Criteria for Adverse Events, v5.0. Responses were evaluated using Response Evaluation Criteria in Solid Tumors (RECIST1.1). Results Thirty-nine patients (22 male, 17 female; median age 15 years; range 1-22 years) received the treatment regimen. The most common diagnoses included bone sarcomas (n=21; 14 Ewing sarcoma, 7 osteosarcoma) and soft tissue sarcomas (n=9; 2 rhabdomyosarcoma, 3 synovial sarcoma, 2 desmoplastic small round cell tumors, and 2 high-grade sarcoma). The most common Grade 3 non-hematologic toxicities included hypertension (2, 5.4%) and hematuria (2, 5.4%). Five patients (13.5%) had a pneumothorax (3 at progressive disease, 1 post lung biopsy, and 1 spontaneous). Common Grade 3/4 hematologic toxicities were lymphopenia (19, 51%) and leukopenia (13, 35%). Sixteen patients (43.2%) developed palmar-plantar erythrodysesthesia Grade 2 or less. A total of 297 cycles were administered. Twenty-three patients required a dose reduction of cyclophosphamide, sorafenib or bevacizumab during therapy, all of whom continued to have clinical benefit following dose modification. One patient (Ewing sarcoma) achieved a complete response after 11 cycles; 2 patients (Ewing sarcoma, high grade sarcoma) achieved a partial response following cycles 2 and 4, respectively and 20 patients had stable disease as a best response. Conclusions Intravenous bevacizumab combined with oral sorafenib and metronomic cyclophosphamide was tolerated and required minimal supportive care or additional clinic visits. Disease stabilization for prolonged time periods was observed in greater than half of the treated patients. Patients with bone sarcoma demonstrated a signal of activity suggesting possible benefit from incorporation of the therapy as a maintenance regimen in upfront setting, or as a palliative regimen.
Collapse
Affiliation(s)
- Jessica Bodea
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Kenneth J Caldwell
- Johns Hopkins All Children's Hospital, Cancer and Blood Disorders Institute, St. Petersburg, FL, United States
| | - Sara M Federico
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
7
|
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
|
8
|
Ollauri-Ibáñez C, Astigarraga I. Use of Antiangiogenic Therapies in Pediatric Solid Tumors. Cancers (Basel) 2021; 13:E253. [PMID: 33445470 PMCID: PMC7827326 DOI: 10.3390/cancers13020253] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/08/2021] [Accepted: 01/08/2021] [Indexed: 12/23/2022] Open
Abstract
Cancer is an important cause of death in childhood. In recent years, scientists have made an important effort to achieve greater precision and more personalized treatments against cancer. But since only a few pediatric patients have identifiable therapeutic targets, other ways to stop the neoplastic cell proliferation and dissemination are needed. Therefore, the inhibition of general processes involved in the growth and behavior of tumors can be a relevant strategy for the development of new cancer therapies. In the case of solid tumors, one of these processes is angiogenesis, essential for tumor growth and generation of metastases. This review summarizes the results obtained with the use of antiangiogenic drugs in the main pediatric malignant solid tumors and also an overview of clinical trials currently underway. It should be noted that due to the rarity and heterogeneity of the different types of pediatric cancer, most studies on antiangiogenic drugs include only a small number of patients or isolated clinical cases, so they are not conclusive and further studies are needed.
Collapse
Affiliation(s)
- Claudia Ollauri-Ibáñez
- Pediatric Oncology Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain;
| | - Itziar Astigarraga
- Pediatric Oncology Group, BioCruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain;
- Pediatrics Department, Hospital Universitario Cruces, 48903 Barakaldo, Spain
- Pediatrics Department, University of the Basque Country UPV/EHU, 48940 Leioa, Spain
| |
Collapse
|
9
|
Maleki Dana P, Hallajzadeh J, Asemi Z, Mansournia MA, Yousefi B. Chitosan applications in studying and managing osteosarcoma. Int J Biol Macromol 2020; 169:321-329. [PMID: 33310094 DOI: 10.1016/j.ijbiomac.2020.12.058] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/22/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
Osteosarcoma has a high prevalence among children and adolescents. Common treatments of this disease are not promising enough. Molecular processes involved in the pathogenesis of osteosarcoma are not fully understood. Besides, the remnants of tumor cells after surgery can cause bone destruction and recurrence of the disease. Thus, there is a need to develop novel drugs or enhancing the currently-used drugs as well as identifying bone-repairing methods. Chitosan is a natural compound produced by the deacetylation of chitin. Research has shown that chitosan can be used in various fields due to its beneficial effects, such as biodegradability and biocompatibility. Regarding cancer, chitosan exerts several anti-tumor activities. Moreover, it can be used in diagnostic techniques, drug delivery systems, and cell culture methods. Herein, we aim to discuss the potential roles of chitosan in studying and treating osteosarcoma. We review the literature on chitosan's applications as a drug delivery system and how it can be combined with other substances to improve its ability of local drug delivery. We take a look into the studies concerning the possible benefits of chitosan in the field of bone tissue engineering and 3D culturing. Furthermore, anti-cancer activities of different compounds of chitosan are reviewed.
Collapse
Affiliation(s)
- Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Jamal Hallajzadeh
- Department of Biochemistry and Nutrition, Research Center for Evidence-Based Health Management, Maragheh University of Medical Sciences, Maragheh, Iran.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Ali Mansournia
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
10
|
Dimethoate Induces DNA Damage and Mitochondrial Dysfunction Triggering Apoptosis in Rat Bone-Marrow and Peripheral Blood Cells. TOXICS 2020; 8:toxics8040080. [PMID: 33019668 PMCID: PMC7712428 DOI: 10.3390/toxics8040080] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/26/2020] [Accepted: 09/28/2020] [Indexed: 12/17/2022]
Abstract
Dimethoate (DM) is an organophosphorus (OP) pesticide with wide use in the pest control. Its persistence in crops and soils could possibly cause adverse health consequences in humans as well as other non-target species. Since molecular studies confirming potential genotoxicity of DM have not been previously reported, the acute in vivo toxicological impact was evaluated in Wistar rats. Significant micronuclei induction and metaphase chromosome abnormalities in bone marrow cells exposed to three different DM doses (20, 40 and 60 mg/kg-bw) at multiple treatment durations (24, 48 and 72 h) indicated positive dose response relationship, confirming its genotoxic and cytotoxic potential. Significant mitotic index decrease was seen in dosed animals compared to vehicle control. The study used peripheral blood comet assay, indicating DM-mediated damage to DNA at all exposure levels in a time responsive manner. These assays were found to be an effective, precise, and fast technique with applied value in biomonitoring studies. Cell cycle and apoptosis along with mitochondrial membrane potential (MMP) in flow cytometric analyses confirmed DM exposure decreased MMP, affected the cell cycle, and inflicted DNA damage, which led to cellular apoptosis of leukocytes culminating into immunotoxic effects. The in silico experiments consequently augmented that DM showed acceptable binding energy value for Cyclin A2, suggesting that it could inhibit the cell cycle progression by inhibiting cyclin A2.
Collapse
|
11
|
Huang F, Liao F, Ma G, Hu Y, Zhang C, Xu P, Xu T, Chang J. TBRG4 Knockdown Suppresses Proliferation and Growth of Human Osteosarcoma Cell Lines MG63 Through PI3K/Akt Pathway. Onco Targets Ther 2020; 13:7271-7281. [PMID: 32801755 PMCID: PMC7394601 DOI: 10.2147/ott.s249477] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/14/2020] [Indexed: 01/02/2023] Open
Abstract
Background The transforming growth factor β regulator 4 (TBRG4) has been proved to be involved in various types of tumor. However, its contribution in human osteosarcoma (OS) is still unclear. Patients and Methods In the present study, immunohistochemistry and quantitative real-time PCR were performed to investigate the expression of TBRG4 in OS tissues obtained from patients and three types of cell lines. The effect of TBRG4 knockdown using lentivirus on tumorigenesis was detected by CCK8, high-content screening analysis, colony formation assay and flow cytometric analysis. Bioinformatics analysis was operated to investigate related signaling pathways following TBRG4 knockdown. Results The results showed that the expression of TBRG4 increased significantly in OS tissues and MG63 cell line. TBRG4 knockdown inhibited cell proliferation, colony and tumor formation, while activating cell apoptosis. Ingenuity Pathway Analysis and Western blot assay further indicated that TBRG4 knockdown may regulate the proliferation of human MG63 cells through PI3K/Akt signaling pathway. Conclusion Our results suggest that TBRG4 may become a promising therapeutic target for the treatment of human OS.
Collapse
Affiliation(s)
- Fei Huang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Faxue Liao
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Guangwen Ma
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Yong Hu
- Department of Orthopaedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Chi Zhang
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Pengfei Xu
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Tangbing Xu
- Department of Orthopaedics, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Jun Chang
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
12
|
Shan H, Li K, Zhao D, Chi C, Tan Q, Wang X, Yu J, Piao M. Locally Controlled Release of Methotrexate and Alendronate by Thermo-Sensitive Hydrogels for Synergistic Inhibition of Osteosarcoma Progression. Front Pharmacol 2020; 11:573. [PMID: 32508628 PMCID: PMC7248331 DOI: 10.3389/fphar.2020.00573] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 04/15/2020] [Indexed: 12/28/2022] Open
Abstract
Osteosarcoma (OS) is a serious primary bone malignant tumor that can easily affect children and adolescents. Chemotherapy is one of the important and feasible clinical treatment strategies for the treatment of OS at present, which is severely limited due to insufficient retention time, poor penetration ability, and serious side effects of current anti-tumor drug preparations. In this work, a novel injectable thermo-sensitive hydrogel (mPEG45-PLV19) loaded with methotrexate and alendronate, and the sustained release at the tumor site synergistically inhibited the progression of OS. The mPEG45-PLV19 shows excellent physical and chemical properties. Compared with other treatment groups, the in vivo treatment of gel+ methotrexate + alendronate effectively inhibited the growth of tumor. More importantly, it significantly reduced bone destruction and lung metastasis caused by OS. Therefore, this injectable thermo-sensitive hydrogel drug delivery system has broad prospects for OS chemotherapy.
Collapse
Affiliation(s)
- Hongli Shan
- Department of Clinical Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Ke Li
- Department of Orthopedics, the Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Duoyi Zhao
- Department of Orthopedics, the Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Changliang Chi
- Department of Urology, the First Hospital of Jilin University, Changchun, China
| | - Qinyuan Tan
- Department of Urology, the First Hospital of Jilin University, Changchun, China
| | - Xiaoqing Wang
- Department of Urology, the First Hospital of Jilin University, Changchun, China
| | - Jinhai Yu
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, China
| | - Meihua Piao
- Department of Anesthesiology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Zhao A, Zhang Z, Zhou Y, Li X, Li X, Ma B, Zhang Q. β-Elemonic acid inhibits the growth of human Osteosarcoma through endoplasmic reticulum (ER) stress-mediated PERK/eIF2α/ATF4/CHOP activation and Wnt/β-catenin signal suppression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 69:153183. [PMID: 32113150 DOI: 10.1016/j.phymed.2020.153183] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Osteosarcoma (OS) is a significant threat to the lives of children and young adults. Although neoadjuvant chemotherapy is the first choice of treatment for OS, it is limited by serious side-effects and cancer metastasis. β-Elemonic acid (β-EA), an active component extracted from Boswellia carterii Birdw., has been reported to exhibit potential anti-inflammatory and anticancer activities. However, the anti-tumor effects and underlying mechanisms on OS as well as pharmacokinetic characteristics of β-EA remain unknown. PURPOSE This study was aimed to investigating the anti-tumor effects of β-EA on human OS, the underlying mechanisms, and the pharmacokinetic and tissue distribution characteristics. STUDY DESIGN AND METHODS Cell viability and colony formation assays were performed to determine the effect of β-EA cell on cell proliferation. Apoptosis rates, mitochondrial membrane potential and cell cycle features were analyzed by flow cytometry. qRT-PCR, Western blot, immunofluorescence and immunohistochemical assays were conducted to evaluate the expression levels of genes or proteins related to the pathways affected by β-EA in vitro and in vivo. Cell migration and invasion were evaluated in wound healing and Transwell chamber assays. The effects and pharmacokinetic characteristics of β-EA in vivo were evaluated by analyzing tumor suppression, pharmacokinetics and tissue distribution. RESULTS Explorations indicated that endoplasmic reticulum (ER) stress conditions provoked by β-EA activated the PERK/eIF2α/ATF4 branch of the unfolded protein reaction (UPR), stimulating C/EBP homologous protein (CHOP)-regulated apoptosis and inducing Ca2+ leakage leading to caspase-dependent apoptosis. Furthermore, β-EA induced G0/G1 cell cycle arrest and inhibited metastasis of HOS and 143B cells by attenuating Wnt/β-catenin signaling effects, which included decreased levels of p-Akt(Ser473), p-Gsk3β (Ser9), Wnt/β-catenin target genes (c-Myc and CyclinD1) along with a decline in nuclear β-catenin accumulation. The fast absorption, short elimination half-life, and linear pharmacokinetic characteristics of β-EA were also revealed. The distribution of β-EA was detected in the tumor and bone tissues. CONCLUSIONS Overall, both in vitro and in vivo investigations showed the potential of β-EA for the treatment of human OS. The pharmacokinetic profile and considerable distribution in the tumor and bone tissues warrant further preclinical or even clinical studies.
Collapse
Affiliation(s)
- Ang Zhao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 210009, People's Republic of China
| | - Zhanjie Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 210009, People's Republic of China
| | - Yanfen Zhou
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 210009, People's Republic of China
| | - Xin Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 210009, People's Republic of China
| | - Xiaotian Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, People's Republic of China
| | - Bo Ma
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 210009, People's Republic of China.
| | - Qi Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 210009, People's Republic of China.
| |
Collapse
|
14
|
Yin F, Wang Z, Jiang Y, Zhang T, Wang Z, Hua Y, Song Z, Liu J, Xu W, Xu J, Cai Z, Ding J. Reduction-responsive polypeptide nanomedicines significantly inhibit progression of orthotopic osteosarcoma. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 23:102085. [DOI: 10.1016/j.nano.2019.102085] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 07/27/2019] [Accepted: 08/09/2019] [Indexed: 12/15/2022]
|
15
|
miR-509-5p Inhibits the Proliferation and Invasion of Osteosarcoma by Targeting TRIB2. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2523032. [PMID: 31930114 PMCID: PMC6942763 DOI: 10.1155/2019/2523032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 11/12/2019] [Indexed: 01/28/2023]
Abstract
Osteosarcoma (OS) is one of the most common malignant bone tumors in adolescents with a poor prognosis. Though miR-509-5p has been reported as a tumor suppressor in several human cancers, the role of miR-509-5p in OS remains unclear. In this study, our result of real-time PCR (RT-PCR) showed that the expression of miR-509-5p was significantly decreased in OS tissues and cell lines. Overexpression of miR-509-5p significantly suppressed cell proliferation and invasion in OS cell lines. Moreover, we identified tribbles homolog 2 (TRIB2) as the direct target of miR-509-5p. Knockdown of TRIB2 could inhibit the malignant capacity of OS cells. At last, we reported that TRIB2 could inhibit the bioactivity of the tumor suppressor gene p21 via blocking its transcriptional activity. Collectively, our study revealed that miR-509-5p functions as a tumor suppressor by targeting TRIB2 in OS and thus could affect the activity of p21, suggesting that miR-509-5p is a novel preventive intervention for OS patients.
Collapse
|
16
|
Xia X, Xia J, Yang H, Li Y, Liu S, Cao Y, Tang L, Yu X. Baicalein blocked cervical carcinoma cell proliferation by targeting CCND1 via Wnt/β-catenin signaling pathway. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2729-2736. [PMID: 31284780 DOI: 10.1080/21691401.2019.1636055] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The purpose of this study was to investigate the inhibitory effect of baicalein on the proliferation of cervical carcinoma cells and stimulate cervical carcinoma cells with baicalein. MTT method was used to observe cell proliferation. Flow cytometry was used to observe cell cycle, and gene technology was used to observe the expression of corresponding genes at the level of gene and protein. β-catenin activity was assessed using Western blot and ChIP. Baicalein suppressed cervical carcinoma cell HeLa proliferation by enhancing the activity of caspase-3. Baicalein blocked cell cycle at G0/G1 stage by inhibiting the expression of some genes. At the same time, it can prevent the nuclear translocation of β-catenin and inhibit the activity of Wnt. When the Wnt signaling pathway is increased, the proliferation of HeLa cells is inhibited, and apoptosis is promoted in this way. In conclusion, it indicated that baicalein inhibits cervical carcinoma progression by targeting CCND1 via Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xiulian Xia
- a Department of Obstetrics and Gynecology, The Affiliated TCM Hospital of South West Medical University , Luzhou , China
| | - Jiyi Xia
- b School of Medical Information and Engineering, Southwest Medical University , Luzhou , China
| | - Hai Yang
- a Department of Obstetrics and Gynecology, The Affiliated TCM Hospital of South West Medical University , Luzhou , China
| | - Yan Li
- c Medicine Experimental Center, The Affiliated Hospital of Southwest Medical University , Luzhou , China
| | - Shengyue Liu
- a Department of Obstetrics and Gynecology, The Affiliated TCM Hospital of South West Medical University , Luzhou , China
| | - Yong Cao
- c Medicine Experimental Center, The Affiliated Hospital of Southwest Medical University , Luzhou , China
| | - Li Tang
- c Medicine Experimental Center, The Affiliated Hospital of Southwest Medical University , Luzhou , China
| | - Xiaolan Yu
- a Department of Obstetrics and Gynecology, The Affiliated TCM Hospital of South West Medical University , Luzhou , China
| |
Collapse
|
17
|
Yoon SJ, Moon YJ, Chun HJ, Yang DH. Doxorubicin·Hydrochloride/Cisplatin-Loaded Hydrogel/Nanosized (2-Hydroxypropyl)-Beta-Cyclodextrin Local Drug-Delivery System for Osteosarcoma Treatment In Vivo. NANOMATERIALS 2019; 9:nano9121652. [PMID: 31766334 PMCID: PMC6956151 DOI: 10.3390/nano9121652] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 12/21/2022]
Abstract
Osteosarcoma (OSA) is a difficult cancer to treat due to its tendency for relapse and metastasis; advanced methods are therefore required for OSA treatment. In this study, we prepared a local drug-delivery system for OSA treatment based on doxorubicin·hydrochloride (DOX·HCl)/cisplatin (CP)-loaded visible light-cured glycol chitosan (GC) hydrogel/(2-hydroxypropyl)-beta-cyclodextrin (GDHCP), and compared its therapeutic efficiency with that of DOX·HCl- and CP-loaded GC hydrogels (GD and GHCP). Because of diffusion driven by concentration gradients in the swollen matrix, the three hydrogels showed sustained releases of DOX·HCl and CP over 7 days, along with initial 3-h bursts. Results of in vitro cell viability and in vivo animal testing revealed that GDHCP had a stronger anticancer effect than GD and GHCP even though there were no significant differences. Body weight measurement and histological evaluations demonstrated that the drug-loaded GC hydrogels had biocompatibility without cardiotoxicity or nephrotoxicity. These results suggested that GDHCP could be a good platform as a local drug-delivery system for clinical use in OSA treatment.
Collapse
Affiliation(s)
- Sun Jung Yoon
- Department of Orthopedic Surgery, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju 54907, Korea;
| | - Young Jae Moon
- Department of Biochemistry and Molecular Biology, Chonbuk National University Medical School, Jeonju 54896, Korea;
| | - Heung Jae Chun
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Dae Hyeok Yang
- Institute of Cell and Tissue Engineering, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence: ; Tel.: +82-2-2258-7497
| |
Collapse
|
18
|
Lei Z, Mengying Z, Yifei G, Xiangtao W, Meihua H. Alendronate-modified polydopamine-coated paclitaxel nanoparticles for osteosarcoma-targeted therapy. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101133] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
19
|
Zhao L, Bi D, Qi X, Guo Y, Yue F, Wang X, Han M. Polydopamine-based surface modification of paclitaxel nanoparticles for osteosarcoma targeted therapy. NANOTECHNOLOGY 2019; 30:255101. [PMID: 30736019 DOI: 10.1088/1361-6528/ab055f] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
In order to achieve the purpose of targeting treatment of osteosarcoma, we developed novel paclitaxel (PTX) nanoparticles (Nps) coated with polydopamine (PDA) and grafted by alendronate (ALN) as ligand. Dopamine can be easily polymerized on various surfaces to form a thin PDA film in alkaline environment, which provided a versatile platform to perform secondary reactions for compounds without functional groups. The targeting Nps had a mean particle size of 290.6 ± 2.2 nm and a zeta potential of -13.4 ± 2.7. It was stable in phosphate buffer saline (PBS, pH 7.4), 5% glucose, plasma and displayed sustained drug release behavior. In vitro assay demonstrated the targeting Nps had stronger cytotoxicity against K7M2 wt osteosarcoma cells than the non-targeting Nps. Furthermore, in vivo distribution study indicated they could accumulate much more in tumor than non-targeting Nps. This is consistent with the in vivo antitumor study, targeting Nps achieved a better therapeutic effect than Taxol (8 mg kg-1, i.v.) (71.85% versus 66.53%) and prominently decreased the side effects of PTX. In general, the PTX-PDA-ALN-Nps may offer a feasible and effective strategy for osteosarcoma targeted therapy.
Collapse
Affiliation(s)
- Lei Zhao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 151, Malianwa North Road, Haidian District, Beijing, People's Republic of China. Life Sciences and Environmental Sciences Center, Harbin University of Commerce, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
20
|
Potential Regulatory Effects of miR-182-3p in Osteosarcoma via Targeting EBF2. BIOMED RESEARCH INTERNATIONAL 2019; 2019:4897905. [PMID: 30993113 PMCID: PMC6434304 DOI: 10.1155/2019/4897905] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 02/20/2019] [Indexed: 02/06/2023]
Abstract
Osteosarcoma (OS) is one of the most common primary malignant bone tumors in adolescents with a high mortality rate. MicroRNA (miRNA) is a kind of noncoding RNAs and has been proved to participate in many physiological processes. Many miRNAs have been reported to act as function regulators in OS. In our study, the miRNA and gene expression profiles of OS were downloaded from GEO Datasets and the differential expression analysis was performed using GEO2R. 58 up- and 126 downregulated miRNAs were found. In the three OS gene profiles, 125 up- and 27 downregulated genes were found to be differentially expressed in at least two profiles. The miRNA-mRNA networks were constructed to predict the potential target genes of 10 most up- and downregulated miRNA. Venn analysis was used to detect the coexpressed differentially expressed genes (DEGs). EBF2, one of the upregulated DEGs, was also a potential target gene of miR-182-3p. Knockdown and overexpression of miR-182-3p resulted in overexpression and downexpression of EBF2 separately. Luciferase reporter gene experiment further verified the binding site of miR-182-3p and EBF2. CCK8 assay showed that miR-182-3p knockdown can further enhance the proliferation activity of OS cells, while overexpressing miR-182-3p can inhibit the proliferation activity of OS cells. Our research indicated that downexpression of miR-182-3p in OS cells results in overexpression of EBF2 and promotes the progression of OS.
Collapse
|
21
|
Wu J, Sun H, Li J, Guo Y, Zhang K, Lang C, Zou C, Ma H. Increased survival of patients aged 0-29 years with osteosarcoma: A period analysis, 1984-2013. Cancer Med 2018; 7:3652-3661. [PMID: 29992762 PMCID: PMC6089162 DOI: 10.1002/cam4.1659] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/11/2018] [Accepted: 06/13/2018] [Indexed: 12/27/2022] Open
Abstract
Purpose Osteosarcoma is the most common primary malignancy of bone, and typically occurs among children and adolescence. This study aims to evaluate treatment outcomes among children, adolescents and young adults with osteosarcoma over the three decades by the changes in the long‐term relative survival. Methods Osteosarcoma incidence and relative survival data from Surveillance, Epidemiology, and End Results (SEER) registries during 1984‐2013 were analyzed. The survival differences over three decades, age, sex, race, and socioeconomic status (SES) were assessed by comparing Kaplan‐Meier curves. Results The overall incidence of osteosarcoma kept relatively stable with 0.4 per 100 000 in the three decades with the peak incidence occurring in the aged 10‐19 group. The 10‐year relative survival rate (RSR) increased from 57.7% to 61.0% in the three decades, with the greatest increase in the aged 0‐9 group from 48.2% to 65.7%. The 10‐year RSR improved from 54.1% to 61.5% in males, and from 62.4% to 63.0% in females, respectively, in the three decades. Furthermore, survival dramatically improved from 30% to 60% in the high‐poverty group over the three decades. Conclusion This study demonstrated that the overall incidence of osteosarcoma remained stable, with an improvement in survival in the three decades. The improved survival was greater in males than in females in the three decades. Furthermore, the survival significantly increased in high‐poverty group, which was attributed to increasing improved health care system and patients with low finance can also have access to receiving effective and consistent treatment without distinction.
Collapse
Affiliation(s)
- Jinna Wu
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Huanhuan Sun
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Jie Li
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuanqing Guo
- Department of Spinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Kuibo Zhang
- Department of Spinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Chuandong Lang
- Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Changye Zou
- Department of Orthopedic Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Haiqing Ma
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| |
Collapse
|
22
|
Yu X, Liu Y, Wang Y, Mao X, Zhang Y, Xia J. Baicalein induces cervical cancer apoptosis through the NF-κB signaling pathway. Mol Med Rep 2018; 17:5088-5094. [PMID: 29393414 PMCID: PMC5865972 DOI: 10.3892/mmr.2018.8493] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 07/25/2017] [Indexed: 01/03/2023] Open
Abstract
To investigate the mechanism of baicalein in inducing human cervical cancer cell line C33A apoptosis. Baicalein (200 µM) was used to treat C33A cells. Cell proliferation was tested by the MTT assay. Cell apoptosis was detected by the TUNEL assay and caspase‑3 activity measurement. Cell cycle was determined by flow cytometry and associated gene expression at mRNA and protein levels. Nuclear factor (NF)‑κB activity was assessed by luciferase assay and western blotting. Baicalein suppressed cervical cancer cell C33A proliferation and induced cell apoptosis by activating caspase‑3 activity. Baicalein blocked cell cycle in G0/G1 phase through regulating the expression of associated genes. Baicalein inhibited NF‑κB activity by repressing nuclear translocation. Baicalein suppressed C33A proliferation and promoted cellular apoptosis by inhibiting NF‑κB signaling pathway. In conclusion, the results indicate that baicalein can inhibit cervical cancer cell proliferation and promote cell apoptosis by affecting NF-κB activity.
Collapse
Affiliation(s)
- Xiaolan Yu
- Department of Obstetrics and Gynecology, The Affiliated TCM Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yuqing Liu
- School of Medical Information and Engineering, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yongzhou Wang
- Department of Obstetrics and Gynecology, The Affiliated TCM Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xiguan Mao
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yujiao Zhang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jiyi Xia
- School of Medical Information and Engineering, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
23
|
Shih YL, Au MK, Liu KL, Yeh MY, Lee CH, Lee MH, Lu HF, Yang JL, Wu RSC, Chung JG. Ouabain impairs cell migration, and invasion and alters gene expression of human osteosarcoma U-2 OS cells. ENVIRONMENTAL TOXICOLOGY 2017; 32:2400-2413. [PMID: 28795476 DOI: 10.1002/tox.22453] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 07/07/2017] [Accepted: 07/13/2017] [Indexed: 06/07/2023]
Abstract
Ouabain, the specific Na+ /K+ -ATPase blocker, has biological activity including anti-proliferative and anti-metastasis effects in cancer cell. There is no study to show ouabain inhibiting cell migration and invasion in human osteosarcoma U-2 OS cells. Thus, we investigated the effect of ouabain on the cell migration and invasion of human osteosarcoma U-2 OS cells. Results indicated that ouabain significantly decreased the percentage of viable cells at 2.5-5.0 μM, thus, we selected 0.25-1.0 μM for inhibiting studies. Ouabain inhibited cell migration, invasion and the enzymatic activities of MMP-2, and also affected the expression of metastasis-associated protein in U-2 OS cells. The cDNA microarray assay indicated that CDH1, TGFBR3, SHC3 and MAP2K6 metastasis-related genes were increased, but CCND1, JUN, CDKN1A, TGFB1, 2 and 3, SMAD4, MMP13, MMP2 and FN1 genes were decreased. These findings provide more information regarding ouabain inhibited cell migration and invasion and associated gene expressions in U-2 OS cells after exposed to ouabain.
Collapse
Affiliation(s)
- Yung-Luen Shih
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei, Taiwan
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Man-Kuan Au
- Department of Orthopedics, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Ko-Lin Liu
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Ming-Yang Yeh
- Office of Director, Cheng-Hsin General Hospital, Taipei, Taiwan
| | - Ching-Hsiao Lee
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli County, Taiwan
| | - Mei-Hui Lee
- Department of Genetic Counseling Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Hsu-Feng Lu
- Restaurant, Hotel and Institutional Management, Fu-Jen Catholic University, New Taipei City, Taiwan
- Department of Clinical Pathology, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Jiun-Long Yang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung, Taiwan
| | | | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Wufeng, Taichung, Taiwan
| |
Collapse
|
24
|
Rello-Varona S, Tirado OM. DNA methylation profiling opens a new phase in the search of targeted therapy against Ewing sarcoma. Pharmacogenomics 2017; 18:1307-1309. [DOI: 10.2217/pgs-2017-0017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Santi Rello-Varona
- Sarcoma Research Group, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona 08908, Spain
| | - Oscar M Tirado
- Sarcoma Research Group, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona 08908, Spain
| |
Collapse
|
25
|
Fernández L, Metais JY, Escudero A, Vela M, Valentín J, Vallcorba I, Leivas A, Torres J, Valeri A, Patiño-García A, Martínez J, Leung W, Pérez-Martínez A. Memory T Cells Expressing an NKG2D-CAR Efficiently Target Osteosarcoma Cells. Clin Cancer Res 2017; 23:5824-5835. [DOI: 10.1158/1078-0432.ccr-17-0075] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 04/27/2017] [Accepted: 06/21/2017] [Indexed: 11/16/2022]
|
26
|
Yun HM, Park KR, Quang TH, Oh H, Hong JT, Kim YC, Kim EC. 4-parvifuran inhibits metastatic and invasive actions through the JAK2/STAT3 pathway in osteosarcoma cells. Arch Pharm Res 2017; 40:601-609. [PMID: 28386742 DOI: 10.1007/s12272-017-0911-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 03/15/2017] [Indexed: 02/04/2023]
Abstract
This study was performed to examine the anticancer and anti-metastatic effects of 4-parvifuran (PVN), a novel flavonoid isolated from the heartwood of Dalbergia odorifera, and to study its underlying signaling pathway in human osteosarcoma cells. In the present study, PVN was found to inhibit cell proliferation in a concentration- and time-dependent manner in the human osteosarcoma cell lines studied (MG-63 and U-2 OS) and induce apoptosis, as evidenced by Annexin V+ and TUNEL+ cells. Cleaved poly (ADP-ribose) polymerase (PARP) and caspase-3 were up-regulated while anti-apoptotic proteins including Bcl-2, Bcl-xL, and survivin were down-regulated after treatment with PVN. Matrigel cell migration assay, invasion assay, and soft agar assay were used to show that PVN effectively suppressed cell migration and invasion and colony formation in osteosarcoma cells. Protein and mRNA levels of MMP-2 and MMP-9 were reduced by PVN in a concentration-dependent manner. Furthermore, PVN inhibited Janus kinase 2 (JAK2)/signal transducers and activators of transcription 3 (STAT3), mitogen-activated protein kinases (MAPKs) including JNK, ERK, p38 kinase, and cAMP response element-binding protein (CREB). Therefore, this is the first study to demonstrate that PVN might be a novel anticancer and anti-metastatic agent for the treatment of osteosarcoma through the inhibition of JAK2/STAT3, MAPKs, and CREB signaling pathways.
Collapse
Affiliation(s)
- Hyung-Mun Yun
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Kyung Hee University, Seoul, 130-701, Republic of Korea
| | - Kyung-Ran Park
- Department of Oral & Maxillofacial Regeneration, Graduate School, Kyung Hee University, Seoul, 130-701, Republic of Korea
| | - Tran Hong Quang
- Institute of Pharmaceutical Research and Development, College of Pharmacy, Wonkwang University, Iksan, 570-749, Republic of Korea
| | - Hyuncheol Oh
- Institute of Pharmaceutical Research and Development, College of Pharmacy, Wonkwang University, Iksan, 570-749, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Chungbuk, 361-763, Republic of Korea
| | - Youn-Chul Kim
- Institute of Pharmaceutical Research and Development, College of Pharmacy, Wonkwang University, Iksan, 570-749, Republic of Korea
| | - Eun-Cheol Kim
- Department of Maxillofacial Tissue Regeneration, School of Dentistry and Research Center for Tooth & Periodontal Regeneration (MRC), Kyung Hee University, Seoul, 130-701, Republic of Korea.
| |
Collapse
|
27
|
Lone MI, Nabi A, Dar NJ, Hussain A, Nazam N, Hamid A, Ahmad W. Toxicogenetic evaluation of dichlorophene in peripheral blood and in the cells of the immune system using molecular and flow cytometric approaches. CHEMOSPHERE 2017; 167:520-529. [PMID: 27764745 DOI: 10.1016/j.chemosphere.2016.08.131] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 08/26/2016] [Accepted: 08/28/2016] [Indexed: 06/06/2023]
Abstract
Dichlorophene; a halogenated phenolic compound with wide applications as a fungicide, bactericide and antiprotozoan. Dichlorophene spray also has therapeutic use in the disease digital dermatitis. In guinea pigs, a few studies obtained mixed results in dicholorophene sensitization tests. In consideration of the fact, that the mechanism of its genotoxicity has not been adequately elucidated lead to present study assessing the acute in vivo toxicological impact in Rattus norvegicus. A systematic research has been made encompassing the use of molecular and flow cytometric approaches. The study was designed on blood cells for comet assay which revealed dichlorophene induced DNA damage in all exposures understandable in time dependent manner. The feasibility of this assay was also established as an effective, fast and accurate method with a great potential in biomonitoring. Contemporary molecular techniques were further engaged using leukocytes for the cell apoptosis/cycle and mitochondrial membrane potential employing propidium iodide staining and rhodamine 123 respectively. The effect on cell cycle phases and mitochondrial membrane permeability was analyzed through flow cytometry. These indicators exposed that dichlorophene decreased the mitochondrial membrane potential, altered the cell cycle and confirmed the DNA damage leading to apoptosis of the cells of the immune system accountable for immunotoxic effects of dichlorophene on rat leukocytes.
Collapse
Affiliation(s)
- Mohammad Iqbal Lone
- Gene-Tox Laboratory, Division of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, 202002, UP, India; Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, 180001, Jammu, India.
| | - Arisa Nabi
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, 180001, Jammu, India
| | - Nawab John Dar
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, 180001, Jammu, India
| | - Aashiq Hussain
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, 180001, Jammu, India
| | - Nazia Nazam
- Gene-Tox Laboratory, Division of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, 202002, UP, India
| | - Abid Hamid
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, 180001, Jammu, India; Department of Dermatology, University of Wisconsin, 1300 University Avenue, Madison, WI 53706, USA
| | - Waseem Ahmad
- Gene-Tox Laboratory, Division of Genetics, Department of Zoology, Aligarh Muslim University, Aligarh, 202002, UP, India
| |
Collapse
|
28
|
Abstract
Rhabdomyosarcoma (RMS) is a myogenic tumor classified as the most frequent soft tissue sarcoma affecting children and adolescents. The histopathological classification includes 5 different histotypes, with 2 most predominant referred as to embryonal and alveolar, the latter being characterized by adverse outcome. The current molecular classification identifies 2 major subsets, those harboring the fused Pax3-Foxo1 transcription factor generating from a recurrent specific translocation (fusion-positive RMS), and those lacking this signature but harboring mutations in the RAS/PI3K/AKT signaling axis (fusion-negative RMS). Since little attention has been devoted to RMS metabolism until now, in this review we summarize the "state of art" of metabolism and discuss how some of the molecular signatures found in this cancer, as observed in other more common tumors, can predict important metabolic challenges underlying continuous cell growth, oxidative stress resistance and metastasis, which could be the subject of future targeted therapies.
Collapse
Affiliation(s)
- Eugenio Monti
- a Department of Molecular and Translational Medicine , University of Brescia , Brescia , Italy
| | - Alessandro Fanzani
- a Department of Molecular and Translational Medicine , University of Brescia , Brescia , Italy.,b Interuniversity Institute of Myology , Rome , Italy
| |
Collapse
|
29
|
Thuault S, Comunale F, Hasna J, Fortier M, Planchon D, Elarouci N, De Reynies A, Bodin S, Blangy A, Gauthier-Rouvière C. The RhoE/ROCK/ARHGAP25 signaling pathway controls cell invasion by inhibition of Rac activity. Mol Biol Cell 2016; 27:2653-61. [PMID: 27413008 PMCID: PMC5007086 DOI: 10.1091/mbc.e16-01-0041] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 07/06/2016] [Indexed: 02/06/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of skeletal muscle origin in children and adolescents. Among RMS subtypes, alveolar rhabdomyosarcoma (ARMS), which is characterized by the presence of the PAX3-FOXO1A or PAX7-FOXO1A chimeric oncogenic transcription factor, is associated with poor prognosis and a strong risk of metastasis compared with the embryonal subtype (ERMS). To identify molecular pathways involved in ARMS aggressiveness, we first characterized the migratory behavior of cell lines derived from ARMS and ERMS biopsies using a three-dimensional spheroid cell invasion assay. ARMS cells were more invasive than ERMS cells and adopted an ellipsoidal morphology to efficiently invade the extracellular matrix. Moreover, the invasive potential of ARMS cells depended on ROCK activity, which is regulated by the GTPase RhoE. Specifically, RhoE expression was low in ARMS biopsies, and its overexpression in ARMS cells reduced their invasion potential. Conversely, ARHGAP25, a GTPase-activating protein for Rac, was up-regulated in ARMS biopsies. Moreover, we found that ARHGAP25 inhibits Rac activity downstream of ROCKII and is required for ARMS cell invasion. Our results indicate that the RhoE/ROCK/ARHGAP25 signaling pathway promotes ARMS invasive potential and identify these proteins as potential therapeutic targets for ARMS treatment.
Collapse
Affiliation(s)
- Sylvie Thuault
- Université de Montpellier, CRBM, CNRS, UMR 5237, 34293 Montpellier, France
| | - Franck Comunale
- Université de Montpellier, CRBM, CNRS, UMR 5237, 34293 Montpellier, France
| | - Jessy Hasna
- Université de Montpellier, CRBM, CNRS, UMR 5237, 34293 Montpellier, France
| | - Mathieu Fortier
- Université de Montpellier, CRBM, CNRS, UMR 5237, 34293 Montpellier, France
| | - Damien Planchon
- Université de Montpellier, CRBM, CNRS, UMR 5237, 34293 Montpellier, France
| | - Nabila Elarouci
- Ligue Nationale Contre le Cancer, Cartes d'Identité des Tumeurs, 75013 Paris, France
| | - Aurélien De Reynies
- Ligue Nationale Contre le Cancer, Cartes d'Identité des Tumeurs, 75013 Paris, France
| | - Stéphane Bodin
- Université de Montpellier, CRBM, CNRS, UMR 5237, 34293 Montpellier, France
| | - Anne Blangy
- Université de Montpellier, CRBM, CNRS, UMR 5237, 34293 Montpellier, France
| | | |
Collapse
|
30
|
Liu J, Wu J, Zhou L, Pan C, Zhou Y, Du W, Chen JM, Zhu X, Shen J, Chen S, Liu RY, Huang W. ZD6474, a new treatment strategy for human osteosarcoma, and its potential synergistic effect with celecoxib. Oncotarget 2016; 6:21341-52. [PMID: 26050198 PMCID: PMC4673269 DOI: 10.18632/oncotarget.4179] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 05/12/2015] [Indexed: 12/23/2022] Open
Abstract
ZD6474, a small molecule VEGFR and EGFR tyrosine kinase inhibitor, has been considered as a promising tumor-targeted drug in various malignancies. EGFR and cyclooxygenase-2 (COX-2) were found overexpressed in osteosarcoma in previous reports, so here we tried to explore the anti-osteosarcoma effect of ZD6474 alone or combination with celecoxib, a COX-2 inhibitor. The data demonstrated that ZD6474 inhibited the growth of osteosarcoma cells, and promoted G1-phase cell cycle arrest and apoptosis by inhibiting the activity of EGFR tyrosine kinase, and consequently suppressing its downstream PI3k/Akt and MAPK/ERK pathway. Additionally, daily administration of ZD6474 produced a dose-dependent inhibition of tumor growth in nude mice. Celecoxib also significantly inhibited the growth of osteosarcoma cells in dose-dependent manner, while combination of ZD6474 and celecoxib displayed a synergistic or additive antitumor effect on osteosarcoma in vitro and in vivo. The possible molecular mechanisms to address the synergism are likely that ZD6474 induces the down-regulation of COX-2 expression through inhibiting ERK phosphorylation, while celecoxib promotes ZD6474-directed inhibition of ERK phosphorylation. In conclusion, ZD6474 exerts direct anti-proliferative effects on osteosarcoma cells, and the synergistic antitumor effect of the combination of ZD6474 with celecoxib may indicate a new strategy of the combinative treatment of human osteosarcoma.
Collapse
Affiliation(s)
- Jiani Liu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.,Department of Oncology, Jingzhou Hospital, Tongji Medical College of Huazhong University of Science and Technology, Jingzhou, Hubei, China
| | - Jiangxue Wu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Ling Zhou
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Changchuan Pan
- Medical Oncology, Sichuan Cancer Hospital and Institute, Second People's Hospital of Sichuan Province, Chengdu, China
| | - Yi Zhou
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Wuying Du
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Jie-Min Chen
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Xiaofeng Zhu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Jingnan Shen
- Musculoskeletal Oncology Department, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shuai Chen
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Ran-Yi Liu
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Wenlin Huang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.,Guangdong Provincial Key Laboratory of Tumor Targeted Drugs and Guangzhou Enterprise Key Laboratory of Gene Medicine, Guangzhou Doublle Bioproducts Co. Ltd., Guangzhou, China
| |
Collapse
|
31
|
Rengaswamy V, Zimmer D, Süss R, Rössler J. RGD liposome-protamine-siRNA (LPR) nanoparticles targeting PAX3-FOXO1 for alveolar rhabdomyosarcoma therapy. J Control Release 2016; 235:319-327. [PMID: 27261335 DOI: 10.1016/j.jconrel.2016.05.063] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 05/26/2016] [Accepted: 05/30/2016] [Indexed: 11/30/2022]
Abstract
Alveolar rhabdomyosarcoma (ARMS) are aggressive soft tissue tumors harboring specific fusion transcripts, notably PAX3-FOXO1 (P3F). Current therapy concepts result in unsatisfactory survival rates making the search for innovative approaches necessary: targeting PAX3-FOXO1 could be a promising strategy. In this study, we developed integrin receptor-targeted Lipid-Protamine-siRNA (LPR) nanoparticles using the RGD peptide and validated target specificity as well as their post-silencing effects. We demonstrate that RGD-LPRs are specific to ARMS in vitro and in vivo. Loaded with siRNA directed against the breakpoint of P3F, these particles efficiently down regulated the fusion transcript and inhibited cell proliferation, but did not induce substantial apoptosis. In a xenograft ARMS model, LPR nanoparticles targeting P3F showed statistically significant tumor growth delay as well as inhibition of tumor initiation when injected in parallel with the tumor cells. These findings suggest that RGD-LPR targeting P3F are promising to be highly effective in the setting of minimal residual disease for ARMS.
Collapse
Affiliation(s)
- Venkatesh Rengaswamy
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Doris Zimmer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Technology and Biopharmacy and Freiburger, Materialforschungszentrum (FMF), University of Freiburg, Germany
| | - Regine Süss
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical Technology and Biopharmacy and Freiburger, Materialforschungszentrum (FMF), University of Freiburg, Germany
| | - Jochen Rössler
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany.
| |
Collapse
|
32
|
Shaikh AB, Li F, Li M, He B, He X, Chen G, Guo B, Li D, Jiang F, Dang L, Zheng S, Liang C, Liu J, Lu C, Liu B, Lu J, Wang L, Lu A, Zhang G. Present Advances and Future Perspectives of Molecular Targeted Therapy for Osteosarcoma. Int J Mol Sci 2016; 17:506. [PMID: 27058531 PMCID: PMC4848962 DOI: 10.3390/ijms17040506] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 03/30/2016] [Indexed: 12/20/2022] Open
Abstract
Osteosarcoma (OS) is a bone cancer mostly occurring in pediatric population. Current treatment regime of surgery and intensive chemotherapy could cure about 60%-75% patients with primary osteosarcoma, however only 15% to 30% can be cured when pulmonary metastasis or relapse has taken place. Hence, novel precise OS-targeting therapies are being developed with the hope of addressing this issue. This review summarizes the current development of molecular mechanisms and targets for osteosarcoma. Therapies that target these mechanisms with updated information on clinical trials are also reviewed. Meanwhile, we further discuss novel therapeutic targets and OS-targeting drug delivery systems. In conclusion, a full insight in OS pathogenesis and OS-targeting strategies would help us explore novel targeted therapies for metastatic osteosarcoma.
Collapse
Affiliation(s)
- Atik Badshah Shaikh
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Fangfei Li
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Min Li
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
- Department of Orthopaedic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China.
| | - Bing He
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Xiaojuan He
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Guofen Chen
- Orthopaedic Surgery Department, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Baosheng Guo
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Defang Li
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Feng Jiang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Lei Dang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Shaowei Zheng
- Department of Orthopaedic Surgery, the First Hospital of Huizhou, Huizhou 516000, China.
| | - Chao Liang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Jin Liu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Cheng Lu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Biao Liu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Jun Lu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Luyao Wang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Aiping Lu
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| | - Ge Zhang
- Institute for Advancing Translational Medicine in Bone & Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong 999077, China.
| |
Collapse
|
33
|
Cao J, Hou D, Lu J, Zhu L, Zhang P, Zhou N, Chen K. Anti-tumor activity of exopolysaccharide from Rhizopus nigricans Ehrenb on S180 tumor-bearing mice. Bioorg Med Chem Lett 2016; 26:2098-104. [DOI: 10.1016/j.bmcl.2016.02.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Revised: 01/24/2016] [Accepted: 02/04/2016] [Indexed: 11/25/2022]
|
34
|
UENO TAKEHISA, UEHARA SHUICHIRO, NAKAHATA KENGO, OKUYAMA HIROOMI. Survivin selective inhibitor YM155 promotes cisplatin-induced apoptosis in embryonal rhabdomyosarcoma. Int J Oncol 2016; 48:1847-54. [DOI: 10.3892/ijo.2016.3438] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 02/26/2016] [Indexed: 11/06/2022] Open
|
35
|
Ciccarelli C, Vulcano F, Milazzo L, Gravina GL, Marampon F, Macioce G, Giampaolo A, Tombolini V, Di Paolo V, Hassan HJ, Zani BM. Key role of MEK/ERK pathway in sustaining tumorigenicity and in vitro radioresistance of embryonal rhabdomyosarcoma stem-like cell population. Mol Cancer 2016; 15:16. [PMID: 26897742 PMCID: PMC4761200 DOI: 10.1186/s12943-016-0501-y] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 02/13/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The identification of signaling pathways that affect the cancer stem-like phenotype may provide insights into therapeutic targets for combating embryonal rhabdomyosarcoma. The aim of this study was to investigate the role of the MEK/ERK pathway in controlling the cancer stem-like phenotype using a model of rhabdospheres derived from the embryonal rhabdomyosarcoma cell line (RD). METHODS Rhabdospheres enriched in cancer stem like cells were obtained growing RD cells in non adherent condition in stem cell medium. Stem cell markers were evaluated by FACS analysis and immunoblotting. ERK1/2, myogenic markers, proteins of DNA repair and bone marrow X-linked kinase (BMX) expression were evaluated by immunoblotting analysis. Radiation was delivered using an x-6 MV photon linear accelerator. Xenografts were obtained in NOD/SCID mice by subcutaneously injection of rhabdosphere cells or cells pretreated with U0126 in stem cell medium. RESULTS MEK/ERK inhibitor U0126 dramatically prevented rhabdosphere formation and down-regulated stem cell markers CD133, CXCR4 and Nanog expression, but enhanced ALDH, MAPK phospho-active p38 and differentiative myogenic markers. By contrast, MAPK p38 inhibition accelerated rhabdosphere formation and enhanced phospho-active ERK1/2 and Nanog expression. RD cells, chronically treated with U0126 and then xeno-transplanted in NOD/SCID mice, delayed tumor development and reduced tumor mass when compared with tumor induced by rhabdosphere cells. U0126 intraperitoneal administration to mice bearing rhabdosphere-derived tumors inhibited tumor growth . The MEK/ERK pathway role in rhabdosphere radiosensitivity was investigated in vitro. Disassembly of rhabdospheres was induced by both radiation or U0126, and further enhanced by combined treatment. In U0126-treated rhabdospheres, the expression of the stem cell markers CD133 and CXCR4 decreased and dropped even more markedly following combined treatment. The expression of BMX, a negative regulator of apoptosis, also decreased following combined treatment, which suggests an increase in radiosensitivity of rhabdosphere cells. CONCLUSIONS Our results indicate that the MEK/ERK pathway plays a prominent role in maintaining the stem-like phenotype of RD cells, their survival and their innate radioresistance. Thus, therapeutic strategies that target cancer stem cells, which are resistant to traditional cancer therapies, may benefit from MEK/ERK inhibition combined with traditional radiotherapy, thereby providing a promising therapy for embryonal rhabdomyosarcoma.
Collapse
Affiliation(s)
- Carmela Ciccarelli
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy.
| | - Francesca Vulcano
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Luisa Milazzo
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy.
| | - Francesco Marampon
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy.
| | - Giampiero Macioce
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Adele Giampaolo
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | | | - Virginia Di Paolo
- Department of Anatomy, Histology, Forensic Medicine and Orthopedic, Section of Histology, Sapienza University of Rome, Rome, Italy.
| | - Hamisa Jane Hassan
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| | - Bianca Maria Zani
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, Coppito 2, 67100, L'Aquila, Italy.
| |
Collapse
|
36
|
Fleuren EDG, Hillebrandt-Roeffen MHS, Flucke UE, Te Loo DMWM, Boerman OC, van der Graaf WTA, Versleijen-Jonkers YMH. The role of AXL and the in vitro activity of the receptor tyrosine kinase inhibitor BGB324 in Ewing sarcoma. Oncotarget 2015; 5:12753-68. [PMID: 25528764 PMCID: PMC4350331 DOI: 10.18632/oncotarget.2648] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 10/27/2014] [Indexed: 11/25/2022] Open
Abstract
New targets for Ewing sarcoma (ES) patients are urgently needed. Therefore, we investigated the expression and genetic aberrations of the oncogenic receptor tyrosine kinase (RTK) AXL in ES and determined the efficacy of AXL targeting on cell viability and migration. First, AXL and Gas6 (ligand) mRNA expression was determined by RT-PCR on 29 ES samples. Low, medium and high AXL mRNA expression was observed in 31% (n = 9), 48% (n = 14) and 21% (n = 6) of samples. Gas6 was abundantly present in all specimens. We next tested AXL protein expression immunohistochemically in 36 tumors (primary, post-chemotherapy, metastasized and relapsed samples) from 25 ES patients. Low, medium and high AXL protein expression was observed in 17% (n = 6), 19% (n = 7) and 36% (n = 13) of samples. In primary tumors (n = 15), high AXL expression correlated significantly with a worse overall survival compared to patients with lower expression (61 vs. 194 months, p = 0.026). No genetic aberrations were detected in the AXL RTK domain (n = 29). The AXL-inhibitor BGB324 affected viability (IC50 0.79-2.13 μmol/L) and migratory potential of all tested ES cell lines in vitro (n = 5-6). BGB324 chemosensitized chemotherapy-resistant ES-4 cells to vincristine and doxorubicin. These data suggest that AXL is a potential novel, druggable therapeutic target in ES.
Collapse
Affiliation(s)
- Emmy D G Fleuren
- Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | | | - Uta E Flucke
- Department of Pathology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - D Maroeska W M Te Loo
- Department of Pediatric Hematology and Oncology, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands
| | | | | |
Collapse
|
37
|
Beccafico S, Morozzi G, Marchetti MC, Riccardi C, Sidoni A, Donato R, Sorci G. Artesunate induces ROS- and p38 MAPK-mediated apoptosis and counteracts tumor growth in vivo in embryonal rhabdomyosarcoma cells. Carcinogenesis 2015; 36:1071-83. [PMID: 26153023 DOI: 10.1093/carcin/bgv098] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 07/01/2015] [Indexed: 12/12/2022] Open
Abstract
Rhabdomyosarcoma represents about 50% of soft-tissue sarcomas and 10% of malignant solid tumors in childhood. Embryonal rhabdomyosarcoma (ERMS) is the most frequent subtype, suggested to have an origin in muscle precursor cells that fail to exit the cell cycle and terminally differentiate mainly because of overexpression of the transcription factor, PAX7, which sustains proliferation, migration and invasiveness in ERMS cells. Artesunate (ARS) is a semi-synthetic derivative of artemisinin (ART), a natural compound well known as an antimalarial drug. However, ART and its derivatives have been found efficacious even as anticancer drugs that induce cell cycle arrest and/or apoptosis in several kinds of cancer. Here, we show that ARS dose-dependently induces DNA damage and apoptosis in ERMS cell lines. Production of reactive oxygen species (ROS) and activation of p38 MAPK have a central role in triggering ARS-mediated apoptosis in ERMS cells; indeed either the antioxidant, N-acetylcysteine or the p38 MAPK inhibitor, SB203580, protects ERMS cells from ARS-induced apoptosis. Moreover, ARS treatment in ERMS cells ROS-dependently induces the expression of the myo-miRs, miR-133a and miR-206, which are down-regulated in RMS, and reduces PAX7 protein levels. Finally, ARS upregulates the expression of the adhesion molecules, NCAM and integrin β1, and reduces migration and invasiveness of ERMS cells in vitro, and ARS treatment reduces of about 50% the growth of ERMS xenografts in vivo. Our results are the first evidence of efficacy of ART derivatives in restraining ERMS growth in vivo, and suggest ARS as a potential candidate for therapeutic treatment of ERMS.
Collapse
Affiliation(s)
- Sara Beccafico
- Department of Experimental Medicine, University of Perugia 06132 Perugia, Italy, Interuniversity Institute of Myology (IIM), Padova, Italy and
| | - Giulio Morozzi
- Department of Experimental Medicine, University of Perugia 06132 Perugia, Italy
| | | | - Carlo Riccardi
- Department of Medicine, University of Perugia 06132 Perugia, Italy
| | - Angelo Sidoni
- Department of Experimental Medicine, University of Perugia 06132 Perugia, Italy
| | - Rosario Donato
- Department of Experimental Medicine, University of Perugia 06132 Perugia, Italy, Interuniversity Institute of Myology (IIM), Padova, Italy and
| | - Guglielmo Sorci
- Department of Experimental Medicine, University of Perugia 06132 Perugia, Italy, Interuniversity Institute of Myology (IIM), Padova, Italy and
| |
Collapse
|
38
|
Chen J, Huo J, Jia Z, Song Y, Li Y, Zhang L. Effects of atrazine on the proliferation and cytotoxicity of murine lymphocytes with the use of carboxyfluorescein succinimidyl ester-based flow cytometric approaches. Food Chem Toxicol 2015; 76:61-9. [DOI: 10.1016/j.fct.2014.11.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 10/24/2014] [Accepted: 11/28/2014] [Indexed: 12/31/2022]
|
39
|
Czifra G, Szöllősi A, Nagy Z, Boros M, Juhász I, Kiss A, Erdődi F, Szabó T, Kovács I, Török M, Kovács L, Blumberg PM, Bíró T. Protein kinase Cδ promotes proliferation and induces malignant transformation in skeletal muscle. J Cell Mol Med 2014; 19:396-407. [PMID: 25283340 PMCID: PMC4407591 DOI: 10.1111/jcmm.12452] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 08/28/2014] [Indexed: 02/06/2023] Open
Abstract
In this paper, we investigated the isoform-specific roles of certain protein kinase C (PKC) isoforms in the regulation of skeletal muscle growth. Here, we provide the first intriguing functional evidence that nPKCδ (originally described as an inhibitor of proliferation in various cells types) is a key player in promoting both in vitro and in vivo skeletal muscle growth. Recombinant overexpression of a constitutively active nPKCδ in C2C12 myoblast increased proliferation and inhibited differentiation. Conversely, overexpression of kinase-negative mutant of nPKCδ (DN-nPKCδ) markedly inhibited cell growth. Moreover, overexpression of nPKCδ also stimulated in vivo tumour growth and induced malignant transformation in immunodeficient (SCID) mice whereas that of DN-nPKCδ suppressed tumour formation. The role of nPKCδ in the formation of rhabdomyosarcoma was also investigated where recombinant overexpression of nPKCδ in human rhabdomyosarcoma RD cells also increased cell proliferation and enhanced tumour formation in mouse xenografts. The other isoforms investigated (PKCα, β, ε) exerted only minor (mostly growth-inhibitory) effects in skeletal muscle cells. Collectively, our data introduce nPKCδ as a novel growth-promoting molecule in skeletal muscles and invite further trials to exploit its therapeutic potential in the treatment of skeletal muscle malignancies.
Collapse
Affiliation(s)
- Gabriella Czifra
- DE-MTA "Lendület" Cellular Physiology Research Group, Department of Physiology, Medical Faculty, University of Debrecen, Research Center for Molecular Medicine, Debrecen, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Fujiwara T, Kunisada T, Takeda K, Uotani K, Yoshida A, Ochiya T, Ozaki T. MicroRNAs in soft tissue sarcomas: overview of the accumulating evidence and importance as novel biomarkers. BIOMED RESEARCH INTERNATIONAL 2014; 2014:592868. [PMID: 25165708 PMCID: PMC4139009 DOI: 10.1155/2014/592868] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 07/09/2014] [Indexed: 12/11/2022]
Abstract
Sarcomas are distinctly heterogeneous tumors and a variety of subtypes have been described. Although several diagnostic explorations in the past three decades, such as identification of chromosomal translocation, have greatly improved the diagnosis of soft tissue sarcomas, the unsolved issues, including the limited useful biomarkers, remain. Emerging reports on miRNAs in soft tissue sarcomas have provided clues to solving these problems. Evidence of circulating miRNAs in patients with soft tissue sarcomas and healthy individuals has been accumulated and is accelerating their potential to develop into clinical applications. Moreover, miRNAs that function as novel prognostic factors have been identified, thereby facilitating their use in miRNA-targeted therapy. In this review, we provide an overview of the current knowledge on miRNA deregulation in soft tissue sarcomas, and discuss their potential as novel biomarkers and therapeutics.
Collapse
Affiliation(s)
- Tomohiro Fujiwara
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 7008558, Japan
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama 7008558, Japan
| | - Toshiyuki Kunisada
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 7008558, Japan
- Department of Medical Materials for Musculoskeletal Reconstruction, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 7008558, Japan
| | - Ken Takeda
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 7008558, Japan
- Department of Intelligent Orthopaedic System, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 7008558, Japan
| | - Koji Uotani
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 7008558, Japan
| | - Aki Yoshida
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 7008558, Japan
| | - Takahiro Ochiya
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo 1040045, Japan
| | - Toshifumi Ozaki
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 7008558, Japan
| |
Collapse
|
41
|
Dumont SN, Yang D, Dumont AG, Reynoso D, Blay JY, Trent JC. Targeted polytherapy in small cell sarcoma and its association with doxorubicin. Mol Oncol 2014; 8:1458-68. [PMID: 24998445 DOI: 10.1016/j.molonc.2014.05.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 05/15/2014] [Accepted: 05/27/2014] [Indexed: 12/19/2022] Open
Abstract
A paradigm shift has occurred in the last decade from chemotherapy to targeted therapy for the management of many patients with advanced sarcoma. This work identifies a combination of targeted agents and doxorubicin that are effective against small cell sarcoma cell lines. Three small cell sarcoma cell lines were studied: RD18 (rhabdomyosarcoma), A204 (undifferentiated sarcoma) and TC 71 (Ewing's sarcoma). Each cell line was exposed to increasing concentrations of vorinostat (HDAC inhibitor), 17-DMAG (HSP90 inhibitor), abacavir (anti-telomerase) or sorafenib (tyrosine kinase inhibitor) alone, combined with one another, or combined with doxorubicin. Cell viability, cell cycle analysis and apoptosis were assessed by MTS assay, propidium iodide-Annexin V staining, and caspase 3/7 activity, respectively. The Chou and Talalay combination index (CI) was used to determine whether the effects were additive (CI = 1), synergistic (CI < 1) or antagonistic (CI > 1). In monotherapy, targeted agents achieved 30-90% reductions in viability, with the exception of abacavir. Dual-targeted combination therapies with vorinostat, sorafenib and 17-DMAG demonstrated synergy. Abacavir was antagonistic with every other drug and was not further studied. Both vorinostat and 17-DMAG synergized with doxorubicin, achieving 60% cell killing compared to 12% with doxorubicin alone. No synergy was observed for sorafenib with doxorubicin. The triple therapy vorinostat, 17-DMAG and doxorubicin did not show synergy, but increased the subG1 population at 24H, from 30% to 70% compared to monotherapies with an increase in apoptosis. This work provides evidence of synergy of combinations of vorinostat, 17-DMAG and sorafenib in small cell sarcoma. In addition to doxorubicin, these combinations enhance doxorubicin cytotoxicity at therapeutically relevant concentrations.
Collapse
Affiliation(s)
- S N Dumont
- University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA; Medical Oncology Department, Saint-Antoine Hospital, Public Assistance of Paris Hospitals, Pierre and Marie Curie University, Paris VI, Paris, France
| | - D Yang
- University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - A G Dumont
- University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - D Reynoso
- University of Texas M. D. Anderson Cancer Center, Houston, TX, USA
| | - J-Y Blay
- Claude Bernard Lyon I University, Centre Léon Bérard, Lyon, France
| | - J C Trent
- University of Miami, Sylvester Comprehensive Cancer Center, Miami, FL, USA.
| |
Collapse
|
42
|
Ferreira de Oliveira JMP, Costa M, Pedrosa T, Pinto P, Remédios C, Oliveira H, Pimentel F, Almeida L, Santos C. Sulforaphane induces oxidative stress and death by p53-independent mechanism: implication of impaired glutathione recycling. PLoS One 2014; 9:e92980. [PMID: 24667842 PMCID: PMC3965485 DOI: 10.1371/journal.pone.0092980] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 02/27/2014] [Indexed: 11/19/2022] Open
Abstract
Sulforaphane (SFN) is a naturally-occurring isothiocyanate best known for its role as an indirect antioxidant. Notwithstanding, in different cancer cell lines, SFN may promote the accumulation of reactive oxygen species (ROS) and cause cell death e.g. by apoptosis. Osteosarcoma often becomes chemoresistant, and new molecular targets to prevent drug resistance are needed. Here, we aimed to determine the effect of SFN on ROS levels and to identify key biomarkers leading to ROS unbalance and apoptosis in the p53-null MG-63 osteosarcoma cell line. MG-63 cells were exposed to SFN for up to 48 h. At 10 μM concentration or higher, SFN decreased cell viability, increased the%early apoptotic cells and increased caspase 3 activity. At these higher doses, SFN increased ROS levels, which correlated with apoptotic endpoints and cell viability decline. In exposed cells, gene expression analysis revealed only partial induction of phase-2 detoxification genes. More importantly, SFN inhibited ROS-scavenging enzymes and impaired glutathione recycling, as evidenced by inhibition of glutathione reductase (GR) activity and combined inhibition of glutathione peroxidase (GPx) gene expression and enzyme activity. In conclusion, SFN induced oxidative stress and apoptosis via a p53-independent mechanism. GPx expression and activity were found associated with ROS accumulation in MG-63 cells and are potential biomarkers for the efficacy of ROS-inducing agents e.g. as co-adjuvant drugs in osteosarcoma.
Collapse
Affiliation(s)
- José Miguel P. Ferreira de Oliveira
- CESAM & Laboratory of Biotechnology and Cytomics, Department of Biology, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Maria Costa
- CESAM & Laboratory of Biotechnology and Cytomics, Department of Biology, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Tiago Pedrosa
- CESAM & Laboratory of Biotechnology and Cytomics, Department of Biology, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Pedro Pinto
- CESAM & Laboratory of Biotechnology and Cytomics, Department of Biology, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Catarina Remédios
- CESAM & Laboratory of Biotechnology and Cytomics, Department of Biology, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Helena Oliveira
- CESAM & Laboratory of Biotechnology and Cytomics, Department of Biology, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| | - Francisco Pimentel
- Lenitudes, Lisboa, Portugal
- Faculty of Health Sciences, University Fernando Pessoa, R. Carlos da Maia 296, Porto, Portugal
- Center for Health Studies & Research, University of Coimbra, Avenida Dias da Silva, Coimbra, Portugal
- Faculdade de Ciências da Saúde - Universidade da Beira Interior, Avenida Infante D. Henrique, Covilhã, Portugal
| | - Luís Almeida
- Luzitin SA, R. Bayer 16, Coimbra, Portugal
- Blueclinical Phase I, R. Sarmento de Beires 153, Porto, Portugal
| | - Conceição Santos
- CESAM & Laboratory of Biotechnology and Cytomics, Department of Biology, University of Aveiro, Campus Universitário de Santiago, Aveiro, Portugal
| |
Collapse
|
43
|
Fleuren EDG, Versleijen-Jonkers YMH, Boerman OC, van der Graaf WTA. Targeting receptor tyrosine kinases in osteosarcoma and Ewing sarcoma: current hurdles and future perspectives. Biochim Biophys Acta Rev Cancer 2014; 1845:266-76. [PMID: 24582852 DOI: 10.1016/j.bbcan.2014.02.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 02/20/2014] [Accepted: 02/22/2014] [Indexed: 12/26/2022]
Abstract
Osteosarcoma (OS) and Ewing sarcoma (ES) are the two most common types of primary bone cancer, which mainly affect children and young adults. Despite intensive multi-modal treatment, the survival of both OS and ES has not improved much during the last decades and new therapeutic options are awaited. One promising approach is the specific targeting of transmembrane receptor tyrosine kinases (RTKs) implicated in these types of bone cancer. However, despite encouraging in vitro and in vivo results, apart from intriguing results of Insulin-like Growth Factor-1 Receptor (IGF-1R) antibodies in ES, clinical studies are limited or disappointing. Primary resistance to RTK inhibitors is frequently observed in OS and ES patients, and even patients that initially respond well eventually develop acquired resistance. There are, however, a few remarks to make concerning the current set-up of clinical trials and about strategies to improve RTK-based treatments in OS and ES. This review provides an overview concerning current RTK-mediated therapies in OS and ES and discusses the problems observed in the clinic. More importantly, we describe several strategies to overcome resistance to RTK inhibitors which may significantly improve outcome of OS and ES patients.
Collapse
Affiliation(s)
- Emmy D G Fleuren
- Department of Medical Oncology, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | | | - Otto C Boerman
- Department of Nuclear Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | | |
Collapse
|
44
|
Zheng L, Zhang D, Zhang Y, Wen Y, Wang Y. mTOR signal transduction pathways contribute to TN-C FNIII A1 overexpression by mechanical stress in osteosarcoma cells. Mol Cells 2014; 37:118-25. [PMID: 24598996 PMCID: PMC3935624 DOI: 10.14348/molcells.2014.2247] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 01/13/2014] [Accepted: 01/14/2014] [Indexed: 11/27/2022] Open
Abstract
Osteosarcoma is the most common primary malignant bone tumor with a very poor prognosis. Treating osteosarcoma remains a challenge due to its high transitivity. Tenascin-C, with large molecular weight variants including different combinations of its alternative spliced FNIII repeats, is specifically over expressed in tumor tissues. This study examined the expression of Tenascin-C FNIIIA1 in osteosarcoma tissues, and estimated the effect of mechanical stimulation on A1 expression in MG-63 cells. Through immunohistochemical analysis, we found that the A1 protein was expressed at a higher level in osteosarcoma tissues than in adjacent normal tissues. By cell migration assay, we observed that there was a significant correlation between A1 expression and MG-63 cell migra-tion. The relation is that Tenascin-C FNIIIA1 can promote MG-63 cell migration. According to our further study into the effect of mechanical stimulation on A1 expression in MG-63 cells, the mRNA and protein levels of A1 were significantly up-regulated under mechanical stress with the mTOR molecule proving indispensable. Meanwhile, 4E-BP1 and S6K1 (downstream molecule of mTOR) are necessary for A1 normal expression in MG-63 cells whether or not mechanical stress has been encountered. We found that Tenascin-C FNIIIA1 is over-expressed in osteosar-coma tissues and can promote MG-63 cell migration. Furthermore, mechanical stress can facilitate MG-63 cell migration though facilitating A1 overexpression with the necessary molecules (mTOR, 4E-BP1 and S6K1). In con-clusion, high expression of A1 may promote the meta-stasis of osteosarcoma by facilitating MG-63 cell migration. Tenascin-C FNIIIA1 could be used as an indicator in metastatic osteosarcoma patients.
Collapse
Affiliation(s)
- Lianhe Zheng
- Department of Orthopaedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, 710032, Shaanxi Province,
China
| | - Dianzhong Zhang
- Department of Orthopaedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, 710032, Shaanxi Province,
China
| | - Yunfei Zhang
- Department of Orthopaedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, 710032, Shaanxi Province,
China
| | - Yanhua Wen
- Department of Orthopaedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, 710032, Shaanxi Province,
China
| | - Yucai Wang
- Department of Orthopaedic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, 710032, Shaanxi Province,
China
| |
Collapse
|
45
|
Kuijjer ML, van den Akker BEWM, Hilhorst R, Mommersteeg M, Buddingh EP, Serra M, Bürger H, Hogendoorn PCW, Cleton-Jansen AM. Kinome and mRNA expression profiling of high-grade osteosarcoma cell lines implies Akt signaling as possible target for therapy. BMC Med Genomics 2014; 7:4. [PMID: 24447333 PMCID: PMC3932036 DOI: 10.1186/1755-8794-7-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 01/14/2014] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND High-grade osteosarcoma is a primary malignant bone tumor mostly occurring in adolescents and young adults, with a second peak at middle age. Overall survival is approximately 60%, and has not significantly increased since the introduction of neoadjuvant chemotherapy in the 1970s. The genomic profile of high-grade osteosarcoma is complex and heterogeneous. Integration of different types of genome-wide data may be advantageous in extracting relevant information from the large number of aberrations detected in this tumor. METHODS We analyzed genome-wide gene expression data of osteosarcoma cell lines and integrated these data with a kinome screen. Data were analyzed in statistical language R, using LIMMA for detection of differential expression/phosphorylation. We subsequently used Ingenuity Pathways Analysis to determine deregulated pathways in both data types. RESULTS Gene set enrichment indicated that pathways important in genomic stability are highly deregulated in these tumors, with many genes showing upregulation, which could be used as a prognostic marker, and with kinases phosphorylating peptides in these pathways. Akt and AMPK signaling were identified as active and inactive, respectively. As these pathways have an opposite role on mTORC1 signaling, we set out to inhibit Akt kinases with the allosteric Akt inhibitor MK-2206. This resulted in inhibition of proliferation of osteosarcoma cell lines U-2 OS and HOS, but not of 143B, which harbors a KRAS oncogenic transformation. CONCLUSIONS We identified both overexpression and hyperphosphorylation in pathways playing a role in genomic stability. Kinome profiling identified active Akt signaling, which could inhibit proliferation in 2/3 osteosarcoma cell lines. Inhibition of PI3K/Akt/mTORC1 signaling may be effective in osteosarcoma, but further studies are required to determine whether this pathway is active in a substantial subgroup of this heterogeneous tumor.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Anne-Marie Cleton-Jansen
- Department of Pathology, Leiden University Medical Center, Albinusdreef 2, 2300RC Leiden, The Netherlands.
| |
Collapse
|
46
|
Ferreira de Oliveira JMP, Remédios C, Oliveira H, Pinto P, Pinho F, Pinho S, Costa M, Santos C. Sulforaphane Induces DNA Damage and Mitotic Abnormalities in Human Osteosarcoma MG-63 Cells: Correlation with Cell Cycle Arrest and Apoptosis. Nutr Cancer 2014; 66:325-34. [DOI: 10.1080/01635581.2014.864777] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
47
|
Wang Y, Yang Z, Meng Z, Cao H, Zhu G, Liu T, Wang X. Knockdown of TRPM8 suppresses cancer malignancy and enhances epirubicin-induced apoptosis in human osteosarcoma cells. Int J Biol Sci 2013; 10:90-102. [PMID: 24391455 PMCID: PMC3879595 DOI: 10.7150/ijbs.7738] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 12/03/2013] [Indexed: 11/05/2022] Open
Abstract
As the function of transient receptor potential melastatin member 8 (TRPM8) in osteosarcoma is still unknown, we aim to investigate the possible effects and potential mechanisms of TRPM8 on cell proliferation, metastasis and chemosensitivity in osteosarcoma cells. We find that TRPM8 is aberrantly over-expressed in human osteosarcoma tissues and cell lines. Knockdown of TRPM8 by siRNA in osteosarcoma cells leads to the impaired regulation of intracellular Ca(2+) concentration and then the Akt-GSK-3β pathway and the phosphorylation of p44/p42 and FAK are suppressed. Knockdown of TRPM8 not only negatively influences the cell proliferation and metastasis but also enhances epirubicin-induced cell apoptosis. Such results reveal that TRPM8 is worthy further investigation for its potential as a clinical biomarker and therapeutic target in osteosarcoma.
Collapse
Affiliation(s)
- Yongzhi Wang
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, China, 430071
| | - Zhonghua Yang
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, China, 430071
| | - Zhe Meng
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, China, 430071
| | - Hong Cao
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, China, 430071
| | - Guangbin Zhu
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, China, 430071
| | - Tao Liu
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, China, 430071
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital, Wuhan University, Wuhan, China, 430071
| |
Collapse
|
48
|
Peters HL, Yan Y, Solheim JC. APLP2 regulates the expression of MHC class I molecules on irradiated Ewing's sarcoma cells. Oncoimmunology 2013; 2:e26293. [PMID: 24353913 PMCID: PMC3862638 DOI: 10.4161/onci.26293] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 08/27/2013] [Accepted: 08/27/2013] [Indexed: 01/06/2023] Open
Abstract
Ewing's sarcoma (EWS) is a pediatric cancer that is conventionally treated by surgery, chemotherapy, and radiation therapy. Innovative immunotherapies to treat EWS are currently under development. Unfortunately for EWS patients, when the disease is found to be resistant to current therapeutic approaches, the prognosis is predictably grim. Radiation therapy and immunotherapy could potentially synergize in the eradication of EWS, as some studies have previously shown that irradiation increases the presence of immune receptors, including MHC class I molecules, on the surface of tumor cells. However, EWS cells have been reported to express low levels of MHC class I molecules, a phenotype that would inhibit T-cell mediated lysis. We have previously demonstrated that the transgene-driven overexpression of amyloid β (A4) precursor-like protein 2 (APLP2) reduces the expression of MHC class I molecules on the surface of human cervical carcinoma HeLa cells. We thus examined whether endogenously expressed APLP2 downregulates MHC class I expression on EWS cells, particularly upon irradiation. We found that irradiation induces the relocalization of APLP2 and MHC class I molecules on the surface of EWS cells, redistributing cells from subpopulations with relatively low APLP2 and high MHC class I into subpopulations with relatively high APLP2 and low MHC class I surface expression. Consistent with these findings, the transfection of an APLP2-targeting siRNA into EWS cells increased MHC class I expression on the cell surface. Furthermore, APLP2 was found by co-immunoprecipitation to bind to MHC class I molecules. Taken together, these findings suggest that APLP2 inhibits MHC class I expression on the surface of irradiated EWS cells by a mechanism that involves APLP2/MHC class I interactions. Thus, therapeutic strategies that limit APLP2 expression may boost the ability of T cells to recognize and eradicate EWS in patients.
Collapse
Affiliation(s)
- Haley L Peters
- Eppley Institute; University of Nebraska Medical Center; Omaha, NE USA
| | - Ying Yan
- Eppley Institute; University of Nebraska Medical Center; Omaha, NE USA
| | - Joyce C Solheim
- Eppley Institute; University of Nebraska Medical Center; Omaha, NE USA ; Department of Biochemistry and Molecular Biology; University of Nebraska Medical Center; Omaha, NE USA ; Department of Pathology and Microbiology; University of Nebraska Medical Center; Omaha, NE USA
| |
Collapse
|
49
|
Lv Z, Yang D, Li J, Hu M, Luo M, Zhan X, Song P, Liu C, Bai H, Li B, Yang Y, Chen Y, Shi Q, Weng Y. Bone morphogenetic protein 9 overexpression reduces osteosarcoma cell migration and invasion. Mol Cells 2013; 36:119-26. [PMID: 23807047 PMCID: PMC3887952 DOI: 10.1007/s10059-013-0043-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 05/08/2013] [Accepted: 05/24/2013] [Indexed: 01/24/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is known to promote tumor migration and invasion. Bone morphogenetic proteins (BMPs) are members of the TGF-β family expressed in a variety of human carcinoma cell lines. The role of bone morphogenetic protein 9 (BMP9), the most powerful osteogenic factor, in osteosarcoma (OS) progression has not been fully clarified. The expression of BMP9 and its receptors in OS cell lines was analyzed by RT-PCR. We found that BMP9 and its receptors were expressed in OS cell lines. We further investigated the influence of BMP9 on the biological behaviors of OS cells. BMP9 overexpression in the OS cell lines 143B and MG63 inhibited in vitro cell migration and invasion. We further investigated the expression of a panel of cancer-related genes and found that BMP9 overexpression increased the phosphorylation of Smad1/5/8 proteins, increased the expression of ID1, and reduced the expression and activity of matrix metalloproteinase 9 (MMP9) in OS cells. BMP9 silencing induced the opposite effects. We also found that BMP9 may not affect the chemokine (C-X-C motif) ligand 12 (CXCL12)/C-X-C chemokine receptor type 4 (CXCR4) axis to regulate the invasiveness and metastatic capacity of OS cells. Interestingly, CXCR4 was expressed in both 143B and MG63 cells, while CXCL12 was only detected in MG63 cells. Taken together, we hypothesize that BMP9 inhibits the migration and invasiveness of OS cells through a Smad-dependent pathway by downregulating the expression and activity of MMP9.
Collapse
Affiliation(s)
- Zilan Lv
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Dandan Yang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Jie Li
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Min Hu
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Min Luo
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Xiaoqin Zhan
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Peipei Song
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Chen Liu
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Huili Bai
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Baolin Li
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Yang Yang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Yingying Chen
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Qiong Shi
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| | - Yaguang Weng
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education and School of Clinical Diagnostic and Laboratory Medicine, Chongqing Medical University, Chongqing 400016,
People’s Republic of China
| |
Collapse
|
50
|
JIANG WENG, YE LIN, JI KE, RUGE FIONA, WU YILING, GAO YONG, JI JIAFU, MASON MALCOLMD. Antitumour effects of Yangzheng Xiaoji in human osteosarcoma: The pivotal role of focal adhesion kinase signalling. Oncol Rep 2013; 30:1405-13. [DOI: 10.3892/or.2013.2586] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 05/22/2013] [Indexed: 11/06/2022] Open
|