1
|
Todosenko N, Khlusov I, Yurova K, Khaziakhmatova O, Litvinova L. Signal Pathways and microRNAs in Osteosarcoma Growth and the Dual Role of Mesenchymal Stem Cells in Oncogenesis. Int J Mol Sci 2023; 24:ijms24108993. [PMID: 37240338 DOI: 10.3390/ijms24108993] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/10/2023] [Accepted: 05/15/2023] [Indexed: 05/28/2023] Open
Abstract
The major challenges in Osteosarcoma (OS) therapy are its heterogeneity and drug resistance. The development of new therapeutic approaches to overcome the major growth mechanisms of OS is urgently needed. The search for specific molecular targets and promising innovative approaches in OS therapy, including drug delivery methods, is an urgent problem. Modern regenerative medicine focuses on harnessing the potential of mesenchymal stem cells (MSCs) because they have low immunogenicity. MSCs are important cells that have received considerable attention in cancer research. Currently, new cell-based methods for using MSCs in medicine are being actively investigated and tested, especially as carriers for chemotherapeutics, nanoparticles, and photosensitizers. However, despite the inexhaustible regenerative potential and known anticancer properties of MSCs, they may trigger the development and progression of bone tumors. A better understanding of the complex cellular and molecular mechanisms of OS pathogenesis is essential to identify novel molecular effectors involved in oncogenesis. The current review focuses on signaling pathways and miRNAs involved in the development of OS and describes the role of MSCs in oncogenesis and their potential for antitumor cell-based therapy.
Collapse
Affiliation(s)
- Natalia Todosenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Igor Khlusov
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
- Laboratory of Cellular and Microfluidic Technologies, Siberian State Medical University, 2, Moskovskii Trakt, 634050 Tomsk, Russia
| | - Kristina Yurova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Olga Khaziakhmatova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
| | - Larisa Litvinova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia
- Laboratory of Cellular and Microfluidic Technologies, Siberian State Medical University, 2, Moskovskii Trakt, 634050 Tomsk, Russia
| |
Collapse
|
2
|
Grundy M, Narendran A. The hepatocyte growth factor/mesenchymal epithelial transition factor axis in high-risk pediatric solid tumors and the anti-tumor activity of targeted therapeutic agents. Front Pediatr 2022; 10:910268. [PMID: 36034555 PMCID: PMC9399617 DOI: 10.3389/fped.2022.910268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/15/2022] [Indexed: 01/04/2023] Open
Abstract
Clinical trials completed in the last two decades have contributed significantly to the improved overall survival of children with cancer. In spite of these advancements, disease relapse still remains a significant cause of death in this patient population. Often, increasing the intensity of current protocols is not feasible because of cumulative toxicity and development of drug resistance. Therefore, the identification and clinical validation of novel targets in high-risk and refractory childhood malignancies are essential to develop effective new generation treatment protocols. A number of recent studies have shown that the hepatocyte growth factor (HGF) and its receptor Mesenchymal epithelial transition factor (c-MET) influence the growth, survival, angiogenesis, and metastasis of cancer cells. Therefore, the c-MET receptor tyrosine kinase and HGF have been identified as potential targets for cancer therapeutics and recent years have seen a race to synthesize molecules to block their expression and function. In this review we aim to summarize the literature that explores the potential and biological rationale for targeting the HGF/c-MET pathway in common and high-risk pediatric solid tumors. We also discuss selected recent and ongoing clinical trials with these agents in relapsed pediatric tumors that may provide applicable future treatments for these patients.
Collapse
Affiliation(s)
- Megan Grundy
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Aru Narendran
- POETIC Laboratory for Preclinical and Drug Discovery Studies, Division of Pediatric Oncology, Alberta Children's Hospital, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
3
|
Hosseini F, Alemi F, Malakoti F, Mahmoodpoor A, Younesi S, Yousefi B, Asemi Z. Targeting Wnt/β-catenin signaling by microRNAs as a therapeutic approach in chemoresistant osteosarcoma. Biochem Pharmacol 2021; 193:114758. [PMID: 34481813 DOI: 10.1016/j.bcp.2021.114758] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/24/2021] [Accepted: 08/31/2021] [Indexed: 12/13/2022]
Abstract
Osteosarcoma (OS) is an adolescent and young adult malignancy that mostly occurs in long bones. The treatment of OS is still a big challenge for clinicians due to increasing chemoresistance, and many efforts are being made today to find more beneficial treatments. In this regard, the use of microRNAs has shown a high capacity to develop promising therapies. By targeting cancer-involved signaling pathways, microRNAs reduce the cellular level of these protein pathways; thereby reducing the growth and invasion of tumors, and even leading cancer cells to apoptosis. One of these oncogenic pathways that play an important role in OS development and can be targeted by microRNAs is the Wnt/β-catenin signaling pathway. Hence, the first goal of this review article is to explain the cross-talk of microRNAs and the Wnt/β-catenin signaling in OS and then discussing recent findings of the use of microRNAs as a therapeutic approach in OS.
Collapse
Affiliation(s)
- Foroogh Hosseini
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Anesthesiology Research Team, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Forough Alemi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faezeh Malakoti
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology and Intensive Care, School of Medicine, Tabriz University of Medical Science and Health Services, Tabriz, Iran; Anesthesiology Research Team, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Simin Younesi
- Schoole of Health and Biomedical Sciences, RMIT University, Melborne, VIC, Australia
| | - Bahman Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
4
|
Prognostic and Therapeutic Utility of Variably Expressed Cell Surface Receptors in Osteosarcoma. Sarcoma 2021; 2021:8324348. [PMID: 33603563 PMCID: PMC7872755 DOI: 10.1155/2021/8324348] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/17/2020] [Accepted: 01/07/2021] [Indexed: 01/05/2023] Open
Abstract
Background Six cell surface receptors, human epidermal growth factor receptor-2 (Her-2), platelet-derived growth factor receptor-β (PDGFR-β), insulin-like growth factor-1 receptor (IGF-1R), insulin receptor (IR), c-Met, and vascular endothelial growth factor receptor-3 (VEGFR-3), previously demonstrated variable expression across varying patient-derived and standard osteosarcoma (OS) cell lines. The current study sought to validate previous expression patterns and evaluate whether these receptors offer prognostic and/or therapeutic value. Methods Patient-derived OS cell lines (n = 52) were labeled with antibodies to Her-2, PDGFR-β, IGF-1R, IR, c-Met, and VEGFR-3. Expression was characterized using flow cytometry. The difference in geometric mean fluorescent intensity (geoMFIdiff = geoMFIpositive - geoMFInegative) was calculated for each receptor across all cell lines. Receptor expression was categorized as low (Q1), intermediate (Q2, Q3), or high (Q4). The event-free survival (EFS) and overall survival for the six cell surface receptors were estimated by the Kaplan-Meier method. Differences in hazard for EFS event and overall survival event for patients in each of the three expression levels in each of the six cell surface receptors were assessed using the log-rank test. Results All 6 receptors were variably expressed in the majority of cell lines. IR and PDGFR-β expressions were found to be significant predictors for EFS amongst patients with nonmetastatic disease (p=0.02 and 0.01, respectively). The hazard ratio for EFS was significantly higher between high IR and intermediate IR expression (HR = 2.66, p=0.02), as well as between high PDGFR-β and intermediate PDGFR-β expression (HR = 5.68, p=0.002). Her-2, c-Met, IGF-1R, and VEGFR-3 were not found to be significant predictors for either EFS or overall survival. Conclusion The six cell surface receptors demonstrated variable expression across the majority of patient-derived OS cell lines tested. Limited prognostic value was offered by IR and PDGFR-β expression within nonmetastatic patients. The remaining receptors do not provide clear prognostic utility. Nevertheless, their consistent, albeit variable, surface expression across a large panel of patient-derived OS cell lines maintains their potential use as future therapeutic targets.
Collapse
|
5
|
Li K, Zhang Q, Niu D, Xing H. Mining miRNAs' Expressions in Glioma Based on GEO Database and Their Effects on Biological Functions. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5637864. [PMID: 33102581 PMCID: PMC7576330 DOI: 10.1155/2020/5637864] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE To mine miR expression in glioma based on the Gene Expression Omnibus (GEO) database and to explore its effects on biological functions. METHODS Differentially expressed miRs in glioma-related chips were found out based on the GEO database. Fifty patients with glioma treated in our hospital from February 2012 to July 2013 (observation group, OG) and a further 50 healthy people undergoing physical examinations (control group, CG) were enrolled. miR-873-5p expression in serum and in U87, T98G, U251, LN-229, and HEK-293T cells was tested by qRT-PCR. T98G and U251 cells were transfected with miR-873-5p-mimics and miR-NC sequences. The expression in the two cells was also tested by qRT-PCR. The proliferation, invasion, and apoptosis of the transfected cells were, respectively, tested by MTT assay, Transwell, and flow cytometry. The patients were followed up for 5 years to observe their survival. RESULTS miR-873-5p expression in OG was remarkably higher than that in CG (p < 0.001). miR-873-5p was closely correlated with the tumor diameter, lymph node metastasis, and TNM staging of the patients (p < 0.05). According to the plotted receiver operating characteristic (ROC) curves, the areas under the curves (AUCs) of miR-873-5p for diagnosing the disease, tumor diameter, lymph node metastasis, and TNM staging were 0.842, 0.706, 0.865, and 0.793, respectively. The 5-year and recurrence-free survival rates in the low expression group were lower than those in the high expression group. According to multivariate Cox regression analysis, tumor diameter, lymph node metastasis, and miR-873-5p were independent prognostic factors for the disease. After transfection, compared with those in the miR-NC group, T98G and U251 cells in the miR-873-5p-mimic group had remarkably higher miR-873-5p expression (p < 0.05), remarkably lower proliferation and invasion rates (p < 0.05), and a remarkably higher apoptotic rate (p < 0.05). CONCLUSIONS miR-873-5p can inhibit glioma cells to proliferate and invade, and promote their apoptosis, so it is expected to become a potential diagnostic index and therapeutic target for glioma.
Collapse
Affiliation(s)
- Ke Li
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou 256603, China
| | - Qi Zhang
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou 256603, China
| | - Duan Niu
- Department of Pediatrics, Binchengqu Shili Hospital, Binzhou 256600, China
| | - Hailong Xing
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou 256603, China
| |
Collapse
|
6
|
Yue N, Ye M, Zhang R, Guo Y. MiR-449b-5p targets lncRNA PSMG3-AS1 to suppress cancer cell proliferation in lung adenocarcinoma. BMC Pulm Med 2020; 20:152. [PMID: 32471413 PMCID: PMC7260832 DOI: 10.1186/s12890-020-01189-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/18/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND PSMG3-AS1 has been characterized as an oncogenic lncRNA in breast cancer, while its role in other cancers is unknown. This study investigated the role of PSMG3-AS1 in lung adenocarcinoma (LUAD). METHODS This study included 64 LUAD patients (42 males and 22 females) who were enrolled between May 2012 and May 2014. RT-qPCR was used to evaluate the expression levels of lncRNA. Cell proliferation analysis was performed using CCK-8 kit. RESULTS We found that upregulation of PSMG3-AS1 in LUAD predicted the poor survival of patients. MiR-449b-5p is downregulated in LUAD and the expression levels of LUAD were inversely correlated with the expression levels of PSMG3-AS1. MiR-449b-5p was predicted to target PSMG3-AS1, and overexpression of miR-449b-5p resulted in the downregulation of PSMG3-AS1 in LUAD cells. Cell proliferation analysis showed that overexpression of PSMG3-AS1 resulted in increased rate of cell proliferation. Overexpression of miR-449b-5p reduced the enhancing effects of PSMG3-AS1 on cell proliferation. CONCLUSIONS Therefore, miR-449b-5p may target PSMG3-AS1 in LUAD to suppress cancer cell proliferation.
Collapse
Affiliation(s)
- Na Yue
- Department of Pathology, the 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital), NO.789 East Suzhou Street, New Urban Area, Urumqi, 830000, Xinjiang, China
| | - Ming Ye
- Department of Pathology, the 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital), NO.789 East Suzhou Street, New Urban Area, Urumqi, 830000, Xinjiang, China
| | - Ran Zhang
- Department of Pathology, the 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital), NO.789 East Suzhou Street, New Urban Area, Urumqi, 830000, Xinjiang, China
| | - Yunquan Guo
- Department of Pathology, the 3rd Affiliated Teaching Hospital of Xinjiang Medical University (Affiliated Cancer Hospital), NO.789 East Suzhou Street, New Urban Area, Urumqi, 830000, Xinjiang, China.
| |
Collapse
|