1
|
Huang X, Zhang D, Zhang D, Guo J, Gu G, Wang Y, Wu G, Wang C, Fu B, Li K. Decoding PTEN: from biological functions to signaling pathways in tumors. Mol Biol Rep 2024; 51:1089. [PMID: 39446204 DOI: 10.1007/s11033-024-10049-y] [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: 08/16/2024] [Accepted: 10/21/2024] [Indexed: 10/25/2024]
Abstract
The tumor suppressor gene Phosphatase and tensin homologue deleted on chromosome 10 (PTEN), possessing both protein and lipid phosphatase activities, is frequently mutated in various human cancers. PTEN aberrations disrupt critical cellular processes like proliferation, apoptosis, migration, and invasion, thereby promoting tumor growth. In the cells, PTEN localizes to the nucleus, cytoplasm, or cell membrane, and its roles depends on the subcellular localization. PTEN is regulated at the transcriptional, post-transcriptional, and post-translational levels, implying that its functions on the tumors are complex. The relationship between PTEN abnormalities and tumors has garnered significant interest in recent years. PTEN regulates essential cellular processes involved in tumorigenesis. Mutations or deletions in the PTEN gene often correlate with unfavorable prognosis and increased cancer recurrence. Numerous studies suggest that PTEN expression levels in tumors could be a valuable biomarker for cancer diagnosis, treatment, and predicting patient outcomes. This paper provides a comprehensive review of the biological function, regulatory mechanisms, and post-translational modifications of PTEN. Furthermore, this review explores the expression and regulation of PTEN in different tumor types, as well as its interactions with environmental factors in tumorigenesis. This comprehensive analysis aims to deepen our understanding of the signaling pathways between PTEN and cancer.
Collapse
Affiliation(s)
- Xueping Huang
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, PR China
| | - Dongyan Zhang
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, PR China
| | - Di Zhang
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, PR China
| | - Jianran Guo
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, PR China
| | - Guohao Gu
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, PR China
| | - Yingying Wang
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, PR China
| | - Guohao Wu
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, PR China
| | - Chuanbao Wang
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong Province, PR China
| | - Bo Fu
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, PR China.
| | - Keyi Li
- Department of Precision Biomedical Key Laboratory, Liaocheng People's Hospital, Shandong Provincial Key Medical and Health Laboratory of Precision Medicine for Aging Intervention and Active Health, Liaocheng, PR China.
- Department of Stomatology, Liaocheng People's Hospital, Liaocheng, Shandong Province, PR China.
| |
Collapse
|
2
|
Wang Y, Hong J, Ge S, Wang T, Mei Z, He M, Liu Y, Fang J, Liu C, Yang L, Yuan Y. 9-O-monoethyl succinate berberine effectively blocks the PI3K/AKT signaling pathway by targeting Wnt5a protein in inhibiting osteosarcoma growth. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155430. [PMID: 39047413 DOI: 10.1016/j.phymed.2024.155430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Osteosarcoma (OS) is the most common primary bone malignancy, mainly affecting children, adolescents, and young adults, followed by the elderly, with a high propensity for local invasion and metastasis. Although surgery combined with chemotherapy has greatly improved the prognosis of patients with OS, the prognosis for metastatic or recurrent OS is still unsatisfactory. The research community has struggled to develop an effective chemotherapy treatment regimen for this tumor. For the creation of an OS drug, our research team has effectively developed and manufactured a new drug named 9-O-monoethyl succinate berberine (B2). PURPOSE In this study, we aimed to investigate the roles and functions of B2 in the treatment of OS. METHODS Human OS cell lines and mouse OS cell lines were used in vitro cell experiments, while BALB/c mice and BALB/c nude mice were used in vivo animal experiments. To investigate the molecular mechanism of B2 treatment, antibody microarray analysis, proteomic analysis, quantitative real-time PCR, immunohistochemical labeling, and western blotting analysis were mostly carried out. We assessed the impact of B2 on OS therapy and the underlying molecular pathways based on in vivo and in vitro studies. RESULTS Our findings demonstrated that B2 has the ability to inhibit the proliferation, migration, and invasion of OS cell lines, while also induce apoptosis in vitro. Additionally, our results suggested that B2 could effectively impede the growth of OS and has less heart and lung damage than cisplatin in vivo. In terms of mechanism, we discovered that the Wnt5a protein is significantly expressed in OS cell lines. Knockdown of Wnt5a can restrict OS cell lines proliferation, and overexpression of Wnt5a had the opposite results. B2 also had a strong affinity with Wnt5a and can inhibit the PI3K/AKT signaling pathway by targeting Wnt5a. Tumor cells proliferation can be inhibited by blocking the PI3K/AKT signaling pathway, and Wnt5a-mediated inactivation of the PI3K/AKT signaling pathway after B2 treatment. In vitro and in vivo experiments with Wnt5a overexpression, B2 significantly inhibited tumor growth, migration, and invasion. Moreover, B2 and Wnt5a also have a strong structural binding ability (binding energy of -7.567 ± 0.084 kcal/mol, binding values of 2.860 ± 0.434 µM), and three hydrogen bonds are generated at the docking positions of amino acids GLN286, ASN288, and ASN292. CONCLUSION In summary, our study confirmed for the first time that the growth of OS is related to abnormal overexpression of Wnt5a protein, and designed a novel small molecule inhibitor named B2 targeting Wnt5a protein, which inhibits OS growth by mediating PI3K/AKT signaling pathway by targeting Wnt5a protein. Our research laid the groundwork for the promotion of B2 as a new anticancer drug and revealed an innovative chemotherapeutic strategy for OS therapy.
Collapse
Affiliation(s)
- Yanquan Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Jinhuan Hong
- Department of Pharmacy (The University Key Laboratory of Drug Research, Heilongjiang Province), The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Shiyu Ge
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Tong Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Zhongting Mei
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Mingyu He
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Ying Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Jiaxin Fang
- Department of Pharmacy (The University Key Laboratory of Drug Research, Heilongjiang Province), The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Chuang Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China
| | - Lei Yang
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery of Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China; NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Ye Yuan
- Department of Pharmacy (The University Key Laboratory of Drug Research, Heilongjiang Province), The Second Affiliated Hospital of Harbin Medical University, Harbin, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone (2019RU070), Chinese Academy of Medical Sciences, Harbin 150081, China; Department of Clinical Pharmacology, College of Pharmacy, Harbin Medical University, Harbin, China.
| |
Collapse
|
3
|
Leuci A, Marano M, Millet M, Lienhart A, Desage S, Chapurlat R, Dargaud Y. Deciphering the circulating microRNA signature of hemophilic arthropathy. Thromb Res 2024; 241:109099. [PMID: 39079229 DOI: 10.1016/j.thromres.2024.109099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/22/2024] [Accepted: 07/17/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Haemophilic arthropathy (HArt) is a serious complication in patients with hemophilia. Early diagnosis and treatment are essential to minimise the development of HArt. The use of biomarkers may improve early diagnosis of HArt. Circulating microRNAs (miRNAs) are small, non-coding RNAsthat regulate gene expression, and are being investigated as promising biomarkers due to their role in joint and bone metabolism. AIMS To investigate differential expression of miRNAs and their relationship to arthropathy in patients with hemophilia A. METHODS miRNA expression was examined in a pilot study followed by a validation study (100 hemophilia A patients with [n = 83] and without HArt [n = 17], 14 controls). Differential miRNA expression was investigated using real-time quantitative PCR. RESULTS The pilot study identified 2 miRNAs differentially expressed in patients with Hart (Pettersson score ≥ 1), after adjusting for the false discovery rate (FDR). The validation study evaluated these 2 miRNAs. The results demonstrated that two miRNAs (miR- 208a-3p and 524-3p) were significantly underexpressed in plasma of patients with HArt compared to patients without arthropathy, with FDR <0.05 (Fig. 1). In addition, 3 miRNAs (130a-3p, miR- and 506-3p) were significantly underexpressed in patients with moderate HArt (Pettersson score 4 to 7). CONCLUSIONS In this proof of concept study we identified a signature of 5 circulating miRNAs associated with Hart with potential as diagnosis tools for HArt. These miRNAs are potential negative regulators of gene expression, suggesting their activity in HArt by interfering with osteoblastic (miR- 208a-3p) and osteoclastic (miR-506-3p) differentiation to impair bone mineralization and remodeling processes, or regulating chondrogenesis (miR-335-5p). miRNAs associated with earlier stages of HArt will be further investigated in a sub-study of the prospective clinical trial PROVE, which will investigate the effects of long-term prophylaxis with simoctocog alfa versus emicizumab in adults with hemophilia A.
Collapse
Affiliation(s)
- Alexandre Leuci
- UR4609 Thrombosis & Haemostasis, University of Lyon Claude Bernard 1 (UCLB1), Lyon, France
| | - Muriel Marano
- UR4609 Thrombosis & Haemostasis, University of Lyon Claude Bernard 1 (UCLB1), Lyon, France
| | - Marjorie Millet
- INSERM UMR 1033, University of Lyon Claude Bernard 1 (UCLB1), Lyon, France
| | - Anne Lienhart
- French Reference Center for Hemophilia, Clinical Haemostasis Unit, Louis Pradel Hospital, Hospices Civils de Lyon, Lyon, France
| | - Stephanie Desage
- French Reference Center for Hemophilia, Clinical Haemostasis Unit, Louis Pradel Hospital, Hospices Civils de Lyon, Lyon, France
| | - Roland Chapurlat
- INSERM UMR 1033, University of Lyon Claude Bernard 1 (UCLB1), Lyon, France; Department of Rheumatology, Edouard Herriot University Hospital, Lyon, France
| | - Yesim Dargaud
- UR4609 Thrombosis & Haemostasis, University of Lyon Claude Bernard 1 (UCLB1), Lyon, France; French Reference Center for Hemophilia, Clinical Haemostasis Unit, Louis Pradel Hospital, Hospices Civils de Lyon, Lyon, France.
| |
Collapse
|
4
|
Wang C, Zhang Y, Kong W, Rong X, Zhong Z, Jiang L, Chen S, Li C, Zhang F, Jiang J. Delivery of miRNAs Using Nanoparticles for the Treatment of Osteosarcoma. Int J Nanomedicine 2024; 19:8641-8660. [PMID: 39188861 PMCID: PMC11346496 DOI: 10.2147/ijn.s471900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 07/31/2024] [Indexed: 08/28/2024] Open
Abstract
Osteosarcoma is the predominant primary malignant bone tumor that poses a significant global health challenge. MicroRNAs (miRNAs) that regulate gene expression are associated with osteosarcoma pathogenesis. Thus, miRNAs are potential therapeutic targets for osteosarcoma. Nanoparticles, widely used for targeted drug delivery, facilitate miRNA-based osteosarcoma treatment. Numerous studies have focused on miRNA delivery using nanoparticles to inhibit the progress of osteosarcoma. Polymer-based, lipid-based, inorganic-based nanoparticles and extracellular vesicles were used to deliver miRNAs for the treatment of osteosarcoma. They can be modified to enhance drug loading and delivery capabilities. Also, miRNA delivery was combined with traditional therapies, for example chemotherapy, to treat osteosarcoma. Consequently, miRNA delivery offers promising therapeutic avenues for osteosarcoma, providing renewed hope for patients. This review emphasizes the studies utilizing nanoparticles for miRNA delivery in osteosarcoma treatment, then introduced and summarized the nanoparticles in detail. And it also discusses the prospects for clinical applications.
Collapse
Affiliation(s)
- Chengran Wang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Yihong Zhang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Weihui Kong
- Department of Stomatology, the First Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Xin’ao Rong
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Ziming Zhong
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Lei Jiang
- Department of Geriatric Medicine, Changchun Central Hospital, Changchun, Jilin Province, People’s Republic of China
| | - Shuhan Chen
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Chuang Li
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Fuqiang Zhang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Jinlan Jiang
- Department of Scientific Research Center, China–Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| |
Collapse
|
5
|
Shao W, Feng Y, Huang J, Li T, Gao S, Yang Y, Li D, Yang Z, Yao Z. Interaction of ncRNAs and the PI3K/AKT/mTOR pathway: Implications for osteosarcoma. Open Life Sci 2024; 19:20220936. [PMID: 39119480 PMCID: PMC11306965 DOI: 10.1515/biol-2022-0936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumor in children and adolescents, and is characterized by high heterogeneity, high malignancy, easy metastasis, and poor prognosis. Recurrence, metastasis, and multidrug resistance are the main problems that limit the therapeutic effect and prognosis of OS. PI3K/AKT/mTOR signaling pathway is often abnormally activated in OS tissues and cells, which promotes the rapid development, metastasis, and drug sensitivity of OS. Emerging evidence has revealed new insights into tumorigenesis through the interaction between the PI3K/AKT/mTOR pathway and non-coding RNAs (ncRNAs). Therefore, we reviewed the interactions between the PI3K/AKT/mTOR pathway and ncRNAs and their implication in OS. These interactions have the potential to serve as cancer biomarkers and therapeutic targets in clinical applications.
Collapse
Affiliation(s)
- Weilin Shao
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Yan Feng
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Jin Huang
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Tingyu Li
- Clinical Oncology Institute, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Shengguai Gao
- Clinical Oncology Institute, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Yihao Yang
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Dongqi Li
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Zuozhang Yang
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Zhihong Yao
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan, 650118, China
- Department of Cancer Research Institute, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan, 650118, China
| |
Collapse
|
6
|
Shi Q, Xu J, Chen C, Hu X, Wang B, Zeng F, Ren T, Huang Y, Guo W, Tang X, Ji T. Direct contact between tumor cells and platelets initiates a FAK-dependent F3/TGF-β positive feedback loop that promotes tumor progression and EMT in osteosarcoma. Cancer Lett 2024; 591:216902. [PMID: 38641310 DOI: 10.1016/j.canlet.2024.216902] [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: 02/10/2024] [Revised: 03/31/2024] [Accepted: 04/15/2024] [Indexed: 04/21/2024]
Abstract
Platelets have received growing attention for their roles in hematogenous tumor metastasis. However, the tumor-platelet interaction in osteosarcoma (OS) remains poorly understood. Here, using platelet-specific focal adhesion kinase (FAK)-deficient mice, we uncover a FAK-dependent F3/TGF-β positive feedback loop in OS. Disruption of the feedback loop by inhibition of F3, TGF-β, or FAK significantly suppresses OS progression. We demonstrate that OS F3 initiated the feedback loop by increasing platelet TGF-β secretion, and platelet-derived TGF-β promoted OS F3 expression in turn and modulated OS EMT process. Immunofluorescence results indicate platelet infiltration in OS niche and we verified it was mediated by platelet FAK. In addition, platelet FAK was proved to mediate platelet adhesion to OS cells, which was vital for the initiation of F3/TGF-β feedback loop. Collectively, these findings provide a rationale for novel therapeutic strategies targeting tumor-platelet interplay in metastatic OS.
Collapse
Affiliation(s)
- Qianyu Shi
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Jiuhui Xu
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Chenglong Chen
- Department of Orthopedics, Beijing Jishuitan Hospital, Beijing, China
| | - Xueyu Hu
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing, China
| | - Boyang Wang
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Fanwei Zeng
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Tingting Ren
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Yi Huang
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Wei Guo
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Xiaodong Tang
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| | - Tao Ji
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, China; Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, China
| |
Collapse
|
7
|
Sun X, Shi C, Dai J, Zhang MQ, Pei DS, Yang L. Targeting the mitochondrial protein YME1L to inhibit osteosarcoma cell growth in vitro and in vivo. Cell Death Dis 2024; 15:346. [PMID: 38769124 PMCID: PMC11106333 DOI: 10.1038/s41419-024-06722-6] [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: 05/03/2023] [Revised: 05/02/2024] [Accepted: 05/03/2024] [Indexed: 05/22/2024]
Abstract
Exploring novel diagnostic and therapeutic biomarkers is extremely important for osteosarcoma. YME1 Like 1 ATPase (YME1L), locating in the mitochondrial inner membrane, is key in regulating mitochondrial plasticity and metabolic activity. Its expression and potential functions in osteosarcoma are studied in the present study. We show that YME1L mRNA and protein expression is significantly elevated in osteosarcoma tissues derived from different human patients. Moreover, its expression is upregulated in various primary and immortalized osteosarcoma cells. The Cancer Genome Atlas database results revealed that YME1L overexpression was correlated with poor overall survival and poor disease-specific survival in sarcoma patients. In primary and immortalized osteosarcoma cells, silencing of YME1L through lentiviral shRNA robustly inhibited cell viability, proliferation, and migration. Moreover, cell cycle arrest and apoptosis were detected in YME1L-silenced osteosarcoma cells. YME1L silencing impaired mitochondrial functions in osteosarcoma cells, causing mitochondrial depolarization, oxidative injury, lipid peroxidation and DNA damage as well as mitochondrial respiratory chain complex I activity inhibition and ATP depletion. Contrarily, forced YME1L overexpression exerted pro-cancerous activity and strengthened primary osteosarcoma cell proliferation and migration. YME1L is important for Akt-S6K activation in osteosarcoma cells. Phosphorylation of Akt and S6K was inhibited after YME1L silencing in primary osteosarcoma cells, but was strengthened with YME1L overexpression. Restoring Akt-mTOR activation by S473D constitutively active Akt1 mitigated YME1L shRNA-induced anti-osteosarcoma cell activity. Lastly, intratumoral injection of YME1L shRNA adeno-associated virus inhibited subcutaneous osteosarcoma xenograft growth in nude mice. YME1L depletion, mitochondrial dysfunction, oxidative injury, Akt-S6K inactivation, and apoptosis were detected in YME1L shRNA-treated osteosarcoma xenografts. Together, overexpressed YME1L promotes osteosarcoma cell growth, possibly by maintaining mitochondrial function and Akt-mTOR activation.
Collapse
Affiliation(s)
- Xu Sun
- Department of Hand and Foot Surgery, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, China
| | - Ce Shi
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Orthopedics, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, China
| | - Jin Dai
- Department of Orthopedics, Suzhou Wujiang District Children's Hospital, Suzhou, China
| | | | - Dong-Sheng Pei
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Lei Yang
- Department of Orthopedics, Wujin Hospital Affiliated with Jiangsu University, Changzhou, China.
| |
Collapse
|
8
|
Di Gregorio J, Di Giuseppe L, Terreri S, Rossi M, Battafarano G, Pagliarosi O, Flati V, Del Fattore A. Protein Stability Regulation in Osteosarcoma: The Ubiquitin-like Modifications and Glycosylation as Mediators of Tumor Growth and as Targets for Therapy. Cells 2024; 13:537. [PMID: 38534381 DOI: 10.3390/cells13060537] [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: 02/14/2024] [Revised: 03/11/2024] [Accepted: 03/16/2024] [Indexed: 03/28/2024] Open
Abstract
The identification of new therapeutic targets and the development of innovative therapeutic approaches are the most important challenges for osteosarcoma treatment. In fact, despite being relatively rare, recurrence and metastatic potential, particularly to the lungs, make osteosarcoma a deadly form of cancer. In fact, although current treatments, including surgery and chemotherapy, have improved survival rates, the disease's recurrence and metastasis are still unresolved complications. Insights for analyzing the still unclear molecular mechanisms of osteosarcoma development, and for finding new therapeutic targets, may arise from the study of post-translational protein modifications. Indeed, they can influence and alter protein structure, stability and function, and cellular interactions. Among all the post-translational modifications, ubiquitin-like modifications (ubiquitination, deubiquitination, SUMOylation, and NEDDylation), as well as glycosylation, are the most important for regulating protein stability, which is frequently altered in cancers including osteosarcoma. This review summarizes the relevance of ubiquitin-like modifications and glycosylation in osteosarcoma progression, providing an overview of protein stability regulation, as well as highlighting the molecular mediators of these processes in the context of osteosarcoma and their possible targeting for much-needed novel therapy.
Collapse
Affiliation(s)
- Jacopo Di Gregorio
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Laura Di Giuseppe
- Department of Clinical, Internal, Anaesthesiological and Cardiovascular Sciences, Sapienza University, 00185 Rome, Italy
| | - Sara Terreri
- Bone Physiopathology Research Unit, Translational Pediatrics and Clinical Genetics Research Division, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Michela Rossi
- Bone Physiopathology Research Unit, Translational Pediatrics and Clinical Genetics Research Division, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Giulia Battafarano
- Bone Physiopathology Research Unit, Translational Pediatrics and Clinical Genetics Research Division, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Olivia Pagliarosi
- Bone Physiopathology Research Unit, Translational Pediatrics and Clinical Genetics Research Division, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Vincenzo Flati
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Andrea Del Fattore
- Bone Physiopathology Research Unit, Translational Pediatrics and Clinical Genetics Research Division, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| |
Collapse
|
9
|
Kong Y, Li X, Zhang H, Fu B, Jiang HY, Yang HL, Dai J. Targeting POLRMT by a first-in-class inhibitor IMT1 inhibits osteosarcoma cell growth in vitro and in vivo. Cell Death Dis 2024; 15:57. [PMID: 38228583 PMCID: PMC10791695 DOI: 10.1038/s41419-024-06444-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 01/18/2024]
Abstract
Osteosarcoma (OS) is a highly aggressive form of bone cancer that predominantly affects adolescents and young adults. In this study, we have undertaken an investigation into the potential anti-OS cell activity of IMT1 (inhibitor of mitochondrial transcription 1), a first-in-class inhibitor of RNA polymerase mitochondrial (POLRMT). IMT1 exhibited a profound inhibitory effect on cell survival, proliferation, cell cycle progression, and migration in primary and immortalized OS cells. Furthermore, this POLRMT inhibitor elicited apoptosis in the OS cells, without, however, inducing cytotoxicity in human osteoblasts or osteoblastic cells. IMT1 disrupted mitochondrial functions in OS cells, resulting in mitochondrial depolarization, oxidative injury, lipid peroxidation, and ATP reduction in OS cells. Silencing POLRMT using targeted shRNA closely mimicked the actions of IMT1 and exerted potent anti-OS cell activity. Importantly, IMT1's effectiveness was diminished in POLRMT-silenced OS cells. Subsequent investigations revealed that IMT1 suppressed the activation of the Akt-mammalian target of rapamycin (mTOR) cascade in OS cells. IMT1 treatment or POLRMT silencing in primary OS cells led to a significant reduction in Akt1-S6K-S6 phosphorylation. Conversely, it was enhanced upon POLRMT overexpression. The restoration of Akt-mTOR activation through the introduction of a constitutively active S473D mutant Akt1 (caAkt1) mitigated IMT1-induced cytotoxicity in OS cells. In vivo, oral administration of IMT1 robustly curtailed the growth of OS xenografts in nude mice. Furthermore, IMT1 suppressed POLRMT activity, impaired mitochondrial function, repressed Akt-mTOR activation, and induced apoptosis within xenograft tissues. Collectively, these findings underscore the potent growth-inhibitory effects attributed to IMT1 via targeted POLRMT inhibition. The utilization of this POLRMT inhibitor carries substantial therapeutic promise in the context of OS treatment.
Collapse
Affiliation(s)
- Yang Kong
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Orthopedics, The First People's Hospital of ChuZhou, ChuZhou, China
| | - Xiangrong Li
- Department of Pharmacy, Kongjiang Hospital of Yangpu District, Shanghai, China
| | - Huanle Zhang
- Department of Radiotherapy, Suzhou Ninth People's Hospital, Suzhou, China
| | - Bin Fu
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hua-Ye Jiang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hui-Lin Yang
- Department of Orthopedics, the First Affiliated Hospital of Soochow University, Suzhou, China.
- Orthopedic Institute, Medical College, Soochow University, Suzhou, China.
| | - Jin Dai
- Department of Orthopedics, Suzhou Wujiang District Children's Hospital, Suzhou, China.
| |
Collapse
|
10
|
Chen Z, Ni R, Hu Y, Yang Y, Tian Y. Arnicolide D Inhibits Proliferation and Induces Apoptosis of Osteosarcoma Cells through PI3K/Akt/mTOR Pathway. Anticancer Agents Med Chem 2024; 24:1288-1294. [PMID: 38967079 DOI: 10.2174/0118715206289595240105082138] [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: 10/27/2023] [Revised: 12/22/2023] [Accepted: 01/01/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Osteosarcoma is considered as the most prevalent form of primary malignant bone cancer, prompting a pressing need for novel therapeutic options. Arnicolide D, a sesquiterpene lactone derived from the traditional Chinese herbal medicine Centipeda minima (known as E Bu Shi Cao in Chinese), showed anticancer efficacy against several kinds of cancers. However, its effect on osteosarcoma remains unclear. OBJECTIVE This study aimed to investigate the anticancer activity of arnicolide D and the underlying molecular mechanism of its action in osteosarcoma cells, MG63 and U2OS. METHODS Cell viability and proliferation were evaluated through MTT assay and colony formation assay following 24 h and 48 h treatment with different concentrations of arnicolide D. Flow cytometry was employed to examine cell cycle progression and apoptosis after 24 h treatment of arnicolide D. Western blotting was performed to determine the expression of the PI3k, Akt and m-TOR and their phosphorylated forms. RESULTS Our findings revealed that arnicolide D treatment resulted in a significant reduction in cell viability, the inhibition of proliferation, and the induction of apoptosis and cell cycle arrest in the G2/M phase. Furthermore, arnicolide D could inhibit the activation of PI3K/Akt/mTOR pathway in osteosarcoma cells. CONCLUSION Based on our results, arnicolide D demonstrated significant anti-osteosarcoma activity and held the potential to be considered as a therapeutic candidate for osteosarcoma in the future.
Collapse
Affiliation(s)
- Zhuo Chen
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| | - Renhua Ni
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| | - Yuanyu Hu
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| | - Yiyuan Yang
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| | - Yun Tian
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| |
Collapse
|
11
|
Abd Elmoneim HM, Huwait HF, Nafady-Hego H, Mohamed FA. PROGNOSTIC IMPLICATIONS OF PD-L1 EXPRESSION AND LOSS OF PTEN IN PATIENTS WITH RHABDOMYOSARCOMA, EWING'S SARCOMA AND OSTEOSARCOMA. Exp Oncol 2023; 45:337-350. [PMID: 38186021 DOI: 10.15407/exp-oncology.2023.03.337] [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: 12/28/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND In children, osteosarcoma (OS), Ewing's sarcoma (ES), and rhabdomyosarcoma (RMS) are the most common sarcomas. A link between the anti-programmed death ligand-1 PD-L1 and the tumor suppressor phosphatase and tensin homologue (PTEN) expression has been described in many tumors. The aim of this work is to determine clinicopathological relationships and the possible prognostic significance of PD-L1 and PTEN expression in rhabdomyosarcoma (RMS), Ewing's sarcoma (ES), and osteosarcoma (OS). MATERIALS AND METHODS Expression of PD-L1 and PTEN were examined by immunohistochemistry in 45 archival RMS, ES, and OS cases. RESULTS The positive expression of PD-L1 was found in 16.7% and 31.6% of ES and OS, respectively. The negative PD-L1 was related to a substantially longer survival in ES cases (p = 0.045), but positive PD-L1 expression was significantly associated with the increased tumor stage and vascular invasion in the OS cases (p = 0.005 and p = 0.002), respectively. On the other hand, PTEN loss was strongly associated with deep tumor, high tumor grade, and recurrence in RMS (p = 0.002, p = 0.045, and p = 0.026, respectively). However, PTEN loss was significantly absent in ES as tumor grade increased (p = 0.031). It is noteworthy that tumor recurrence, the loss of PTEN, and positive PD-L1 were all considered predictive factors in OS patients (p = 0.045, p = 0.032, and p = 0.02, respectively). CONCLUSIONS In children, OS and ES have positive PD-L1 expression, which has an independent unfavorable prognostic effect and raises the possibility of using PD-L1 as a therapeutic target. OS, ES, and RMS prognosis are all predicted by PTEN loss.
Collapse
Affiliation(s)
- H M Abd Elmoneim
- Department of Pathology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
- Department of Pathology, Faculty of Medicine, Minia University, Minia, Egypt
| | - H F Huwait
- Department of Pathology, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - H Nafady-Hego
- Microbiology and Immunology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
- Laboratory Department, Al Tahrir Medical Center, Doha, Qatar
| | - Fez A Mohamed
- Department of Pathology, Faculty of Medicine, Minia University, Minia, Egypt
- Department of Pathological Sciences, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia
| |
Collapse
|
12
|
Fu L, Xu S, Zhou Y, Huang J, Qiu J, Huang P. Knockdown of LncRNA DICER1-AS1 arrests the cell cycle, inhibits cell proliferation, and induces cell apoptosis by regulating CDC5L nuclear transfer in osteosarcoma. Connect Tissue Res 2023; 64:519-531. [PMID: 37310074 DOI: 10.1080/03008207.2023.2223289] [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] [Received: 07/29/2022] [Revised: 12/05/2022] [Accepted: 12/14/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND DICER1-AS1 is reported to promote the progression and disturb the cell cycle in osteosarcoma; however, its mechanism has rarely been studied. MATERIALS AND METHODS DICER1-AS1 expression levels were evaluated by qPCR and fluorescence in situ hybridization (FISH). The total, nuclear, and cytosolic levels of CDC5L were measured by western blotting and immunofluorescence (IF). Cell proliferation, apoptosis, and cell cycle analyses were conducted using the colony formation, CCK-8 assay, terminal transferase-mediated UTP nick end-labeling kit (TUNEL) assay, and flow cytometry. Levels of cell proliferation-, cell cycle-, and cell apoptosis-related proteins were determined by western blotting. RNA immunoprecipitation (RIP) and RNA pull-down assays were conducted to evaluate the relationship between DICER1-AS1 and CDC5L. RESULTS LncRNA DICER1-AS1 was highly expressed in samples of osteosarcoma tissue and in osteosarcoma cell lines. DICER1-AS1 knockdown inhibited cell proliferation, promoted cell apoptosis, and disturbed the cell cycle. Moreover, DICER1-AS1 was found to bind with CDC5L, and knockdown of DICER-AS1 inhibited the nuclear transfer of CDC5L. DICER1-AS1 knockdown also reversed the effects of CDC5L overexpression on cell proliferation, apoptosis, and the cell cycle. Moreover, CDC5L inhibition suppressed cell proliferation, promoted cell apoptosis, and disturbed the cell cycle, and those effects were further enhanced by DICER1-AS1 knockdown. Finally, DICER1-AS knockdown inhibited tumor growth and proliferation, and promoted cell apoptosis in vivo. CONCLUSION LncRNA DICER1-AS1 knockdown inhibits the nuclear transfer of CDC5L protein, arrests the cell cycle, and induces apoptosis to suppress the development of osteosarcoma. Our results suggest a novel target (DICER1-AS1) for treatment of osteosarcoma.
Collapse
Affiliation(s)
- Laihua Fu
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Songfeng Xu
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Yang Zhou
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Jingyang Huang
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Jin Qiu
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Pengzhou Huang
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| |
Collapse
|
13
|
Sadrkhanloo M, Paskeh MDA, Hashemi M, Raesi R, Bahonar A, Nakhaee Z, Entezari M, Beig Goharrizi MAS, Salimimoghadam S, Ren J, Nabavi N, Rashidi M, Dehkhoda F, Taheriazam A, Tan SC, Hushmandi K. New emerging targets in osteosarcoma therapy: PTEN and PI3K/Akt crosstalk in carcinogenesis. Pathol Res Pract 2023; 251:154902. [PMID: 37922723 DOI: 10.1016/j.prp.2023.154902] [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] [Received: 08/21/2023] [Revised: 10/14/2023] [Accepted: 10/19/2023] [Indexed: 11/07/2023]
Abstract
Osteosarcoma (OS) is a malignant bone carcinoma that affects people in childhood and adulthood. The heterogeneous nature and chromosomal instability represent certain characteristics of OS cells. These cancer cells grow and migrate abnormally, making the prognosis undesirable for patients. Conventional and current treatments fail to completely eradicate tumor cells, so new therapeutics targeting genes may be considered. PI3K/Akt is a regulator of events such as growth, cell death, migration, and differentiation, and its expression changes during cancer progression. PTEN reduces PI3K/Akt expression, and its mutations and depletions have been reported in various tumors. Experimental evidence shows that there is upregulation of PI3K/Akt and downregulation of PTEN in OS. Increasing PTEN expression may suppress PI3K/Akt to minimize tumorigenesis. In addition, PI3K/Akt shows a positive association with growth, metastasis, EMT and metabolism of OS cells and inhibits apoptosis. Importantly, overexpression of PI3K/Akt causes drug resistance and radio-resistance and its level can be modulated by miRNAs, lncRNAs and circRNAs. Silencing PI3K/Akt by compounds and drugs can suppress OS. Here, we review in detail the function of the PTEN/PI3K/Akt in OS, revealing its biological function, function in tumor progression, resistance to therapy, and pharmacological significance.
Collapse
Affiliation(s)
| | - Mahshid Deldar Abad Paskeh
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Rasoul Raesi
- Department of Health Services Management, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical-Surgical Nursing, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Bahonar
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Zahra Nakhaee
- Medical School, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6 Vancouver, BC, Canada
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Farshid Dehkhoda
- Department of Orthopedics, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
14
|
Mokhtari Y, Yousefi AM, Bashash D. Inhibition of PI3K Signaling Intensified the Antileukemic Effects of Pioglitazone: New Insight into the Application of PPARγ Stimulators in Acute Lymphoblastic Leukemia. Indian J Hematol Blood Transfus 2023; 39:546-556. [PMID: 37786817 PMCID: PMC10542079 DOI: 10.1007/s12288-023-01650-5] [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: 09/30/2022] [Accepted: 03/20/2023] [Indexed: 04/05/2023] Open
Abstract
Over the past two decades, molecular targeted therapy has revolutionized the landscape of cancer treatment due to lower side effects as well as higher anticancer effects. Peroxisome proliferator-activated receptor gamma (PPARγ) is a member of the nuclear hormone receptor which plays a crucial role in cell proliferation and death and the efficacy of PPARγ ligands either as monotherapy or in combination with traditional chemotherapy drugs has been proved by recent studies. In this study, we aimed to investigate the effects of pioglitazone, a well-known PPARγ stimulator, in ALL-derived NALM6 cells by using trypan blue assay, MTT assay, and flow cytometry analysis. Moreover, to investigate the molecular mechanism action of pioglitazone in these cells, we assessed the possible alterations in the expression of some target genes which regulate cell proliferation, apoptosis, and autophagy system. Our result demonstrated that pioglitazone induced a remarkable antileukemic effect on NALM6 cells through a PTEN-mediated manner. Based on the fact that PI3K hyperactivation is one of the main properties of ALL cells, the effects of PI3K inhibition using CAL-101 on pioglitazone-induced cytotoxicity were evaluated by combinatorial experiments. Moreover, the result of cell cycle assay and qRT-PCR demonstrated that pioglitazone-CAL-101 induced antileukemic effect mainly through induction of p21 and p27-mediated G1 arrest. Additionally, our result showed that inhibition of proteasome and autophagy system, two main cellular processes, increased the antileukemic effects of the agents. Taken together, we suggest a novel therapeutic application for PPARγ stimulators as a single agent or in combination with PI3K inhibitors that should be clinically evaluated in ALL patients.
Collapse
Affiliation(s)
- Yazdan Mokhtari
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir-Mohammad Yousefi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Huang R, Xu M, Guo W, Cheng M, Dong R, Tu J, Xu S, Zou C. Network pharmacology and experimental verification-based strategy for exploring the mechanisms of luteolin in the treatment of osteosarcoma. Cancer Cell Int 2023; 23:213. [PMID: 37749554 PMCID: PMC10521544 DOI: 10.1186/s12935-023-03046-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 08/29/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND Luteolin is an active ingredient in various traditional Chinese medicines for the treatment of multiple tumors. However, the mechanisms of its inhibitory effect on osteosarcoma proliferation and metastasis remain unclear. PURPOSE To elucidate the anti-osteosarcoma mechanisms of luteolin based on network pharmacology and experimental verification. STUDY DESIGN Integrate network pharmacology predictions, scRNA-seq analysis, molecular docking, and experimental validation. METHODS Luteolin-related targets and osteosarcoma-associated targets were collected from several public databases. The luteolin against osteosarcoma targets were screened and a PPI network was constructed to identify the hub targets. The GO and KEGG enrichment of osteosarcoma-associated targets and luteolin against osteosarcoma targets were performed. And scRNA-seq analysis was performed to determine the distribution of the core target expression in OS tissues. Molecular docking, cell biological assays, and osteosarcoma orthotopic mouse model was performed to validate the inhibitory effect and mechanisms of luteolin on osteosarcoma proliferation and metastasis. RESULTS Network pharmacology showed that 251 luteolin against osteosarcoma targets and 8 hub targets including AKT1, ALB, CASP3, IL6, JUN, STAT3, TNF, and VEGFA, and the PI3K-AKT signaling pathway might play an important role in anti-osteosarcoma of luteolin. Analysis of public data revealed that AKT1, IL6, JUN, STAT3, TNF, and VEGFA expression in OS tissue was significantly higher than that in normal bones, and the diagnostic value of VEGFA for overall survival and metastasis was increased over time. scRNA-seq analysis revealed significantly higher expression of AKT1, STAT3, and VEGFA in MYC+ osteoblastic OS cells, especially in primary samples. Moreover, the docking activity between luteolin and the hub targets was excellent, as verified by molecular docking. Experimental results showed that luteolin could inhibit cell viability and significantly decrease the expression of AKT1, STAT3, IL6, TNF, and VEGFA, and luteolin could also inhibit osteosarcoma proliferation and metastasis in osteosarcoma orthotopic mouse model. CONCLUSION This study shows that luteolin may regulate multiple signaling pathways by targeting various genes like AKT1, STAT3, IL6, TNF, and VEGFA to inhibit osteosarcoma proliferation and metastasis.
Collapse
Affiliation(s)
- Renxuan Huang
- Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Guangzhou, 510080, China
| | - Mingxian Xu
- Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Guangzhou, 510080, China
| | - Weitang Guo
- Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Guangzhou, 510080, China
| | - Mingzhe Cheng
- Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Guangzhou, 510080, China
| | - Rui Dong
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Sciences, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, 510080, China
| | - Jian Tu
- Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Guangzhou, 510080, China
| | - Shao Xu
- Department of Stomatology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Road, Guangzhou, 510630, China.
| | - Changye Zou
- Musculoskeletal Oncology Center, The First Affiliated Hospital of Sun Yat-Sen University, No. 58, 2nd Zhongshan Road, Guangzhou, 510080, China.
| |
Collapse
|
16
|
Tong Y, Jiang L, Cui Y, Pi Y, Gong Y, Zhao D. Clinical characteristic-assisted surgical benefit stratification for resection of primary tumor in patients with advanced primary malignant bone neoplasms: a population-based propensity score-matched analysis. Front Oncol 2023; 13:960502. [PMID: 37746283 PMCID: PMC10512233 DOI: 10.3389/fonc.2023.960502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
Background Primary tumor resection (PTR) is the standard treatment for patients with primary malignant bone neoplasms (PMBNs). However, it remains unclear whether patients with advanced PMBNs still benefit from PTR. This study aimed to develop a prediction model to estimate the beneficial probability of PTR for this population. Methods This study extracted data from patients diagnosed with advanced PMBNs, as recorded in the Surveillance, Epidemiology, and End Results (SEER) database, with the period from 2004 to 2015. The patient cohort was then bifurcated into two groups: those who underwent surgical procedures and the non-surgery group. Propensity score matching (PSM) was utilized to mitigate any confounding factors in the study. The survival rates of patients from both the surgical and non-surgery groups were evaluated using Kaplan-Meier (K-M) curves analysis. Moreover, the study used this method to assess the capacity of the nomogram to distinguish patients likely to derive benefits from surgical intervention. The study was grounded in the hypothesis that patients who underwent PTR and survived beyond the median overall survival (OS) time would potentially benefit from the surgery. Subsequently, logistic regression analysis was performed to ascertain significant predictors, facilitating the development of a nomogram. This nomogram was subjected to both internal and external validation using receiver operating characteristic curves, area under the curve analysis, calibration plots, and decision curve analysis. Results The SEER database provided a total of 839 eligible patients for the study, among which 536 (63.9%) underwent PTR. Following a 2:1 PSM analysis, patients were classified into two groups: 364 patients in the surgery group and 182 patients in the non-surgery group. Both K-M curves and multivariate Cox regression analysis revealed that patients who received PTR had a longer survival duration, observed both before and after PSM. Crucial factors such as age, M stage, and tumor size were identified to be significantly correlated with surgical benefits in patients with advanced PMBNs. Subsequently, a nomogram was developed that uses these independent predictors. The validation of this predictive model confirmed its high accuracy and excellent discrimination ability of the nomogram to distinguish patients who would most likely benefit from surgical intervention. Conclusion In this study, we devised a user-friendly nomogram to forecast the likehood of surgical benefits for patients diagnosed with advanced PMBNs. This tool facilitates the identification of the most suitable candidates for PTR, thus promoting more discerning and effective use of surgical intervention in this patient population.
Collapse
Affiliation(s)
- Yuexin Tong
- Department of Orthopedics, The China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Liming Jiang
- Department of Orthopedics, The China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Yuekai Cui
- The Second Clinical Medical School of The Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yangwei Pi
- Department of Orthopedics, The China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Yan Gong
- Department of Orthopedics, The China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Dongxu Zhao
- Department of Orthopedics, The China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
17
|
Wei Z, Xia K, Zhou B, Zheng D, Guo W. Zyxin Inhibits the Proliferation, Migration, and Invasion of Osteosarcoma via Rap1-Mediated Inhibition of the MEK/ERK Signaling Pathway. Biomedicines 2023; 11:2314. [PMID: 37626810 PMCID: PMC10452081 DOI: 10.3390/biomedicines11082314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/02/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Zyxin (ZYX) is an actin-interacting protein with unknown biological functions in patients with osteosarcoma. This research sought to understand how ZYX affects the biological behavior of osteosarcoma cells and to identify the associated mechanism. Firstly, ZYX expression was decreased in osteosarcoma, and its higher expression indicated better outcomes in patients with osteosarcoma. ZYX overexpression significantly inhibited the proliferation, migration, and invasion of osteosarcoma cells, whereas ZYX silencing resulted in the opposite trend. Subsequently, we found that the Rap1 signaling pathway was significantly correlated with ZYX expression as reported in The Cancer Genome Atlas's database using bioinformatic analysis. Moreover, we found that ZYX overexpression regulated the Rap1/MEK/ERK axis, and osteosarcoma cell growth, migration, and invasion were consequently restrained. Additionally, by administering tumor cells subcutaneously to nude mice, a mouse model of transplanted tumors was created. Compared to the control group, the ZYX overexpression group's tumors were lighter and smaller, and the ZYX/Rap1 axis was activated in the ZYX overexpression group. Taken together, our results suggest that ZYX inhibits osteosarcoma cell proliferation, migration, and invasion by regulating the Rap1/MEK/ERK signaling pathway. ZYX might be crucial in the clinical management of osteosarcoma and is a promising novel therapeutic target in patients with this disease.
Collapse
Affiliation(s)
- Zhun Wei
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Kezhou Xia
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Bin Zhou
- Department of Orthopedics, Ezhou Central Hospital, Ezhou 436000, China
| | - Di Zheng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Weichun Guo
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
18
|
Xia K, Zheng D, Wei Z, Liu W, Guo W. TRIM26 inhibited osteosarcoma progression through destabilizing RACK1 and thus inactivation of MEK/ERK signaling. Cell Death Dis 2023; 14:529. [PMID: 37591850 PMCID: PMC10435491 DOI: 10.1038/s41419-023-06048-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/21/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023]
Abstract
Osteosarcoma is a highly aggressive malignant tumor that is common in the pediatric population and has a high rate of disability and mortality. Recent studies have suggested that the tripartite motif-containing family genes (TRIMs) play critical roles in oncogenesis in several cancers. TRIM26, one of the TRIMs family genes, was more frequently reported to exert a tumor-suppressive role, while its detailed functional roles in the osteosarcoma progression were still unknown and require further investigation. Herein, we found that TRIM26 was markedly downregulated in osteosarcoma tissues and cells. Survival analysis revealed that higher expression of TRIM26 was associated with better prognosis and its expression was an independent protective factor in osteosarcoma. Functional analysis demonstrated that overexpression of TRIM26 inhibited osteosarcoma cell proliferation and invasion via inhibiting the EMT process and MEK/ERK signaling. In contrast, the silence of TRIM26 caused the opposite effect. RACK1, a member of the Trp-Asp repeat protein family, was identified as a novel target of TRIM26. TRIM26 could interact with RACK1 and accelerate the degradation of RACK1, thus inactivation of MEK/ERK signaling. Overexpression of RACK1 could attenuate the inhibitory effect of TRIM26 overexpression on p-MEK1/2 and p-ERK1/2, and silence of RACK1 could partly impair the effect of TRIM26 knockdown-induced upregulation of p-MEK1/2 and p-ERK1/2. Further, a series of gain- and loss-of-function experiments showed that decreased malignant behaviors including cell proliferation and invasion in TRIM26-upregulated cells were reversed when RACK1 was overexpressed, whereas RACK1 knockdown diminished the increased malignant phenotypes in TRIM26-silenced osteosarcoma cells. In conclusion, our study indicated that TRIM26 inhibited osteosarcoma progression via promoting proteasomal degradation of RACK1, thereby resulting in inactivation of MEK/ERK signaling, and impeding the EMT process.
Collapse
Affiliation(s)
- Kezhou Xia
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Di Zheng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhun Wei
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wenda Liu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Weichun Guo
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
19
|
Liu J, Pan Y, Liu Y, Wei W, Hu X, Xin W, Chen N. The regulation of PTEN: Novel insights into functions as cancer biomarkers and therapeutic targets. J Cell Physiol 2023; 238:1693-1715. [PMID: 37334436 DOI: 10.1002/jcp.31053] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023]
Abstract
This review summarizes the implications of the primary tumor suppressor protein phosphatase and tensin homolog (PTEN) in aggressive cancer development. PTEN interacts with other cellular proteins or factors suggesting the existence of an intricate molecular network that regulates their oncogenic function. Accumulating evidence has shown that PTEN exists and plays a role in the cytoplasmic organelles and in the nucleus. PTEN blocks phosphoinositide 3-kinases (PI3K)-protein kinase B-mammalian target of rapamycin signaling pathway by dephosphorylating phosphatidylinositol (PI)-3,4,5-triphosphate to PI-4,5-bisphosphate thus counteracting PI3K function. Studies have shown that PTEN expression is tightly regulated at transcriptional, posttranscriptional, and posttranslational levels (including protein-protein interactions and posttranslational modifications). Despite recent advances in PTEN research, the regulation and function of the PTEN gene remain largely unknown. How mutation or loss of specific exons in the PTEN gene occurs and involves in cancer development is not clear. This review illustrates the regulatory mechanisms of PTEN expression and discusses how PTEN participates in tumor development and/or suppression. Future prospects for the clinical applications are also highlighted.
Collapse
Affiliation(s)
- Jie Liu
- Department of Dermatology, Skin Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yongli Pan
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Yuheng Liu
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Wei Wei
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Xiaoping Hu
- Department of Dermatology, Skin Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Wenqiang Xin
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Nan Chen
- Department of Gastroenterology, Liaocheng People's Hospital, Liaocheng, China
| |
Collapse
|
20
|
Sarver AL, Mills LJ, Makielski KM, Temiz NA, Wang J, Spector LG, Subramanian S, Modiano JF. Distinct mechanisms of PTEN inactivation in dogs and humans highlight convergent molecular events that drive cell division in the pathogenesis of osteosarcoma. Cancer Genet 2023; 276-277:1-11. [PMID: 37267683 DOI: 10.1016/j.cancergen.2023.05.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/02/2023] [Accepted: 05/19/2023] [Indexed: 06/04/2023]
Abstract
A hallmark of osteosarcoma in both human and canine tumors is somatic fragmentation and rearrangement of chromosome structure which leads to recurrent increases and decreases in DNA copy number. The PTEN gene has been implicated as an important tumor suppressor in osteosarcoma via forward genetic screens. Here, we analyzed copy number changes, promoter methylation and transcriptomes to better understand the role of PTEN in canine and human osteosarcoma. Reduction in PTEN copy number was observed in 23 of 95 (25%) of the canine tumors examined leading to corresponding decreases in PTEN transcript levels from RNA-Seq samples. Unexpectedly, canine tumors with an intact PTEN locus had higher levels of PTEN transcripts than human tumors. This variation in transcript abundance was used to evaluate the role of PTEN in osteosarcoma biology. Decreased PTEN copy number and transcript level was observed in - and likely an important driver of - increases in cell cycle transcripts in four independent canine transcriptional datasets. In human osteosarcoma, homozygous copy number loss was not observed, instead increased methylation of the PTEN promoter was associated with increased cell cycle transcripts. Somatic modification of PTEN, either by homozygous deletion in dogs or by promoter methylation in humans, is clinically relevant to osteosarcoma, because the cell cycle related transcripts are associated with patient outcomes. The PTEN gene is part of a syntenic rearrangement unique to the canine genome, making it susceptible to somatic loss of both copies of distal chromosome 26 which also includes the FAS death receptor. SIGNIFICANCE STATEMENT: PTEN function is abrogated by different mechanisms in canine and human osteosarcoma tumors leading to uncontrolled cell cycling. Somatic loss of this canine specific syntenic region may help explain why the canine genome appears to be uniquely susceptible to osteosarcoma. Syntenic arrangement, in the context of copy number change, may lead to synergistic interactions that in turn modify species specific cancer risk. Comparative models of tumorigenesis may utilize different driver mechanisms.
Collapse
Affiliation(s)
- Aaron L Sarver
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA; Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA.
| | - Lauren J Mills
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Kelly M Makielski
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA
| | - Nuri A Temiz
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jinhua Wang
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Institute for Health Informatics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Logan G Spector
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA
| | - Subbaya Subramanian
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; Department of Surgery, University of Minnesota School of Medicine, Minneapolis, MN 55455, USA
| | - Jaime F Modiano
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA; Animal Cancer Care and Research Program, University of Minnesota, St. Paul, MN 55108, USA; Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, MN 55108, USA; Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA; Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA; Center for Engineering and Medicine, University of Minnesota, Minneapolis, MN 55455, USA; Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
21
|
Ji Z, Shen J, Lan Y, Yi Q, Liu H. Targeting signaling pathways in osteosarcoma: Mechanisms and clinical studies. MedComm (Beijing) 2023; 4:e308. [PMID: 37441462 PMCID: PMC10333890 DOI: 10.1002/mco2.308] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 07/15/2023] Open
Abstract
Osteosarcoma (OS) is a highly prevalent bone malignancy among adolescents, accounting for 40% of all primary malignant bone tumors. Neoadjuvant chemotherapy combined with limb-preserving surgery has effectively reduced patient disability and mortality, but pulmonary metastases and OS cells' resistance to chemotherapeutic agents are pressing challenges in the clinical management of OS. There has been an urgent need to identify new biomarkers for OS to develop specific targeted therapies. Recently, the continued advancements in genomic analysis have contributed to the identification of clinically significant molecular biomarkers for diagnosing OS, acting as therapeutic targets, and predicting prognosis. Additionally, the contemporary molecular classifications have revealed that the signaling pathways, including Wnt/β-catenin, PI3K/AKT/mTOR, JAK/STAT3, Hippo, Notch, PD-1/PD-L1, MAPK, and NF-κB, have an integral role in OS onset, progression, metastasis, and treatment response. These molecular classifications and biological markers have created new avenues for more accurate OS diagnosis and relevant treatment. We herein present a review of the recent findings for the modulatory role of signaling pathways as possible biological markers and treatment targets for OS. This review also discusses current OS therapeutic approaches, including signaling pathway-based therapies developed over the past decade. Additionally, the review covers the signaling targets involved in the curative effects of traditional Chinese medicines in the context of expression regulation of relevant genes and proteins through the signaling pathways to inhibit OS cell growth. These findings are expected to provide directions for integrating genomic, molecular, and clinical profiles to enhance OS diagnosis and treatment.
Collapse
Affiliation(s)
- Ziyu Ji
- School of Integrated Traditional Chinese and Western MedicineSouthwest Medical UniversityLuzhouSichuanChina
| | - Jianlin Shen
- Department of OrthopaedicsAffiliated Hospital of Putian UniversityPutianFujianChina
| | - Yujian Lan
- School of Integrated Traditional Chinese and Western MedicineSouthwest Medical UniversityLuzhouSichuanChina
| | - Qian Yi
- Department of PhysiologySchool of Basic Medical ScienceSouthwest Medical UniversityLuzhouSichuanChina
| | - Huan Liu
- Department of OrthopaedicsThe Affiliated Traditional Chinese Medicine Hospital of Southwest Medical UniversityLuzhouSichuanChina
| |
Collapse
|
22
|
Han Q, Yan P, Song R, Liu F, Tian Q. HOXC13-driven TIMM13 overexpression promotes osteosarcoma cell growth. Cell Death Dis 2023; 14:398. [PMID: 37407582 DOI: 10.1038/s41419-023-05910-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 06/11/2023] [Accepted: 06/19/2023] [Indexed: 07/07/2023]
Abstract
TIMM13 (translocase of inner mitochondrial membrane 13) located at the mitochondrial intermembrane space is vital for the integrity and function of mitochondria. We found that the mitochondrial protein TIMM13 is upregulated in human OS tissues and cells. In patient-derived primary OS cells and established cell lines, TIMM13 shRNA or knockout provoked mitochondrial dysfunction, causing mitochondrial depolarization, reactive oxygen species production, and oxidative injury, as well as lipid peroxidation, DNA damage, and ATP depletion. Moreover, TIMM13 depletion provoked OS cell apoptosis and inhibited cell proliferation and migration. Conversely, ectopic TIMM13 overexpression increased ATP contents, enhancing OS cell proliferation and migration. Moreover, we discovered that Akt-mTOR activation was inhibited with TIMM13 depletion in primary OS cells. Further studies revealed that HOXC13 (Homeobox C13)-dependent TIMM13 transcription was significantly increased in OS tissues and cells. Whereas TIMM13 transcription and expression were decreased following HOXC13 silencing in primary OS cells. In vivo, TIMM13 KO potently inhibited OS xenograft growth in the proximal tibia of nude mice. TIMM13 KO also induced Akt-mTOR inactivation, ATP depletion, oxidative injury, and apoptosis in the in situ OS tumors. Together, upregulation of the mitochondrial protein TIMM13 is important for OS cell growth, representing a novel and promising therapeutic target.
Collapse
Affiliation(s)
- Qicai Han
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Penghui Yan
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruipeng Song
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Feifei Liu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qing Tian
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
23
|
Ji T, Shi Q, Mei S, Xu J, Liang H, Xie L, Ren T, Sun K, Li D, Tang X, Zhang P, Guo W. Integrated analysis of single-cell and bulk RNA sequencing data reveals an immunostimulatory microenvironment in tumor thrombus of osteosarcoma. Oncogenesis 2023; 12:31. [PMID: 37244923 DOI: 10.1038/s41389-023-00474-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 04/24/2023] [Accepted: 04/27/2023] [Indexed: 05/29/2023] Open
Abstract
Tumor thrombus of bone sarcomas represents a unique reservoir for various types of cancer and immune cells, however, the investigation of tumor thrombus at a single-cell level is very limited. And it is still an open question to identify the thrombus-specific tumor microenvironment that is associated with the tumor-adaptive immune response. Here, by analyzing bulk tissue and single-cell level transcriptome from the paired thrombus and primary tumor samples of osteosarcoma (OS) patients, we define the immunostimulatory microenvironment in tumor thrombus of OS with a higher proportion of tumor-associated macrophages with M1-like states (TAM-M1) and TAM-M1 with high expression of CCL4. OS tumor thrombus is found to have upregulated IFN-γ and TGF-β signalings that are related to immune surveillance of circulating tumor cells in blood circulation. Further multiplexed immunofluorescence staining of the CD3/CD4/CD8A/CD68/CCL4 markers validates the immune-activated state in the tumor thrombus samples. Our study first reports the transcriptome differences at a single-cell level between tumor thrombus and primary tumor in sarcoma.
Collapse
Affiliation(s)
- Tao Ji
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Qianyu Shi
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Song Mei
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiuhui Xu
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Haijie Liang
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Lu Xie
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Tingting Ren
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Kunkun Sun
- Department of Pathology, People's Hospital, Peking University, Beijing, 100044, China
| | - Dasen Li
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Xiaodong Tang
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Rare Disease Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| | - Wei Guo
- Department of Musculoskeletal Tumor, People's Hospital, Peking University, Beijing, 100044, China.
| |
Collapse
|
24
|
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: 5.0] [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
|
25
|
Martins-Neves SR, Sampaio-Ribeiro G, Gomes CMF. Self-Renewal and Pluripotency in Osteosarcoma Stem Cells' Chemoresistance: Notch, Hedgehog, and Wnt/β-Catenin Interplay with Embryonic Markers. Int J Mol Sci 2023; 24:8401. [PMID: 37176108 PMCID: PMC10179672 DOI: 10.3390/ijms24098401] [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: 03/31/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Osteosarcoma is a highly malignant bone tumor derived from mesenchymal cells that contains self-renewing cancer stem cells (CSCs), which are responsible for tumor progression and chemotherapy resistance. Understanding the signaling pathways that regulate CSC self-renewal and survival is crucial for developing effective therapies. The Notch, Hedgehog, and Wnt/β-Catenin developmental pathways, which are essential for self-renewal and differentiation of normal stem cells, have been identified as important regulators of osteosarcoma CSCs and also in the resistance to anticancer therapies. Targeting these pathways and their interactions with embryonic markers and the tumor microenvironment may be a promising therapeutic strategy to overcome chemoresistance and improve the prognosis for osteosarcoma patients. This review focuses on the role of Notch, Hedgehog, and Wnt/β-Catenin signaling in regulating CSC self-renewal, pluripotency, and chemoresistance, and their potential as targets for anti-cancer therapies. We also discuss the relevance of embryonic markers, including SOX-2, Oct-4, NANOG, and KLF4, in osteosarcoma CSCs and their association with the aforementioned signaling pathways in overcoming drug resistance.
Collapse
Affiliation(s)
- Sara R. Martins-Neves
- iCBR—Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.R.M.-N.)
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Gabriela Sampaio-Ribeiro
- iCBR—Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.R.M.-N.)
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-548 Coimbra, Portugal
- CACC—Clinical Academic Center of Coimbra, 3000-075 Coimbra, Portugal
| | - Célia M. F. Gomes
- iCBR—Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (S.R.M.-N.)
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3000-548 Coimbra, Portugal
- CACC—Clinical Academic Center of Coimbra, 3000-075 Coimbra, Portugal
| |
Collapse
|
26
|
Li T, Gao M, Wu Z, Yang J, Mo B, Yu S, Gong X, Liu J, Wang W, Luo S, Li R. Tantalum-Zirconium Co-Doped Metal-Organic Frameworks Sequentially Sensitize Radio-Radiodynamic-Immunotherapy for Metastatic Osteosarcoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206779. [PMID: 36739599 PMCID: PMC10074130 DOI: 10.1002/advs.202206779] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/03/2023] [Indexed: 06/18/2023]
Abstract
Due to radiation resistance and the immunosuppressive microenvironment of metastatic osteosarcoma, novel radiosensitizers that can sensitize radiotherapy (RT) and antitumor immunity synchronously urgently needed. Here, the authors developed a nanoscale metal-organic framework (MOF, named TZM) by co-doping high-atomic elements Ta and Zr as metal nodes and porphyrinic molecules (tetrakis(4-carboxyphenyl)porphyrin (TCPP)) as a photosensitizing ligand. Given the 3D arrays of ultra-small heavy metals, porous TZM serves as an efficient attenuator absorbing X-ray energy and sensitizing hydroxyl radical generation for RT. Ta-Zr co-doping narrowed the highest occupied molecular orbital-lowest unoccupied molecular orbital (HOMO-LUMO) energy gap and exhibited close energy levels between the singlet and triplet photoexcited states, facilitating TZM transfer energy to the photosensitizer TCPP to sensitize singlet oxygen (1 O2 ) generation for radiodynamic therapy (RDT). The sensitized RT-RDT effects of TZM elicit a robust antitumor immune response by inducing immunogenic cell death, promoting dendritic cell maturation, and upregulating programmed cell death protein 1 (PD-L1) expression via the cGAS-STING pathway. Furthermore, a combination of TZM, X-ray, and anti-PD-L1 treatments amplify antitumor immunotherapy and efficiently arrest osteosarcoma growth and metastasis. These results indicate that TZM is a promising radiosensitizer for the synergistic RT and immunotherapy of metastatic osteosarcoma.
Collapse
Affiliation(s)
- Tao Li
- Institute of Combined InjuryState Key Laboratory of TraumaBurns and Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineThird Military Medical University (Army Medical University)Chongqing400038China
- Center for Joint SurgerySouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Mingquan Gao
- Institute of Combined InjuryState Key Laboratory of TraumaBurns and Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineThird Military Medical University (Army Medical University)Chongqing400038China
- Department of Radiation OncologySichuan Cancer Hospital & InstituteSichuan Key Laboratory of Radiation OncologyChengduSichuan610041China
| | - Zifei Wu
- Institute of Combined InjuryState Key Laboratory of TraumaBurns and Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineThird Military Medical University (Army Medical University)Chongqing400038China
- Department of Radiation OncologySichuan Cancer Hospital & InstituteSichuan Key Laboratory of Radiation OncologyChengduSichuan610041China
| | - Junjun Yang
- Center for Joint SurgerySouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Banghui Mo
- Department of OncologySouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Songtao Yu
- Department of OncologySouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Xiaoyuan Gong
- Center for Joint SurgerySouthwest HospitalThird Military Medical University (Army Medical University)Chongqing400038China
| | - Jing Liu
- Institute of Combined InjuryState Key Laboratory of TraumaBurns and Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineThird Military Medical University (Army Medical University)Chongqing400038China
| | - Weidong Wang
- Department of Radiation OncologySichuan Cancer Hospital & InstituteSichuan Key Laboratory of Radiation OncologyChengduSichuan610041China
| | - Shenglin Luo
- Institute of Combined InjuryState Key Laboratory of TraumaBurns and Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineThird Military Medical University (Army Medical University)Chongqing400038China
| | - Rong Li
- Institute of Combined InjuryState Key Laboratory of TraumaBurns and Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineThird Military Medical University (Army Medical University)Chongqing400038China
| |
Collapse
|
27
|
Cheng S, Liu S, Chen B, Du C, Xiao P, Luo X, Wei L, Lei Y, Zhao C, Huang W. Psoralidin inhibits osteosarcoma growth and metastasis by downregulating ITGB1 expression via the FAK and PI3K/Akt signaling pathways. Chin Med 2023; 18:34. [PMID: 37004120 PMCID: PMC10064721 DOI: 10.1186/s13020-023-00740-w] [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: 01/17/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
BACKGROUND Psoralea corylifolia is a medicinal leguminous plant that has long been used to treat various diseases. Psoralidin (PSO) is the main extract compound of P. corylifolia and exhibits antibacterial, antitumor, anti-inflammatory, antioxidant, and other pharmacological activities. PSO has demonstrated inhibitory effects in several cancers; however, its inhibitory effect on osteosarcoma has not been reported. This study aimed to evaluate the inhibitory effect of PSO on osteosarcoma and elucidate the underlying molecular mechanisms. METHODS Crystal violet, cell counting kit-8 (CCK8), and 5-Ethynyl-2'-deoxyuridine (EdU) staining assays were used to assess the inhibitory effect of PSO on the proliferation of 143B and MG63 osteosarcoma cells. Wound healing and Transwell assays were conducted to evaluate the effects of PSO on osteosarcoma cell migration and invasion. The cell cycle and apoptosis were analyzed using flow cytometry. To determine the possible molecular mechanisms, RNA-sequencing was performed and protein expression was analyzed by western blotting. The inhibitory effect of PSO on osteosarcoma in vivo was analyzed using a mouse model of orthotopic osteosarcoma and immunohistochemistry. RESULTS PSO inhibited osteosarcoma cell proliferation in a concentration-dependent manner, inhibited cell migration and invasion, and induced cell-cycle arrest and apoptosis. Mechanistically, PSO treatment significantly inhibited the focal adhesion kinase (FAK) and phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathways by downregulating ITGB1 expression in both MG63 and 143B cells. Furthermore, we demonstrated that PSO restrained osteosarcoma growth in vivo. CONCLUSION PSO may suppress osteosarcoma via the FAK and PI3K/Akt signaling pathways by downregulating ITGB1 expression.
Collapse
Affiliation(s)
- Shengwen Cheng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Senrui Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Bowen Chen
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chengcheng Du
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Pengcheng Xiao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Xuefeng Luo
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Li Wei
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiting Lei
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Chen Zhao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Wei Huang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
28
|
Jiang J, Qu H, Zhan X, Liu D, Liang T, Chen L, Huang S, Sun X, Chen J, Chen T, Li H, Yao Y, Liu C. Identification of osteosarcoma m6A-related prognostic biomarkers using artificial intelligence: RBM15. Sci Rep 2023; 13:5255. [PMID: 37002245 PMCID: PMC10066227 DOI: 10.1038/s41598-023-28739-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 01/24/2023] [Indexed: 04/03/2023] Open
Abstract
Osteosarcoma has the worst prognosis among malignant bone tumors, and effective biomarkers are lacking. Our study aims to explore m6A-related and immune-related biomarkers. Gene expression profiles of osteosarcoma and healthy controls were downloaded from multiple public databases, and their m6A-based gene expression was utilized for tumor typing using bioinformatics. Subsequently, a prognostic model for osteosarcoma was constructed using the least absolute shrinkage and selection operator and multivariate Cox regression analysis, and its immune cell composition was calculated using the CIBERSORTx algorithm. We also performed drug sensitivity analysis for these two genes. Finally, analysis was validated using immunohistochemistry. We also examined the RBM15 gene by qRT-PCR in an in vitro experiment. We collected routine blood data from 1738 patients diagnosed with osteosarcoma and 24,344 non-osteosarcoma patients and used two independent sample t tests to verify the accuracy of the CIBERSORTx analysis for immune cell differences. The analysis based on m6A gene expression tumor typing was most reliable using the two typing methods. The prognostic model based on the two genes constituting RNA-binding motif protein 15 (RBM15) and YTDC1 had a much lower survival rate for patients in the high-risk group than those in the low-risk group (P < 0.05). CIBERSORTx immune cell component analysis demonstrated that RBM15 showed a negative and positive correlation with T cells gamma delta and activated natural killer cells, respectively. Drug sensitivity analysis showed that these two genes showed varying degrees of correlation with multiple drugs. The results of immunohistochemistry revealed that the expression of these two genes was significantly higher in osteosarcoma than in paraneoplastic tissues. The results of qRT-PCR experiments showed that the expression of RBM15 was significantly higher in both osteosarcomas than in the control cell lines. Absolute lymphocyte value, lymphocyte percentage, hematocrit and erythrocyte count were lower in osteosarcoma than in the control group (P < 0.001). RBM15 and YTHDC1 can serve as potential prognostic biomarkers associated with m6A in osteosarcoma.
Collapse
Affiliation(s)
- Jie Jiang
- The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Haishun Qu
- Department of Traditional Chinese Medicine, The People's Hospital of Guangxi Zhuang Autonmous Region, Nanning, 530016, People's Republic of China
| | - Xinli Zhan
- The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Dachang Liu
- The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Tuo Liang
- The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Liyi Chen
- The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Shengsheng Huang
- The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Xuhua Sun
- The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Jiarui Chen
- The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Tianyou Chen
- The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Hao Li
- The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Yuanlin Yao
- The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China
| | - Chong Liu
- The First Clinical Affiliated Hospital of Guangxi Medical University, Nanning, 530021, People's Republic of China.
| |
Collapse
|
29
|
Lai X, Zhong J, Zhang B, Zhu T, Liao R. Exosomal Non-Coding RNAs: Novel Regulators of Macrophage-Linked Intercellular Communication in Lung Cancer and Inflammatory Lung Diseases. Biomolecules 2023; 13:536. [PMID: 36979471 PMCID: PMC10046066 DOI: 10.3390/biom13030536] [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: 02/07/2023] [Revised: 03/08/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Macrophages are innate immune cells and often classified as M1 macrophages (pro-inflammatory states) and M2 macrophages (anti-inflammatory states). Exosomes are cell-derived nanovesicles that range in diameter from 30 to 150 nm. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs), are abundant in exosomes and exosomal ncRNAs influence immune responses. Exosomal ncRNAs control macrophage-linked intercellular communication via their targets or signaling pathways, which can play positive or negative roles in lung cancer and inflammatory lung disorders, including acute lung injury (ALI), asthma, and pulmonary fibrosis. In lung cancer, exosomal ncRNAs mediated intercellular communication between lung tumor cells and tumor-associated macrophages (TAMs), coordinating cancer proliferation, migration, invasion, metastasis, immune evasion, and therapy resistance. In inflammatory lung illnesses, exosomal ncRNAs mediate macrophage activation and inflammation to promote or inhibit lung damage. Furthermore, we also discussed the possible applications of exosomal ncRNA-based therapies for lung disorders.
Collapse
Affiliation(s)
- Xingning Lai
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Research Unit for Perioperative Stress Assessment and Clinical Decision, Chinese Academy of Medical Sciences (2018RU012), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Zhong
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Research Unit for Perioperative Stress Assessment and Clinical Decision, Chinese Academy of Medical Sciences (2018RU012), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Boyi Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Research Unit for Perioperative Stress Assessment and Clinical Decision, Chinese Academy of Medical Sciences (2018RU012), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Research Unit for Perioperative Stress Assessment and Clinical Decision, Chinese Academy of Medical Sciences (2018RU012), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ren Liao
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu 610041, China
- Research Unit for Perioperative Stress Assessment and Clinical Decision, Chinese Academy of Medical Sciences (2018RU012), West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
30
|
Nowwarote N, Osathanon T, Fournier BPJ, Theerapanon T, Yodsanga S, Kamolratanakul P, Porntaveetus T, Shotelersuk V. PTEN regulates proliferation and osteogenesis of dental pulp cells and adipogenesis of human adipose-derived stem cells. Oral Dis 2023; 29:735-746. [PMID: 34558757 DOI: 10.1111/odi.14030] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/08/2021] [Accepted: 09/18/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To investigate the role of phosphatase and tensin homolog (PTEN) in dental pulp cells (hDPs) and adipose-derived mesenchymal stem cells (hADSCs). MATERIALS AND METHODS Genetic variant was identified with exome sequencing. The hDPs isolated from a patient with Cowden syndrome were investigated for their proliferation, osteogenesis, adipogenesis, and gene expression compared with controls. The normal hDPs and hADSCs were treated with the PTEN inhibitor, VO-OHpic trihydrate (VOT), to investigate the effect of PTEN inhibition. RESULTS A heterozygous nonsense PTEN variant, c.289C>T (p.Gln97*), was identified in the Cowden patient's blood and intraoral lipomas. The mutated hDPs showed significantly decreased proliferation, but significantly upregulated RUNX2 and OSX expression and mineralization, indicating enhanced osteogenic ability in mutated cells. The normal hDPs treated with VOT showed the decreases in proliferation, colony formation, osteogenic marker genes, alkaline phosphatase activity, and mineral deposition, suggesting that PTEN inhibition diminishes proliferation and osteogenic potential of hDPs. Regarding adipogenesis, the VOT-treated hADSCs showed a reduced number of cells containing lipid droplets, suggesting that PTEN inhibition might compromise adipogenic ability of hADSCs. CONCLUSIONS PTEN regulates proliferation, enhances osteogenesis of hDPs, and induces adipogenesis of hADSCs. The gain-of-function PTEN variant, p.Gln97*, enhances osteogenic ability of PTEN in hDPs.
Collapse
Affiliation(s)
- Nunthawan Nowwarote
- Dental Stem Cell Biology Research Unit and Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.,Centre de Recherche des Cordeliers, Universite de Paris, Sorbonne Universite, Paris, France.,Dental Faculty Garanciere, Oral Biology Department, Universite de Paris, Paris, France
| | - Thanaphum Osathanon
- Dental Stem Cell Biology Research Unit and Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Benjamin P J Fournier
- Centre de Recherche des Cordeliers, Universite de Paris, Sorbonne Universite, Paris, France.,Dental Faculty Garanciere, Oral Biology Department, Universite de Paris, Paris, France
| | - Thanakorn Theerapanon
- Center of Excellence in Genomics and Precision Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Somchai Yodsanga
- Department of Oral Pathology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Paksinee Kamolratanakul
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Thantrira Porntaveetus
- Center of Excellence in Genomics and Precision Dentistry, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Vorasuk Shotelersuk
- Center of Excellence for Medical Genomics, Medical Genomics Cluster, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Excellence Center for Genomics and Precision Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| |
Collapse
|
31
|
Hui L, Ziyue Z, Chao L, Bin Y, Aoyu L, Haijing W. Epigenetic Regulations in Autoimmunity and Cancer: from Basic Science to Translational Medicine. Eur J Immunol 2023; 53:e2048980. [PMID: 36647268 DOI: 10.1002/eji.202048980] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/25/2022] [Accepted: 01/10/2023] [Indexed: 01/18/2023]
Abstract
Epigenetics, as a discipline that aims to explain the differential expression of phenotypes arising from the same gene sequence and the heritability of epigenetic expression, has received much attention in medicine. Epigenetic mechanisms are constantly being discovered, including DNA methylation, histone modifications, noncoding RNAs and m6A. The immune system mainly achieves an immune response through the differentiation and functional expression of immune cells, in which epigenetic modification will have an important impact. Because of immune infiltration in the tumor microenvironment, immunotherapy has become a research hotspot in tumor therapy. Epigenetics plays an important role in autoimmune diseases and cancers through immunology. An increasing number of drugs targeting epigenetic mechanisms, such as DNA methyltransferase inhibitors, histone deacetylase inhibitors, and drug combinations, are being evaluated in clinical trials for the treatment of various cancers (including leukemia and osteosarcoma) and autoimmune diseases (systemic lupus erythematosus, rheumatoid arthritis, systemic sclerosis). This review summarizes the progress of epigenetic regulation for cancers and autoimmune diseases to date, shedding light on potential therapeutic strategies.
Collapse
Affiliation(s)
- Li Hui
- Department of Orthopedics, Second Xiangya Hospital of Central South University, Changsha, Hunan, P. R. China
| | - Zhao Ziyue
- Department of Orthopedics, Second Xiangya Hospital of Central South University, Changsha, Hunan, P. R. China
| | - Liu Chao
- Department of Orthopedics, Second Xiangya Hospital of Central South University, Changsha, Hunan, P. R. China
| | - Yu Bin
- Department of Orthopedics, Second Xiangya Hospital of Central South University, Changsha, Hunan, P. R. China
| | - Li Aoyu
- Department of Orthopedics, Second Xiangya Hospital of Central South University, Changsha, Hunan, P. R. China
| | - Wu Haijing
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, Second Xiangya Hospital of Central South University, Changsha, Hunan, P. R. China
| |
Collapse
|
32
|
Bai J, Zhang X, Jiang F, Shan H, Gao X, Bo L, Zhang Y. A Feedback Loop of LINC00665 and the Wnt Signaling Pathway Expedites Osteosarcoma Cell Proliferation, Invasion, and Epithelial-Mesenchymal Transition. Orthop Surg 2022; 15:286-300. [PMID: 36387061 PMCID: PMC9837296 DOI: 10.1111/os.13532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 08/09/2022] [Accepted: 09/06/2022] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVES Osteosarcoma (OS) is a malignant tumor with frequent occurrence among teenagers. Long non-coding RNAs (lncRNAs) play pro-cancer roles in many tumors. The purpose of this study was to figure out the functional role of a novel lncRNA long intergenic non-protein coding RNA 665 (LINC00665) in OS by observing the OS cell behaviors. METHODS Quantitative reverse transcription polymerase chain reaction (RT-qPCR) was used to analyze LINC00665 expression in OS cells. Cell function assays assessed the impacts of LINC00665 on OS cell phenotype. Immunofluorescence and western blot analyzed the function of LINC00665 on epithelial-mesenchymal transition (EMT) in OS. Moreover, mechanistic assays analyzed the downstream mechanism of LINC00665 in OS cells. RESULTS LINC00665 was significantly up-regulated in OS cells. LINC00665 silence facilitated OS cell proliferation, migration, invasion, and EMT while inhibiting cell apoptosis. Mechanically, LINC00665 acted as a competing endogenous RNA (ceRNA) to sponge miR-1249-5p and thereby modulated Wnt family member 2B (WNT2B) to activate Wnt pathway. Wnt pathway activated LINC00665 expression transcriptionally. CONCLUSIONS Our study uncovered the cancer-promoting role of LINC00665 in OS, and the feedback loop of LINC00665/miR-1249-5p/WNT2B/Wnt might be a potential target for OS treatment.
Collapse
Affiliation(s)
- Jinyu Bai
- Department of Orthopaedicsthe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xiao Zhang
- Department of Traditional Chinese Medicine Orthopaedicsthe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Fengxian Jiang
- Department of Orthopaedicsthe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Huajian Shan
- Department of Orthopaedicsthe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Xiang Gao
- Department of Orthopaedicsthe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Lin Bo
- Department of Rheumatologythe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yingzi Zhang
- Department of Orthopaedicsthe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
33
|
Hu Z, Wen S, Huo Z, Wang Q, Zhao J, Wang Z, Chen Y, Zhang L, Zhou F, Guo Z, Liu H, Zhou S. Current Status and Prospects of Targeted Therapy for Osteosarcoma. Cells 2022; 11:3507. [PMID: 36359903 PMCID: PMC9653755 DOI: 10.3390/cells11213507] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 09/26/2023] Open
Abstract
Osteosarcoma (OS) is a highly malignant tumor occurring in bone tissue with a high propensity to metastasize, and its underlying mechanisms remain largely elusive. The OS prognosis is poor, and improving the survival of OS patients remains a challenge. Current treatment methods such as surgical approaches, chemotherapeutic drugs, and immunotherapeutic drugs remain ineffective. As research progresses, targeted therapy is gradually becoming irreplaceable. In this review, several treatment modalities for osteosarcoma, such as surgery, chemotherapy, and immunotherapy, are briefly described, followed by a discussion of targeted therapy, the important targets, and new technologies for osteosarcoma treatment.
Collapse
Affiliation(s)
- Zunguo Hu
- Department of Joint Surgery, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang 261061, China
| | - Shuang Wen
- Department of Joint Surgery, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang 261061, China
| | - Zijun Huo
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Qing Wang
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Jiantao Zhao
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Zihao Wang
- Department of Joint Surgery, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang 261061, China
| | - Yanchun Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Weifang Medical University, Weifang 261053, China
| | - Lingyun Zhang
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Fenghua Zhou
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Zhangyu Guo
- Neurologic Disorders and Regenerative Repair Laboratory, Weifang Medical University, Weifang 261053, China
| | - Huancai Liu
- Department of Joint Surgery, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang 261061, China
| | - Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
34
|
Fan L, Zhong Z, Lin Y, Li J. Non-coding RNAs as potential biomarkers in osteosarcoma. Front Genet 2022; 13:1028477. [PMID: 36338952 PMCID: PMC9627036 DOI: 10.3389/fgene.2022.1028477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/07/2022] [Indexed: 11/24/2022] Open
Abstract
Osteosarcoma (OS) is a primary solid malignant tumor that occurs most frequently in the metaphysis of long bones. More likely to happen to children and adolescents. OS has high mortality and disability rate. However, the etiology and pathogenesis of OS have not been fully understood till now. Due to the lack of effective biomarkers, OS cannot be precisely detected in the early stage. With the application of next-generation and high-throughput sequencing, more and more abnormally expressed non-coding RNAs(ncRNAs) have been identified in OS. Growing evidences have suggested the ncRNAs, such as microRNAs (miRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs), have played an important role in the tumorigenesis and progression of OS. Thus, they can be served as novel biomarkers for diagnosis, treatment and prognosis. This review summarized the application of ncRNA as biomarkers in OS in detail, and discussed the limitation and future improvement of the potential biomarkers.
Collapse
Affiliation(s)
- Lijuan Fan
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, Henan, China
- Luoyang Postgraduate Training Department, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Zhenhao Zhong
- Department of Spinal Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Yubo Lin
- School of Clinical Medicine, Guilin Medical University, Guilin, Guangxi, China
| | - Jitian Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, Henan, China
- Luoyang Postgraduate Training Department, Henan University of Chinese Medicine, Zhengzhou, Henan, China
- The First College for Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- *Correspondence: Jitian Li,
| |
Collapse
|
35
|
Yue J, Chen ZS, Xu XX, Li S. Functions and therapeutic potentials of exosomes in osteosarcoma. ACTA MATERIA MEDICA 2022; 1:552-562. [PMID: 36710945 PMCID: PMC9879305 DOI: 10.15212/amm-2022-0024] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Osteosarcoma is a primary malignant tumor of the skeleton with the morbidity of 2.5 in 1 million. The regularly on-set is in the epiphysis of the extremities with a high possibility of early metastasis, rapid progression, and poor prognosis. The survival rate of patients with metastatic or recurrent osteosarcoma remains low, and novel diagnostic and therapeutic methods are urgently needed. Exosomes are extracellular vesicles 30-150 nm in diameter secreted by various cells that are widely present in various body fluids. Exosomes are abundant in biologically active components such as proteins, nucleic acids, and lipids. Exosomes participate in numerous physiological and pathological processes via intercellular substance exchange and signaling. This review presents the novel findings of exosomes in osteosarcoma in diagnosis, prognosis, and therapeutic aspects.
Collapse
Affiliation(s)
- Jiaji Yue
- Department of Bone and Joint Surgery, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518000, PR China
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John’s University, Queens, NY,United States
| | - Xiang-Xi Xu
- Department of Radiation Oncology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, PR China
| |
Collapse
|
36
|
Human Bone Marrow Mesenchymal Stem Cell (hBMSCs)-Derived miR-29a-3p-Containing Exosomes Impede Laryngocarcinoma Cell Malignant Phenotypes by Inhibiting PTEN. Stem Cells Int 2022; 2022:8133632. [PMID: 36338027 PMCID: PMC9635976 DOI: 10.1155/2022/8133632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 08/29/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
Although microRNA-29a-3p was reported to inhibit laryngocarcinoma progression, the potential mechanisms have not been explored clearly. Laryngocarcinoma tissues were collected for analyzing the levels of miR-29a-3p and phosphatase and tensin homolog (PTEN). The miR mimics or inhibitor was transfected into laryngocarcinoma cell lines M4E and Hep2 for the investigation of the biological functions (proliferative, invasion, migratory rates, and apoptotic rates) of this miRNA. The exosomes (Exo) from human bone marrow mesenchymal stem cells (hBMSCs) after the transfection of miR mimics/inhibitor/si-PTEN were isolated and used to stimulate M4E and Hep2 cells. The in vivo mouse model was constructed to verify our findings. The miR-29a-3p level was decreased, and PTEN level was elevated in laryngocarcinoma tissues and the cancer cell lines. MiR mimics could inhibit proliferative, invasive migratory rates while promoting apoptotic rates of M4E and Hep2 cells. MiR-29a-3p was enriched in hBMSC-derived Exo, and the Exo from miR-29a-3p mimics transfected hBMSCs could inhibit laryngocarcinoma cell malignant phenotypes in vitro and prevent tumor progression in vivo. In addition, the direct binding relationship between miR-29a-3p and PTEN in this disease was determined. In conclusion, hBMSC-derived Exo with upregulated miR-29a-3p inhibited laryngocarcinoma progression via regulating PTEN, providing a potential diagnostic and therapeutic target in this disease.
Collapse
|
37
|
Gao X, Gao B, Li S. Extracellular vesicles: A new diagnostic biomarker and targeted drug in osteosarcoma. Front Immunol 2022; 13:1002742. [PMID: 36211364 PMCID: PMC9539319 DOI: 10.3389/fimmu.2022.1002742] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/12/2022] [Indexed: 11/25/2022] Open
Abstract
Osteosarcoma (OS) is a primary bone cancer that is highly prevalent among adolescents and adults below the age of 20 years. The prognostic outcome of metastatic OS or relapse is extremely poor; thus, developing new diagnostic and therapeutic strategies for treating OS is necessary. Extracellular vesicles (EVs) ranging from 30–150 nm in diameter are commonly produced in different cells and are found in various types of body fluids. EVs are rich in biologically active components like proteins, lipids, and nucleic acids. They also strongly affect pathophysiological processes by modulating the intercellular signaling pathways and the exchange of biomolecules. Many studies have found that EVs influence the occurrence, development, and metastasis of osteosarcoma. The regulation of inflammatory communication pathways by EVs affects OS and other bone-related pathological conditions, such as osteoarthritis and rheumatoid arthritis. In this study, we reviewed the latest findings related to diagnosis, prognosis prediction, and the development of treatment strategies for OS from the perspective of EVs.
Collapse
Affiliation(s)
- Xiaozhuo Gao
- Department of Pathology, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, China
| | - Bo Gao
- Department of Pathology, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, China
| | - Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, China
- *Correspondence: Shenglong Li, ;
| |
Collapse
|
38
|
Al-Rawashde FA, Al-wajeeh AS, Vishkaei MN, Saad HKM, Johan MF, Taib WRW, Ismail I, Al-Jamal HAN. Thymoquinone Inhibits JAK/STAT and PI3K/Akt/ mTOR Signaling Pathways in MV4-11 and K562 Myeloid Leukemia Cells. Pharmaceuticals (Basel) 2022; 15:ph15091123. [PMID: 36145344 PMCID: PMC9504933 DOI: 10.3390/ph15091123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Constitutive activation of Janus tyrosine kinase-signal transducer and activator of transcription (JAK/STAT) and Phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathways plays a crucial role in the development of acute myeloid leukemia (AML) and chronic myeloid leukemia (CML). Thymoquinone (TQ), one of the main constituents of Nigella sativa, has shown anti-cancer activities in several cancers. However, the inhibitory effect mechanism of TQ on leukemia has not been fully understood. Therefore, this study aimed to investigate the effect of TQ on JAK/STAT and PI3K/Akt/mTOR pathways in MV4-11 AML cells and K562 CML cells. FLT3-ITD positive MV4-11 cells and BCR-ABL positive K562 cells were treated with TQ. Cytotoxicity assay was assessed using WSTs-8 kit. The expression of the target genes was evaluated using RT-qPCR. The phosphorylation status and the levels of proteins involved in JAK/STAT and PI3K/Akt/mTOR pathways were investigated using Jess western analysis. TQ induced a dose and time dependent inhibition of K562 cells proliferation. TQ significantly downregulated PI3K, Akt, and mTOR and upregulated PTEN expression with a significant inhibition of JAK/STAT and PI3K/Akt/mTOR signaling. In conclusion, TQ reduces the expression of PI3K, Akt, and mTOR genes and enhances the expression of PTEN gene at the mRNA and protein levels. TQ also inhibits JAK/STAT and PI3K/Akt/mTOR pathways, and consequently inhibits proliferation of myeloid leukemia cells, suggesting that TQ has potential anti-leukemic effects on both AML and CML cells.
Collapse
Affiliation(s)
- Futoon Abedrabbu Al-Rawashde
- School of Biomedicine, Faculty of Health Sciences, Universiti Sultan Zainal Abidin (UniSZA), Terengganu 21300, Malaysia
- Department of Anatomy and Histology, Faculty of Medicine, Mutah University, Al-Karak 61710, Jordan
| | | | | | - Hanan Kamel M. Saad
- School of Biomedicine, Faculty of Health Sciences, Universiti Sultan Zainal Abidin (UniSZA), Terengganu 21300, Malaysia
| | - Muhammad Farid Johan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan 16150, Malaysia
| | - Wan Rohani Wan Taib
- School of Biomedicine, Faculty of Health Sciences, Universiti Sultan Zainal Abidin (UniSZA), Terengganu 21300, Malaysia
| | - Imilia Ismail
- School of Biomedicine, Faculty of Health Sciences, Universiti Sultan Zainal Abidin (UniSZA), Terengganu 21300, Malaysia
| | - Hamid Ali Nagi Al-Jamal
- School of Biomedicine, Faculty of Health Sciences, Universiti Sultan Zainal Abidin (UniSZA), Terengganu 21300, Malaysia
- Correspondence: ; Tel.: +60-174729012
| |
Collapse
|
39
|
Zheng W, Li S, Huang J, Dong Y, Zhang H, Zheng J. Down-Regulation of Ubiquitin-Specific Peptidase 9X Inhibited Proliferation, Migration and Invasion of Osteosarcoma <i>via</i> ERK1/2 and PI3K/Akt Signaling Pathways. Biol Pharm Bull 2022; 45:1283-1290. [DOI: 10.1248/bpb.b22-00198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Wendi Zheng
- Department of Orthopedics, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital
| | - Shuang Li
- Department of Pathology, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital
| | - Jincheng Huang
- Department of Orthopedics, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital
| | - Yonghui Dong
- Department of Orthopedics, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital
| | - Hongjun Zhang
- Department of Orthopedics, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital
| | - Jia Zheng
- Department of Orthopedics, Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital
| |
Collapse
|
40
|
Lai HT, Naumova N, Marchais A, Gaspar N, Geoerger B, Brenner C. Insight into the interplay between mitochondria-regulated cell death and energetic metabolism in osteosarcoma. Front Cell Dev Biol 2022; 10:948097. [PMID: 36072341 PMCID: PMC9441498 DOI: 10.3389/fcell.2022.948097] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Osteosarcoma (OS) is a pediatric malignant bone tumor that predominantly affects adolescent and young adults. It has high risk for relapse and over the last four decades no improvement of prognosis was achieved. It is therefore crucial to identify new drug candidates for OS treatment to combat drug resistance, limit relapse, and stop metastatic spread. Two acquired hallmarks of cancer cells, mitochondria-related regulated cell death (RCD) and metabolism are intimately connected. Both have been shown to be dysregulated in OS, making them attractive targets for novel treatment. Promising OS treatment strategies focus on promoting RCD by targeting key molecular actors in metabolic reprogramming. The exact interplay in OS, however, has not been systematically analyzed. We therefore review these aspects by synthesizing current knowledge in apoptosis, ferroptosis, necroptosis, pyroptosis, and autophagy in OS. Additionally, we outline an overview of mitochondrial function and metabolic profiles in different preclinical OS models. Finally, we discuss the mechanism of action of two novel molecule combinations currently investigated in active clinical trials: metformin and the combination of ADI-PEG20, Docetaxel and Gemcitabine.
Collapse
Affiliation(s)
- Hong Toan Lai
- CNRS, Institut Gustave Roussy, Aspects métaboliques et systémiques de l’oncogénèse pour de nouvelles approches thérapeutiques, Université Paris-Saclay, Villejuif, France
| | - Nataliia Naumova
- CNRS, Institut Gustave Roussy, Aspects métaboliques et systémiques de l’oncogénèse pour de nouvelles approches thérapeutiques, Université Paris-Saclay, Villejuif, France
| | - Antonin Marchais
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Nathalie Gaspar
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Birgit Geoerger
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Catherine Brenner
- CNRS, Institut Gustave Roussy, Aspects métaboliques et systémiques de l’oncogénèse pour de nouvelles approches thérapeutiques, Université Paris-Saclay, Villejuif, France
- *Correspondence: Catherine Brenner,
| |
Collapse
|
41
|
Yang S, Qu Y, Wang J, Gao F, Ji M, Xie P, Zhu A, Tan B, Wang X, Zhu G. Anshen Dingzhi prescription in the treatment of PTSD in mice: Investigation of the underlying mechanism from the perspective of hippocampal synaptic function. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 101:154139. [PMID: 35523115 DOI: 10.1016/j.phymed.2022.154139] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/19/2022] [Accepted: 04/28/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Anshen Dingzhi prescription (ADP) is an important prescription for the treatment of mental diseases in traditional Chinese medicine and is widely used to treat neuropsychiatric disorders. PURPOSE To explore the ameliorative effect of ADP on post-traumatic stress disorder (PTSD)-like behaviors in mice and determine the underlying mechanism. METHODS The constituents of ADP were analyzed by UPLC-Q-TOF/MS. The PTSD-like behaviors of mice subjected to single prolonged stress (SPS) were evaluated using behavioral tests. Potential pathological changes in the hippocampus were assessed by hematoxylin and eosin (H&E) staining. Western blotting and immunohistochemistry (IHC) were employed to detect the expression of proteins involved in relevant signaling pathways. RESULTS Five quality control markers (ginsenoside Rg1, ginsenoside Rb1, tenuifolin, poricoic acid B, and α-asarone) were detected in the ADP solution. The ginsenoside Rg1 content in ADP was found to be 0.114 mg/g. Mice subjected to SPS showed obvious fear generalization and anxiety-like behaviors. ADP treatment prevented the behavioral changes caused by exposure to SPS. Compared with control animals, the number of normal pyramidal cells in the hippocampal CA1 region of mice exposed to SPS was decreased and the number of degenerating pyramidal cells was increased; however, ADP administration could counteract these effects. Furthermore, the protein expression of BDNF, p-TrkB, μ-calpain, PSD95, GluN2A, GluA1, p-AKT, p-mTOR, and ARC was decreased, while that of PTEN and GluN2B was increased in the hippocampus of mice subjected to SPS compared with that in control animals; however, these changes in protein expression were reversed following ADP treatment. Importantly, the ameliorative effect of ADP on PTSD-like behaviors and synaptic protein expression were inhibited by rapamycin administration. CONCLUSIONS ADP administration improves PTSD-like behaviors in mice and this effect may be mediated through an mTOR-dependent improvement in synaptic function in the hippocampus.
Collapse
Affiliation(s)
- Shaojie Yang
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Yan Qu
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Juan Wang
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Feng Gao
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Manman Ji
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Pan Xie
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Aisong Zhu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Hangzhou, Zhejiang, 310053, China
| | - Bei Tan
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Hangzhou, Zhejiang, 310053, China
| | - Xuncui Wang
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China.
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China.
| |
Collapse
|
42
|
Huang SX, Mei HB, Liu K, Tang J, Wu JY, Zhu GH, Ye WH. CircPVT1 promotes the tumorigenesis and metastasis of osteosarcoma via mediation of miR-26b-5p/CCNB1 axis. J Bone Miner Metab 2022; 40:581-593. [PMID: 35648221 DOI: 10.1007/s00774-022-01326-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/09/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Osteosarcoma (OS) is the most aggressive malignancy among the bone tumors in the world. Circular RNAs (circRNAs) have been reported to be participated in multiple cancers, including OS. Meanwhile, circPVT1 has been proved to be upregulated in OS. However, the mechanism by which circPVT1 mediates the tumorigenesis of OS remains to be further explored. MATERIALS AND METHODS Protein and gene expressions in OS cells were measured by western blot and RT-qPCR, respectively. Cell growth was assessed by flow cytometry and colony formation, respectively. In addition, cell migration was assessed by wound healing, and invasion was evaluated by Transwell assay. Meanwhile, the correlation among circPVT1, miR-26b-5p and CCNB1 was explored by RNA pull-down and dual luciferase assay. Finally, in vivo model was established to explore the role of circPVT1 in OS in vivo. RESULTS CircPVT1 and CCNB1 were significantly upregulated in OS cells, while miR-26b-5p was downregulated. Knockdown of circPVT1 notably inhibited proliferation and induced apoptosis of OS cells. CircPVT1 shRNA significantly suppressed the OS cell invasion and migration. Meanwhile, circPVT1 sponged miR-26b-5p and CCNB1 was found to be the direct target of miR-26b-5p. Furthermore, silencing of circPVT1 inhibited the growth and metastasis of OS in vivo. CONCLUSION Silencing of circPVT1 notably suppressed the tumorigenesis and metastasis of OS via miR-26b-5p/CCNB1 axis. Therefore, circPVT1 might be used as a target for OS treatment.
Collapse
Affiliation(s)
- Sheng-Xiang Huang
- Department of Pediatric Orthopedics, Hunan Children's Hospital, No. 86, Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Hai-Bo Mei
- Department of Pediatric Orthopedics, Hunan Children's Hospital, No. 86, Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Kun Liu
- Department of Pediatric Orthopedics, Hunan Children's Hospital, No. 86, Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Jin Tang
- Department of Pediatric Orthopedics, Hunan Children's Hospital, No. 86, Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Jiang-Yan Wu
- Department of Pediatric Orthopedics, Hunan Children's Hospital, No. 86, Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Guang-Hui Zhu
- Department of Pediatric Orthopedics, Hunan Children's Hospital, No. 86, Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China
| | - Wei-Hua Ye
- Department of Pediatric Orthopedics, Hunan Children's Hospital, No. 86, Ziyuan Road, Changsha, 410007, Hunan, People's Republic of China.
| |
Collapse
|
43
|
Xing CY, Zhang YZ, Hu W, Zhao LY. LINC00313 facilitates osteosarcoma carcinogenesis and metastasis through enhancing EZH2 mRNA stability and EZH2-mediated silence of PTEN expression. Cell Mol Life Sci 2022; 79:382. [PMID: 35751755 PMCID: PMC11073393 DOI: 10.1007/s00018-022-04376-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 05/13/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Osteosarcoma is one of the five leading causes of cancer death among all pediatric malignancies. Recent advances in non-coding RNAs suggested that many long noncoding RNAs (lncRNAs) are dysregulated in cancer tissues and play important roles in carcinogenesis. We aimed to further explore the mechanisms of Long Intergenic Non-Protein Coding RNA 313 (LINC00313)-promoted malignant phenotypes of osteosarcoma. METHODS The mRNA expressions were determined by quantitative real-time PCR. Protein levels were detected using Western blotting or immunohistochemistry staining. Protein binding to genomic DNA and RNA were measured using chromatin and RNA immunoprecipitation assay, respectively. CCK-8 and EdU incorporation assay were adopted to detect cell proliferation. Transwell assay was employed to assess the capacity of cell migration and invasion. The roles of LINC00313 and its target genes in tumorigenesis and metastasis of osteosarcoma were evaluated using subcutaneous xenograft models and tail vein inoculation models. RESULTS LINC00313 was elevated in osteosarcoma tissues compared with adjacent normal tissues. Higher LINC00313 was associated with advanced grades of osteosarcoma. LINC00313 promoted cell proliferation, migration, invasion in vitro and tumor growth as well as metastasis in vivo through inhibiting PTEN expression to promote AKT phosphorylation. Mechanistically, LINC00313 favored the interaction between FUS and EZH2, leading to the prolonged half-life of EZH2 mRNA, thereby in turn up-regulating EZH2 proteins and increasing EZH2-mediated epigenetic silence of PTEN. CONCLUSION LINC00313 exerted oncogene-like actions through increasing EZH2 mRNA stability, leading to PTEN deficiency in osteosarcoma.
Collapse
Affiliation(s)
- Chun-Yang Xing
- Department of Orthopedics, First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79, Qingchun Road, Hangzhou, 310003, Zhejiang Province, China.
| | - Yu-Zhu Zhang
- Department of Orthopedics, First Affiliated Hospital, College of Medicine, Zhejiang University, No. 79, Qingchun Road, Hangzhou, 310003, Zhejiang Province, China
| | - Wei Hu
- Department of Radiotherapy, The People's Hospital of Changxing County, Huzhou, 313199, Zhejiang Province, China
| | - Li-Yuan Zhao
- Department of Emergency, Qilu Hospital, Shandong University, Jinan, 250063, Shandong Province, China
| |
Collapse
|
44
|
Exosomal CTCF Confers Cisplatin Resistance in Osteosarcoma by Promoting Autophagy via the IGF2-AS/miR-579-3p/MSH6 Axis. JOURNAL OF ONCOLOGY 2022; 2022:9390611. [PMID: 35693981 PMCID: PMC9175095 DOI: 10.1155/2022/9390611] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/21/2022] [Indexed: 11/18/2022]
Abstract
Cancer-derived exosomes participate in carcinogenesis and progression of cancers, including metastasis and drug-resistance. Of note, CTCF has been suggested to induce drug resistance in various cancers. Herein, we aim to investigate the role of cisplatin- (CDDP-) resistant osteosarcoma- (OS-) derived exosomal CTCF in OS cell resistance to CDDP and its mechanistic basis. Differentially expressed transcription factors, long noncoding RNAs (lncRNAs), miRNAs, and genes in OS were retrieved using bioinformatics approaches. Exosomes were extracted from CDDP-resistant OS cells and then cocultured with parental OS cells, followed by lentiviral transduction to manipulate the expression of CTCF, IGF2-AS, miR-579-3p, and MSH6. We assessed the in vitro and in vivo effects on malignant phenotypes, autophagy, CDDP sensitivity, and tumor formation of OS cells. It was established that CTCF and IGF2-AS were highly expressed in CDDP-resistant OS cells, and the CDDP-resistant OS cell-derived exosomal CTCF enhanced IGF2-AS transcription. CDDP-resistant OS-derived exosomes transmitted CTCF to OS cells and increased CDDP resistance in OS cells by activating an autophagy-dependent pathway. Mechanistically, CTCF activated IGF2-AS transcription and IGF2-AS competitively bound to miR-579-3p to upregulate MSH6 expression. Additionally, the promoting function of exosomal CTCF-mediated IGF2-AS/miR-579-3p/MSH6 in OS cell resistance to CDDP was confirmed in vivo. Taken together, CDDP-resistant OS-derived exosomal CTCF enhanced resistance of OS cells to CDDP via activating the autophagy-dependent pathway, providing a potential therapeutic consideration for OS treatment.
Collapse
|
45
|
Ling S, Luo X, Lv B, Wang H, Xie M, Huang K, Sun J. Effect of miR-144-3p-Targeted Regulation of PTEN on Proliferation, Apoptosis, and Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells under Stretch. Emerg Med Int 2022; 2022:5707504. [PMID: 35592654 PMCID: PMC9113913 DOI: 10.1155/2022/5707504] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/15/2022] [Indexed: 12/04/2022] Open
Abstract
Objective To investigate the effects of miR-144-3p-targeted regulation of phosphatase and tensin homolog deleted on chromosome ten (PTEN) gene on proliferation, apoptosis, and osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) under retraction force. Methods The BMSCs of rats were randomly divided into the tension MSC group with detrusor stimulation and the MSC group without detrusor stimulation, after which osteogenic differentiation of BMSCs was induced in both groups. Alkaline phosphatase (ALP) staining and alizarin red staining were used to detect the osteogenic differentiation ability of the two groups of cells. Real-time quantitative reverse transcription PCR (qRT-PCR) was used to detect the expression of miR-144-3p and PTEN in the two groups of cells after osteogenic differentiation. Bioinformatics website and dual luciferase reporter were used to detect the relationship between miR-144-3p and PTEN. The tension MSC group was used as a control group, and miR-144-3p mimics (miR-144-3p mimic group), mimic controls (mimic-NC group), PTEN interferers (si-PTEN group), and interference controls (si-NC group) were transfected into BMSCs. The BMSCs were then continuously stimulated for 24 h using a Flexercell in vitro cellular mechanics loading device, applying a draft force at a frequency of 1 Hz and a deformation rate of 18%. The cell proliferation was detected by Cell Counting Kit-8 (CCK-8) colorimetric assay; the expression levels of cyclin, cyclin-dependent kinases (CDK), BCL2-associated X (BAX), B-cell lymphoma-2 (BCL-2), and other cell cycle and apoptosis related proteins were detected by western blot (WB); and the osteogenic differentiation ability of MSC cells was detected by ALP staining and alizarin red staining. Results Compared with the MSC group, the level of miR-144-3p was significantly lower and the level of PTEN was significantly higher in the tension MSC group. ALP staining showed normal activity in the MSC group and decreased ALP activity in the tension MSC group compared to the MSC group. Alizarin red staining in the MSC group showed scattered calcium nodule formation, and alizarin red staining showed red nodules with a more uniform color distribution. Compared to the MSC group, the tension MSC group showed fewer, smaller, and lighter staining mineralized nodules. Compared with the tension group and mimic-NC group (si-NC group), the proliferation rate of cells in the miR-144-3p mimic group (si-PTEN group) was significantly higher; the expression levels of PTEN and BAX were significantly lower; and the expression levels of cyclin, CDK, and BCL-2 protein were significantly higher. ALP staining results revealed that the miR-144-3p mimic group (si-PTEN group) showed significantly higher osteogenic differentiation ability and ALP activity of MSC than the tension group and mimic-NC group (si-NC group). Conclusion miR-144-3p may inhibit apoptosis and promote proliferation and osteogenic differentiation of BMSCs under tension by targeting and regulating PTEN.
Collapse
Affiliation(s)
- Shiyong Ling
- Department of Orthopedics, Zhabei Central Hospital, Jing'an District, Shanghai 200070, China
| | - Xi Luo
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Bo Lv
- Department of Orthopedics, Zhabei Central Hospital, Jing'an District, Shanghai 200070, China
| | - Hua Wang
- Department of Orthopedics, Zhabei Central Hospital, Jing'an District, Shanghai 200070, China
| | - Mengzhi Xie
- Department of Radiology, Zhabei Central Hospital, Jing'an District, Shanghai 200070, China
| | - Kai Huang
- Department of Orthopedics, Zhabei Central Hospital, Jing'an District, Shanghai 200070, China
| | - Jingchuan Sun
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| |
Collapse
|
46
|
Zhang Z, Hu J, Ishihara M, Sharrow AC, Flora K, He Y, Wu L. The miRNA-21-5p Payload in Exosomes from M2 Macrophages Drives Tumor Cell Aggression via PTEN/Akt Signaling in Renal Cell Carcinoma. Int J Mol Sci 2022; 23:3005. [PMID: 35328425 PMCID: PMC8949275 DOI: 10.3390/ijms23063005] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/06/2022] [Accepted: 03/08/2022] [Indexed: 12/15/2022] Open
Abstract
M2 macrophages in the tumor microenvironment are important drivers of cancer metastasis. Exosomes play a critical role in the crosstalk between different cells by delivering microRNAs or other cargos. Whether exosomes derived from pro-tumorigenic M2 macrophages (M2-Exos) could modulate the metastatic behavior of renal cell carcinoma (RCC) is unclear. This study found that M2-Exos promotes migration and invasion in RCC cells. Inhibiting miR-21-5p in M2-Exos significantly reversed their pro-metastatic effects on RCC cells in vitro and in the avian embryo chorioallantoic membrane in vivo tumor model. We further found that the pro-metastatic mechanism of miR-21-5p in M2-Exos is by targeting PTEN-3'UTR to regulate PTEN/Akt signaling. Taken together, our results demonstrate that M2-Exos carries miR-21-5p promote metastatic features of RCC cells through PTEN/Akt signaling. Reversing this could serve as a novel approach to control RCC metastasis.
Collapse
Affiliation(s)
- Zhicheng Zhang
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; (Z.Z.); (J.H.); (M.I.); (A.C.S.); (K.F.)
| | - Junhui Hu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; (Z.Z.); (J.H.); (M.I.); (A.C.S.); (K.F.)
| | - Moe Ishihara
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; (Z.Z.); (J.H.); (M.I.); (A.C.S.); (K.F.)
| | - Allison C. Sharrow
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; (Z.Z.); (J.H.); (M.I.); (A.C.S.); (K.F.)
| | - Kailey Flora
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; (Z.Z.); (J.H.); (M.I.); (A.C.S.); (K.F.)
| | - Yao He
- Department of Microbiology, Immunology and Molecular Genetics, UCLA, Los Angeles, CA 90095, USA;
| | - Lily Wu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; (Z.Z.); (J.H.); (M.I.); (A.C.S.); (K.F.)
- Department of Urology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
47
|
Biglycan Interacts with Type I Insulin-like Receptor (IGF-IR) Signaling Pathway to Regulate Osteosarcoma Cell Growth and Response to Chemotherapy. Cancers (Basel) 2022; 14:cancers14051196. [PMID: 35267503 PMCID: PMC8909324 DOI: 10.3390/cancers14051196] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Osteosarcoma (OS) is an aggressive, primary bone cancer. OS cells produce altered osteoid whose components participate in signaling correlated to the development of this cancer. Biglycan (BGN), a proteoglycan, is correlated to aggressive OS type and resistance to chemotherapy. A constitutive signaling of insulin-like growth factor receptor I (IGF-IR) signaling in sarcoma progression was established. We showed that biglycan binds IGF-IR resulting in prolonged IGF-IR activation, nuclear translocation, and growth response of the poorly-differentiated MG63 cells correlated to increased aggressiveness markers expression and enhanced chemoresistance. This mechanism is not valid in moderately and well-differentiated, biglycan non-expressing U-2OS and Saos-2 OS cells. Abstract Osteosarcoma (OS) is a mesenchymally derived, aggressive bone cancer. OS cells produce an aberrant nonmineralized or partly mineralized extracellular matrix (ECM) whose components participate in signaling pathways connected to specific pathogenic phenotypes of this bone cancer. The expression of biglycan (BGN), a secreted small leucine-rich proteoglycan (SLRP), is correlated to aggressive OS phenotype and resistance to chemotherapy. A constitutive signaling of IGF-IR signaling input in sarcoma progression has been established. Here, we show that biglycan activates the IGF-IR signaling pathway to promote MG63 biglycan-secreting OS cell growth by forming a complex with the receptor. Computational models of IGF-IR and biglycan docking suggest that biglycan binds IGF-IR dimer via its concave surface. Our binding free energy calculations indicate the formation of a stable complex. Biglycan binding results in prolonged IGF-IR activation leading to protracted IGF-IR-dependent cell growth response of the poorly-differentiated MG63 cells. Moreover, biglycan facilitates the internalization (p ≤ 0.01, p ≤ 0.001) and sumoylation-enhanced nuclear translocation of IGF-IR (p ≤ 0.05) and its DNA binding in MG63 cells (p ≤ 0.001). The tyrosine kinase activity of the receptor mediates this mechanism. Furthermore, biglycan downregulates the expression of the tumor-suppressor gene, PTEN (p ≤ 0.01), and increases the expression of endothelial–mesenchymal transition (EMT) and aggressiveness markers vimentin (p ≤ 0.01) and fibronectin (p ≤ 0.01) in MG63 cells. Interestingly, this mechanism is not valid in moderately and well-differentiated, biglycan non-expressing U-2OS and Saos-2 OS cells. Furthermore, biglycan exhibits protective effects against the chemotherapeutic drug, doxorubicin, in MG63 OS cells (p ≤ 0.01). In conclusion, these data indicate a potential direct and adjunct therapeutical role of biglycan in osteosarcoma.
Collapse
|
48
|
Sun X, Shan HJ, Yin G, Zhang XY, Huang YM, Li HJ. The anti-osteosarcoma cell activity by the sphingosine kinase 1 inhibitor SKI-V. Cell Death Dis 2022; 8:48. [PMID: 35115496 PMCID: PMC8814198 DOI: 10.1038/s41420-022-00838-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 01/03/2022] [Accepted: 01/14/2022] [Indexed: 01/04/2023]
Abstract
Sphingosine kinase 1 (SphK1) expression and activity are elevated in human osteosarcoma (OS) and is a promising target of therapy. SKI-V is a non-competitive and highly-efficient non-lipid SphK1 inhibitor. The potential anti-OS cell activity by the SphK1 inhibitor was studied here. In primary OS cells and immortalized cell lines, SKI-V robustly suppressed cell survival, growth and proliferation as well as cell mobility, and inducing profound OS cell death and apoptosis. The SphK1 inhibitor was however non-cytotoxic nor pro-apoptotic in human osteoblasts. SKI-V robustly inhibited SphK1 activation and induced accumulation of ceramides, without affecting SphK1 expression in primary OS cells. The SphK1 activator K6PC-5 or sphingosine-1-phosphate partially inhibited SKI-V-induced OS cell death. We showed that SKI-V concurrently blocked Akt-mTOR activation in primary OS cells. A constitutively-active Akt1 (ca-Akt1, S473D) construct restored Akt-mTOR activation and mitigated SKI-V-mediated cytotoxicity in primary OS cells. In vivo, daily injection of SKI-V potently suppressed OS xenograft tumor growth in nude mice. In SKI-V-administrated OS xenograft tissues, SphK1 inhibition, ceramide increase and Akt-mTOR inhibition were detected. Together, SKI-V exerts significant anti-OS activity by inhibiting SphK1 and Akt-mTOR cascades in OS cells.
Collapse
Affiliation(s)
- Xu Sun
- Department of Hand and Foot Surgery, Hospital Affiliated 5 to Nantong University, Taizhou People's Hospital, Taizhou, China
| | - Hua-Jian Shan
- Department of Orthopaedics, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Gang Yin
- Department of Orthopaedics, Wujin Hospital Affiliated to Jiangsu University, Changzhou, China
| | - Xiang-Yang Zhang
- Department of Orthopaedics, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Min Huang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Hai-Jun Li
- Department of Hand and Foot Surgery, Hospital Affiliated 5 to Nantong University, Taizhou People's Hospital, Taizhou, China.
| |
Collapse
|
49
|
Jiang ZY, Liu JB, Wang XF, Ma YS, Fu D. Current Status and Prospects of Clinical Treatment of Osteosarcoma. Technol Cancer Res Treat 2022; 21:15330338221124696. [PMID: 36128851 PMCID: PMC9500272 DOI: 10.1177/15330338221124696] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Osteosarcoma, one of the common malignant tumors in the skeletal system, originates in mesenchymal tissue, and the most susceptible area of occurrence is the metaphysis with its abundant blood supply. Tumors are characterized by highly malignant spindle stromal cells that can produce bone-like tissue. Most of the osteosarcoma are primary, and a few are secondary. Osteosarcoma occurs primarily in children and adolescents undergoing vigorous bone growth and development. Most cases involve rapid tumor development and early blood metastasis. In recent years, research has grown in the areas of molecular biology, imaging medicine, biological materials, applied anatomy, surgical techniques, biomechanics, and comprehensive treatment of tumors. With developments in molecular biology and tissue bioengineering, treatment methods have also made great progress, especially in comprehensive limb salvage treatment, which significantly enhances the quality of life after surgery and improves the 5-year survival rate of patients with malignant tumors. This article provides a review of limb salvage, immunotherapy, gene therapy, and targeted therapy from traditional amputation to neoadjuvant chemotherapy, providing a reference for current clinical treatments for osteosarcoma.
Collapse
Affiliation(s)
- Zong-Yuan Jiang
- Department of Hand Surgery, 380381Shenzhen Longhua District People's Hospital, Shenzhen, China
| | - Ji-Bin Liu
- Institute of Oncology, Nantong UniversityAffiliated Tumor Hospital of Nantong University, Nantong, China
| | - Xiao-Feng Wang
- Department of Orthopedics, Zhongshan Hospital, 12478Fudan University, Shanghai, China
| | - Yu-Shui Ma
- Cancer Institute, 74754Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Da Fu
- Department of General Surgery, Ruijin Hospital, 12474Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
50
|
Liu R, Hu Y, Liu T, Wang Y. Profiles of immune cell infiltration and immune-related genes in the tumor microenvironment of osteosarcoma cancer. BMC Cancer 2021; 21:1345. [PMID: 34922489 PMCID: PMC8684084 DOI: 10.1186/s12885-021-09042-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 11/18/2021] [Indexed: 11/23/2022] Open
Abstract
Backgrounds Osteosarcomas are one of the most common primary malignant tumors of bone. It primarily occurs in children and adolescents, with the second highest incidence among people over 50 years old. Although there were immense improvements in the survival of patients with osteosarcoma in the past 30 years, targetable mutations and agents of osteosarcomas still have been generally not satisfactory. Therefore, it is of great importance to further explore the highly specialized immune environment of bone, genes related to macrophage infiltration and potential therapeutic biomarkers and targets. Methods The 11 expression data sets of OS tissues and the 11 data sets of adjacent non-tumorous tissues available in the GEO database GSE126209 were used to conduct immune infiltration analysis. Then, through WGCNA analysis, we acquired the co-expression modules related to Mast cells activated and performed the GO and KEGG enrichment analysis. Next, we did the survival prognosis analysis and plotted a survival curve. Finally, we analyzed the COX multivariate regression of gene expression on clinical parameters and drew forest maps for visualization by the forest plot package. Results OS disease-related immune cell populations, mainly Mast cells activated, have higher cell content (p = 0.006) than the normal group. Then, we identified co-expression modules related to Mast cells activated. In sum, a total of 822 genes from the top three strongest positive correlation module MEbrown4, MEdarkslateblue and MEnavajowhite2 and the strongest negative correlation module MEdarkturquoise. From that, we identified nine genes with different levels in immune cell infiltration related to osteosarcoma, eight of which including SORBS2, BAIAP2L2, ATAD2, CYGB, PAMR1, PSIP1, SNAPC3 and ZDHHC21 in their low abundance have higher disease-free survival probability than the group in their high abundances. Conclusion These results could assist clinicians to select targets for immunotherapies and individualize treatment strategies for patients with OS. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-09042-6.
Collapse
Affiliation(s)
- Ruixuan Liu
- Department of Spine Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Yuhang Hu
- Department of Spine Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Tianyi Liu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Yansong Wang
- Department of Spine Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.
| |
Collapse
|