1
|
Gao Q, Wang M, Hou X, Li M, Li L. Substrate stiffness modulates osteogenic and adipogenic differentiation of osteosarcoma through PIEZO1 mediated signaling pathway. Cell Signal 2025; 127:111601. [PMID: 39798771 DOI: 10.1016/j.cellsig.2025.111601] [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: 08/01/2024] [Revised: 12/29/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Most osteosarcoma (OS) cases exhibit poor differentiation at the histopathological level. Disruption of the normal osteogenic differentiation process results in the unregulated proliferation of precursor cells, which is a critical factor in the development of OS. Differentiation therapy aims to slow disease progression by restoring the osteogenic differentiation process of OS cells and is considered a new approach to treating OS. However, there are currently few studies on the mechanism of differentiation of OS, which puts the development of differentiation therapeutic drugs into a bottleneck. Substrate stiffness can regulate differentiation in mesenchymal stem cells. Evidence supports that mesenchymal stem cells and osteoblast precursors are the origin of OS. In this study, we simulated different stiffnesses in vitro to investigate the mechanism of substrate stiffness affecting differentiation of OS. We demonstrate that Piezo type mechanosensitive ion channel component 1 (PIEZO1) plays a critical regulatory role in sensing substrate stiffness in osteogenic and adipogenic differentiation of OS. When OS cells are cultured on the stiff substrate, integrin subunit beta 1 (ITGB1) increases and cooperates with PIEZO1 to promote Yes-Associated Protein (YAP) entering the nucleus, and may inhibit EZH2, thereby inhibiting H3K27me3 and increasing RUNX2 expression, and cells differentiate toward osteogenesis. Our results provide new insights for research on differentiation treatment of OS and are expected to help identify new targets for future drug design.
Collapse
Affiliation(s)
- Qingyuan Gao
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China
| | - Meijing Wang
- Department of Pathology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Xiangyi Hou
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China
| | - Meiying Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, Jilin Province, China.
| |
Collapse
|
2
|
Zhang J, Lai Z, Zhang Z, Zheng S. Fluorescent monitoring osteogenic differentiation of osteosarcoma cells with an aggregation-induced emission probe. Heliyon 2024; 10:e31664. [PMID: 38828353 PMCID: PMC11140698 DOI: 10.1016/j.heliyon.2024.e31664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/19/2024] [Accepted: 05/20/2024] [Indexed: 06/05/2024] Open
Abstract
Osteosarcoma is widely believed to be an osteogenic differentiation disorder. In recent years, to further understand this disease, a lot resources were poured into the potential link between differentiation defects and tumorigenesis. Long-term monitoring of the differentiation progress of osteosarcoma cells is of great importance. In order to better promote the research, we have developed a novel fluorescent probe called PTB-EDTA, which exhibits remarkable bio-compatibility and demonstrates high selectivity towards osteosarcoma cells. Not only is the PTB-EDTA is capable of live cell imaging while conventional histology requires to kill the cells, its fluorescence is also enhanced as the osteogenic differentiation proceeding. These properties make PTB-EDTA a promising tool for monitoring osteosarcoma cells.
Collapse
Affiliation(s)
- Junxiong Zhang
- Department of Orthopaedics, The First Affiliated Hospital, Southern Medical University, Academy of Orthopaedics, Guangdong Province, Guangzhou, 510515, China
| | - Zhongming Lai
- Department of Orthopaedics, The First Affiliated Hospital, Southern Medical University, Academy of Orthopaedics, Guangdong Province, Guangzhou, 510515, China
| | - Zhongmin Zhang
- Department of Orthopaedics, The First Affiliated Hospital, Southern Medical University, Academy of Orthopaedics, Guangdong Province, Guangzhou, 510515, China
| | - Shuai Zheng
- Department of Orthopaedics, The First Affiliated Hospital, Southern Medical University, Academy of Orthopaedics, Guangdong Province, Guangzhou, 510515, China
| |
Collapse
|
3
|
Shi Y, Kang Q, Zhou H, Yue X, Bi Y, Luo Q. Aberrant LETM1 elevation dysregulates mitochondrial functions and energy metabolism and promotes lung metastasis in osteosarcoma. Genes Dis 2024; 11:100988. [PMID: 38292199 PMCID: PMC10825238 DOI: 10.1016/j.gendis.2023.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/10/2023] [Accepted: 05/25/2023] [Indexed: 02/01/2024] Open
Abstract
Osteosarcoma is a differentiation-deficient disease, and despite the unique advantages and great potential of differentiation therapy, there are only a few known differentiation inducers, and little research has been done on their targets. Cell differentiation is associated with an increase in mitochondrial content and activity. The metabolism of some tumor cells is characterized by impaired oxidative phosphorylation, as well as up-regulation of aerobic glycolysis and pentose phosphate pathways. Leucine-containing zipper and EF-hand transmembrane protein 1 (LETM1) is involved in the maintenance of mitochondrial morphology and is closely associated with tumorigenesis and progression, as well as cancer cell stemness. We found that MG63 and 143B osteosarcoma cells overexpress LETM1 and exhibit abnormalities in mitochondrial structure and function. Knockdown of LETM1 partially restored the mitochondrial structure and function, inhibited the pentose phosphate pathway, promoted oxidative phosphorylation, and led to osteogenic differentiation. It also inhibited spheroid cell formation, proliferation, migration, and invasion in an in vitro model. When LETM1 was knocked down in vivo, there was reduced tumor formation and lung metastasis. These data suggest that mitochondria are aberrant in LETM1-overexpressing osteosarcoma cells, and knockdown of LETM1 partially restores the mitochondrial structure and function, inhibits the pentose phosphate pathway, promotes oxidative phosphorylation, and increases osteogenic differentiation, thereby reducing malignant biological behavior of the cells.
Collapse
Affiliation(s)
- Yulu Shi
- Stem Cell Biology and Therapy Laboratory, The Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Quan Kang
- Department of Pediatric Surgery, The Children’s Hospital of Chongqing Medical University, Chongqing 400014, China
| | - Hong Zhou
- Stem Cell Biology and Therapy Laboratory, The Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Xiaohan Yue
- Stem Cell Biology and Therapy Laboratory, The Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Yang Bi
- Stem Cell Biology and Therapy Laboratory, The Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Qing Luo
- Stem Cell Biology and Therapy Laboratory, The Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| |
Collapse
|
4
|
Zhang F, Chen J, Luo W, Wen C, Mao W, Yang Y, Liu C, Xu Y, Chen W, Wen L. Mitochondria targeted biomimetic platform for chemo/photodynamic combination therapy against osteosarcoma. Int J Pharm 2024; 652:123865. [PMID: 38286195 DOI: 10.1016/j.ijpharm.2024.123865] [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: 11/07/2023] [Revised: 01/13/2024] [Accepted: 01/26/2024] [Indexed: 01/31/2024]
Abstract
Clinical treatment for osteosarcoma (OS) is still lacking effective means, and no significant progress in OS treatment have been made in recent years. Single chemotherapy has serious side effects and can produce drug resistance easily, resulting poor therapeutic effect. As a modern and non-invasive treatment form, photodynamic therapy (PDT) is widely used to treat diverse cancers. Chemotherapy in combination with PDT is a particularly efficient antitumor method that could overcome the defects of monotherapies. Since mitochondria is a key subcellular organelle involved in cell apoptosis regulation, targeting tumor cells mitochondria for drug delivery has become an important entry point for anti-tumor therapy. Herein, we rationally designed a core-shell structured biomimetic nanoplatform, i.e., D@SLNP@OSM-IR780, to achieve tumor homologous targeting and mitochondria targeted drug release for chemotherapy combined with PDT against OS. Upon 808 nm laser irradiation, D@SLNP@OSM-IR780 exhibited excellent photo-cytotoxicity in vitro. The excellent targeting effect of D@SLNP@OSM-IR780 in tumor tissues produced a tumor inhibition rate of 98.9% in vivo. We further indicated that synergistic chemo-photodynamic effect induced by D@SLNP@OSM-IR780 could activate mitochondria-mediated apoptosis pathway, along with host immune response and potential photothermal effect. On the whole, D@SLNP@OSM-IR780 is revealed to be a promising platform for OS targeted combination therapeutics.
Collapse
Affiliation(s)
- Fengtian Zhang
- Department of Orthopaedics, Second Affiliated Hospital of Soochow University Osteoporosis Research Institute of Soochow University, 1055 Sanxiang Road, Suzhou 215000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China; Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Jinling East Avenue, Zhanggong District, Ganzhou 341000, People's Republic of China
| | - Jiaoting Chen
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Weihong Luo
- College of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Changlong Wen
- Department of Infectious Diseases, Ganzhou People's Hospital, 17 Hongqi Avenue, Zhanggong District, Ganzhou 341000, People's Republic of China
| | - Wei Mao
- College of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Yutian Yang
- College of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Chunting Liu
- People's Hospital of Shicheng County, Xihua Middle Road, Qinjiang Town, Ganzhou 342700, People's Republic of China
| | - Youjia Xu
- Department of Orthopaedics, Second Affiliated Hospital of Soochow University Osteoporosis Research Institute of Soochow University, 1055 Sanxiang Road, Suzhou 215000, People's Republic of China.
| | - Weiliang Chen
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China; College of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China.
| | - Lijuan Wen
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China; College of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China.
| |
Collapse
|
5
|
Su H, Lv Y, Zhu L, Wang Y. Roles of PTEN gene methylation in Se-CQDs induced mitochondrial apoptosis of osteosarcoma cells. Colloids Surf B Biointerfaces 2024; 234:113757. [PMID: 38241895 DOI: 10.1016/j.colsurfb.2024.113757] [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: 10/19/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/21/2024]
Abstract
Biocompatible carbon quantum dots (CQDs) containing anti-osteosarcoma elements are intriguing therapeutics promising for bioimaging and tumor therapy. However, how the anti-osteosarcoma element doped in the structure of such CQDs triggers tumor inhibition remains unclear. Here, selenium-doped CQDs (Se-CQDs) are developed via a one-step hydrothermal route using discarded orange peel as a carbon source and structurally characterized by various physicochemical techniques. The biocompatibility and anti-osteosarcoma efficacy are deeply evaluated using animal and cell models. The resulting spherical Se-CQDs, with a 3-7 nm diameter, possess green-yellow tunable luminescence and excellent biocompatibility. Cell experiments show that Se-CQDs can be up-taken by osteosarcoma U2OS cells and activate the mitochondrial apoptosis pathway triggered by increased reactive oxygen species. They can arrest the cell cycle at the G2/S phase and promote cellular apoptosis with reduced invasion and migration. Molecularly, Se-CQDs can down-regulate the expression of DNMT1 while up-regulating the expression of PTEN due to the decreased promoter methylation. Notably, Se-incorporated CQDs are more effective in inhibiting the proliferation, migration, and invasion of osteosarcoma than Se-free CQDs. It is feasible to use Se-CQDs as candidates for the potential application of early monitoring and treatment of osteosarcoma.
Collapse
Affiliation(s)
- Huahua Su
- The Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Yan Lv
- The Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Lixian Zhu
- The Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang 443002, China
| | - Yanhua Wang
- Department of Morphology, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China; The Third-Grade Pharmacological Laboratory on Chinese Medicine (Approved by State Administration of Traditional Chinese Medicine), China Three Gorges University, Yichang 443002, China.
| |
Collapse
|
6
|
Huang L, Liang W, Cai W, Peng H. Circadian rhythm-associated lncRNA RP11-414H17.5 as a key therapeutic target in osteosarcoma affects the tumor immune microenvironment and enhances malignancy. J Orthop Surg Res 2023; 18:947. [PMID: 38071320 PMCID: PMC10710728 DOI: 10.1186/s13018-023-04442-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND It has previously been proven that circadian rhythm disruption is associated with the incidence and deterioration of several tumors, which potentially leads to increased tumor susceptibility and a worse prognosis for tumor-bearing patients. However, their potential role in osteosarcoma has yet to be sufficiently investigated. METHODS Transcriptomic and clinical data of 84 osteosarcoma samples and 70 normal bone tissue samples were obtained from the TARGET and GTEx databases, circadian rhythm-related genes were obtained from Genecards, and circadian rhythm-related lncRNAs (CRLs) were obtained by Pearson correlation analysis, differential expression analysis, and protein-protein interaction (PPI) analysis. COX regression and LASSO regression were performed on the CRLs in order to construct a circadian rhythm-related prognostic prediction signature (CRPS). CRPS reliability was verified by Kaplan-Meier (KM), principal component analysis (PCA), nomogram, and receiver operating characteristic (ROC) curve. CRPS effects on the immune microenvironment of osteosarcoma were explored by enrichment analysis and immune infiltration analysis, and the effect of critical gene RP11-414H17.5 on osteosarcoma was experimentally verified. RESULT CRPS consisting of three CRLs was constructed and its area under the curve (AUC) values predicted that osteosarcoma prognosis reached 0.892 in the training group and 0.843 in the test group, with a p value of < 0.05 for the KM curve and stable performance across different clinical subgroups. PCA analysis found that CRPS could significantly distinguish between different risk subgroups, and exhibited excellent performance in the prediction of the immune microenvironment. The experiment verified that RP11-414H17.5 can promote metastasis and inhibit apoptosis of osteosarcoma cells. CONCLUSION The study revealed that circadian rhythm plays a crucial role in osteosarcoma progression and identified the impact of the key gene RP11-414H17.5 on osteosarcoma, which provides novel insights into osteosarcoma diagnosis and therapy.
Collapse
Affiliation(s)
- Liangkun Huang
- Department of Orthopedics Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wanting Liang
- Department of Clinical Medicine, Xiamen Medical College, Xiamen, 310058, China
| | - Wenxiang Cai
- Department of Orthopedics Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Hao Peng
- Department of Orthopedics Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
7
|
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
|
8
|
He C, Jiang Y, Guo Y, Wu Z. Amplified Ferroptosis and Apoptosis Facilitated by Differentiation Therapy Efficiently Suppress the Progression of Osteosarcoma. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2302575. [PMID: 37394717 DOI: 10.1002/smll.202302575] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/20/2023] [Indexed: 07/04/2023]
Abstract
Osteosarcoma (OS) is the most frequent osseous neoplasm among young people aged 10-20. Currently, the leading treatment for osteosarcoma is a combination of surgery and chemotherapy. However, the mortality remains high due to chemoresistance, metastasis, and recurrence, attributing to the existence of cancer stem cells (CSCs) as reported. To target CSCs, differentiation therapy attracts increasing attention, inducing CSCs to bulk tumor cells with elevated reactive oxygen species (ROS) levels and less chemoresistance. Moreover, increasing studies have implied that ferroptosis is a promising approach to eliminating cancer cells through eliciting oxidative damage and subsequent apoptosis, effectively bypassing chemoresistance. Here, a cancer-cell-membrane-decorated biocompatible formulation (GA-Fe@CMRALi liposome) is constructed to combat OS efficiently by combining distinct differentiation and ferroptosis therapies through magnified ROS-triggered ferroptosis and apoptosis with homologous target capability to tumor sites. The combinational approach exhibited favorable therapeutic efficacy against OS in vitro and in vivo. Impressively, the potential mechanisms are revealed by mRNA sequencing. This study provides a tactical design and typical paradigm of the synergized differentiation and ferroptosis therapies to combat heterogeneous OS.
Collapse
Affiliation(s)
- Chao He
- Department of Orthopedic Surgery, Translational Research Center of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Yuhang Jiang
- Department of Orthopedic Surgery, Translational Research Center of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Yuan Guo
- Department of Orthopedic Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| | - Zenghui Wu
- Department of Orthopedic Surgery, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China
| |
Collapse
|
9
|
Pan B, Pan Y, Wang S, Bai Y, Hu X, Yang Y, Wu L, Liu J. ANXA2 and Rac1 negatively regulates autophagy and osteogenic differentiation in osteosarcoma cells to confer CDDP resistance. Biochem Biophys Res Commun 2023; 676:198-206. [PMID: 37536195 DOI: 10.1016/j.bbrc.2023.07.006] [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: 06/02/2023] [Revised: 06/19/2023] [Accepted: 07/03/2023] [Indexed: 08/05/2023]
Abstract
BACKGROUND Cisplatin (CDDP) is a mainstay chemotherapeutic agent for OS treatment, but drug resistance has become a hurdle to limit its clinical effect. Autophagy plays an important role in CDDP resistance in OS, and in the present study we explored the role of ANXA2 and Rac1 in dictating CDDP sensitivity in OS cells. METHODS ANXA2 and Rac1 expression levels were examined by Western blot and autophagy induction was detected by transmission electron miscroscope (TEM) in the clinical samples and OS cell lines. CDDP resistant cells were established by exposing OS cells to increasing doses of CDDP. The effects of ANXA2 and Rac1 knockdown on CDDP sensitivity were evaluated in the cell and animal models. RESULTS Reduced autophagy was associated with the increased expression of ANXA2 and Rac1 in CDDP resistant OS tumor samples and cells. Autophagy suppression promoted CDDP resistance and inducing autophagy re-sensitized the resistant cells to CDDP treatment in vitro and in vivo. Further, knocking down ANXA2 or Rac1 re-activated autophagy and attenuated CDDP resistance in OS cells. We further demonstrated that CDDP resistant OS cells displayed a poorer osteogenic differentiation state when compared to the parental cell lines, which was significantly reversed by autophagy re-activation and ANXA2 or Rac1 silencing. CONCLUSION Our findings revealed a complicated interplay of ANXA2/Rac1, autophagy induction, and osteogenic differentiation in dictating CDDP resistance in OS cells, suggesting ANXA2 and Rac1 as promising targets to modulate autophagy and overcome CDDP resistance in OS cells.
Collapse
Affiliation(s)
- Baolong Pan
- Health Examination Center, Sixth Affiliated Hospital of Kunming Medical University, Yuxi, 653100, Yunnan, China
| | - Yanyu Pan
- College of Basic Medical Sciences, Naval Medical University, Shanghai, 200433, China
| | - Shuangneng Wang
- Health Examination Center, Sixth Affiliated Hospital of Kunming Medical University, Yuxi, 653100, Yunnan, China
| | - Yingying Bai
- Health Examination Center, Sixth Affiliated Hospital of Kunming Medical University, Yuxi, 653100, Yunnan, China
| | - Xuemei Hu
- Health Examination Center, Sixth Affiliated Hospital of Kunming Medical University, Yuxi, 653100, Yunnan, China
| | - Yang Yang
- Health Examination Center, Sixth Affiliated Hospital of Kunming Medical University, Yuxi, 653100, Yunnan, China
| | - Ling Wu
- Department of Blood Composition Production, Central Blood Station of Yuxi City, Yuxi, 653100, Yunnan, China.
| | - Jianping Liu
- Research Management Department, Sixth Affiliated Hospital of Kunming Medical University, Yuxi, 653100, Yunnan, China.
| |
Collapse
|
10
|
Chen W, Li Z, Yu N, Zhang L, Li H, Chen Y, Gong F, Lin W, He X, Wang S, Wu Y, Ji G. Bone-targeting exosome nanoparticles activate Keap1 / Nrf2 / GPX4 signaling pathway to induce ferroptosis in osteosarcoma cells. J Nanobiotechnology 2023; 21:355. [PMID: 37775799 PMCID: PMC10541697 DOI: 10.1186/s12951-023-02129-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 09/25/2023] [Indexed: 10/01/2023] Open
Abstract
BACKGROUND In recent years, the development of BMSCs-derived exosomes (EXO) for the treatment of osteosarcoma (OS) is a safe and promising modality for OS treatment, which can effectively deliver drugs to tumor cells in vivo. However, the differences in the drugs carried, and the binding of EXOs to other organs limit their therapeutic efficacy. Therefore, improving the OS-targeting ability of BMSCs EXOs and developing new drugs is crucial for the clinical application of targeted therapy for OS. RESULTS In this study, we constructed a potential therapeutic nano platform by modifying BMSCs EXOs using the bone-targeting peptide SDSSD and encapsulated capreomycin (CAP) within a shell. These constructed nanoparticles (NPs) showed the ability of homologous targeting and bone-targeting exosomes (BT-EXO) significantly promotes cellular endocytosis in vitro and tumor accumulation in vivo. Furthermore, our results revealed that the constructed NPs induced ferroptosis in OS cells by prompting excessive accumulation of reactive oxygen species (ROS), Fe2+ aggregation, and lipid peroxidation and further identified the potential anticancer molecular mechanism of ferroptosis as transduced by the Keap1/Nrf2/GPX4 signaling pathway. Also, these constructed NP-directed ferroptosis showed significant inhibition of tumor growth in vivo with no significant side effects. CONCLUSION These results suggest that these constructed NPs have superior anticancer activity in mouse models of OS in vitro and in vivo, providing a new and promising strategy for combining ferroptosis-based chemotherapy with targeted therapy for OS.
Collapse
Affiliation(s)
- Wenkai Chen
- Department of Orthopedic Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zongguang Li
- Department of Orthopedic Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Naichun Yu
- Department of Orthopedic Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Linlin Zhang
- Department of Orthopedic Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Hongyu Li
- Department of Orthopedic Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yongjie Chen
- Department of Orthopedic Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Fengqing Gong
- Department of Orthopedic Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wenping Lin
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Xu He
- Department of Spine Surgery, Shenzhen Pingle Orthopedic Hospital, Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Siyuan Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Yue Wu
- Department of Pathology, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Guangrong Ji
- Department of Orthopedic Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
- Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, Xiamen Key Laboratory of Regeneration Medicine, Organ Transplantation Institute of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
11
|
Dong Z, Wang H, Wang G. Research progress on the pharmacological effects and chemical constituents of Pien Tze Huang and its potential Q-markers. Chin J Nat Med 2023; 21:658-669. [PMID: 37777316 DOI: 10.1016/s1875-5364(23)60400-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Indexed: 10/02/2023]
Abstract
Pien Tze Huang (PTH) was documented as an imperial prescription composed of Notoginseng Radix, Calculus Bovis, Snake Gallbladder, and Musk. It is famous in China and Asian countries due to its excellent effects in heat clearing, detoxifying, swelling reduction, and pain relieving. Modern pharmacological studies demonstrate that PTH shows excellent effects against various inflammatory diseases, liver diseases, and cancers. This review summaries the pharmacological effects, clinical applications, and mainchemical components of PTH. More importantly, its potential quality markers (Q-markers) were then analyzed based on the "five principles" of Q-markers under the guidance of Traditional Chinese Medicine theory, including transfer and traceability, specificity, efficacy, compatibility, and measurability. As a result, ginsenosides Rb1, ginsenoside Rg1, ginsenoside Rd, ginsenoside Re, notoginsenoside R1, dencichine, bilirubin, biliverdin, taurocholic acid, and muscone are considered as the Q-markers of PTH. These findings will provide guidance and assistance for the construction of a quality control system for PTH.
Collapse
Affiliation(s)
- Zhaomin Dong
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, Research Unit of PK-PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Hong Wang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, Research Unit of PK-PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Guangji Wang
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, Research Unit of PK-PD Based Bioactive Components and Pharmacodynamic Target Discovery of Natural Medicine of Chinese Academy of Medical Sciences, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
12
|
Han GD, Dai J, Hui HX, Zhu J. ALOX5AP suppresses osteosarcoma progression via Wnt/β-catenin/EMT pathway and associates with clinical prognosis and immune infiltration. J Orthop Surg Res 2023; 18:446. [PMID: 37344882 DOI: 10.1186/s13018-023-03919-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/08/2023] [Indexed: 06/23/2023] Open
Abstract
Osteosarcoma (OS) is one of the most common malignant neoplasms in children and adolescents. Immune infiltration into the microenvironment of the tumor has a positive correlation with overall survival in patients with OS. The purpose of this study was to search for potential diagnostic markers that are involved in immune cell infiltration for OS. Patients with OS who acquired metastases within 5 years (n = 34) were compared to patients who did not develop metastases within 5 years (n = 19). Differentially expressed genes (DEGs) were tested for in both patient groups. To discover possible biomarkers, the LASSO regression model and the SVM-RFE analysis were both carried out. With the assistance of CIBERSORT, the compositional patterns of the 22 different types of immune cell fraction in OS were estimated. In this research, a total of 33 DEGs were obtained: 33 genes were significantly downregulated. Moreover, we identified six critical genes, including ALOX5AP, HLA-DOA, HLA-DMA, HLA-DRB4, HCLS1 and LOC647450. ROC assays confirmed their diagnostic value with AUC > 0.7. In addition, we found that the six critical genes were associated with immune infiltration. Then, we confirmed the expression of ALOX5AP was distinctly decreased in OS specimens and cell lines. High expression of ALOX5AP predicted an advanced clinical stage and overall survival of OS patients. Functionally, we found that overexpression of ALOX5AP distinctly suppressed the proliferation, migration, invasion and EMT via modulating Wnt/β-catenin signaling. Overall, we found that ALOX5AP overexpression inhibits OS development via regulation of Wnt/β-catenin signaling pathways, suggesting ALOX5AP as a novel molecular biomarker for enhanced therapy of OS.
Collapse
Affiliation(s)
- Guo-Dong Han
- Department of Orthopedics, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Jian Dai
- Department of Orthopedics, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Hong-Xia Hui
- Department of Medical Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Jing Zhu
- Department of Medical Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China.
| |
Collapse
|
13
|
Zhang Y, Gan W, Ru N, Xue Z, Chen W, Chen Z, Wang H, Zheng X. Comprehensive multi-omics analysis reveals m7G-related signature for evaluating prognosis and immunotherapy efficacy in osteosarcoma. J Bone Oncol 2023; 40:100481. [PMID: 37139222 PMCID: PMC10149372 DOI: 10.1016/j.jbo.2023.100481] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 05/05/2023] Open
Abstract
Background Osteosarcoma is one of the most prevalent bone malignancies with a poor prognosis. The N7-methylguanosine (m7G) modification facilitates the modification of RNA structure and function tightly associated with cancer. Nonetheless, there is a lack of joint exploration of the relationship between m7G methylation and immune status in osteosarcoma. Methods With the support of TARGET and GEO databases, we performed consensus clustering to characterize molecular subtypes based on m7G regulators in all osteosarcoma patients. The least absolute shrinkage and selection operator (LASSO) method, Cox regression, and receiver operating characteristic (ROC) curves were employed to construct and validate m7G-related prognostic features and derived risk scores. In addition, GSVA, ssGSEA, CIBERSORT, ESTIMATE, and gene set enrichment analysis were conducted to characterize biological pathways and immune landscapes. We explored the relationship between risk scores and drug sensitivity, immune checkpoints, and human leukocyte antigens by correlation analysis. Finally, the roles of EIF4E3 in cell function were verified through external experiments. Results Two molecular isoforms based on regulator genes were identified, which presented significant discrepancies in terms of survival and activated pathways. Moreover, the six m7G regulators most associated with prognosis in osteosarcoma patients were identified as independent predictors for the construction of prognostic signature. The model was well stabilized and outperformed traditional clinicopathological features to reliably predict 3-year (AUC = 0.787) and 5-year (AUC = 0.790) survival in osteosarcoma cohorts. Patients with increased risk scores had a poorer prognosis, higher tumor purity, lower checkpoint gene expression, and were in an immunosuppressive microenvironment. Furthermore, enhanced expression of EIF4E3 indicated a favorable prognosis and affected the biological behavior of osteosarcoma cells. Conclusions We identified six prognostic relevant m7G modulators that may provide valuable indicators for the estimation of overall survival and the corresponding immune landscape in patients with osteosarcoma.
Collapse
|
14
|
Rossi F, Picone G, Cappadone C, Sorrentino A, Columbaro M, Farruggia G, Catelli E, Sciutto G, Prati S, Oliete R, Pasini A, Pereiro E, Iotti S, Malucelli E. Shedding Light on Osteosarcoma Cell Differentiation: Impact on Biomineralization and Mitochondria Morphology. Int J Mol Sci 2023; 24:ijms24108559. [PMID: 37239904 DOI: 10.3390/ijms24108559] [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: 04/21/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumor and its etiology has recently been associated with osteogenic differentiation dysfunctions. OS cells keep a capacity for uncontrolled proliferation showing a phenotype similar to undifferentiated osteoprogenitors with abnormal biomineralization. Within this context, both conventional and X-ray synchrotron-based techniques have been exploited to deeply characterize the genesis and evolution of mineral depositions in a human OS cell line (SaOS-2) exposed to an osteogenic cocktail for 4 and 10 days. A partial restoration of the physiological biomineralization, culminating with the formation of hydroxyapatite, was observed at 10 days after treatment together with a mitochondria-driven mechanism for calcium transportation within the cell. Interestingly, during differentiation, mitochondria showed a change in morphology from elongated to rounded, indicating a metabolic reprogramming of OS cells possibly linked to an increase in glycolysis contribution to energy metabolism. These findings add a dowel to the genesis of OS giving new insights on the development of therapeutic strategies able to restore the physiological mineralization in OS cells.
Collapse
Affiliation(s)
- Francesca Rossi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Giovanna Picone
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Concettina Cappadone
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Andrea Sorrentino
- Mistral Beamline, ALBA Synchrotron Light Source, Cerdanyola del Valles, 08290 Barcelona, Spain
| | - Marta Columbaro
- Piattaforma di Microscopia Elettronica, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Giovanna Farruggia
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
- National Institute of Biostructures and Biosystems (NIBB), 00136 Rome, Italy
| | - Emilio Catelli
- Department of Chemistry "G. Ciamician", Università di Bologna, Ravenna Campus, Via Guaccimanni, 42, 48121 Ravenna, Italy
| | - Giorgia Sciutto
- Department of Chemistry "G. Ciamician", Università di Bologna, Ravenna Campus, Via Guaccimanni, 42, 48121 Ravenna, Italy
| | - Silvia Prati
- Department of Chemistry "G. Ciamician", Università di Bologna, Ravenna Campus, Via Guaccimanni, 42, 48121 Ravenna, Italy
| | - Robert Oliete
- Mistral Beamline, ALBA Synchrotron Light Source, Cerdanyola del Valles, 08290 Barcelona, Spain
| | - Alice Pasini
- Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi" (DEI), University of Bologna, Via dell'Università 50, 47522 Cesena, Italy
| | - Eva Pereiro
- Mistral Beamline, ALBA Synchrotron Light Source, Cerdanyola del Valles, 08290 Barcelona, Spain
| | - Stefano Iotti
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
- National Institute of Biostructures and Biosystems (NIBB), 00136 Rome, Italy
| | - Emil Malucelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
15
|
Luo P, Zhou P. The Relationship between PBLS and Osteosarcoma Distribution in Different Subgroups and the Survival and Prognosis of Osteosarcoma. JOURNAL OF ONCOLOGY 2023; 2023:3893134. [PMID: 37064862 PMCID: PMC10104740 DOI: 10.1155/2023/3893134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/08/2022] [Indexed: 04/18/2023]
Abstract
Objective To analyze the differences in the distribution of lymphocytes (PBLS) in different subgroups of osteosarcoma (OS) and the predictive value of related parameters on the survival prognosis of OS. Methods For retrospective analysis, 80 patients with malignant OS diagnosed and treated in our hospital from June 2016 to June 2017 were selected as the observation group, and 80 patients with benign bone tumors during the same period were selected as the control group. Patients in the observation group were followed up for three years and grouped according to the tumor diameter, stage, metastasis, and prognosis. Fasting venous blood was collected from each group and the levels of CD3+, CD3+CD4+, and CD3+CD8+ were detected. Meanwhile, the ratio of CD4+/CD8+, CD4+/CD3+, and CD8+/CD3+ was calculated and compared. Kaplan-Meier survival curve was used to analyze the relationship between PBLS parameters and OS survival. The area under the curve (AUC), sensitivity, and specificity of each entry index were analyzed by the receiver operating characteristic curve (ROC curve). Results The CD3+CD8+ level and CD4+/CD3+ ratio in the observation group were significantly higher than those in the control group (P < 0.05). The level of CD3+CD8+ in the patients with tumor diameter ≥ 11 cm was observably higher than that in the patients with tumor diameter <11 cm (P < 0.05). The levels of CD3+CD4+ and the ratio of CD4+/CD8 and CD4+/CD3+ of patients in stage III were markedly lower than those of patients in stage II, while the ratio of CD8+/CD3+ and the levels of CD3+CD8+ were prominently higher than those of patients in stage II (P < 0.05). The CD3+CD4+ level and CD4+/CD3+ ratio of patients in the metastatic group before treatment, the metastatic group after treatment, and the nonmetastatic group after treatment increased successively, while the ratio of CD4+/CD8+ and CD8+/CD3+ and the level of CD3+CD8+ decreased successively (P < 0.05). The CD3+CD4+ level and CD4+/CD3+ ratio in the poor prognosis group were significantly higher than those in the good prognosis group, whereas the ratio of CD8+/CD3+ and CD4+/CD8+ and the level of CD3+CD8+ were significantly lower than those in the poor prognosis group (P < 0.05). ROC curve analysis showed that the AUC of CD4+/CD8+ and CD4+/CD3+ in predicting poor prognosis in patients with OS was notably higher than other indicators, which were 0.818 and 0.866, respectively (P < 0.05). Kaplan-Meier survival curve results revealed that patients with CD3+CD4+ ≤ 5.15, CD3+CD8+ > 3.85, CD4+/CD8+ ≤ 1.42, CD4+/CD3+ ≤ 0.50, and CD8+/CD3+ > 0.38 had longer survival. Conclusion The distribution of PBLS parameters varied widely among different subgroups of OS. Patients with poor prognosis had a higher ratio of CD4+/CD8+ and CD4+/CD3+, which were related to the survival of patients with OS. Moreover, both the ratio of CD4+/CD8+ and CD4+/CD3+ had certain predictive values in terms of survival and prognosis of OS. Therefore, regular clinical monitoring of patients' immune function could help predict disease changes and assess prognosis.
Collapse
Affiliation(s)
- Peng Luo
- Department of Orthopedics, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan 430015, Hubei, China
| | - Peng Zhou
- Department of Orthopedics and Joints, The Third Affiliated Hospital of Soochow University, Changzhou 213000, Jiangsu, China
| |
Collapse
|
16
|
The Role of Tumor Microenvironment in Regulating the Plasticity of Osteosarcoma Cells. Int J Mol Sci 2022; 23:ijms232416155. [PMID: 36555795 PMCID: PMC9788144 DOI: 10.3390/ijms232416155] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/07/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Osteosarcoma (OS) is a malignancy that is becoming increasingly common in adolescents. OS stem cells (OSCs) form a dynamic subset of OS cells that are responsible for malignant progression and chemoradiotherapy resistance. The unique properties of OSCs, including self-renewal, multilineage differentiation and metastatic potential, 149 depend closely on their tumor microenvironment. In recent years, the likelihood of its dynamic plasticity has been extensively studied. Importantly, the tumor microenvironment appears to act as the main regulatory component of OS cell plasticity. For these reasons aforementioned, novel strategies for OS treatment focusing on modulating OS cell plasticity and the possibility of modulating the composition of the tumor microenvironment are currently being explored. In this paper, we review recent studies describing the phenomenon of OSCs and factors known to influence phenotypic plasticity. The microenvironment, which can regulate OSC plasticity, has great potential for clinical exploitation and provides different perspectives for drug and treatment design for OS.
Collapse
|
17
|
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: 16] [Impact Index Per Article: 5.3] [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
|
18
|
Dihydrotanshinone I Enhances Cell Adhesion and Inhibits Cell Migration in Osteosarcoma U-2 OS Cells through CD44 and Chemokine Signaling. Molecules 2022; 27:molecules27123714. [PMID: 35744840 PMCID: PMC9231138 DOI: 10.3390/molecules27123714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/06/2022] [Accepted: 06/06/2022] [Indexed: 12/10/2022] Open
Abstract
In the screening of novel natural products against cancer using an in vitro cancer cell model, we recently found that tanshinones from a traditional Chinese medicine, the rhizome of Salvia miltiorrhiza Bunge (Danshen), had potent effects on cell proliferation and migration. Especially for human osteosarcoma U−2 OS cells, tanshinones significantly enhanced the cell adherence, implying a possible role in cell adhesion and cell migration inhibition. In this work, therefore, we aimed to provide a new insight into the possible molecule mechanisms of dihydrotanshinone I, which had the strongest effects on cell adhesion among several candidate tanshinones. RNA−sequencing-based transcriptome analysis and several biochemical experiments indicated that there were comprehensive signals involved in dihydrotanshinone I-treated U−2 OS cells, such as cell cycle, DNA replication, thermogenesis, tight junction, oxidative phosphorylation, adherens junction, and focal adhesion. First, dihydrotanshinone I could potently inhibit cell proliferation and induce cell cycle arrest in the G0/G1 phase by downregulating the expression of CDK4, CDK2, cyclin D1, and cyclin E1 and upregulating the expression of p21. Second, it could significantly enhance cell adhesion on cell plates and inhibit cell migration, involving the hyaluronan CD44−mediated CXCL8–PI3K/AKT–FOXO1, IL6–STAT3–P53, and EMT signaling pathways. Thus, the increased expression of CD44 and lengthened protrusions around the cell yielded a significant increase in cell adhesion. In summary, these results suggest that dihydrotanshinone I might be an interesting molecular therapy for enhancing human osteosarcoma U−2 OS cell adhesion and inhibiting cell migration and proliferation.
Collapse
|
19
|
De Luca A, Bellavia D, Raimondi L, Carina V, Costa V, Fini M, Giavaresi G. Multiple Effects of Resveratrol on Osteosarcoma Cell Lines. Pharmaceuticals (Basel) 2022; 15:342. [PMID: 35337142 PMCID: PMC8956103 DOI: 10.3390/ph15030342] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 02/04/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary bone sarcoma affecting the life of pediatric patients. The clinical treatment faces numerous difficulties, including the adverse effects of chemotherapies, chemoresistance, and recurrences. In this study, the effects of resveratrol (RSV), a natural polyphenol, on OS cell lines were investigated to evaluate its action as an adjuvant therapy to the current chemotherapy regimens. RSV exhibited multiple tumor-suppressing activities on OS cell lines, inducing a series of critical events. We found (1) a cell growth inhibition due to an increase in cell distress, which was, in part, due to the involvement of the AKT and caspase-3 pathways, (2) an increase in cellular differentiation due to major gene expression levels of the osteoblastic differentiation genes, (3) an inhibition of IL-6 secretion due to an epigenetic effect on the IL-6 promoter, and (4) an inhibition of OS cells migration related to the decrease in IL-8 secretion levels due to an epigenetic effect on its promoter. Finally, the cotreatment of RSV with doxorubicin and cisplatin increased their cytotoxic effect on OS cells. Although further investigations are mandatory, it seems RSV might be a promising therapeutic adjuvant agent for OS cell treatment, exerting an antitumor effect when combined with chemotherapy.
Collapse
Affiliation(s)
- Angela De Luca
- IRCCS Istituto Ortopedico Rizzoli, CS Surgical Sciences and Technologies—SS Omics Science Platform for Personalized Orthopedics, 40136 Bologna, Italy; (D.B.); (L.R.); (V.C.); (V.C.); (M.F.); (G.G.)
| | | | | | | | | | | | | |
Collapse
|
20
|
Sun X, Zheng D, Guo W. Comprehensive Analysis of a Zinc Finger Protein Gene–Based Signature with Regard to Prognosis and Tumor Immune Microenvironment in Osteosarcoma. Front Genet 2022; 13:835014. [PMID: 35281811 PMCID: PMC8914066 DOI: 10.3389/fgene.2022.835014] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
Osteosarcoma is the most common malignant bone tumor that seriously threatens the lives of teenagers and children. Zinc finger (ZNF) protein genes encode the largest transcription factor family in the human genome. Aberrant expressions of ZNF protein genes widely occur in osteosarcoma, and these genes are therefore attractive biomarker candidates for prognosis prediction. In this study, we conducted a comprehensive analysis of ZNF protein genes in osteosarcoma and identified prognosis-related ZNF protein genes. Then, we constructed a prognostic signature based on seven prognosis-related ZNF protein genes and stratified patients into high- and low-risk groups. The seven genes included MKRN3, ZNF71, ZNF438, ZNF597, ATMIN, ZNF692, and ZNF525. After validation of the prognostic signature in internal and external cohorts, we constructed a nomogram including clinical features such as sex and age and the relative risk score based on the risk signature. Functional enrichment analysis of the risk-related differentially expressed genes revealed that the prognostic signature was closely associated with immune-related biological processes and signaling pathways. Moreover, we found significant differences between the high- and low-risk groups for the scores of diverse immune cell subpopulations, including CD8+ T cells, neutrophils, Th1 cells, and TILs. Regarding immune function, APC co-inhibition, HLA, inflammation promotion, para-inflammation, T-cell co-inhibition, and the type I IFN response were significantly different between the high- and low-risk groups. Of the seven ZNF protein genes, lower expressions of ATMIN, MKRN3, ZNF71, ZNF438, and ZNF597 were correlated with a high risk, while higher expressions of ZNF525 and ZNF692 were associated with a high risk. The Kaplan–Meier survival analysis suggested that lower expressions of ATMIN, ZNF438, and ZNF597 and the higher expression of ZNF692 were associated with worse overall survival in osteosarcoma. In conclusion, our ZNF protein gene–based signature was a novel and clinically useful prognostic biomarker for osteosarcoma patients.
Collapse
|
21
|
Reconstruction of Temporomandibular Joint and Skull Base Defect Following Osteosarcoma Resection. J Craniofac Surg 2022; 33:e667-e669. [DOI: 10.1097/scs.0000000000008564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/14/2022] [Indexed: 11/26/2022] Open
|
22
|
Liu G, Mao H, Liu Y, Zhang Z, Ha S, Zhang X. miR-148b-3p, as a tumor suppressor, targets son of sevenless homolog 1 to regulate the malignant progression in human osteosarcoma. Bioengineered 2022; 13:4271-4284. [PMID: 35152853 PMCID: PMC8974129 DOI: 10.1080/21655979.2022.2031389] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Affiliation(s)
- Guodong Liu
- Department of Orthopedics, Baogang Hospital (The Third Affiliated Hospital of Inner Mongolia Medical University), Baotou, Inner Mongolia, People’s Republic of China
| | - Honggang Mao
- Department of Orthopedics, Baogang Hospital (The Third Affiliated Hospital of Inner Mongolia Medical University), Baotou, Inner Mongolia, People’s Republic of China
| | - Yan Liu
- Department of Orthopedics, Baogang Hospital (The Third Affiliated Hospital of Inner Mongolia Medical University), Baotou, Inner Mongolia, People’s Republic of China
| | - Zun Zhang
- Department of Orthopedics, Baogang Hospital (The Third Affiliated Hospital of Inner Mongolia Medical University), Baotou, Inner Mongolia, People’s Republic of China
| | - Si Ha
- Department of Orthopedics, Baogang Hospital (The Third Affiliated Hospital of Inner Mongolia Medical University), Baotou, Inner Mongolia, People’s Republic of China
| | - Xiaoyan Zhang
- Department of Orthopedics, Baogang Hospital (The Third Affiliated Hospital of Inner Mongolia Medical University), Baotou, Inner Mongolia, People’s Republic of China
| |
Collapse
|
23
|
Han G, Zhang Y, Liu T, Li J, Li H. The anti-osteosarcoma effect from panax notoginseng saponins by inhibiting the G 0 / G 1 phase in the cell cycle and affecting p53-mediated autophagy and mitochondrial apoptosis. J Cancer 2021; 12:6383-6392. [PMID: 34659528 PMCID: PMC8489146 DOI: 10.7150/jca.54602] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/04/2021] [Indexed: 12/26/2022] Open
Abstract
Osteosarcoma is the most common primary bone malignancy, and current chemotherapy sessions against it often induce severe complications in patients. Thus, it is necessary to develop new and effective antineoplastic agents with fewer side effects. Panax notoginseng saponins (PNS) are the active components in panax notoginseng and were reported to be capable of inhibiting the growth of several tumors both in vitro and in vivo. However, its effects on osteosarcoma have not been studied yet, which is addressed in this study for the first time. Our results indicated that PNS can inhibit proliferation, invasion and migration of the osteosarcoma cells, while promoting their apoptosis simultaneously. Specifically, PNS caused an increase in mitochondrial membrane potential and the amount of reactive oxygen species. The cell cycle in osteosarcoma cells was arrested in the G0 / G1 phase after PNS treatment. The expression of p53 and other apoptosis-related mitochondrial proteins were promoted. Nevertheless, it was observed that autophagy became less active in osteosarcoma cells when PNS was administered. In a word, PNS were of potential therapeutic significance for osteosarcoma.
Collapse
Affiliation(s)
- Guangtao Han
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yubiao Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Ting Liu
- Department of Orthopedics, Hospital of Shenmu, Shenmu, Shaanxi, 719300, P.R. China
| | - Jianping Li
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Haohuan Li
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
24
|
Ji SF, Wen SL, Sun Y, Huang PW, Wu H, He ML. The biological function and clinical significance of STIL in osteosarcoma. Cancer Cell Int 2021; 21:218. [PMID: 33858425 PMCID: PMC8051131 DOI: 10.1186/s12935-021-01922-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 04/07/2021] [Indexed: 12/19/2022] Open
Abstract
Background SCL/TAL1 interrupting locus (STIL) is associated with the progression of several tumors; however, the biological role of STIL in osteosarcoma remains poorly understood. Methods In this study, the clinical significance of STIL in osteosarcoma was analyzed by gene chip data recorded in public databases. STIL expression was silenced in osteosarcoma cell lines to observe the effects on proliferation, apoptosis, invasion, and migration. Differentially expressed genes (DEGs) in the osteosarcoma chip were analyzed using The Limma package, and STIL co-expressed genes were obtained via the Pearson correlation coefficient. The potential molecular mechanism of STIL in osteosarcoma was further explored by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Results Osteosarcoma was associated with higher STIL expression compared to the control samples, and the standardized mean difference (SMD) was 1.52. STIL also had a good ability to distinguish osteosarcoma from non-osteosarcoma samples [area under the curve (AUC) = 0.96]. After silencing STIL, osteosarcoma cell proliferation decreased, apoptosis increased, and the migratory and invasion ability decreased. A total of 294 STIL differentially co-expressed genes were screened, and a bioinformatics analysis found that differentially co-expressed genes were primarily enriched in the cell signaling pathways. The protein-protein interaction (PPI) network indicated that the hub differentially co-expressed genes of STIL were CDK1, CCNB2, CDC20, CCNA2, BUB1, and AURKB. Conclusions STIL is associated with osteosarcoma proliferation and invasion, and may be promote the progression of osteosarcoma by regulating the expression of CDK1, CCNB2, CDC20, CCNA2, BUB1 and AURKB.
Collapse
Affiliation(s)
- Shu-Fan Ji
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.,Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.,Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Sheng-Lian Wen
- Department of Radiology, The First Affiliated Hospital, Guangxi Medical University, Shuangyong Road 6, Guangxi Zhuang Autonomous Region, 530021, Nanning, People's Republic of China
| | - Yu Sun
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Pi-Wei Huang
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Hao Wu
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Mao-Lin He
- Division of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road 6, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China. .,Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China. .,Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment, Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China.
| |
Collapse
|
25
|
Mechanisms of Resistance to Conventional Therapies for Osteosarcoma. Cancers (Basel) 2021; 13:cancers13040683. [PMID: 33567616 PMCID: PMC7915189 DOI: 10.3390/cancers13040683] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary bone tumor, mainly occurring in children and adolescents. Current standard therapy includes tumor resection associated with multidrug chemotherapy. However, patient survival has not evolved for the past decades. Since the 1970s, the 5-year survival rate is around 75% for patients with localized OS but dramatically drops to 20% for bad responders to chemotherapy or patients with metastases. Resistance is one of the biological processes at the origin of therapeutic failure. Therefore, it is necessary to better understand and decipher molecular mechanisms of resistance to conventional chemotherapy in order to develop new strategies and to adapt treatments for patients, thus improving the survival rate. This review will describe most of the molecular mechanisms involved in OS chemoresistance, such as a decrease in intracellular accumulation of drugs, inactivation of drugs, improved DNA repair, modulations of signaling pathways, resistance linked to autophagy, disruption in genes expression linked to the cell cycle, or even implication of the micro-environment. We will also give an overview of potential therapeutic strategies to circumvent resistance development.
Collapse
|
26
|
Effects of Inonotus obliquus Polysaccharides on Proliferation, Invasion, Migration, and Apoptosis of Osteosarcoma Cells. ACTA ACUST UNITED AC 2020; 2020:4282036. [PMID: 33282634 PMCID: PMC7685809 DOI: 10.1155/2020/4282036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 10/28/2020] [Accepted: 10/30/2020] [Indexed: 12/15/2022]
Abstract
Objectives To observe the effect of Inonotus obliquus polysaccharide (IOP) on the proliferation, invasion, migration, and apoptosis of osteosarcoma cells and to elucidate its underlying molecular mechanism. Methods IOP was extracted from Inonotus obliquus, human osteosarcoma MG-63 cells and U2OS cells were cultured in vitro, and the effects of IOP on the proliferation, migration, invasion, and apoptosis of MG-63 cells and U2OS cells were determined by CCK-8 assays, cell scratch assays, transwell assays, and flow cytometry, respectively. Western blot was used to detect the expression of related proteins in the Akt/mTOR and NF-κB signaling pathways. Results Compared with the control group, MG-63 cells and U2OS cells treated with IOP of 80 μg/ml, 160 μg/ml, and 320 μ g/ml in the experimental group had significantly lower proliferation activity, decreased migration and invasion ability, and increased apoptosis rate (P < 0.05). Furthermore, IOP could significantly inhibit the activation of the Akt/mTOR and NF-κB signaling pathway (P < 0.05). Conclusion IOP can regulate the proliferation, migration, invasion, and apoptosis of osteosarcoma cells by inhibiting the activation of the Akt/mTOR signaling pathway. It has antitumor activity on osteosarcoma and has the potential of clinical application in osteosarcoma treatment.
Collapse
|
27
|
Yang C, Zhang L, Huang H, Yuan X, Zhang P, Ye C, Wei M, Huang Y, Luo X, Luo J. Alantolactone inhibits proliferation, metastasis and promotes apoptosis of human osteosarcoma cells by suppressing Wnt/β-catenin and MAPKs signaling pathways. Genes Dis 2020; 9:466-478. [PMID: 35224161 PMCID: PMC8843874 DOI: 10.1016/j.gendis.2020.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/17/2020] [Accepted: 07/31/2020] [Indexed: 12/25/2022] Open
Abstract
Although there are many therapeutic strategies such as surgery and chemotherapy, the prognosis of osteosarcoma (OS) is still far from being satisfactory. It is urgent to develop more effective, tolerable and safe drugs for the treatment of OS. In the present study, we investigated the anti-OS activity of Alantolactone (ALT), a natural eucalyptone sesquiterpene lactone mainly exists in Inula helenium, and probed the possible mechanism involved. We demonstrated that ALT significantly inhibited cell proliferation of various human OS cell lines while had relative lower cytotoxicity against normal cells. Then, we validated that ALT reduced migration, decreased invasion possibly through reversing epithelial mesenchymal transition (EMT) process and suppressing Matrix metalloproteinases (MMPs). Moreover, we confirmed that ALT promoted apoptosis and arrested cell cycle at G2/M phase of human OS cells in vitro. In addition, we confirmed that ALT restrained tumor growth and metastasis of OS 143 cells in a xenograft model in vivo. Mechanistically, ALT inhibited the activity of Wnt/β-catenin and p38, ERK1/2 and JNK Mitogen Activated Protein Kinases (MAPKs) signal pathway. Notably, the combination of ALT and Wnt/β-catenin inhibitor, as well as the combination of ALT and MAPKs inhibitors resulted in a synergistically effect on inhibiting the proliferation, migration and invasion of OS cells. Collectively, our results validate the ALT may inhibit proliferation, metastasis and promotes apoptosis of human OS cells possibly through suppressing Wnt/β-Catenin and MAPKs signaling pathways.
Collapse
Affiliation(s)
- Chunmei Yang
- School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Lulu Zhang
- School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Huakun Huang
- School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Xiaohui Yuan
- School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Ping Zhang
- School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Caihong Ye
- School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Mengqi Wei
- School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Yanran Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, PR China
| | - Xiaoji Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, PR China
| | - Jinyong Luo
- School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, PR China
- Corresponding author. School of Laboratory Medicine, Chongqing Medical University, No. 1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, PR China.
| |
Collapse
|
28
|
Stearoyl-CoA Desaturase-1 Attenuates the High Shear Force Damage Effect on Human MG63 Osteosarcoma Cells. Int J Mol Sci 2020; 21:ijms21134720. [PMID: 32630668 PMCID: PMC7369751 DOI: 10.3390/ijms21134720] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 06/24/2020] [Accepted: 07/02/2020] [Indexed: 12/02/2022] Open
Abstract
Mechanical regulation is known as an important regulator in cancer progression and malignancy. High shear force has been found to inhibit the cell cycle progression and result in cell death in various cancer cells. Stearoyl-CoA desaturase (SCD)-1, one of the important lipogenic enzymes, has recently been indicated as a potential pharmaceutical target in cancer therapy. In this study, we determined whether the cell fate control of shear force stimulation is through regulating the SCD-1 expression in cancer cells. Human MG63 osteosarcoma cells were used in this study. 2 and 20 dynes/cm2 shear forces were defined as low and high intensities, respectively. A SCD-1 upregulation in human MG63 osteosarcoma cells under 20, but not 2, dynes/cm2 shear force stimulation was shown, and this induction was regulated by Smad1/5 and peroxisome proliferator-activated receptor δ (PPARδ) signaling. Moreover, gene knockdown of PPARδ and SCD-1 in human MG63 osteosarcoma cells attenuated the differentiation inhibition and resulted in much more cell death of high shear force initiation. The present study finds a possible auto-protective role of SCD-1 upregulation in high shear force-damaged human MG63 osteosarcoma cells. However, its detailed regulation in the cancer fate decision of high shear force should be further examined.
Collapse
|
29
|
Picone G, Cappadone C, Pasini A, Lovecchio J, Cortesi M, Farruggia G, Lombardo M, Gianoncelli A, Mancini L, Ralf H. M, Donato S, Giordano E, Malucelli E, Iotti S. Analysis of Intracellular Magnesium and Mineral Depositions during Osteogenic Commitment of 3D Cultured Saos2 Cells. Int J Mol Sci 2020; 21:ijms21072368. [PMID: 32235449 PMCID: PMC7177893 DOI: 10.3390/ijms21072368] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/20/2020] [Accepted: 03/26/2020] [Indexed: 12/23/2022] Open
Abstract
In this study, we explore the behaviour of intracellular magnesium during bone phenotype modulation in a 3D cell model built to mimic osteogenesis. In addition, we measured the amount of magnesium in the mineral depositions generated during osteogenic induction. A two-fold increase of intracellular magnesium content was found, both at three and seven days from the induction of differentiation. By X-ray microscopy, we characterized the morphology and chemical composition of the mineral depositions secreted by 3D cultured differentiated cells finding a marked co-localization of Mg with P at seven days of differentiation. This is the first experimental evidence on the presence of Mg in the mineral depositions generated during biomineralization, suggesting that Mg incorporation occurs during the bone forming process. In conclusion, this study on the one hand attests to an evident involvement of Mg in the process of cell differentiation, and, on the other hand, indicates that its multifaceted role needs further investigation.
Collapse
Affiliation(s)
- Giovanna Picone
- Department of Pharmacy and Biotechnology, University of Bologna, 33-40126 Bologna, Italy; (G.P.)
| | - Concettina Cappadone
- Department of Pharmacy and Biotechnology, University of Bologna, 33-40126 Bologna, Italy; (G.P.)
| | - Alice Pasini
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi” (DEI), University of Bologna 50, 47522 Cesena, Italy
| | - Joseph Lovecchio
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi” (DEI), University of Bologna 50, 47522 Cesena, Italy
| | - Marilisa Cortesi
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi” (DEI), University of Bologna 50, 47522 Cesena, Italy
| | - Giovanna Farruggia
- Department of Pharmacy and Biotechnology, University of Bologna, 33-40126 Bologna, Italy; (G.P.)
- National Institute of Biostructures and Biosystems (NIBB), 00136 Rome, Italy
| | - Marco Lombardo
- Department of Chemistry “G. Ciamician”, Alma Mater Studiorum–Università di Bologna, via Selmi 2, I-40126 Bologna, Italy
| | | | - Lucia Mancini
- Elettra-Sincrotrone Trieste S.C.p.A., Trieste, 34149 Basovizza, Italy
| | - Menk Ralf H.
- Elettra-Sincrotrone Trieste S.C.p.A., Trieste, 34149 Basovizza, Italy
- INFN section of Trieste, 2-34127 Trieste, Italy
- Department of Medical Imaging, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
| | - Sandro Donato
- Elettra-Sincrotrone Trieste S.C.p.A., Trieste, 34149 Basovizza, Italy
- Department of Physics, University of Calabria, 87036 Arcavacata di Rende (CS), Italy
| | - Emanuele Giordano
- Department of Electrical, Electronic and Information Engineering “Guglielmo Marconi” (DEI), University of Bologna 50, 47522 Cesena, Italy
| | - Emil Malucelli
- Department of Pharmacy and Biotechnology, University of Bologna, 33-40126 Bologna, Italy; (G.P.)
- Correspondence: ; Tel.: +39-051-209-5414
| | - Stefano Iotti
- Department of Pharmacy and Biotechnology, University of Bologna, 33-40126 Bologna, Italy; (G.P.)
- National Institute of Biostructures and Biosystems (NIBB), 00136 Rome, Italy
| |
Collapse
|