1
|
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
|
2
|
Liu S, Liu C, Wang Y, Chen J, He Y, Hu K, Li T, Yang J, Peng J, Hao L. The role of programmed cell death in osteosarcoma: From pathogenesis to therapy. Cancer Med 2024; 13:e7303. [PMID: 38800967 PMCID: PMC11129166 DOI: 10.1002/cam4.7303] [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: 01/11/2024] [Revised: 04/01/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
Osteosarcoma (OS) is a prevalent bone solid malignancy that primarily affects adolescents, particularly boys aged 14-19. This aggressive form of cancer often leads to deadly lung cancer due to its high migration ability. Experimental evidence suggests that programmed cell death (PCD) plays a crucial role in the development of osteosarcoma. Various forms of PCD, including apoptosis, ferroptosis, autophagy, necroptosis, and pyroptosis, contribute significantly to the progression of osteosarcoma. Additionally, different signaling pathways such as STAT3/c-Myc signal pathway, JNK signl pathway, PI3k/AKT/mTOR signal pathway, WNT/β-catenin signal pathway, and RhoA signal pathway can influence the development of osteosarcoma by regulating PCD in osteosarcoma cell. Therefore, targeting PCD and the associated signaling pathways could offer a promising therapeutic approach for treating osteosarcoma.
Collapse
Affiliation(s)
- Suqing Liu
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
- Queen Marry CollegeNanchang UniversityNanchangChina
| | - Chengtao Liu
- Shandong Wendeng Osteopathic HospitalWeihaiChina
| | - Yian Wang
- Queen Marry CollegeNanchang UniversityNanchangChina
| | - Jiewen Chen
- Queen Marry CollegeNanchang UniversityNanchangChina
| | - Yujin He
- Queen Marry CollegeNanchang UniversityNanchangChina
| | - Kaibo Hu
- The Second Clinical Medical College, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Ting Li
- The Second Clinical Medical College, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Junmei Yang
- The Second Clinical Medical College, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Jie Peng
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
- The Second Clinical Medical College, Jiangxi Medical CollegeNanchang UniversityNanchangChina
- Department of Sports Medicine, Huashan HospitalFudan UniversityShanghaiChina
| | - Liang Hao
- Department of Orthopedics, The Second Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| |
Collapse
|
3
|
Casciati A, Taddei AR, Rampazzo E, Persano L, Viola G, Cani A, Bresolin S, Cesi V, Antonelli F, Mancuso M, Merla C, Tanori M. Involvement of Mitochondria in the Selective Response to Microsecond Pulsed Electric Fields on Healthy and Cancer Stem Cells in the Brain. Int J Mol Sci 2024; 25:2233. [PMID: 38396911 PMCID: PMC10889160 DOI: 10.3390/ijms25042233] [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: 11/17/2023] [Revised: 02/02/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
In the last few years, pulsed electric fields have emerged as promising clinical tools for tumor treatments. This study highlights the distinct impact of a specific pulsed electric field protocol, PEF-5 (0.3 MV/m, 40 μs, 5 pulses), on astrocytes (NHA) and medulloblastoma (D283) and glioblastoma (U87 NS) cancer stem-like cells (CSCs). We pursued this goal by performing ultrastructural analyses corroborated by molecular/omics approaches to understand the vulnerability or resistance mechanisms triggered by PEF-5 exposure in the different cell types. Electron microscopic analyses showed that, independently of exposed cells, the main targets of PEF-5 were the cell membrane and the cytoskeleton, causing membrane filopodium-like protrusion disappearance on the cell surface, here observed for the first time, accompanied by rapid cell swelling. PEF-5 induced different modifications in cell mitochondria. A complete mitochondrial dysfunction was demonstrated in D283, while a mild or negligible perturbation was observed in mitochondria of U87 NS cells and NHAs, respectively, not sufficient to impair their cell functions. Altogether, these results suggest the possibility of using PEF-based technology as a novel strategy to target selectively mitochondria of brain CSCs, preserving healthy cells.
Collapse
Affiliation(s)
- Arianna Casciati
- Division of Health Protection Technologies, Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (V.C.); (F.A.); (M.M.)
| | - Anna Rita Taddei
- Great Equipment Center-Section of Electron Microscopy, University of Tuscia, Largo dell’Università snc, 01100 Viterbo, Italy;
| | - Elena Rampazzo
- Department of Women’s and Children’s Health (SDB), University of Padova, Via Giustiniani 3, 35128 Padova, Italy; (E.R.); (L.P.); (G.V.); (A.C.); (S.B.)
- Pediatric Research Institute (IRP), Corso Stati Uniti 4, 35127 Padova, Italy
| | - Luca Persano
- Department of Women’s and Children’s Health (SDB), University of Padova, Via Giustiniani 3, 35128 Padova, Italy; (E.R.); (L.P.); (G.V.); (A.C.); (S.B.)
- Pediatric Research Institute (IRP), Corso Stati Uniti 4, 35127 Padova, Italy
| | - Giampietro Viola
- Department of Women’s and Children’s Health (SDB), University of Padova, Via Giustiniani 3, 35128 Padova, Italy; (E.R.); (L.P.); (G.V.); (A.C.); (S.B.)
- Pediatric Research Institute (IRP), Corso Stati Uniti 4, 35127 Padova, Italy
| | - Alice Cani
- Department of Women’s and Children’s Health (SDB), University of Padova, Via Giustiniani 3, 35128 Padova, Italy; (E.R.); (L.P.); (G.V.); (A.C.); (S.B.)
- Pediatric Research Institute (IRP), Corso Stati Uniti 4, 35127 Padova, Italy
| | - Silvia Bresolin
- Department of Women’s and Children’s Health (SDB), University of Padova, Via Giustiniani 3, 35128 Padova, Italy; (E.R.); (L.P.); (G.V.); (A.C.); (S.B.)
- Pediatric Research Institute (IRP), Corso Stati Uniti 4, 35127 Padova, Italy
| | - Vincenzo Cesi
- Division of Health Protection Technologies, Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (V.C.); (F.A.); (M.M.)
| | - Francesca Antonelli
- Division of Health Protection Technologies, Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (V.C.); (F.A.); (M.M.)
| | - Mariateresa Mancuso
- Division of Health Protection Technologies, Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (V.C.); (F.A.); (M.M.)
| | - Caterina Merla
- Division of Health Protection Technologies, Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (V.C.); (F.A.); (M.M.)
| | - Mirella Tanori
- Division of Health Protection Technologies, Italian National Agency for Energy New Technologies and Sustainable Economic Development (ENEA), Via Anguillarese 301, 00123 Rome, Italy; (A.C.); (V.C.); (F.A.); (M.M.)
| |
Collapse
|
4
|
Yang J, Jiang J. Gasdermins: a dual role in pyroptosis and tumor immunity. Front Immunol 2024; 15:1322468. [PMID: 38304430 PMCID: PMC10830654 DOI: 10.3389/fimmu.2024.1322468] [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] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/04/2024] [Indexed: 02/03/2024] Open
Abstract
The gasdermin (GSDM) protein family plays a pivotal role in pyroptosis, a process critical to the body's immune response, particularly in combatting bacterial infections, impeding tumor invasion, and contributing to the pathogenesis of various inflammatory diseases. These proteins are adept at activating inflammasome signaling pathways, recruiting immune effector cells, creating an inflammatory immune microenvironment, and initiating pyroptosis. This article serves as an introduction to the GSDM protein-mediated pyroptosis signaling pathways, providing an overview of GSDMs' involvement in tumor immunity. Additionally, we explore the potential applications of GSDMs in both innovative and established antitumor strategies.
Collapse
Affiliation(s)
- Jiayi Yang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| |
Collapse
|
5
|
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
|
6
|
Yang J, Jiang H, Fu Q, Qin H, Li Y, Liu M. Blue light photobiomodulation induced apoptosis by increasing ROS level and regulating SOCS3 and PTEN/PI3K/AKT pathway in osteosarcoma cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2023; 249:112814. [PMID: 37956614 DOI: 10.1016/j.jphotobiol.2023.112814] [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: 09/19/2023] [Revised: 10/23/2023] [Accepted: 11/06/2023] [Indexed: 11/15/2023]
Abstract
Blue light photobiomodulation (PBM) has attracted great attention in diminishing proliferation and inducing death of cancer cells recently. Osteosarcoma (OS) primarily occurring in children and adolescents, the limitations of drug resistance and limb salvage make it urgent to develop and identify new adjuvant therapeutic strategies. In this work, we attempted to research the anticancer effects and biological mechanisms of blue light PBM in human OS MG63 cells. The effects of various blue light parameters on MG63 cells indicated that suppressed cell proliferation and cell migration, induced cell apoptosis which are experimentally assessed using multiple assays including CCK, LDH, wound healing assay and Hoechst staining. Concurrently, the increases of ROS level and the inhibition of PI3K and AKT expression were identified under high-dose blue light PBM in MG63 cells. Meanwhile, SOCS3 is a major inducible anti-tumor molecule, we also found that blue light LED substantially promoted its expression. Thus, this study proposed that bule light PBM may be a hopeful therapeutic approach in OS clinical treatment in the future.
Collapse
Affiliation(s)
- Jiali Yang
- School of information science and technology, Fudan University, 2005th Songhu Road, Shanghai 200438, China
| | - Hui Jiang
- Academy for Engineering and Technology, Fudan University, 220th Handan Road, Shanghai 200433, China
| | - Qiqi Fu
- School of information science and technology, Fudan University, 2005th Songhu Road, Shanghai 200438, China
| | - Haokuan Qin
- Academy for Engineering and Technology, Fudan University, 220th Handan Road, Shanghai 200433, China
| | - Yinghua Li
- Shanghai Fifth People's Hospital, Fudan University, 801th Heqing Road, Shanghai 200240, China
| | - Muqing Liu
- School of information science and technology, Fudan University, 2005th Songhu Road, Shanghai 200438, China; Zhongshan Fudan Joint Innovation Center, 6th Xiangxing Road, Zhongshan 28403, China.
| |
Collapse
|
7
|
Zhao X, Zhang J, Liu J, Chen Q, Cai C, Miao X, Wu T, Cheng X. Identification of mitochondrial-related signature and molecular subtype for the prognosis of osteosarcoma. Aging (Albany NY) 2023; 15:12794-12816. [PMID: 37976137 DOI: 10.18632/aging.205143] [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: 06/01/2023] [Accepted: 09/26/2023] [Indexed: 11/19/2023]
Abstract
Mitochondria play a vital role in osteosarcoma. Therefore, the purpose of this study was to investigate the potential role of mitochondrial-related genes (MRGs) in osteosarcoma. Based on 92 differentially expressed MRGs, osteosarcoma samples were divided into two subtypes using the nonnegative matrix factorization (NMF). Ultimately, a univariate, least absolute shrinkage and selection operator (LASSO), and multivariate Cox analysis were performed to construct a prognostic risk model. The single-sample gene set enrichment analysis assessed the immune infiltration characteristics of osteosarcoma patients. Finally, we identified an osteosarcoma biomarker, malonyl-CoA decarboxylase (MLYCD), which showed downregulation. Osteosarcoma cells proliferation, migration, and invasion were effectively inhibited by the overexpression of MLYCD. Our findings will help us to further understand the molecular mechanisms of osteosarcoma and contribute to the discovery of new diagnostic biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Xiaokun Zhao
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Jian Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Jiahao Liu
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Qi Chen
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Changxiong Cai
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Xinxin Miao
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Tianlong Wu
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Jiangxi Key Laboratory of Intervertebral Disc Disease, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Xigao Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Jiangxi Key Laboratory of Intervertebral Disc Disease, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Institute of Minimally Invasive Orthopedics, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| |
Collapse
|
8
|
Zhang L, Wu S, Huang J, Shi Y, Yin Y, Cao X. A mitochondria-related signature for predicting immune microenvironment and therapeutic response in osteosarcoma. Front Oncol 2022; 12:1085065. [PMID: 36531021 PMCID: PMC9751795 DOI: 10.3389/fonc.2022.1085065] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Osteosarcoma remains to be the most devastating malignant tumor in children and teenagers. Mitochondria have also been proven to play critical roles in osteosarcoma. However, a mitochondria-related signature has been established in osteosarcoma to comprehensively evaluate the pathogenic roles and regulatory roles of mitochondria in osteosarcoma. METHODS In this study, osteosarcoma samples' transcriptome data and clinical information were collected from Therapeutically Applicable Research to Generate Effective Treatments (TARGET) and Gene Expression Omnibus (GEO) databases. A comprehensive bioinformatics analysis was performed on the samples at the bulk RNA sequencing level and single-cell RNA sequencing (scRNA-seq) level. EdU, Transwell, and immunohistochemistry (IHC) were performed on PCCB. RESULTS A mitochondria-related signature was constructed in osteosarcoma patients. The prognostic value of the mitochondria-related signature was explored. The predictive value of the mitochondria-related signature in the immune microenvironment and chemotherapy agents was explored. The association between mitochondria and immunity in the tumor microenvironment of osteosarcoma at the scRNA-seq level was investigated. The tumorigenic role of the critical mitochondria-related gene, PCCB, was verified by in vitro validation. CONCLUSION In conclusion, a mitochondria-related signature was developed in osteosarcoma with solid predictive values in the immune microenvironment, chemotherapy agents, and prognosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Xu Cao
- Department of Orthopaedics, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|