1
|
Wang B, Yu RZ, Zhang XY, Ren Y, Zhen YW, Han L. Polo-like kinase 4 accelerates glioma malignant progression and vasculogenic mimicry by phosphorylating EphA2. Cancer Lett 2024:217397. [PMID: 39694224 DOI: 10.1016/j.canlet.2024.217397] [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: 08/21/2024] [Revised: 12/10/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Vasculogenic mimicry (VM), which involved the formation of vascular-like structures by highly invasive tumor cells, had been identified as one of the mechanisms contributing to resistance against anti-angiogenic therapy in patients with glioblastoma (GBM). Therefore, inhibition of VM formation may serve as an effective therapeutic strategy against angiogenesis resistance. Polo-like kinase 4 (PLK4), a protein kinase, had been linked to the progression of glioblastoma and was associated with an unfavorable prognosis. The integration of proteomics and phosphoproteomics revealed that PLK4 directly activated the PI3K-Akt and MAPK signaling cascades by phosphorylating the Ser901 and Ser897 of EphA2. In addition, EphA2 Ser901 phosphorylating catalyzed by PLK4 significantly enhanced the phosphorylation of its own Ser897 site, which is a hallmark of EphA2 activation. The PI3K-Akt signaling was intricately associated with the progression of VM. Thus, PLK4 influenced malignant progression and VM formation via stimulation of the EphA2 signal transduction. Moreover, the expression level of PLK4 protein positively correlated with the level of EphA2 phosphorylation in glioma tissues. These results highlighted the crucial significance of PLK4 phosphorylating EphA2 in the malignant progression and VM formation in GBM.
Collapse
Affiliation(s)
- Bo Wang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Run-Ze Yu
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xiao-Yang Zhang
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yu Ren
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| | - Ying-Wei Zhen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China.
| | - Lei Han
- Tianjin Neurological Institute, Key Laboratory of Post-Neuro injury, Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
2
|
Liu X, Shi Z, Liu X, Cao Y, Yang X, Liu J, Xu T, Yang W, Chen L, Zou Z, Jia Q, Li M. The role of PDCD6 in stemness maintenance of Glioblastoma. Pathol Res Pract 2024; 264:155727. [PMID: 39561536 DOI: 10.1016/j.prp.2024.155727] [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: 11/11/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Glioblastoma (GBM) poses formidable challenges due to its high malignancy and therapeutic resistance and still exhibits dismal 5-year survival rates, high recurrence propensity, and limited treatment modalities. There is an acute need for innovative treatments for recurrent glioblastoma due to the lack of established protocols. This necessity is driving research into the cellular underpinnings that initiate and drive the disease forward, aiming to discover groundbreaking targets for therapy that could enhance the efficacy of medical interventions. METHODS Patient-derived glioblastoma stem cells (GSCs) were harvested and isolated. Subsequently, PDCD6 expression was quantified through both western blotting (WB) and real-time PCR (RT-PCR) techniques. The stem-like properties of the GSCs were evaluated using sphere-forming assays. All gathered data, inclusive of TCGA datasets, were analyzed using SPSS (IBM) version 23.0. RESULTS Elevated PDCD6 expression characterized classical GBM tumor tissues. PDCD6 overexpression significantly correlated with diminished overall survival in GBM patients, emerging as an independent prognostic indicator. Notably, primary GBM cells exhibited heightened PDCD6 levels in GSCs compared to NSTCs. Moreover, alterations in stemness markers paralleled PDCD6 modulation, where PDCD6 knockdown attenuated tumor size in GSCs. CONCLUSION Our findings illuminate PDCD6's role in fostering stemness within classical GBM, hinting at its potential as a therapeutic target.
Collapse
Affiliation(s)
- Xiyu Liu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Zuolin Shi
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Xuantong Liu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Yuan Cao
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Xinyu Yang
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Jiaming Liu
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Tianqi Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Weiyi Yang
- Department of Neurology, Xi'an Daxing Hospital, Xi'an, China
| | - Ligang Chen
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Zheng Zou
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China.
| | - Qingge Jia
- Department of Reproductive Medicine, Xi'an International Medical Center Hospital, Northwest University, Xi'an, China.
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
3
|
Dong L, Zhu Y, Zhang H, Gao L, Zhang Z, Xu X, Ying L, Zhang L, Li Y, Yun Z, Zhu D, Han C, Xu T, Yang H, Ju S, Chen X, Zhang H, Xie J. Open-Source Throttling of CD8 + T Cells in Brain with Low-Intensity Focused Ultrasound-Guided Sequential Delivery of CXCL10, IL-2, and aPD-L1 for Glioblastoma Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407235. [PMID: 39264011 DOI: 10.1002/adma.202407235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/14/2024] [Indexed: 09/13/2024]
Abstract
Improving clinical immunotherapy for glioblastoma (GBM) relies on addressing the immunosuppressive tumor microenvironment (TME). Enhancing CD8+ T cell infiltration and preventing its exhaustion holds promise for effective GBM immunotherapy. Here, a low-intensity focused ultrasound (LIFU)-guided sequential delivery strategy is developed to enhance CD8+ T cells infiltration and activity in the GBM region. The sequential delivery of CXC chemokine ligand 10 (CXCL10) to recruit CD8+ T cells and interleukin-2 (IL-2) to reduce their exhaustion is termed an "open-source throttling" strategy. Consequently, up to 3.39-fold of CD8+ T cells are observed with LIFU-guided sequential delivery of CXCL10, IL-2, and anti-programmed cell death 1 ligand 1 (aPD-L1), compared to the free aPD-L1 group. The immune checkpoint inhibitors (ICIs) therapeutic efficacy is substantially enhanced by the reversed immunosuppressive TME due to the expansion of CD8+ T cells, resulting in the elimination of tumor, prolonged survival time, and long-term immune memory establishment in orthotopic GBM mice. Overall, LIFU-guided sequential cytokine and ICIs delivery offers an "open-source throttling" strategy of CD8+ T cells, which may present a promising strategy for brain-tumor immunotherapy.
Collapse
Affiliation(s)
- Lei Dong
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology; Department of Oncology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Yini Zhu
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu, 210009, China
| | - Haoge Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Lin Gao
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Zhiqi Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Xiaoxuan Xu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Leqian Ying
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology; Department of Oncology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Lu Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology; Department of Oncology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Yue Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, 999078, China
| | - Zhengcheng Yun
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology; Department of Oncology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Danqi Zhu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Chang Han
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Tingting Xu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology; Department of Oncology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Hui Yang
- Department of Biochemistry and Molecular Biology, Medical School of Southeast University, Nanjing, China
| | - Shenghong Ju
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Xiaoyuan Chen
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | - Haijun Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology; Department of Oncology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao, Nanjing, 210009, China
| | - Jinbing Xie
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Basic Medicine Research and Innovation Center of Ministry of Education, State Key Laboratory of Digital Medical Engineering, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| |
Collapse
|
4
|
Zhu H, Allwin C, Bassous MG, Pouliopoulos AN. Focused ultrasound-mediated enhancement of blood-brain barrier permeability for brain tumor treatment: a systematic review of clinical trials. J Neurooncol 2024; 170:235-252. [PMID: 39207625 PMCID: PMC11538134 DOI: 10.1007/s11060-024-04795-z] [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: 07/01/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE Brain tumors, particularly glioblastoma multiforme (GBM), present significant prognostic challenges despite multimodal therapies, including surgical resection, chemotherapy, and radiotherapy. One major obstacle is the limited drug delivery across the blood-brain barrier (BBB). Focused ultrasound (FUS) combined with systemically administered microbubbles has emerged as a non-invasive, targeted, and reversible approach to transiently open the BBB, thus enhancing drug delivery. This review examines clinical trials employing BBB opening techniques to optimise pharmacotherapy for brain tumors, evaluates current challenges, and proposes directions for further research. METHODS A systematic literature search was conducted in PubMed and ClinicalTrials.gov up to November 2023, searching for "ultrasound" AND "brain tumor". The search yielded 1446 results. After screening by title and abstract, followed by full-text screening (n = 48), 35 studies were included in the analysis. RESULTS Our analysis includes data from 11 published studies and 24 ongoing trials. The predominant focus of these studies is on glioma, including GMB and astrocytoma. One paper investigated brain metastasis from breast cancer. Evidence indicates that FUS facilitates BBB opening and enhances drug uptake following sonication. Exploration of FUS in the pediatric population is limited, with no published studies and only three ongoing trials dedicated to this demographic. CONCLUSION FUS is a promising strategy to safely disrupt the BBB, enabling precise and non-invasive lesion targeting, and enhance drug delivery. However, pharmacokinetic studies are required to quantitatively assess improvements in drug uptake. Most studies are phase I clinical trials, and long-term follow-up investigating patient outcomes is essential to evaluate the clinical benefit of this treatment approach. Further studies involving diverse populations and pathologies will be beneficial.
Collapse
Affiliation(s)
- Honglin Zhu
- Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Caitlin Allwin
- Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Monica G Bassous
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | | |
Collapse
|
5
|
Meng X, Wang Z, Yang Q, Liu Y, Gao Y, Chen H, Li A, Li R, Wang J, Sun G. Intracellular C5aR1 inhibits ferroptosis in glioblastoma through METTL3-dependent m6A methylation of GPX4. Cell Death Dis 2024; 15:729. [PMID: 39368999 PMCID: PMC11455874 DOI: 10.1038/s41419-024-06963-5] [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: 01/15/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 10/07/2024]
Abstract
Glioblastoma (GBM) is the most common primary intracranial malignant tumor. Recent literature suggests that induction of programmed death has become a mainstream cancer treatment strategy, with ferroptosis being the most widely studied mode. Complement C5a receptor 1 (C5aR1) is associated with both tumorigenesis and tumor-related immunity. However, knowledge regarding the role of C5aR1 in GBM progression is limited. In the present study, we observed significant upregulation of C5aR1 in glioma tissue. In addition, C5aR1 expression was found to be closely associated with patient prognosis and survival. Subsequent experimental verification demonstrated that C5aR1 promoted the progression of GBM mainly by suppressing ferroptosis induction, inhibiting the accumulation of lipid peroxides, and stabilizing the expression of the core antiferroptotic factor glutathione peroxidase 4 (GPX4). Aberrant N6-methyladenosine (m6A) modification of GPX4 mRNA contributes significantly to epigenetic tumorigenesis, and here, we report that selective methyltransferase-like 3 (METTL3)-dependent m6A methylation of GPX4 plays a key role in C5AR1 knockdown-induced ferroptosis induction. Mechanistically, ERK1/2 signaling pathway activation increases the METTL3 protein abundance in GBM cells. This activation then increases the stability of METTL3-mediated m6A modifications on GPX4, enabling it to fulfill its transcriptional function. More importantly, in an intracranial xenograft mouse model, PMX205, a C5aR1 inhibitor, promoted alterations in ferroptosis in GBM cells and inhibited GBM progression. In conclusion, our findings suggest that C5aR1 inhibits ferroptosis in GBM cells and promotes MettL3-dependent GPX4 expression through ERK1/2, thereby promoting glioma progression. Our study reveals a novel mechanism by which the intracellular complement receptor C5aR1 suppresses ferroptosis induction and promotes GBM progression. These findings may facilitate the identification of a potential therapeutic target for glioma.
Collapse
Affiliation(s)
- Xiangrui Meng
- Department of Neurosurgery, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng, China
- Yancheng Medical Research Center of Nanjing University Medical School, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People's Hospital of Yancheng, Yancheng, China
| | - Zixuan Wang
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Nanjing, China
- Postgraduate College, Xuzhou Medical University, Xuzhou, China
| | - Qingqing Yang
- Yancheng Medical Research Center of Nanjing University Medical School, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People's Hospital of Yancheng, Yancheng, China
| | - Yawei Liu
- Department of Neurosurgery, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng, China
- Yancheng Medical Research Center of Nanjing University Medical School, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People's Hospital of Yancheng, Yancheng, China
| | - Yisu Gao
- Department of Neurosurgery, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng, China
| | - Hefei Chen
- Yancheng Medical Research Center of Nanjing University Medical School, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People's Hospital of Yancheng, Yancheng, China
| | - Ang Li
- Yancheng Medical Research Center of Nanjing University Medical School, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People's Hospital of Yancheng, Yancheng, China
| | - Rongqing Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Jun Wang
- Department of Neurosurgery, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng, China.
| | - Guan Sun
- Department of Neurosurgery, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng, China.
- Yancheng Medical Research Center of Nanjing University Medical School, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, The First People's Hospital of Yancheng, Yancheng, China.
| |
Collapse
|
6
|
Liu L, Li F, Zhang L, Cheng Y, Wu L, Tie R, Jiang X, Gao W, Liu B, Wei Y, Chang P, Xu J, Zhao H, Zhang L. Cysteine and glycine-rich protein 2 is crucial for maintaining the malignant phenotypes of gliomas through its action on Notch signalling cascade. Toxicol Appl Pharmacol 2024; 487:116969. [PMID: 38744347 DOI: 10.1016/j.taap.2024.116969] [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: 03/10/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024]
Abstract
Cysteine and glycine-rich protein 2 (CSRP2) is expressed differently in numerous cancers and plays a key role in carcinogenesis. However, the role of CSRP2 in glioma is unknown. This study sought to determine the expression profile and clinical significance of CSRP2 in glioma and explore its biological functions and mechanisms via lentivirus-mediated CSRP2 silencing experiments. Increased CSRP2 was frequently observed in gliomas, which was associated with clinicopathological characteristics and an unfavourable prognosis. Decreasing CSRP2 led to the suppression of malignant proliferation, metastasis and stemness in glioma cells while causing hypersensitivity to chemotherapeutic drugs. Mechanistic investigations revealed that CSRP2 plays a role in mediating the Notch signalling cascade. Silencing CSRP2 decreased the levels of Notch1, cleaved Notch1, HES1 and HEY1, suppressing the Notch signalling cascade. Reactivation of Notch markedly diminished the tumour-inhibiting effects of CSRP2 silencing on the malignant phenotypes of glioma cells. Notably, CSRP2-silencing glioma cells exhibited reduced potential in the formation of xenografts in nude mice in vivo, which was associated with an impaired Notch signalling cascade. These results showed that CSRP2 is overexpressed in glioma and has a crucial role in sustaining the malignant phenotypes of glioma, suggesting that targeting CSRP2 could be a promising strategy for glioma treatment.
Collapse
Affiliation(s)
- Lingtong Liu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, No.32 West Second Section First Ring Road, Chengdu 610072, China
| | - Fei Li
- Department of Neurosurgery, Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an 710038, China
| | - Lingxue Zhang
- Department of Neurosurgery, Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an 710038, China
| | - Yingying Cheng
- Department of Neurosurgery, Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an 710038, China
| | - Lin Wu
- Central Laboratory, Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an 710038, China
| | - Ru Tie
- Central Laboratory, Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an 710038, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an 710038, China
| | - Wenwen Gao
- Department of Neurosurgery, Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an 710038, China
| | - Bochuan Liu
- Department of Neurosurgery, Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an 710038, China
| | - Yao Wei
- Department of Neurosurgery, Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an 710038, China
| | - Pan Chang
- Central Laboratory, Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an 710038, China
| | - Jun Xu
- Department of Neurosurgery, Xi'an Daxing Hospital, No. 353 Laodong North Road, Lianhu District, Xi'an 710016, China
| | - Haikang Zhao
- Department of Neurosurgery, Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an 710038, China.
| | - Liang Zhang
- Department of Neurosurgery, Second Affiliated Hospital of Xi'an Medical University, No. 167 Fangdong Street, Xi'an 710038, China; Department of Neurosurgery, Xi'an Daxing Hospital, No. 353 Laodong North Road, Lianhu District, Xi'an 710016, China; Northwest University, No. 1 Xuefu Street, Guodu Education and Technology Industrial Zone, Chang'an District, Xi'an 710127, China.
| |
Collapse
|
7
|
Zhou Z, Leng H. Deciphering the causal relationship between plasma and cerebrospinal fluid metabolites and glioblastoma multiforme: a Mendelian Randomization study. Aging (Albany NY) 2024; 16:8306-8319. [PMID: 38742944 PMCID: PMC11131984 DOI: 10.18632/aging.205818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 04/10/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND Glioblastoma Multiforme (GBM) is one of the most aggressive and fatal brain cancers. The study of metabolites could be crucial for understanding GBM's biology and reveal new treatment strategies. METHODS The GWAS data for GBM were sourced from the FinnGen database. A total of 1400 plasma metabolites were collected from the GWAS Catalog dataset. The cerebrospinal fluid (CSF) metabolites data were collected from subsets of participants in the WADRC and WRAP studies. We utilized the inverse variance weighting (IVW) method as the primary tool to explore the causal relationship between metabolites in plasma and CSF and glioblastoma, ensuring the exclusion of instances with horizontal pleiotropy. Additionally, four supplementary analytical methods were applied to reinforce our findings. Aberrant results were identified and omitted based on the outcomes of the leave-one-out sensitivity analysis. Conclusively, a reverse Mendelian Randomization analysis was also conducted to further substantiate our results. RESULTS The study identified 69 plasma metabolites associated with GBM. Of these, 40 metabolites demonstrated a significant positive causal relationship with GBM, while 29 exhibited a significant negative causal association. Notably, Trimethylamine N-oxide (TMAO) levels in plasma, not CSF, were found to be a significant exposure factor for GBM (OR = 3.1627, 95% CI = (1.6347, 6.1189), P = 0.0006). The study did not find a reverse causal relationship between GBM and plasma TMAO levels. CONCLUSIONS This research has identified 69 plasma metabolites potentially associated with the incidence of GBM, among which TMAO stands out as a promising candidate for an early detectable biomarker for GBM.
Collapse
Affiliation(s)
- Zhiwei Zhou
- Department of Neurosurgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan 415003, People’s Republic of China
| | - Haibin Leng
- Department of Neurosurgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People’s Hospital of Changde City), Changde, Hunan 415003, People’s Republic of China
| |
Collapse
|
8
|
Dong N, Qi W, Wu L, Li J, Zhang X, Wu H, Zhang W, Jiang J, Zhang S, Fu W, Liu Q, Qi G, Wang L, Lu Y, Luo J, Kong Y, Liu Y, Zhao RC, Wang J. LINC00606 promotes glioblastoma progression through sponge miR-486-3p and interaction with ATP11B. J Exp Clin Cancer Res 2024; 43:139. [PMID: 38725030 PMCID: PMC11080186 DOI: 10.1186/s13046-024-03058-z] [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: 02/05/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND LncRNAs regulate tumorigenesis and development in a variety of cancers. We substantiate for the first time that LINC00606 is considerably expressed in glioblastoma (GBM) patient specimens and is linked with adverse prognosis. This suggests that LINC00606 may have the potential to regulate glioma genesis and progression, and that the biological functions and molecular mechanisms of LINC00606 in GBM remain largely unknown. METHODS The expression of LINC00606 and ATP11B in glioma and normal brain tissues was evaluated by qPCR, and the biological functions of the LINC00606/miR-486-3p/TCF12/ATP11B axis in GBM were verified through a series of in vitro and in vivo experiments. The molecular mechanism of LINC00606 was elucidated by immunoblotting, FISH, RNA pulldown, CHIP-qPCR, and a dual-luciferase reporter assay. RESULTS We demonstrated that LINC00606 promotes glioma cell proliferation, clonal expansion and migration, while reducing apoptosis levels. Mechanistically, on the one hand, LINC00606 can sponge miR-486-3p; the target gene TCF12 of miR-486-3p affects the transcriptional initiation of LINC00606, PTEN and KLLN. On the other hand, it can also regulate the PI3K/AKT signaling pathway to mediate glioma cell proliferation, migration and apoptosis by binding to ATP11B protein. CONCLUSIONS Overall, the LINC00606/miR-486-3p/TCF12/ATP11B axis is involved in the regulation of GBM progression and plays a role in tumor regulation at transcriptional and post-transcriptional levels primarily through LINC00606 sponging miR-486-3p and targeted binding to ATP11B. Therefore, our research on the regulatory network LINC00606 could be a novel therapeutic strategy for the treatment of GBM.
Collapse
Affiliation(s)
- Naijun Dong
- School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
- School of Medicine, Shanghai University, Shanghai, China
| | - Wenxin Qi
- School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
- School of Medicine, Shanghai University, Shanghai, China
| | - Lingling Wu
- School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
- School of Medicine, Shanghai University, Shanghai, China
| | - Jie Li
- Shanghai Institute of Phage, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xueqi Zhang
- School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Hao Wu
- School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Wen Zhang
- School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Jiawen Jiang
- School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Shibo Zhang
- School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Wenjun Fu
- School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Qian Liu
- School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Guandong Qi
- Residential College, Shanghai University, Shanghai, China
| | - Lukai Wang
- Residential College, Shanghai University, Shanghai, China
| | - Yanyuan Lu
- Residential College, Shanghai University, Shanghai, China
| | - Jingyi Luo
- Residential College, Shanghai University, Shanghai, China
| | - Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihao Liu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
| | - Robert Chunhua Zhao
- School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking, Union Medical College, Beijing, China.
- Centre of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China.
| | - Jiao Wang
- School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| |
Collapse
|
9
|
Liu J, Yang F, Hu J, Zhang X. Nanoparticles for efficient drug delivery and drug resistance in glioma: New perspectives. CNS Neurosci Ther 2024; 30:e14715. [PMID: 38708806 PMCID: PMC11071172 DOI: 10.1111/cns.14715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 05/07/2024] Open
Abstract
Gliomas are the most common primary tumors of the central nervous system, with glioblastoma multiforme (GBM) having the highest incidence, and their therapeutic efficacy depends primarily on the extent of surgical resection and the efficacy of postoperative chemotherapy. The role of the intracranial blood-brain barrier and the occurrence of the drug-resistant gene O6-methylguanine-DNA methyltransferase have greatly limited the efficacy of chemotherapeutic agents in patients with GBM and made it difficult to achieve the expected clinical response. In recent years, the rapid development of nanotechnology has brought new hope for the treatment of tumors. Nanoparticles (NPs) have shown great potential in tumor therapy due to their unique properties such as light, heat, electromagnetic effects, and passive targeting. Furthermore, NPs can effectively load chemotherapeutic drugs, significantly reduce the side effects of chemotherapeutic drugs, and improve chemotherapeutic efficacy, showing great potential in the chemotherapy of glioma. In this article, we reviewed the mechanisms of glioma drug resistance, the physicochemical properties of NPs, and recent advances in NPs in glioma chemotherapy resistance. We aimed to provide new perspectives on the clinical treatment of glioma.
Collapse
Affiliation(s)
- Jiyuan Liu
- Department of Neurosurgerythe First Hospital of China Medical UniversityShenyangChina
| | - Fan Yang
- Department of Cardiologythe Fourth Affiliated Hospital of China Medical UniversityShenyangChina
| | - Jinqu Hu
- Department of Neurosurgerythe First Hospital of China Medical UniversityShenyangChina
| | - Xiuchun Zhang
- Department of Neurologythe First Hospital of China Medical UniversityShenyangChina
| |
Collapse
|
10
|
Wang J, Zhu H, Gan J, Liang G, Li L, Zhao Y. Engineered mRNA Delivery Systems for Biomedical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308029. [PMID: 37805865 DOI: 10.1002/adma.202308029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/05/2023] [Indexed: 10/09/2023]
Abstract
Messenger RNA (mRNA)-based therapeutic strategies have shown remarkable promise in preventing and treating a staggering range of diseases. Optimizing the structure and delivery system of engineered mRNA has greatly improved its stability, immunogenicity, and protein expression levels, which has led to a wider range of uses for mRNA therapeutics. Herein, a thorough analysis of the optimization strategies used in the structure of mRNA is first provided and delivery systems are described in great detail. Furthermore, the latest advancements in biomedical engineering for mRNA technology, including its applications in combatting infectious diseases, treating cancer, providing protein replacement therapy, conducting gene editing, and more, are summarized. Lastly, a perspective on forthcoming challenges and prospects concerning the advancement of mRNA therapeutics is offered. Despite these challenges, mRNA-based therapeutics remain promising, with the potential to revolutionize disease treatment and contribute to significant advancements in the biomedical field.
Collapse
Affiliation(s)
- Ji Wang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Haofang Zhu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jingjing Gan
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Gaofeng Liang
- Institute of Organoids on Chips Translational Research, Henan Academy of Sciences, Zhengzhou, 450009, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Institute of Organoids on Chips Translational Research, Henan Academy of Sciences, Zhengzhou, 450009, China
| |
Collapse
|
11
|
Pieczyńska A, Zasadzka E, Pilarska A, Procyk D, Adamska K, Hojan K. Rehabilitation Exercises Supported by Monitor-Augmented Reality for Patients with High-Grade Glioma Undergoing Radiotherapy: Results of a Randomized Clinical Trial. J Clin Med 2023; 12:6838. [PMID: 37959303 PMCID: PMC10648373 DOI: 10.3390/jcm12216838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/27/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Exercise has been shown to improve quality of life (QoL) and even treatment outcomes in cancer patients. However, the evidence to support the benefits of exercise in patients with high-grade glioma (HGG) is limited. Therefore, we performed a randomized clinical trial (RCT) to examine the effect of augmented-reality-based rehabilitation exercises on physical and functional fitness, cognitive function, fatigue, mood, QoL, selected blood parameters, brain derived neurotrophic factor (BDNF), and S100 protein in patients with HGG. METHODS Adult patients with HGG scheduled to undergo radiotherapy after tumor resection were randomized to participate in an exercise program (experimental group, n = 25) or to receive usual care (controls, n = 22). Physical and mental fitness was measured at baseline, after the completion of radiotherapy, and at 3 months. The following tests were administered: Handgrip Strength Test; 6-Minute Walk Test; Time Up and Go test; Functional Independent Measure scale; Addenbrooke's Cognitive Examination III (ACE III); Hospital Anxiety and Depression Scale; Functional Cancer Therapy Assessment-Brain; and Functional Assessment of Chronic Illness Therapy-Fatigue. We also measured blood parameters, BDNF, and S100 protein levels. RESULTS No significant changes were observed in the exercise group. However, the controls experienced a significant decrease in HGS and in the ACE III attention domain. No significant changes were observed in QoL, fatigue, BDNF, or S100 levels in either group. CONCLUSIONS Augmented-reality-based exercise during radiation therapy may prevent loss of muscle strength and attention in patients with HGG.
Collapse
Affiliation(s)
- Anna Pieczyńska
- Department of Occupational Therapy, Poznan University of Medical Sciences, 61-781 Poznan, Poland; (A.P.); (K.H.)
- Department of Rehabilitation, Greater Poland Cancer Centre, 61-866 Poznan, Poland;
| | - Ewa Zasadzka
- Department of Occupational Therapy, Poznan University of Medical Sciences, 61-781 Poznan, Poland; (A.P.); (K.H.)
| | - Agnieszka Pilarska
- Department of Rehabilitation, Greater Poland Cancer Centre, 61-866 Poznan, Poland;
| | - Danuta Procyk
- Central Laboratory, Greater Poland Cancer Centre, 15, 61-866 Poznan, Poland;
| | - Krystyna Adamska
- Chair and Department of Electroradiology, Poznan University of Medical Science, 61-781 Poznan, Poland;
- 3rd Radiotherapy Department, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Katarzyna Hojan
- Department of Occupational Therapy, Poznan University of Medical Sciences, 61-781 Poznan, Poland; (A.P.); (K.H.)
- Department of Rehabilitation, Greater Poland Cancer Centre, 61-866 Poznan, Poland;
| |
Collapse
|
12
|
Xie Y, Ye J, Luo H. HOXC Cluster Antisense RNA 3, a Novel Long Non-Coding RNA as an Oncological Biomarker and Therapeutic Target in Human Malignancies. Onco Targets Ther 2023; 16:849-865. [PMID: 37899986 PMCID: PMC10612484 DOI: 10.2147/ott.s425523] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 10/10/2023] [Indexed: 10/31/2023] Open
Abstract
HOXC cluster antisense RNA 3 (HOXC-AS3) is a novel long noncoding RNA (lncRNA) that exhibits aberrant expression patterns in various cancer types. Its expression is closely related to clinicopathological features, demonstrating significant clinical relevance across multiple tumors. And HOXC-AS3 plays multifaceted roles in tumor progression, impacting cell proliferation, apoptosis, migration, invasion, epithelial-mesenchymal transition (EMT), autophagy, senescence, tumor growth, and metastasis. In this review, we summarized and comprehensively analyzed the expression and clinical significance of HOXC-AS3 as a diagnostic and prognostic biomarker for malignancies. Additionally, we presented an in-depth update on HOXC-AS3's functions and regulatory mechanisms in cancer pathogenesis. This narrative review underscores the importance of HOXC-AS3 as a promising lncRNA candidate in cancer research and its potential as a predictive biomarker and therapeutic target in clinical applications.
Collapse
Affiliation(s)
- Yunhe Xie
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330008, People’s Republic of China
- Department of General Surgery, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332007, People’s Republic of China
| | - Jiarong Ye
- Queen Mary School, Nanchang University, Nanchang, Jiangxi, 330038, People’s Republic of China
| | - Hongliang Luo
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330008, People’s Republic of China
| |
Collapse
|