1
|
Wu Q, Zhang X, Qin M, Shi D, Li Y. Dual roles of LncRNA RNA143598: a biomarker for rheumatoid arthritis and its implications in cancer. Clin Rheumatol 2025:10.1007/s10067-025-07448-2. [PMID: 40279008 DOI: 10.1007/s10067-025-07448-2] [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: 11/29/2024] [Revised: 02/24/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
OBJECTIVE Rheumatoid arthritis (RA) is a chronic autoimmune disease with complex pathological mechanisms, including immune system dysregulation and chronic inflammation. Recent studies indicate that long non-coding RNAs (LncRNAs) play key roles in immune regulation and have been implicated in the pathogenesis of multiple diseases, including RA and various types of cancer. Understanding the involvement of LncRNAs in RA and their potential transcriptional effects in cancer could provide novel insights into disease mechanisms and therapeutic targets. METHODS Using the GSE94519 dataset, we analyzed serum LncRNA profiles from RA patients and healthy controls. Differential expression genes (DEGs) were identified using GEO2R, and findings were validated via quantitative polymerase chain reaction (qPCR) in 39 RA and 53 healthy samples. Receiver operating characteristic (ROC) analysis was performed to evaluate diagnostic performance. A pan-cancer analysis of MTRNR2L1 was conducted using TCGA data, with immune infiltration assessed via ssGSEA. RESULTS RNA143598 was significantly upregulated in RA patients, and qPCR confirmed its diagnostic potential (AUC = 0.77). Pan cancer analysis shows that MTRNR2L1 is highly expressed in glioblastoma (GBM) and lowly expressed in head and neck squamous cell carcinoma (HNSC), with high GBM expression linked to poor prognosis. Immune infiltration analysis showed MTRNR2L1 correlated with CD8 + T cells, macrophages, and dendritic cells in GBM. CONCLUSION RNA143598 is a promising RA biomarker, and its transcription gene MTRNR2L1 demonstrates potential in cancer prognosis and immune regulation. These findings provide a foundation for future research on targeted therapies for RA and cancer. Key Points • RNA143598 is identified as a significant biomarker for diagnosing rheumatoid arthritis (RA), showing promise for clinical application. • Quantitative PCR validation demonstrates the diagnostic potential of RNA143598, with an area under the curve (AUC) of 0.77. • MTRNR2L1, which is RNA143598 transcribed gene, exhibits differential expression in different cancer types, with high levels associated with poor prognosis in glioblastoma (GBM). • Immune infiltration analysis links MTRNR2L1 expression to the presence of CD8 + T cells, macrophages, and dendritic cells, suggesting its role in immune regulation in GBM.
Collapse
Affiliation(s)
- Qiuhua Wu
- Department of Clinical Laboratory, First Affiliated Hospital of Huzhou University, The First People's Hospital of Huzhou City, Huzhou, 313000, Zhejiang, China
| | - Xiaoxia Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Huzhou University, The First People's Hospital of Huzhou City, Huzhou, 313000, Zhejiang, China
| | - Meiyun Qin
- Department of Clinical Laboratory, First Affiliated Hospital of Huzhou University, The First People's Hospital of Huzhou City, Huzhou, 313000, Zhejiang, China
| | - Danfei Shi
- Department of Pathology, First Affiliated Hospital of Huzhou University, The First People's Hospital of Huzhou City, Huzhou, 313000, Zhejiang, China
| | - Yong Li
- Department of Clinical Laboratory, First Affiliated Hospital of Huzhou University, The First People's Hospital of Huzhou City, Huzhou, 313000, Zhejiang, China.
| |
Collapse
|
2
|
Li GW, Jin YP, Sheng MF. The miR-192/EGR1-HOXB9 loop inhibits immune evasion in glioma by arresting their NSC phenotypes. Int Immunopharmacol 2025; 152:114453. [PMID: 40090079 DOI: 10.1016/j.intimp.2025.114453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/18/2025]
Abstract
OBJECTIVE To elucidate the mechanism by which the miR-192-related axis regulates glioma immune evasion. METHODS Factor expression was measured by PCR/Western Blotting (WB). Immunofluorescence, FISH and DLRTM were used to elucidate target regulation mechanisms. Mitophagy was observed by TEM. We performed BrdU/Transwell assays to evaluate malignant phenotypes and WB to measure stemness-related protein expression in glioma. Immune chemokine levels were measured by ELISA, and M2-TAM/CD8+ T-cell proportions were measured by immune-fluorescence. Growth curves/tumor volume in vivo and tumor weight in vitro were assessed to evaluate tumor growth in vivo. An immune microenvironment was established in nude mice via tail vein injection of immune cells. Masson's trichrome staining was performed to explore the degree of fibrosis in relevant tissues. RESULTS MiR-192 expression was negatively correlated with glioma malignancy. The expression of downstream regulator (EGR1/HOXB9) of miR-192 was positively correlated with malignant phenotypes. MiR-192 inhibited EGR1/HOXB9 through targeted binding. The miR-192/EGR1-HOXB9 loop induced mitophagy, thus inhibited glioma cell proliferation/invasion. Moreover, this loop inhibited MT, weakening glioma cell stemness. This pathway reduced M2-TAM numbers and weakened their inhibitory effect on CD8+ T-cells by regulating immune chemokines. In vivo, miR-192 inhibited glioma proliferation and induced immune infiltration into glioma through this loop. CONCLUSION The miR-192/EGR1-HOXB9 loop inhibited glioma stemness phenotypes, weakening glioma malignancy by regulating mitophagy. Moreover, this loop affected chemokine secretion by TAMs, weakened their inhibitory effect on CD8+ T-cells and reduced immune evasion in glioma by regulating MT.
Collapse
Affiliation(s)
- Guo-Wei Li
- School of Rehabilitation Science, Nanjing Normal University of Special Education, Nanjing, Jiangsu 210038, China
| | - Yan-Ping Jin
- School of Nursing, Jiangsu Health Vocational College, Nanjing, Jiangsu 211899, China.
| | - Min-Feng Sheng
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| |
Collapse
|
3
|
Sun P, Liu F, Huo K, Wang J, Cheng Y, Shang S, Ma W, Yu J, Han J. Adiponectin facilitates the cell cycle, inhibits cell apoptosis and induces temozolomide resistance in glioblastoma via the Akt/mTOR pathway. Oncol Lett 2025; 29:127. [PMID: 39807099 PMCID: PMC11726000 DOI: 10.3892/ol.2025.14875] [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: 07/02/2024] [Accepted: 10/22/2024] [Indexed: 01/16/2025] Open
Abstract
Adiponectin (ADN) regulates DNA synthesis, cell apoptosis and cell cycle to participate in the pathology and progression of glioblastoma. The present study aimed to further explore the effect of ADN on temozolomide (TMZ) resistance in glioblastoma and the underlying mechanism of action. Glioblastoma cell lines (U251 and U87-MG cells) were treated with ADN and TMZ at different concentrations; subsequently, 3.0 µg/ml ADN and 1.0 mM TMZ were selected as the optimal concentrations for the experimental conditions. LY294002 (a PI3K inhibitor) was added to ADN or ADN + TMZ-treated glioblastoma cell lines. Cell growth rate was determined using the Cell Counting Kit-8 assay, the apoptotic rate and cell cycle were evaluated using Annexin V/propidium iodide and cell cycle assays, and p-Akt (Thr308), p-Akt (Ser473), Akt, p-mTOR, c-caspase 3, caspase 3, Bax, cyclin B1 and cyclin D1 expression was determined by western blotting. Adiponectin receptor (ADIPOR) 1 and ADIPOR2 were expressed in glioblastoma cell lines. The glioblastoma cell line growth rate was increased by ADN in a concentration- and time-dependent manner. ADN inhibited glioblastoma cell line apoptosis and facilitated cell cycle. Of note, ADN activated the Akt/mTOR pathway and the addition of LY294002 reversed the effect of ADN, indicating that ADN activated the Akt/mTOR pathway to suppress apoptosis and promote cell cycle in glioblastoma cell lines. Notably, TMZ inhibited glioblastoma cell line growth, promoted apoptosis and increased G2 phase cell cycle arrest. However, the addition of ADN reversed the effect of TMZ in glioblastoma cell lines, disclosing that ADN induced TMZ resistance. Markedly, ADN-mediated TMZ resistance was further attenuated by LY294002, suggesting that ADN activated the Akt/mTOR pathway to induce TMZ resistance in glioblastoma cell lines. In conclusion, ADN activated the Akt/mTOR pathway to facilitate cell cycle, inhibit cell apoptosis and induce TMZ resistance in glioblastoma.
Collapse
Affiliation(s)
- Peng Sun
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Fude Liu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Kang Huo
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jianyi Wang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yawen Cheng
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Suhang Shang
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wenlong Ma
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jia Yu
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jianfeng Han
- Department of Neurology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
4
|
Zhou Y, He Q, Huang G, Ouyang P, Wang H, Deng J, Chen P, Liang X, Hong Z, Zhang X, Qi S, Li Y. Malignant Cells Beyond the Tumor Core: The Non-Negligible Factor to Overcome the Refractory of Glioblastoma. CNS Neurosci Ther 2025; 31:e70333. [PMID: 40104956 PMCID: PMC11920816 DOI: 10.1111/cns.70333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Glioblastoma (GBM) is one of the most aggressive primary brain tumors in adults. Over 95% of GBM patients experience recurrence in the peritumoral brain tissue or distant regions, indicating the presence of critical factors in these areas that drive tumor recurrence. Current clinical treatments primarily focus on tumor cells from the tumor core (TC), while the role of neoplastic cells beyond the TC has been largely neglected. METHODS We conducted a comprehensive review of existing literature and studies on GBM, focusing on the identification and characterization of questionable cells (Q cells). Advanced imaging techniques, such as diffusion tensor imaging (DTI), magnetic resonance spectroscopy (MRS), and positron emission tomography (PET), were utilized to identify Q cells beyond the tumor core. We also analyzed the functional properties, cellular microenvironment, and physical characteristics of Q cells, as well as their implications for surgical resection. RESULTS Our review revealed that Q cells exhibit unique functional attributes, including enhanced invasiveness, metabolic adaptations, and resistance mechanisms. These cells reside in a distinct cellular microenvironment and are influenced by physical properties such as solid stress and stiffness. Advanced imaging techniques have improved the identification of Q cells, enabling more precise surgical resection. Targeting Q cells in therapeutic strategies could significantly reduce the risk of GBM recurrence. CONCLUSION The presence of Q cells in the peritumoral brain zone (PBZ) and beyond is a critical factor in GBM recurrence. Current treatments, which primarily target tumor cells in the TC, are insufficient to prevent recurrence due to the neglect of Q cells. Future research should focus on understanding the mechanisms influencing Q cells and developing targeted therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Yuyang Zhou
- Department of Neurosurgery, Institute of Brain Disease, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Qilin He
- Department of Neurosurgery, Institute of Brain Disease, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Guanglong Huang
- Department of Neurosurgery, Institute of Brain Disease, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Pei Ouyang
- Department of Neurosurgery, Institute of Brain Disease, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Hai Wang
- Department of Neurosurgery, Institute of Brain Disease, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Jiapeng Deng
- Department of Neurosurgery, Institute of Brain Disease, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Pengyu Chen
- Department of Neurosurgery, Institute of Brain Disease, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Xuan Liang
- Department of Neurosurgery, Institute of Brain Disease, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Zhisheng Hong
- Department of Neurosurgery, Institute of Brain Disease, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Xian Zhang
- Department of Neurosurgery, Institute of Brain Disease, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Songtao Qi
- Department of Neurosurgery, Institute of Brain Disease, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Yaomin Li
- Department of Neurosurgery, Institute of Brain Disease, Nanfang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| |
Collapse
|
5
|
Docrat TF, Eltahir AOE, Hussein AA, Marnewick JL. Green synthesis of metal nanocarriers: A perspective for targeting glioblastoma. Drug Discov Today 2024; 29:104219. [PMID: 39476945 DOI: 10.1016/j.drudis.2024.104219] [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: 05/22/2024] [Revised: 10/05/2024] [Accepted: 10/24/2024] [Indexed: 11/11/2024]
Abstract
Glioblastoma, the most aggressive brain cancer, is challenging to treat owing to the difficulty of crossing the blood-brain barrier, high recurrence rates and significant mortality. This review highlights the potential of green synthesis methods in developing metal nanoparticles (MNPs) as a sustainable solution for drug delivery systems targeting glioblastoma. We explore the unique properties and modes of action of MNPs synthesised through eco-friendly processes by focusing on their bioavailability and precision in brain targeting, and discuss the potential of MNPs to target glioblastoma at the molecular level. Integrating green synthesis into cancer therapeutics represents a novel paradigm shift towards treatments with higher efficacy and lower environmental impact, offering hope in the fight against glioblastoma.
Collapse
Affiliation(s)
- Taskeen F Docrat
- Applied Microbial and Health Biotechnology Institute, Cape Peninsula University of Technology, Bellville 7535, South Africa.
| | - Ali O E Eltahir
- Department of Chemistry, Cape Peninsula University of Technology, Bellville 7535, South Africa; Permanent address: Department of Chemistry, Omdurman Islamic University, Omdurman, P.O. Box 382, Khartoum, Sudan
| | - Ahmed A Hussein
- Department of Chemistry, Cape Peninsula University of Technology, Bellville 7535, South Africa
| | - Jeanine L Marnewick
- Applied Microbial and Health Biotechnology Institute, Cape Peninsula University of Technology, Bellville 7535, South Africa
| |
Collapse
|
6
|
Wang X, Sun Q, Liu T, Lu H, Lin X, Wang W, Liu Y, Huang Y, Huang G, Sun H, Chen Q, Wang J, Tian D, Yuan F, Liu L, Wang B, Gu Y, Liu B, Chen L. Single-cell multi-omics sequencing uncovers region-specific plasticity of glioblastoma for complementary therapeutic targeting. SCIENCE ADVANCES 2024; 10:eadn4306. [PMID: 39576855 PMCID: PMC11584018 DOI: 10.1126/sciadv.adn4306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 10/23/2024] [Indexed: 11/24/2024]
Abstract
Glioblastoma (GBM) cells are highly heterogeneous and invasive, leading to treatment resistance and relapse. However, the molecular regulation in and distal to tumors remains elusive. Here, we collected paired tissues from the tumor core (TC) and peritumoral brain (PTB) for integrated snRNA-seq and snATAC-seq analyses. Tumor cells infiltrating PTB from TC behave more like oligodendrocyte progenitor cells than astrocytes at the transcriptome level. Dual-omics analyses further suggest that the distal regulatory regions in the tumor genome and specific transcription factors are potential determinants of regional heterogeneity. Notably, while activator protein 1 (AP-1) is active in all GBM states, its activity declines from TC to PTB, with another transcription factor, BACH1, showing the opposite trend. Combined inhibition of AP-1 and BACH1 more efficiently attenuates the tumor progression in mice and prolongs survival than either single-target treatment. Together, our work reveals marked molecular alterations of infiltrated GBM cells and a synergy of combination therapy targeting intratumor heterogeneity in and distal to GBM.
Collapse
Affiliation(s)
- Xin Wang
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
- BGI Research, Hangzhou 310030, China
| | - Qian Sun
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | | | - Haoran Lu
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Xuyi Lin
- BGI Research, Hangzhou 310030, China
| | - Weiwen Wang
- China National GeneBank, BGI Research, Shenzhen 518120, China
| | - Yang Liu
- BGI Research, Hangzhou 310030, China
| | - Yunting Huang
- China National GeneBank, BGI Research, Shenzhen 518120, China
| | | | - Haixi Sun
- BGI Research, Shenzhen 518083, China
- BGI Research, Beijing 102601, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qianxue Chen
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Junmin Wang
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Daofeng Tian
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Fan'en Yuan
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | | | - Bo Wang
- China National GeneBank, BGI Research, Shenzhen 518120, China
- BGI Research, Shenzhen 518083, China
| | - Ying Gu
- BGI Research, Hangzhou 310030, China
- BGI Research, Shenzhen 518083, China
- BGI Research, Beijing 102601, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen 518083, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Baohui Liu
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Liang Chen
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
- BGI Research, Hangzhou 310030, China
| |
Collapse
|
7
|
Chen WF, Chuang JMJ, Yang SN, Chen NF, Bhattacharya M, Liu HT, Dhama K, Chakraborty C, Wen ZH. Gene expression profiling and the isocitrate dehydrogenase mutational landscape of temozolomide‑resistant glioblastoma. Oncol Lett 2024; 28:378. [PMID: 38939621 PMCID: PMC11209862 DOI: 10.3892/ol.2024.14511] [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: 09/21/2023] [Accepted: 04/09/2024] [Indexed: 06/29/2024] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain cancer that occurs more frequently than other brain tumors. The present study aimed to reveal a novel mechanism of temozolomide resistance in GBM using bioinformatics and wet lab analyses, including meta-Z analysis, Kaplan-Meier survival analysis, protein-protein interaction (PPI) network establishment, cluster analysis of co-expressed gene networks, and hierarchical clustering of upregulated and downregulated genes. Next-generation sequencing and quantitative PCR analyses revealed downregulated [tyrosine kinase with immunoglobulin and epidermal growth factor homology domains 1 (TIE1), calcium voltage-gated channel auxiliary subunit α2Δ1 (CACNA2D1), calpain 6 (CAPN6) and a disintegrin and metalloproteinase with thrombospondin motifs 6 (ADAMTS6)] and upregulated [serum amyloid (SA)A1, SAA2, growth differentiation factor 15 (GDF15) and ubiquitin specific peptidase 26 (USP26)] genes. Different statistical models were developed for these genes using the Z-score for P-value conversion, and Kaplan-Meier plots were constructed using several patient cohorts with brain tumors. The highest number of nodes was observed in the PPI network was for ADAMTS6 and TIE1. The PPI network model for all genes contained 35 nodes and 241 edges. Immunohistochemical staining was performed using isocitrate dehydrogenase (IDH)-wild-type or IDH-mutant GBM samples from patients and a significant upregulation of TIE1 (P<0.001) and CAPN6 (P<0.05) protein expression was demonstrated in IDH-mutant GBM in comparison with IDH-wild-type GBM. Structural analysis revealed an IDH-mutant model demonstrating the mutant residues (R132, R140 and R172). The findings of the present study will help the future development of novel biomarkers and therapeutics for brain tumors.
Collapse
Affiliation(s)
- Wu-Fu Chen
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan, R.O.C
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, R.O.C
| | - Jimmy Ming-Jung Chuang
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan, R.O.C
| | - San-Nan Yang
- Department of Pediatrics, E-DA Hospital, School of Medicine, College of Medicine I-Shou University, Kaohsiung 82445, Taiwan, R.O.C
- School of Medicine for International Students, College of Medicine, I-Shou University, Kaohsiung 82445, Taiwan, R.O.C
| | - Nan-Fu Chen
- Division of Neurosurgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan, R.O.C
- Center for General Education, Cheng Shiu University, Kaohsiung 833301, Taiwan, R.O.C
| | | | - Hsin-Tzu Liu
- Department of Medical Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970374, Taiwan, R.O.C
| | - Kuldeep Dhama
- Division of Pathology, Indian Council of Agriculture Research-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh 243122, India
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, R.O.C
| |
Collapse
|
8
|
Loginova N, Aniskin D, Timashev P, Ulasov I, Kharwar RK. GBM Immunotherapy: Macrophage Impacts. Immunol Invest 2024; 53:730-751. [PMID: 38634572 DOI: 10.1080/08820139.2024.2337022] [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] [Indexed: 04/19/2024]
Abstract
BACKGROUND Glioblastoma (GBM) is an extremely aggressive form of brain tumor with low survival rates. Current treatments such as chemotherapy, radiation, and surgery are problematic due to tumor growth, invasion, and tumor microenvironment. GBM cells are resistant to these standard treatments, and the heterogeneity of the tumor makes it difficult to find a universal approach. Progression of GBM and acquisition of resistance to therapy are due to the complex interplay between tumor cells and the TME. A significant portion of the TME consists of an inflammatory infiltrate, with microglia and macrophages being the predominant cells. METHODS Analysis of the literature data over a course of 5 years suggest that the tumor-associated macrophages (TAMs) are capable of releasing cytokines and growth factors that promote tumor proliferation, survival, and metastasis while inhibiting immune cell function at the same time. RESULTS Thus, immunosuppressive state, provided with this intensively studied kind of TME cells, is supposed to promote GBM development through TAMs modulation of tumor treatment-resistance and aggressiveness. Therefore, TAMs are an attractive therapeutic target in the treatment of glioblastoma. CONCLUSION This review provides a comprehensive overview of the latest research on the nature of TAMs and the development of therapeutic strategies targeting TAMs, focusing on the variety of macrophage properties, being modulated, as well as molecular targets.
Collapse
Affiliation(s)
- Nina Loginova
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Denis Aniskin
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Peter Timashev
- World-Class Research Centre "Digital Biodesign and Personalized Healthcare", Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, World-Class Research Centre "Digital Biodesign and Personalized Healthcare", I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Rajesh Kumar Kharwar
- Endocrine Research Laboratory, Department of Zoology, University of Lucknow, Lucknow, India
| |
Collapse
|
9
|
Uthanaphun T, Manochantr S, Tantrawatpan C, Tantikanlayaporn D, Kheolamai P. PL-hMSC and CH-hMSC derived soluble factors inhibit proliferation but improve hGBM cell migration by activating TGF-β and inhibiting Wnt signaling. Biosci Rep 2024; 44:BSR20231964. [PMID: 38687607 PMCID: PMC11130542 DOI: 10.1042/bsr20231964] [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: 03/18/2024] [Accepted: 04/30/2024] [Indexed: 05/02/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and aggressive brain tumors. GBM resists most chemotherapeutic agents, resulting in a high mortality rate in patients. Human mesenchymal stem cells (hMSCs), which are parts of the cancer stroma, have been shown to be involved in the development and progression of GBM. However, different sources of hMSCs might affect GBM cells differently. In the present study, we established hMSCs from placenta (PL-hMSC) and chorion (CH-hMSC) to study the effects of their released soluble factors on the proliferation, migration, invasion, gene expression, and survival of human GBM cells, U251. We found that the soluble factors derived from CH-hMSCs and PL-hMSCs suppressed the proliferation of U251 cells in a dose-dependent manner. In contrast, soluble factors derived from both hMSC sources increased U251 migration without affecting their invasive property. The soluble factors derived from these hMSCs decreased the expression levels of CyclinD1, E2Fs and MYC genes that promote GBM cell proliferation but increased the expression level of TWIST gene, which promotes EMT and GBM cell migration. The functional study suggests that both hMSCs might exert their effects, at least in part, by activating TGF-β and suppressing Wnt/β-catenin signaling in U251 cells. Our study provides a better understanding of the interaction between GBM cells and gestational tissue-derived hMSCs. This knowledge might be used to develop safer and more effective stem cell therapy that improves the survival and quality of life of patients with GBM by manipulating the interaction between hMSCs and GBM cells.
Collapse
Affiliation(s)
- Tanawat Uthanaphun
- Master of Science Program in Stem Cell and Molecular Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Sirikul Manochantr
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Chairat Tantrawatpan
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Duangrat Tantikanlayaporn
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| | - Pakpoom Kheolamai
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
- Division of Cell Biology, Faculty of Medicine, Thammasat University, Pathumthani 12120, Thailand
| |
Collapse
|
10
|
Zhang P, Gao C, Guo Q, Yang D, Zhang G, Lu H, Zhang L, Zhang G, Li D. Single-cell RNA sequencing reveals the evolution of the immune landscape during perihematomal edema progression after intracerebral hemorrhage. J Neuroinflammation 2024; 21:140. [PMID: 38807233 PMCID: PMC11131315 DOI: 10.1186/s12974-024-03113-8] [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: 02/28/2024] [Accepted: 04/26/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Perihematomal edema (PHE) after post-intracerebral hemorrhage (ICH) has complex pathophysiological mechanisms that are poorly understood. The complicated immune response in the post-ICH brain constitutes a crucial component of PHE pathophysiology. In this study, we aimed to characterize the transcriptional profiles of immune cell populations in human PHE tissue and explore the microscopic differences between different types of immune cells. METHODS 9 patients with basal ganglia intracerebral hemorrhage (hematoma volume 50-100 ml) were enrolled in this study. A multi-stage profile was developed, comprising Group1 (n = 3, 0-6 h post-ICH, G1), Group2 (n = 3, 6-24 h post-ICH, G2), and Group3 (n = 3, 24-48 h post-ICH, G3). A minimal quantity of edematous tissue surrounding the hematoma was preserved during hematoma evacuation. Single cell RNA sequencing (scRNA-seq) was used to map immune cell populations within comprehensively resected PHE samples collected from patients at different stages after ICH. RESULTS We established, for the first time, a comprehensive landscape of diverse immune cell populations in human PHE tissue at a single-cell level. Our study identified 12 microglia subsets and 5 neutrophil subsets in human PHE tissue. What's more, we discovered that the secreted phosphoprotein-1 (SPP1) pathway served as the basis for self-communication between microglia subclusters during the progression of PHE. Additionally, we traced the trajectory branches of different neutrophil subtypes. Finally, we also demonstrated that microglia-produced osteopontin (OPN) could regulate the immune environment in PHE tissue by interacting with CD44-positive cells. CONCLUSIONS As a result of our research, we have gained valuable insight into the immune-microenvironment within PHE tissue, which could potentially be used to develop novel treatment modalities for ICH.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Clinical Medicine, Jining Medical University, Jining, China
| | - Cong Gao
- Department of Clinical Medicine, Jining Medical University, Jining, China
| | - Qiang Guo
- Department of Emergency Stroke, Affiliated Hospital of Jining Medical University, Jining, China
| | - Dongxu Yang
- Department of Emergency Stroke, Affiliated Hospital of Jining Medical University, Jining, China
| | - Guangning Zhang
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, China
| | - Hao Lu
- Department of Emergency Stroke, Affiliated Hospital of Jining Medical University, Jining, China
| | - Liman Zhang
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Guorong Zhang
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Daojing Li
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining, China.
| |
Collapse
|
11
|
Guo M, Zhang J, Han J, Hu Y, Ni H, Yuan J, Sun Y, Liu M, Gao L, Liao W, Ma C, Liu Y, Li S, Li N. VEGFR2 blockade inhibits glioblastoma cell proliferation by enhancing mitochondrial biogenesis. J Transl Med 2024; 22:419. [PMID: 38702818 PMCID: PMC11067099 DOI: 10.1186/s12967-024-05155-1] [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: 12/06/2023] [Accepted: 04/02/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Glioblastoma is an aggressive brain tumor linked to significant angiogenesis and poor prognosis. Anti-angiogenic therapies with vascular endothelial growth factor receptor 2 (VEGFR2) inhibition have been investigated as an alternative glioblastoma treatment. However, little is known about the effect of VEGFR2 blockade on glioblastoma cells per se. METHODS VEGFR2 expression data in glioma patients were retrieved from the public database TCGA. VEGFR2 intervention was implemented by using its selective inhibitor Ki8751 or shRNA. Mitochondrial biogenesis of glioblastoma cells was assessed by immunofluorescence imaging, mass spectrometry, and western blot analysis. RESULTS VEGFR2 expression was higher in glioma patients with higher malignancy (grade III and IV). VEGFR2 inhibition hampered glioblastoma cell proliferation and induced cell apoptosis. Mass spectrometry and immunofluorescence imaging showed that the anti-glioblastoma effects of VEGFR2 blockade involved mitochondrial biogenesis, as evidenced by the increases of mitochondrial protein expression, mitochondria mass, mitochondrial oxidative phosphorylation (OXPHOS), and reactive oxygen species (ROS) production, all of which play important roles in tumor cell apoptosis, growth inhibition, cell cycle arrest and cell senescence. Furthermore, VEGFR2 inhibition exaggerated mitochondrial biogenesis by decreased phosphorylation of AKT and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α), which mobilized PGC1α into the nucleus, increased mitochondrial transcription factor A (TFAM) expression, and subsequently enhanced mitochondrial biogenesis. CONCLUSIONS VEGFR2 blockade inhibits glioblastoma progression via AKT-PGC1α-TFAM-mitochondria biogenesis signaling cascade, suggesting that VEGFR2 intervention might bring additive therapeutic values to anti-glioblastoma therapy.
Collapse
Affiliation(s)
- Min Guo
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Junhao Zhang
- Department of Medicine-Solna, Division of Cardiovascular Medicine, Karolinska University Hospital, Solna, 171 76, Stockholm, Sweden
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiang Han
- Department of Biopharmaceutical Sciences and National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yingyue Hu
- Department of Biopharmaceutical Sciences and National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Hao Ni
- Department of Medicine-Solna, Division of Cardiovascular Medicine, Karolinska University Hospital, Solna, 171 76, Stockholm, Sweden
- Department of Gynaecology and Obstetrics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Juan Yuan
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Yang Sun
- Department of Immunology and Shandong University-Karolinska Institutet Collaborative Laboratory, Shandong University Cheeloo Medical College, School of Basic Medicine, Jinan, China
| | - Meijuan Liu
- Department of Biopharmaceutical Sciences and National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Lifen Gao
- Department of Immunology and Shandong University-Karolinska Institutet Collaborative Laboratory, Shandong University Cheeloo Medical College, School of Basic Medicine, Jinan, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chunhong Ma
- Department of Immunology and Shandong University-Karolinska Institutet Collaborative Laboratory, Shandong University Cheeloo Medical College, School of Basic Medicine, Jinan, China
| | - Yaou Liu
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuijie Li
- Department of Biopharmaceutical Sciences and National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China.
| | - Nailin Li
- Department of Medicine-Solna, Division of Cardiovascular Medicine, Karolinska University Hospital, Solna, 171 76, Stockholm, Sweden.
| |
Collapse
|
12
|
Zhou L, Xiang H, Liu S, Chen H, Yang Y, Zhang J, Cai W. Folic Acid Functionalized AQ4N/Gd@PDA Nanoplatform with Real-Time Monitoring of Hypoxia Relief and Enhanced Synergistic Chemo/Photothermal Therapy in Glioma. Int J Nanomedicine 2024; 19:3367-3386. [PMID: 38617794 PMCID: PMC11012807 DOI: 10.2147/ijn.s451921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/27/2024] [Indexed: 04/16/2024] Open
Abstract
Purpose Hypoxia is often associated with glioma chemoresistance, and alleviating hypoxia is also crucial for improving treatment efficacy. However, although there are already some methods that can improve efficacy by alleviating hypoxia, real-time monitoring that can truly achieve hypoxia relief and efficacy feedback still needs to be explored. Methods AQ4N/Gd@PDA-FA nanoparticles (AGPF NPs) were synthesized using a one-pot method and were characterized. The effects of AGPF NPs on cell viability, cellular uptake, and apoptosis were investigated using the U87 cell line. Moreover, the effectiveness of AGPF NPs in alleviating hypoxia was explored in tumor-bearing mice through photoacoustic imaging. In addition, the diagnosis and treatment effect of AGPF NPs were evaluated by magnetic resonance imaging (MRI) and bioluminescent imaging (BLI) on orthotopic glioma mice respectively. Results In vitro experiments showed that AGPF NPs had good dispersion, stability, and controlled release. AGPF NPs were internalized by cells through endocytosis, and could significantly reduce the survival rate of U87 cells and increase apoptosis under irradiation. In addition, we monitored blood oxygen saturation at the tumor site in real-time through photoacoustic imaging (PAI), and the results showed that synergistic mild-photothermal therapy/chemotherapy effectively alleviated tumor hypoxia. Finally, in vivo anti-tumor experiments have shown that synergistic therapy can effectively alleviate hypoxia and inhibit the growth of orthotopic gliomas. Conclusion This work not only provides an effective means for real-time monitoring of the dynamic feedback between tumor hypoxia relief and therapeutic efficacy, but also offers a potential approach for the clinical treatment of gliomas.
Collapse
Affiliation(s)
- Longjiang Zhou
- Department of Neurology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, People’s Republic of China
| | - Haitao Xiang
- Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine, Suzhou, 215028, People’s Republic of China
| | - Susu Liu
- School of Life Science and Technology, Xidian University and Engineering Research Center of Molecular and Neuro Imaging, Ministry of Education, Xi’an, 710126, People’s Republic of China
| | - Honglin Chen
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| | - Yuanwei Yang
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| | - Jianyong Zhang
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| | - Wei Cai
- Department of Neurosurgery, Suqian First Hospital, Suqian, 223800, People’s Republic of China
| |
Collapse
|
13
|
Li GW, Jin YP, Qiu JP, Lu XF. ITGB2 fosters the cancerous characteristics of ovarian cancer cells through its role in mitochondrial glycolysis transformation. Aging (Albany NY) 2024; 16:3007-3020. [PMID: 38345576 PMCID: PMC10911379 DOI: 10.18632/aging.205529] [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: 09/29/2023] [Accepted: 12/27/2023] [Indexed: 02/20/2024]
Abstract
Related studies have shown that ITGB2 mediates mitochondrial glycolytic transformation in cancer-associated fibroblasts and participates in tumor occurrence, metastasis and invasion of cancer cells. Based on these studies, we tried to construct a mitochondrial glycolysis regulatory network and explored its effect on mitochondrial homeostasis and ovarian cancer cells' cancerous characteristics. Our research revealed a distinct increase in the expression of ITGB2 and associated signaling pathway elements (PI3K-AKT-mTOR) in cases of ovarian cancer. ITGB2 might control mTOR expression via the PI3K-AKT pathway, thus promote mitochondrial glycolysis transformation and cell energy supply in ovarian cancer. This pathway could also inhibit mitophagy, maintain mitochondrial stability, and enhance the cancerous characteristics in case of ovarian cancer cells by mediating mitochondrial glycolytic transformation. Thus, we concluded that ITGB2-associated signaling route (PI3K-AKT-mTOR) may contribute to the progression of cancerous traits in ovarian cancer via mediating mitochondrial glycolytic transformation.
Collapse
Affiliation(s)
- Guo-Wei Li
- Department of Rehabilitation Science, Nanjing Normal University of Special Education, Nanjing, Jiangsu 210000, China
| | - Yan-Ping Jin
- Department of Obstetrics and Gynecology, Zhongda Hospital Jiangbei Branch, School of Medicine, Southeast University, Nanjing, Jiangsu 210000, China
| | - Jian-Ping Qiu
- Department of Obstetrics and Gynecology, Suzhou Municipal Hospital North, Suzhou, Jiangsu 215000, China
| | - Xiu-Fang Lu
- Department of Obstetrics and Gynecology, Suzhou Municipal Hospital North, Suzhou, Jiangsu 215000, China
| |
Collapse
|
14
|
Yang G, Cai S, Hu M, Li C, Yang L, Zhang W, Sun J, Sun F, Xing L, Sun X. Spatial features of specific CD103 +CD8 + tissue-resident memory T cell subsets define the prognosis in patients with non-small cell lung cancer. J Transl Med 2024; 22:27. [PMID: 38183111 PMCID: PMC10770937 DOI: 10.1186/s12967-023-04839-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 12/26/2023] [Indexed: 01/07/2024] Open
Abstract
BACKGROUND Tissue-resident memory T (TRM) cells can reside in the tumor microenvironment and are considered the primary response cells to immunotherapy. Heterogeneity in functional status and spatial distribution may contribute to the controversial role of TRM cells but we know little about it. METHODS Through multiplex immunofluorescence (mIF) (CD8, CD103, PD-1, Tim-3, GZMB, CK), the quantity and spatial location of TRM cell subsets were recognized in the tissue from 274 patients with NSCLC after radical surgery. By integrating multiple machine learning methods, we constructed a TRM-based spatial immune signature (TRM-SIS) to predict the prognosis. Furthermore, we conducted a CD103-related gene set enrichment analysis (GSEA) and verified its finding by another mIF panel (CD8, CD103, CK, CD31, Hif-1α). RESULTS The density of TRM cells was significantly correlated with the expression of PD-1, Tim-3 and GZMB. Four types of TRM cell subsets was defined, including TRM1 (PD-1-Tim-3-TRM), TRM2 (PD-1+Tim-3-TRM), TRM3 (PD-1-Tim-3+TRM) and TRM4 (PD-1+Tim-3+TRM). The cytotoxicity of TRM2 was the strongest while that of TRM4 was the weakest. Compare with TRM1 and TRM2, TRM3 and TRM4 had better infiltration and stronger interaction with cancer cells. The TRM-SIS was an independent prognostic factor for disease-free survival [HR = 2.43, 95%CI (1.63-3.60), P < 0.001] and showed a better performance than the TNM staging system for recurrence prediction. Furthermore, by CD103-related GSEA and mIF validation, we found a negative association between tumor angiogenesis and infiltration of TRM cells. CONCLUSIONS These findings reveal a significant heterogeneity in the functional status and spatial distribution of TRM cells, and support it as a biomarker for the prognosis of NSCLC patients. Regulating TRM cells by targeting tumor angiogenesis may be a potential strategy to improve current immunotherapy.
Collapse
Affiliation(s)
- Guanqun Yang
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Siqi Cai
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mengyu Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Chaozhuo Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Liying Yang
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wei Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jujie Sun
- Department of Pathology, Shandong Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Fenghao Sun
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ligang Xing
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiaorong Sun
- Shandong University Cancer Center, Shandong University, Jinan, Shandong, China.
- Department of Nuclear Medicine, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
15
|
Yakubov E, Schmid S, Hammer A, Chen D, Dahlmanns JK, Mitrovic I, Zurabashvili L, Savaskan N, Steiner HH, Dahlmanns M. Ferroptosis and PPAR-gamma in the limelight of brain tumors and edema. Front Oncol 2023; 13:1176038. [PMID: 37554158 PMCID: PMC10406130 DOI: 10.3389/fonc.2023.1176038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/04/2023] [Indexed: 08/10/2023] Open
Abstract
Human malignant brain tumors such as gliomas are devastating due to the induction of cerebral edema and neurodegeneration. A major contributor to glioma-induced neurodegeneration has been identified as glutamate. Glutamate promotes cell growth and proliferation in variety of tumor types. Intriguently, glutamate is also an excitatory neurotransmitter and evokes neuronal cell death at high concentrations. Even though glutamate signaling at the receptor and its downstream effectors has been extensively investigated at the molecular level, there has been little insight into how glutamate enters the tumor microenvironment and impacts on metabolic equilibration until recently. Surprisingly, the 12 transmembrane spanning tranporter xCT (SLC7A11) appeared to be a major player in this process, mediating glutamate secretion and ferroptosis. Also, PPARγ is associated with ferroptosis in neurodegeneration, thereby destroying neurons and causing brain swelling. Although these data are intriguing, tumor-associated edema has so far been quoted as of vasogenic origin. Hence, glutamate and PPARγ biology in the process of glioma-induced brain swelling is conceptually challenging. By inhibiting xCT transporter or AMPA receptors in vivo, brain swelling and peritumoral alterations can be mitigated. This review sheds light on the role of glutamate in brain tumors presenting the conceptual challenge that xCT disruption causes ferroptosis activation in malignant brain tumors. Thus, interfering with glutamate takes center stage in forming the basis of a metabolic equilibration approach.
Collapse
Affiliation(s)
- Eduard Yakubov
- Department of Neurosurgery, Paracelsus Medical University, Nuremberg, Germany
| | - Sebastian Schmid
- Department of Trauma, Orthopaedics, Plastic and Hand Surgery, University Hospital Augsburg, Augsburg, Germany
| | - Alexander Hammer
- Department of Neurosurgery, Paracelsus Medical University, Nuremberg, Germany
- Center for Spine and Scoliosis Therapy, Malteser Waldkrankenhaus St. Marien, Erlangen, Germany
| | - Daishi Chen
- Department of Otorhinolaryngology, Shenzhen People's Hospital, Jinan University, Shenzhen, China
| | - Jana Katharina Dahlmanns
- Institute for Physiology and Pathophysiology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Ivana Mitrovic
- Department of Cardiac Surgery, Bogenhausen Hospital, Munich, Germany
| | | | - Nicolai Savaskan
- Department of Neurosurgery, University Medical School Hospital Universitätsklinikum Erlangen (UKER), Friedrich-Alexander Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Department of Public Health Neukölln, District Office Neukölln of Berlin Neukölln, Berlin, Germany
| | | | - Marc Dahlmanns
- Institute for Physiology and Pathophysiology, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
16
|
Kumari S, Kumar P. Identification and characterization of putative biomarkers and therapeutic axis in Glioblastoma multiforme microenvironment. Front Cell Dev Biol 2023; 11:1236271. [PMID: 37538397 PMCID: PMC10395518 DOI: 10.3389/fcell.2023.1236271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 06/23/2023] [Indexed: 08/05/2023] Open
Abstract
Non-cellular secretory components, including chemokines, cytokines, and growth factors in the tumor microenvironment, are often dysregulated, impacting tumorigenesis in Glioblastoma multiforme (GBM) microenvironment, where the prognostic significance of the current treatment remains unsatisfactory. Recent studies have demonstrated the potential of post-translational modifications (PTM) and their respective enzymes, such as acetylation and ubiquitination in GBM etiology through modulating signaling events. However, the relationship between non-cellular secretory components and post-translational modifications will create a research void in GBM therapeutics. Therefore, we aim to bridge the gap between non-cellular secretory components and PTM modifications through machine learning and computational biology approaches. Herein, we highlighted the importance of BMP1, CTSB, LOX, LOXL1, PLOD1, MMP9, SERPINE1, and SERPING1 in GBM etiology. Further, we demonstrated the positive relationship between the E2 conjugating enzymes (Ube2E1, Ube2H, Ube2J2, Ube2C, Ube2J2, and Ube2S), E3 ligases (VHL and GNB2L1) and substrate (HIF1A). Additionally, we reported the novel HAT1-induced acetylation sites of Ube2S (K211) and Ube2H (K8, K52). Structural and functional characterization of Ube2S (8) and Ube2H (1) have identified their association with protein kinases. Lastly, our results found a putative therapeutic axis HAT1-Ube2S(K211)-GNB2L1-HIF1A and potential predictive biomarkers (CTSB, HAT1, Ube2H, VHL, and GNB2L1) that play a critical role in GBM pathogenesis.
Collapse
|