1
|
Adamecz DI, Veres É, Papp C, Árva H, Rónavári A, Marton A, Vizler C, Gácser A, Kónya Z, Igaz N, Kiricsi M. Gold and Silver Nanoparticles Efficiently Modulate the Crosstalk Between Macrophages and Cancer Cells. Int J Nanomedicine 2025; 20:4777-4802. [PMID: 40255669 PMCID: PMC12009049 DOI: 10.2147/ijn.s508171] [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: 12/10/2024] [Accepted: 04/02/2025] [Indexed: 04/22/2025] Open
Abstract
Background Macrophages, polarized into pro-inflammatory M1 or anti-inflammatory M2 states, are essential cellular elements of innate immunity. In the tumor microenvironment, owing to a paracrine manipulative program by cancerous cells, tumor-associated macrophages (TAMs) evolve, which can shift between M1-like and M2-like phenotypes. Since it is fairly unknown how the promising anticancer agents, silver (AgNPs) and gold nanoparticles (AuNPs) affect the bidirectional communication and reprogramming in the tumor stroma, we examined the behavior, the tumor-supporting functions, and the expression of polarization and functional marker genes of TAMs to reveal how these are modulated upon interaction with nanoparticle-exposed cancer cells. Methods We established co-cultures of murine immortalized J774 or primary bone marrow-derived macrophages with 4T1 breast cancer cells treated with AuNPs or AgNPs or with none of the nanoparticles. We assessed the expression of macrophage polarization and functional markers using RT-qPCR and Proteome Profiler Array and evaluated macrophage migration and matrix metalloproteinase activity by specific assays. Results Protein and mRNA levels of most examined factors - except tumor necrosis factor-alpha - such as C-C-motif chemokine ligands 2 and 22, interleukin-23, inducible nitric oxide synthase, cyclooxygenase-2, the macrophage mannose receptor CD206, transforming growth factor-beta, and chitinase-like-3 protein decreased, and the expression of polarization markers revealed a shift towards M1-like phenotype in macrophages co-cultured with AgNP- or AuNP-treated 4T1 cells. Both nanoparticle treatments reduced the levels and activity of cell migration-related factors, such as C-C motif chemokine ligand 3, matrix metalloproteinases, and suppressed macrophage migration. Conclusion Both AuNPs and AgNPs showed a remarkable ability to influence macrophage-cancer cell communication, suppressed indirectly M2-like TAM polarization, and perturbed the migration behavior of TAMs that is critical for tumor invasion, indicating modulated immunological functions and debilitated cancer-promoting capabilities of TAMs in this microenvironment.
Collapse
Affiliation(s)
- Dóra Izabella Adamecz
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Éva Veres
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
- Department of Biotechnology and Microbiology, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Pathogen Fungi Research Group, University of Szeged, Szeged, Hungary
| | - Csaba Papp
- Department of Biotechnology and Microbiology, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Pathogen Fungi Research Group, University of Szeged, Szeged, Hungary
| | - Hédi Árva
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Andrea Rónavári
- Department of Applied and Environmental Chemistry, University of Szeged, Szeged, Hungary
| | - Annamária Marton
- Laboratory of Tumor Immunology and Pharmacology, Centre of Excellence of the European Union, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Csaba Vizler
- Laboratory of Tumor Immunology and Pharmacology, Centre of Excellence of the European Union, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Attila Gácser
- Department of Biotechnology and Microbiology, University of Szeged, Szeged, Hungary
- HCEMM-SZTE Pathogen Fungi Research Group, University of Szeged, Szeged, Hungary
| | - Zoltán Kónya
- Department of Applied and Environmental Chemistry, University of Szeged, Szeged, Hungary
| | - Nóra Igaz
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| | - Mónika Kiricsi
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| |
Collapse
|
2
|
Hao T, Pei Z, Hu S, Zhao Z, He W, Wang J, Jiang L, Ariben J, Wu L, Yang X, Wang L, Wu Y, Chen X, Li Q, Yang H, Li S, Wang X, Sun M, Zhang B. Identification of osteoarthritis-associated chondrocyte subpopulations and key gene-regulating drugs based on multi-omics analysis. Sci Rep 2025; 15:12448. [PMID: 40216809 PMCID: PMC11992032 DOI: 10.1038/s41598-025-90694-w] [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: 10/08/2024] [Accepted: 02/14/2025] [Indexed: 04/14/2025] Open
Abstract
The mechanism by which chondrocytes respond to mechanical stress in joints significantly affects the balance and function of cartilage. This study aims to characterize osteoarthritis-associated chondrocyte subpopulations and key gene targets for regulatory drugs. To begin, single-cell and transcriptome datasets were obtained from the Gene Expression Omnibus (GEO) database. Cell communication and pseudo-temporal analysis, as well as High-dimensional Weighted Gene Co-expression Network Analysis (hdWGCNA), were conducted on the single-cell data to identify key chondrocyte subtypes and module genes. Subsequently, Consensus Cluster Plus analysis was utilized to identify distinct disease subgroups within the osteoarthritis (OA) training dataset based on the key module genes. Furthermore, differential gene expression analysis and GO/KEGG pathway enrichment analysis were performed on the identified subgroups. To screen for hub genes associated with OA, a combination of 10 machine learning algorithms and 113 algorithm compositions was integrated. Additionally, the immune and pathway scores of the training dataset samples were evaluated using the ESTIMATE, MCP-counter, and ssGSEA algorithms to establish the relationship between the hub genes and immune and pathways. Following this, a network depicting the interaction between the hub genes and transcription factors was constructed based on the Network Analyst database. Moreover, the hub genes were subjected to drug prediction and molecular docking using the RNAactDrug database and AutoDockTools. Finally, real-time fluorescence quantitative PCR (RT-qPCR) was employed to detect the expression of hub genes in the plasma samples collected from osteoarthritis patients and healthy adults. In the OA sample, there is a significant increase in the proportion of prehypertrophic chondrocytes (preHTC), particularly in subgroups 6, 7, and 9. We defined these subgroups as OA_PreHTC subgroups. The OA_PreHTC subgroup exhibits a higher communication intensity with proliferative-related pathways such as ANGPTL and TGF-β. Furthermore, two OA disease subgroups were identified in the training set samples. This led to the identification of 411 differentially expressed genes (DEGs) related to osteoarthritis, 2485 DEGs among subgroups, as well as 238 intersecting genes and 5 hub genes (MMP13, FAM26F, CHI3L1, TAC1, and CKS2). RT-qPCR results indicate significant differences in the expression levels of five hub genes and their related TFs in the clinical blood samples of OA patients compared to the healthy control group (NC). Moreover, these five hub genes are positively associated with inflammatory pathways such as TNF-α, JAK-STAT3, and inflammatory response, while being negatively associated with proliferation pathways like WNT and KRAS. Additionally, the five hub genes are positively associated with neutrophils, activated CD4 T cell, gamma delta T cell, and regulatory T cell, while being negatively associated with CD56dim natural killer cell and Type 17T helper cell. Molecular docking results reveal that CAY10603, Tenulin, T0901317, and Nonactin exhibit high binding activity to CHI3L1, suggesting their potential as therapeutic drugs for OA. The OA_PreHTC subgroups plays a crucial role in the occurrence and development of osteoarthritis (OA). Five hub genes may exert their effects on OA through interactions with PreHTC cells, other chondrocytes, and immune cells, playing a role in inhibiting cell proliferation and stimulating inflammation, thus having high diagnostic value for OA. Additionally, CAY10603, Tenulin, T0901317, and Nonactin have potential therapeutic effects for OA patients.
Collapse
Affiliation(s)
- Ting Hao
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Zhiwei Pei
- Tianjin Hospital, Tianjin University, Jiefang Nan Road 406, Hexi District, Tianjin, 300211, People's Republic of China
| | - Sile Hu
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Zhenqun Zhao
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Wanxiong He
- Sanya People's Hospital, No. 558 Jiefang Road, Sanya City, Hainan Province, People's Republic of China
| | - Jing Wang
- Baotou Medical College Bayannur Clinical Medical College, Bayannur City, 015000, Inner Mongolia, People's Republic of China
| | - Liuchang Jiang
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Jirigala Ariben
- Bayannur City Hospital, Bayannur City, 015000, Inner Mongolia, People's Republic of China
| | - Lina Wu
- Aier Eye Hospital, Tianjin University, No. 102 Fukang Road, Tianjin, 300000, People's Republic of China
| | - Xiaolong Yang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Leipeng Wang
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Yonggang Wu
- Bayannur City Hospital, Bayannur City, 015000, Inner Mongolia, People's Republic of China
| | - Xiaofeng Chen
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Qiang Li
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Haobo Yang
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
| | - Siqin Li
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China
- Bayannur City Hospital, Bayannur City, 015000, Inner Mongolia, People's Republic of China
| | - Xing Wang
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China.
- Bayannur City Hospital, Bayannur City, 015000, Inner Mongolia, People's Republic of China.
| | - Mingqi Sun
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China.
| | - Baoxin Zhang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China.
- Tianjin Hospital, Tianjin University, Jiefang Nan Road 406, Hexi District, Tianjin, 300211, People's Republic of China.
- Inner Mongolia Medical University, Hohhot, 010050, Inner Mongolia, People's Republic of China.
| |
Collapse
|
3
|
Li Y, Shao Z, Jiang J, Wang H, Zhang M. Transcription factor NFKB1 mediates TUBB6 to promote the proliferation and suppress apoptosis in glioma via Wnt/β-catenin signaling pathway. Discov Oncol 2025; 16:444. [PMID: 40169445 PMCID: PMC11961833 DOI: 10.1007/s12672-025-02268-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/28/2025] [Indexed: 04/03/2025] Open
Abstract
Glioma remains one of the most challenging brain tumors with poor prognosis. In this study, we aimed to elucidate the role of TUBB6 in glioma and its potential as a diagnostic and prognostic biomarker. Analysis of the GSE42656 and TCGA datasets revealed that TUBB6 was significantly upregulated in glioma tissues compared to normal tissues. The diagnostic value of TUBB6 was demonstrated with an area under the curve (AUC) of 0.702, suggesting that it could be used as a biomarker to differentiate gliomas Correlation analyses revealed that high TUBB6 expressions were associated with advanced WHO grades, IDH mutation status, and histological types of glioma. Further investigation identified NFKB1 as a key transcription factor that binds to the promoter region of TUBB6, upregulating its expression in glioma cells. Elevated levels of NFKB1 were associated with poor overall survival and disease-specific survival in glioma patients. Knockdown of NFKB1 resulted in reduced TUBB6 expression in glioma cells, confirming the regulatory roles of NFKB1 in TUBB6 expression. Prognostic analysis using TCGA and CGGA datasets demonstrated that high TUBB6 expression was associated with poorer overall survival (OS) and disease-specific survival (DSS) in glioma patients. TUBB6 was identified as an independent prognostic factor for both OS and DSS. Additionally, pan-cancer analysis revealed that TUBB6 was dysregulated in various tumor types and showed prognostic value across multiple cancers. Functional enrichment analysis of TUBB6-associated differentially expressed genes indicated involvement in immune response, extracellular matrix remodeling, and cytokine signaling pathways. In vitro experiments showed that TUBB6 knockdown suppressed glioma cell proliferation and promoted apoptosis by regulating the canonical Wnt/β-catenin signaling pathway. Our findings suggest that TUBB6 contributes to glioma malignancy through its effects on the Wnt/β-catenin pathway. In conclusion, TUBB6 emerges as a promising biomarker for glioma diagnosis and prognosis. Its regulation by NFKB1 and involvement in key signaling pathways underscore its potential as a therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Yan Li
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ziyu Shao
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jun Jiang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hongyan Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mei Zhang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, China.
| |
Collapse
|
4
|
Yu M, Liu J, Zhou W, Gu X, Yu S. MRI radiomics based on machine learning in high-grade gliomas as a promising tool for prediction of CD44 expression and overall survival. Sci Rep 2025; 15:7433. [PMID: 40032983 PMCID: PMC11876340 DOI: 10.1038/s41598-025-90128-7] [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: 06/22/2024] [Accepted: 02/11/2025] [Indexed: 03/05/2025] Open
Abstract
We aimed to predict CD44 expression and assess its prognostic significance in patients with high-grade gliomas (HGG) using non-invasive radiomics models based on machine learning. Enhanced magnetic resonance imaging, along with the corresponding gene expression and clinicopathological data, was downloaded from online database. Kaplan-Meier survival curves, univariate and multivariate COX analyses, and time-dependent receiver operating characteristic were used to assess the prognostic value of CD44. Following the screening of radiomic features using repeat least absolute shrinkage and selection operator, two radiomics models were constructed utilizing logistic regression and support vector machine for validation purposes. The results indicated that CD44 protein levels were higher in HGG compared to normal brain tissues, and CD44 expression emerged as an independent biomarker of diminished overall survival (OS) in patients with HGG. Moreover, two predictive models based on seven radiomic features were built to predict CD44 expression levels in HGG, achieving areas under the curves (AUC) of 0.809 and 0.806, respectively. Calibration and decision curve analysis validated the fitness of the models. Notably, patients with high radiomic scores presented worse OS (p < 0.001). In summary, our results indicated that the radiomics models effectively differentiate CD44 expression level and OS in patients with HGG.
Collapse
Affiliation(s)
- Mingjun Yu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
- Department of Medical Affairs, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Jinliang Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Wen Zhou
- Department of Pain Management, Dalian Municipal Central Hospital, Dalian, 116033, People's Republic of China
| | - Xiao Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.
| | - Shijia Yu
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China.
| |
Collapse
|
5
|
Li J, Zhang M, Sun Q, Li X, Du F, Cheng Y, Li S, Zhang J. CENPF interaction with PLA2G4A promotes glioma growth by modulating mTORC1 and NF-κB pathways. Cancer Cell Int 2025; 25:73. [PMID: 40025532 PMCID: PMC11871623 DOI: 10.1186/s12935-025-03700-6] [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: 09/29/2024] [Accepted: 02/17/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Glioma is the most common primary malignant tumor of the central nervous system, and due to the limited effectiveness of traditional single-target therapies, there is an urgent need for new therapeutic targets. Centromere protein F (CENPF) belongs to the centromere protein family and is mainly involved in the regulation of the cell cycle. CENPF has recently been found to play a key role in tumorigenesis and tumor progression, but its role in gliomas has not been well studied. METHODS The expression level and clinical information of CENPF were obtained by analyzing the TCGA, CGGA and GEO databases. Immunohistochemistry and western blot analysis were used to quantitatively detect the expression of CENPF in glioma tissues and cell lines. Gene set enrichment analysis (GSEA) of TCGA and GSE16011 datasets was used to explore the molecular mechanism of the CENPF. CENPF-interacting proteins were detected by molecular docking and co-immunoprecipitation (Co-IP). After silencing CENPF, CCK-8 assay, Transwell assay and flow cytometry were used to detect changes in cell proliferation, invasion, cell cycle and apoptosis, and Western blot was used to detect changes in signaling pathway protein levels. RESULTS Bioinformatics analysis showed that CENPF was generally highly expressed in gliomas and was associated with poor prognosis. This result was confirmed in glioma samples from our hospital. Multivariate Cox regression analysis showed that CENPF was an independent prognostic marker for gliomas. Western blot analysis in vitro showed that CENPF was overexpressed in the U251 and LN229 cell lines; therefore, these two cell lines were selected for subsequent experiments. GSEA analysis showed that CENPF was mainly involved in the G2/M phase-mediated cell cycle and P53 signaling pathway. Flow cytometry analysis confirmed that silencing CENPF induced G2/M phase arrest and increased apoptosis in glioma cells. Subsequent experiments confirmed that CENPF influences the epithelial-mesenchymal transition (EMT) process through the mTORC1 signaling pathway. Molecular docking and Co-IP assay revealed that CENPF exerts its effects by interacting with PLA2G4A promoting the downstream signaling pathway. Finally, we found that silencing CENPF combined with a PLA2G4A inhibitor (AACOCF3) induced glioma cell apoptosis and exhibited anti-glioma effects. CONCLUSIONS This study found that CENPF plays a key role in promoting tumorigenesis through its interaction with PLA2G4A. This study provides a theoretical foundation for advancing multi-targeted therapies in glioma and for developing strategies to overcome tumor drug resistance.
Collapse
Affiliation(s)
- Junhong Li
- Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province, 276000, China
| | - Moxuan Zhang
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| | - Qiang Sun
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
| | - Xinglan Li
- Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province, 276000, China
| | - Fei Du
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
| | - Yanhao Cheng
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong Province, 276000, China
| | - Shuzhi Li
- Department of Neurosurgery, Gaomi Traditional Chinese Medicine Hospital, Weifang, Shandong Province, 261500, China.
| | - Jian Zhang
- Department of Neurosurgery, Linyi People's Hospital, Linyi, Shandong Province, 276000, China.
| |
Collapse
|
6
|
Xia B, Yu C, Liu J, Lin J, Lyu J, Wang X, Zhu L. DANCR knockdown alleviates neuroinflammation and functional recovery after spinal cord injury via regulating the ACTN4 / STAT3 axis. Arch Biochem Biophys 2025; 765:110293. [PMID: 39798643 DOI: 10.1016/j.abb.2025.110293] [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: 12/08/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Polarization of microglia following spinal cord injury (SCI) is a pivotal pathological process of secondary injury. Although differentiation antagonistic nonprotein coding RNA (DANCR) has been implicated in immune and inflammatory responses across various diseases, its role in SCI still unclear. This research aimed to clarify the underlying mechanisms of DANCR in SCI recovery by investigating its expression pattern in microglia. Our findings indicate that the DANCR level in microglia is increased after SCI and that its knockdown can promote microglial M2-type polarization; suppress inflammatory cytokines, oxidative stress, and neuronal apoptosis; and facilitate nerve regeneration as well as spinal cord functional recovery. Further investigations suggest that DANCR's effects are mediated through the ACTN4/STAT3 axis. These results provide potential targets for enhancing functional recovery following SCI.
Collapse
Affiliation(s)
- Bin Xia
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China; Department of Orthopedics, Chengdu Seventh People's Hospital (Afliated Cancer Hospital of Chengdu Medical College), Chengdu, 610299, China.
| | - Cheng Yu
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Jin Liu
- Department of Orthopedics, Chengdu Seventh People's Hospital (Afliated Cancer Hospital of Chengdu Medical College), Chengdu, 610299, China.
| | - Jiezhao Lin
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Jiao Lyu
- Department of Orthopedics, Chengdu Seventh People's Hospital (Afliated Cancer Hospital of Chengdu Medical College), Chengdu, 610299, China.
| | - Xin Wang
- Department of Surgery, MAO County Hospital of Traditional Chinese Medicine, Ngawa Tibetan and Qiang Autonomous Prefecture, 623200, China.
| | - Lixin Zhu
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
7
|
Gong Z, Guo D, Lin Y, Liu Z, Lv M, Liu X, Yao Y, Wang S, Wang Y, Wang Z. A single-cell transcriptome analysis reveals astrocyte heterogeneity and identifies CHI3L1 as a diagnostic biomarker in Parkinson's disease. Heliyon 2025; 11:e42051. [PMID: 39931480 PMCID: PMC11808505 DOI: 10.1016/j.heliyon.2025.e42051] [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: 08/18/2024] [Revised: 01/15/2025] [Accepted: 01/15/2025] [Indexed: 02/13/2025] Open
Abstract
Background Parkinson's disease (PD) is the second most common neurodegenerative disease, characterized by motor and non-motor symptoms. It has been reported that astrocytes play a critical role in the pathogenesis and progression of PD. Here, we aimed to identify the heterogeneity of astrocytes and investigate genes associated with astrocyte differentiation trajectories in PD. Methods The single-cell transcriptomic profiles of PD samples were collected from the GEO database. We have identified subsets of astrocytes and analyzed their functions. The differentiation trajectory of astrocyte subtypes was explored using Monocle2. Inflammatory response scores were determined using AUCell. The levels of CHI3L1 mRNA and protein expressions in astrocytes were analyzed using qRT-PCR and Western Blot assay, respectively. Results We characterized seven cell types within the substantia nigra region of both PD and normal samples. Our analysis revealed that astrocytes comprised the second-highest proportion of cell types. Additionally, we identified three distinct subpopulations of astrocytes: Astro-C0, Astro-C1, and Astro-C2. Notably, Astro-C0 was associated with inflammatory signaling pathways. Trajectory analysis indicated that Astro-C0 occupies an intermediate stage of differentiation. The astrocyte-related gene CHI3L1 was found to be highly expressed in the Astro-C0 subpopulation. Furthermore, we observed increased levels of CHI3L1 mRNA and protein in LPS-induced astrocytes. Astrocytes exhibiting elevated CHI3L1 levels demonstrated interactions with microglia in PD patients. Lastly, we discovered that CHI3L1 was significantly overexpressed in PD patients and exhibited strong diagnostic potential for the disease. Conclusion This study clarified the heterogeneity of astrocytes in PD based on the single-cell transcriptomic profiles and found that CHI3L1 may be a diagnostic biomarker for PD.
Collapse
Affiliation(s)
- Zhongying Gong
- Department of Neurology, Tianjin First Central Hospital, School of Medcine, Nankai University, Tianjin, 300192, China
| | - Dan Guo
- Department of Neurology, Tianjin First Central Hospital, School of Medcine, Nankai University, Tianjin, 300192, China
| | - Yufeng Lin
- Department of Neurology, Tianjin First Central Hospital, School of Medcine, Nankai University, Tianjin, 300192, China
| | - Zhiwei Liu
- Department of Neurology, Tianjin First Central Hospital, School of Medcine, Nankai University, Tianjin, 300192, China
| | - Mengdi Lv
- Department of Neurology, Tianjin First Central Hospital, School of Medcine, Nankai University, Tianjin, 300192, China
| | - Xinxin Liu
- Department of Neurology, Tianjin First Central Hospital, School of Medcine, Nankai University, Tianjin, 300192, China
| | - Yang Yao
- Department of Neurology, Tianjin First Central Hospital, School of Medcine, Nankai University, Tianjin, 300192, China
| | - Sijia Wang
- Department of Neurology, Tianjin First Central Hospital, School of Medcine, Nankai University, Tianjin, 300192, China
| | - Yuan Wang
- Department of Neurology, Tianjin First Central Hospital, School of Medcine, Nankai University, Tianjin, 300192, China
| | - Zhiyun Wang
- Department of Neurology, Tianjin First Central Hospital, School of Medcine, Nankai University, Tianjin, 300192, China
| |
Collapse
|
8
|
Yu W, Gui S, Peng L, Luo H, Xie J, Xiao J, Yilamu Y, Sun Y, Cai S, Cheng Z, Tao Z. STAT3-controlled CHI3L1/SPP1 positive feedback loop demonstrates the spatial heterogeneity and immune characteristics of glioblastoma. Dev Cell 2025:S1534-5807(25)00034-6. [PMID: 39933531 DOI: 10.1016/j.devcel.2025.01.014] [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: 08/05/2024] [Revised: 10/30/2024] [Accepted: 01/17/2025] [Indexed: 02/13/2025]
Abstract
Proneural-mesenchymal transition (PMT) is a phenotypic alteration and contributes to the malignant progression of glioblastoma (GBM). Macrophages, as a main infiltrating component of the tumor immune microenvironment (TIM), control the biological processes of PMT; however, the mechanisms driving this process remain largely unknown. Here, the overall landscape of tumor and nontumor cells was described by scMulti-omics technology. Then, we demonstrated that chitinase-3-like protein 1 (CHI3L1) played a critical role in maintaining mesenchymal (MES) status and reprogramming macrophage phenotype using C57BL/6 and NSG mice models derived from PN20 cells. Mechanistically, osteopontin (OPN)/ITGB1 maintained the activation of nuclear factor κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3) pathways by establishing a positive feedback loop with the CHI3L1-STAT3 axis, resulting in PMT. CHI3L1 enhanced the phosphorylation, nuclear localization, and transcriptional activity of STAT3 via directly binding its coiled-coil domain (CCD). Importantly, we screened and validated that hygromycin B (HB), an inhibitor of the STAT3-CCD domain, disrupted the CHI3L1-STAT3 interaction, thereby reducing the tumor burden in vitro and in vivo.
Collapse
Affiliation(s)
- Wanli Yu
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang 330006, Jiangxi, China; JXHC key Laboratory of Neurological Medicine, Nanchang University, Nanchang 330006, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Shikai Gui
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang 330006, Jiangxi, China; JXHC key Laboratory of Neurological Medicine, Nanchang University, Nanchang 330006, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Lunshan Peng
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang 330006, Jiangxi, China; JXHC key Laboratory of Neurological Medicine, Nanchang University, Nanchang 330006, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Haitao Luo
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Jiabao Xie
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang 330006, Jiangxi, China; JXHC key Laboratory of Neurological Medicine, Nanchang University, Nanchang 330006, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Juexian Xiao
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Yimuran Yilamu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Yi Sun
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China
| | - Shihao Cai
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China; Jiangxi Province Key Laboratory of Neurological Diseases, Nanchang University, Nanchang 330006, Jiangxi, China; JXHC key Laboratory of Neurological Medicine, Nanchang University, Nanchang 330006, Jiangxi, China; Institute of Neuroscience, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Zujue Cheng
- Department of Neurosurgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China.
| | - Zhennan Tao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, Jiangsu, China; Neurosurgical Institute, Nanjing University, Nanjing 210008, Jiangsu, China.
| |
Collapse
|
9
|
Yu TH, Ding YY, Zhao SG, Zhao JH, Gu Y, Chen DH, Zhang F, Hong WM. Single-cell sequencing uncovers the mechanistic role of DAPK1 in glioma and its diagnostic and prognostic implications. Front Immunol 2025; 15:1463747. [PMID: 39926603 PMCID: PMC11802534 DOI: 10.3389/fimmu.2024.1463747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 12/31/2024] [Indexed: 02/11/2025] Open
Abstract
Background We conducted an investigation into the characteristics of single-cell differentiation data in gliomas, with a focus on developing DAPK1-based prognostic markers to predict patient outcomes. Dysregulated expression of DAPK1 has been associated with the invasive behavior of various malignancies, including gliomas. However, the precise role and underlying mechanisms of DAPK1 in gliomas remain inadequately understood. Methods We performed analyses on RNA-seq and microarray datasets from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), in addition to single-cell RNA sequencing (scRNA-seq) data from glioma patients available in GEO. Utilizing the Seurat R package, we identified gene clusters associated with survival from the scRNA-seq data. Prognostic models were developed using LASSO and stepwise regression algorithms. Furthermore, we assessed the predictive potential of these genes within the immune microenvironment and their relevance in immunotherapy contexts. Results Our scRNA-seq data analysis revealed 32 distinct cell clusters corresponding to 10 cell types. Through dimensionality reduction and clustering, we identified three glial cell subpopulations based on their differentiation trajectories. DAPK1, serving as a marker gene for the terminal subpopulation, exhibited an association with poor prognosis. Conclusions DAPK1-based prognostic models show promise for accurately predicting outcomes in glioblastoma and glioma. An in-depth examination of DAPK1's specific mechanisms in glioblastoma could elucidate its role in immunotherapy response. Targeting the DAPK1 gene may offer therapeutic benefits for glioma patients.
Collapse
Affiliation(s)
- Tian-Hang Yu
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yan-Yu Ding
- Department of Immunology, School of Basic Medicine, Anhui Medical University, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Center, Hefei, China
| | - Si-Guo Zhao
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jie-Hui Zhao
- School of Nursing, Anhui Medical University, Hefei, China
| | - Yu Gu
- School of Nursing, Anhui Medical University, Hefei, China
| | - Dong-Hui Chen
- Department of Neurosurgery, Lu’an People’s Hospital, Luan, China
| | - Fang Zhang
- School of Nursing, Anhui Medical University, Hefei, China
| | - Wen-Ming Hong
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
- Open Project of Key Laboratory of Dermatology, Ministry of Education, Anhui Medical University, Hefei, China
| |
Collapse
|
10
|
Wang Z, Zhang K, Zhong C, Zhu Z, Zheng X, Yang P, Che B, Lu Y, Zhang Y, Xu T. Plasma Human Cartilage Glycoprotein-39 and Cognitive Impairment After Acute Ischemic Stroke. J Am Heart Assoc 2025; 14:e036790. [PMID: 39819010 DOI: 10.1161/jaha.124.036790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 12/05/2024] [Indexed: 01/19/2025]
Abstract
BACKGROUND Our study aimed at evaluating the association between plasma human cartilage glycoprotein-39 (YKL-40) and cognitive impairment at 3 months among patients with acute ischemic stroke. METHODS AND RESULTS Plasma YKL-40 levels were measured in 604 participants from the China Antihypertensive Trial in Acute Ischemic Stroke. Cognitive impairment outcomes were assessed at 3 months poststroke using the Mini-Mental State Examination and the Montreal Cognitive Assessment. According to the Mini-Mental State Examination score, patients in the highest quartile of YKL-40 had a 2.01-fold (95% CI, 1.23-3.29; P for trend=0.009) risk of poststroke cognitive impairment compared with those in the lowest quartile. Each 1 SD difference of logarithm-transformed YKL-40 was associated with a 28% (95% CI, 7-53) increased risk for the outcome. The multiple-adjusted spline regression model confirmed dose-response relationships between YKL-40 and poststroke cognitive impairment (P for linearity=0.01). Adding YKL-40 to a model containing conventional risk factors significantly improved the discriminatory power (area under the receiver operating characteristic curve improved by 0.02, P=0.03). When cognitive impairment was defined using the Montreal Cognitive Assessment score, similar findings were observed. CONCLUSIONS Elevated YKL-40 levels were associated with an increased risk of cognitive impairment at 3 months among patients with acute ischemic stroke. REGISTRATION URL: clinicaltrials.gov; Unique Identifier: NCT01840072.
Collapse
Affiliation(s)
- Ziyi Wang
- Department of Neurology, Affiliated Hospital of Nantong University Medical School of Nantong University Nantong China
| | - Kaixin Zhang
- Department of Clinical Research Center Wuxi No. 2 People's Hospital (Jiangnan University Medical Center) Wuxi China
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology Suzhou Medical College of Soochow University Suzhou China
| | - Chongke Zhong
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology Suzhou Medical College of Soochow University Suzhou China
| | - Zhengbao Zhu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology Suzhou Medical College of Soochow University Suzhou China
| | - Xiaowei Zheng
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology Suzhou Medical College of Soochow University Suzhou China
| | - Pinni Yang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology Suzhou Medical College of Soochow University Suzhou China
| | - Bizhong Che
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology Suzhou Medical College of Soochow University Suzhou China
| | - Yaling Lu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology Suzhou Medical College of Soochow University Suzhou China
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Major Chronic Non-communicable Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology Suzhou Medical College of Soochow University Suzhou China
| | - Tian Xu
- Department of Neurology Affiliated Hospital of Nantong University Nantong China
| |
Collapse
|
11
|
Mwale PF, Hsieh CT, Yen TL, Jan JS, Taliyan R, Yang CH, Yang WB. Chitinase-3-like-1: a multifaceted player in neuroinflammation and degenerative pathologies with therapeutic implications. Mol Neurodegener 2025; 20:7. [PMID: 39827337 PMCID: PMC11742494 DOI: 10.1186/s13024-025-00801-8] [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: 09/24/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
Chitinase-3-like-1 (CHI3L1) is an evolutionarily conserved protein involved in key biological processes, including tissue remodeling, angiogenesis, and neuroinflammation. It has emerged as a significant player in various neurodegenerative diseases and brain disorders. Elevated CHI3L1 levels have been observed in neurological conditions such as traumatic brain injury (TBI), Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), Creutzfeldt-Jakob disease (CJD), multiple sclerosis (MS), Neuromyelitis optica (NMO), HIV-associated dementia (HAD), Cerebral ischemic stroke (CIS), and brain tumors. This review explores the role of CHI3L1 in the pathogenesis of these disorders, with a focus on its contributions to neuroinflammation, immune cell infiltration, and neuronal degeneration. As a key regulator of neuroinflammation, CHI3L1 modulates microglia and astrocyte activity, driving the release of proinflammatory cytokines that exacerbate disease progression. In addition to its role in disease pathology, CHI3L1 has emerged as a promising biomarker for the diagnosis and monitoring of brain disorders. Elevated cerebrospinal fluid (CSF) levels of CHI3L1 have been linked to disease severity and cognitive decline, particularly in AD and MS, highlighting its potential for clinical diagnostics. Furthermore, therapeutic strategies targeting CHI3L1, such as small-molecule inhibitors and neutralizing antibodies, have shown promise in preclinical studies, demonstrating reduced neuroinflammation, amyloid plaque accumulation, and improved neuronal survival. Despite its therapeutic potential, challenges remain in developing selective and safe CHI3L1-targeted therapies, particularly in ensuring effective delivery across the blood-brain barrier and mitigating off-target effects. This review addresses the complexities of targeting CHI3L1, highlights its potential in precision medicine, and outlines future research directions aimed at unlocking its full therapeutic potential in treating neurodegenerative diseases and brain pathologies.
Collapse
Affiliation(s)
- Pharaoh Fellow Mwale
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei, 110, Taiwan
| | - Cheng-Ta Hsieh
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei, 110, Taiwan
- Division of Neurosurgery, Department of Surgery, Cathay General Hospital, Taipei City, 106438, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan
- Department of Medicine, School of Medicine, Fu Jen Catholic University, New Taipei City, 24205, Taiwan
| | - Ting-Lin Yen
- Department of Medical Research, Cathay General Hospital, Taipei, 22174, Taiwan
| | - Jing-Shiun Jan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei, 110, Taiwan
| | - Rajeev Taliyan
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, Pilani, Rajasthan, India
| | - Chih-Hao Yang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei, 110, Taiwan.
- Research Center for Neuroscience, Taipei Medical University, Taipei, Taiwan.
| | - Wen-Bin Yang
- Research Center for Neuroscience, Taipei Medical University, Taipei, Taiwan.
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
12
|
Song Z, Feng Z, Wang X, Li J, Zhang D. NFKB1 as a key player in Tumor biology: from mechanisms to therapeutic implications. Cell Biol Toxicol 2025; 41:29. [PMID: 39797972 PMCID: PMC11724797 DOI: 10.1007/s10565-024-09974-2] [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: 08/28/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025]
Abstract
NFKB1, a core transcription factor critical in various biological process (BP), is increasingly studied for its role in tumors. This research combines literature reviews, meta-analyses, and bioinformatics to systematically explore NFKB1's involvement in tumor initiation and progression. A unique focus is placed on the NFKB1-94 ATTG promoter polymorphism, highlighting its association with cancer risk across diverse genetic models and ethnic groups, alongside comprehensive analysis of pan-cancer expression patterns and drug sensitivity. The study reveals the intricate connections between NFKB1 and tumors, highlighting its significant roles in invasion, metastasis, genomic stability, and metabolic changes. Through meta-analysis, it is evidenced that tumor specimens exhibit increased NFKB1 expression when compared to non-tumor specimens, although its association with cancer incidence requires further investigation. Analysis from the Gene Expression Omnibus (GEO) database suggests that high NFKB1 gene expression may not markedly impact tumor patient prognosis. The noticeable correlation between the NFKB1-94 ATTG promoter polymorphic sequence and elevated cancer susceptibility is highlighted across different genetic models. Furthermore, bioinformatics analysis uncovers NFKB1's association with the sensitivity to various anticancer drugs and its central involvement in crucial BP like the cell cycle, cytoskeleton assembly, and cellular senescence. Overall, NFKB1's expression and polymorphisms are significantly linked to tumor risk, prognosis, and treatment response, highlighting its prospect as a forthcoming aim for cancer treatment. This study offers a robust foundation for further exploration of NFKB1's mechanisms and the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Zixuan Song
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang , Liaoning Province, China
| | - Zheng Feng
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaoxue Wang
- Department of Health Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jingying Li
- Department of Health Management, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dandan Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang , Liaoning Province, China.
| |
Collapse
|
13
|
YANG YUSHI, HU CHUJIAO, LEI SHAN, BAO XIN, ZENG ZHIRUI, CAO WENPENG. Using Multi-Omics Analysis to Explore Diagnostic Tool and Optimize Drug Therapy Selection for Patients with Glioma Based on Cross-Talk Gene Signature. Oncol Res 2024; 32:1921-1934. [PMID: 39574472 PMCID: PMC11576925 DOI: 10.32604/or.2024.046191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/28/2024] [Indexed: 11/24/2024] Open
Abstract
Background The heterogeneity of prognosis and treatment benefits among patients with gliomas is due to tumor microenvironment characteristics. However, biomarkers that reflect microenvironmental characteristics and predict the prognosis of gliomas are limited. Therefore, we aimed to develop a model that can effectively predict prognosis, differentiate microenvironment signatures, and optimize drug selection for patients with glioma. Materials and Methods The CIBERSORT algorithm, bulk sequencing analysis, and single-cell RNA (scRNA) analysis were employed to identify significant cross-talk genes between M2 macrophages and cancer cells in glioma tissues. A predictive model was constructed based on cross-talk gene expression, and its effect on prognosis, recurrence prediction, and microenvironment characteristics was validated in multiple cohorts. The effect of the predictive model on drug selection was evaluated using the OncoPredict algorithm and relevant cellular biology experiments. Results A high abundance of M2 macrophages in glioma tissues indicates poor prognosis, and cross-talk between macrophages and cancer cells plays a crucial role in shaping the tumor microenvironment. Eight genes involved in the cross-talk between macrophages and cancer cells were identified. Among them, periostin (POSTN), chitinase 3 like 1 (CHI3L1), serum amyloid A1 (SAA1), and matrix metallopeptidase 9 (MMP9) were selected to construct a predictive model. The developed model demonstrated significant efficacy in distinguishing patient prognosis, recurrent cases, and characteristics of high inflammation, hypoxia, and immunosuppression. Furthermore, this model can serve as a valuable tool for guiding the use of trametinib. Conclusions In summary, this study provides a comprehensive understanding of the interplay between M2 macrophages and cancer cells in glioma; utilizes a cross-talk gene signature to develop a predictive model that can predict the differentiation of patient prognosis, recurrence instances, and microenvironment characteristics; and aids in optimizing the application of trametinib in glioma patients.
Collapse
Affiliation(s)
- YUSHI YANG
- Department of Pathology and Pathophysiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - CHUJIAO HU
- Guizhou Provincial Engineering Technology Research Center for Chemical Drug R&D; State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
| | - SHAN LEI
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - XIN BAO
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - ZHIRUI ZENG
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550025, China
| | - WENPENG CAO
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Key Laboratory of Human Brain Bank for Functions and Diseases of Department of Education of Guizhou Province, Guizhou Medical University, Guiyang, 550025, China
| |
Collapse
|
14
|
Cao W, Zeng Z, Sun J, Chen Y, Kuang F, Luo S, Lan J, Lei S. Exosome-derived circ-001422 promotes tumor-associated macrophage M2 polarization to accelerate the progression of glioma. Commun Biol 2024; 7:1504. [PMID: 39538012 PMCID: PMC11561164 DOI: 10.1038/s42003-024-07134-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Cytokines, tumor cells, and tumor-associated macrophages play crucial roles in the composition of glioma tissue. Studies have demonstrated that certain cytokines can induce M2 polarization of tumor-associated macrophages and contribute to the progression of glioma. Nonetheless, the intricate molecular interactions among cytokines, glioma cells, and tumor-associated macrophages remain largely unexplored. To investigate this cross-talk, a combination of RNA-sequencing, chromatin immunoprecipitation, immunoprecipitation, exosome isolation, and biological experiments were employed. Treatment with IL-6 significantly increased circ-001422 expression in glioma cells. A poorer prognosis was associated with elevated levels of circ-001422 in glioma tissues. Circ-001422 was transcribed directly by STAT3 through binding to its promoter. Circ-001422 exerted cancer-promoting functions when co-cultured with M2 macrophages. Furthermore, glioma cells were found to transfer circ-001422 to macrophages via an exosomal pathway, promoting M2 polarization. Mechanically, circ-001422 interacted with p300, resulting in STAT3 acetylation, thus promoting nuclear localization and transcriptional activity of STAT3/NF-κB and M2 macrophage polarization. In conclusion, glioma cells released exosomes enriched with circ-001422, which in turn induce M2 macrophage polarization by activating the STAT3/NF-κB pathway, thereby enhancing the aggressive characteristics of glioma cells. Targeting circ-001422 may represent a potential therapeutic approach for glioma.
Collapse
Affiliation(s)
- Wenpeng Cao
- Key Laboratory of Human Brain bank for Functions and Diseases of Department of Education of Guizhou Province, Guizhou Medical University, Guiyang, 550009, Guizhou, China.
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, 550009, Guizhou, China.
| | - Zhirui Zeng
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550009, Guizhou, China
- Postdoctoral workstation, Affiliated Hospital of Guizhou Medical University, Guiyang, 550009, Guizhou, China
| | - JianFei Sun
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Guizhou Medical University, Guiyang, 550009, Guizhou, China
| | - Yunhua Chen
- Key Laboratory of Human Brain bank for Functions and Diseases of Department of Education of Guizhou Province, Guizhou Medical University, Guiyang, 550009, Guizhou, China
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, 550009, Guizhou, China
| | - FaGuang Kuang
- Key Laboratory of Human Brain bank for Functions and Diseases of Department of Education of Guizhou Province, Guizhou Medical University, Guiyang, 550009, Guizhou, China
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, 550009, Guizhou, China
| | - Shipeng Luo
- Key Laboratory of Human Brain bank for Functions and Diseases of Department of Education of Guizhou Province, Guizhou Medical University, Guiyang, 550009, Guizhou, China
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, 550009, Guizhou, China
| | - Jinzhi Lan
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550009, Guizhou, China
| | - Shan Lei
- Department of Physiology, School of Basic Medicine, Guizhou Medical University, Guiyang, 550009, Guizhou, China.
- Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550009, Guizhou, China.
| |
Collapse
|
15
|
Chen X, Wang Z, Zhu B, Deng M, Qiu J, Feng Y, Ding N, Huang C. Metabolic Reprogramming Induced by Aging Modifies the Tumor Microenvironment. Cells 2024; 13:1721. [PMID: 39451239 PMCID: PMC11506685 DOI: 10.3390/cells13201721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Aging is an important risk factor for tumorigenesis. Metabolic reprogramming is a hallmark of both aging and tumor initiation. However, the manner in which the crosstalk between aging and metabolic reprogramming affects the tumor microenvironment (TME) to promote tumorigenesis was poorly explored. We utilized a computational approach proposed by our previous work, MMP3C (Modeling Metabolic Plasticity by Pathway Pairwise Comparison), to characterize aging-related metabolic plasticity events using pan-cancer bulk RNA-seq data. Our analysis revealed a high degree of metabolically organized heterogeneity across 17 aging-related cancer types. In particular, a higher degree of several energy generation pathways, i.e., glycolysis and impaired oxidative phosphorylation, was observed in older patients. Similar phenomena were also found via single-cell RNA-seq analysis. Furthermore, those energy generation pathways were found to be weakened in activated T cells and macrophages, whereas they increased in exhausted T cells, immunosuppressive macrophages, and Tregs in older patients. It was suggested that aging-induced metabolic switches alter glucose utilization, thereby influencing immune function and resulting in the remodeling of the TME. This work offers new insights into the associations between tumor metabolism and the TME mediated by aging, linking with novel strategies for cancer therapy.
Collapse
Affiliation(s)
- Xingyu Chen
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Kay Laboratory of Quality Research in Chinese Medicine & Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR 999078, China; (X.C.); (Z.W.); (B.Z.); (J.Q.); (Y.F.); (N.D.)
| | - Zihan Wang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Kay Laboratory of Quality Research in Chinese Medicine & Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR 999078, China; (X.C.); (Z.W.); (B.Z.); (J.Q.); (Y.F.); (N.D.)
| | - Bo Zhu
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Kay Laboratory of Quality Research in Chinese Medicine & Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR 999078, China; (X.C.); (Z.W.); (B.Z.); (J.Q.); (Y.F.); (N.D.)
| | - Min Deng
- Faculty of Health Sciences, University of Macau, Taipa, Macau SAR 999078, China;
| | - Jiayue Qiu
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Kay Laboratory of Quality Research in Chinese Medicine & Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR 999078, China; (X.C.); (Z.W.); (B.Z.); (J.Q.); (Y.F.); (N.D.)
| | - Yunwen Feng
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Kay Laboratory of Quality Research in Chinese Medicine & Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR 999078, China; (X.C.); (Z.W.); (B.Z.); (J.Q.); (Y.F.); (N.D.)
| | - Ning Ding
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Kay Laboratory of Quality Research in Chinese Medicine & Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR 999078, China; (X.C.); (Z.W.); (B.Z.); (J.Q.); (Y.F.); (N.D.)
| | - Chen Huang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, State Kay Laboratory of Quality Research in Chinese Medicine & Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR 999078, China; (X.C.); (Z.W.); (B.Z.); (J.Q.); (Y.F.); (N.D.)
| |
Collapse
|
16
|
Zou B, Li M, Zhang J, Gao Y, Huo X, Li J, Fan Y, Guo Y, Liu X. Application of a risk score model based on glycosylation-related genes in the prognosis and treatment of patients with low-grade glioma. Front Immunol 2024; 15:1467858. [PMID: 39445005 PMCID: PMC11496118 DOI: 10.3389/fimmu.2024.1467858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 09/20/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction Low-grade gliomas (LGG) represent a heterogeneous and complex group of brain tumors. Despite significant progress in understanding and managing these tumors, there are still many challenges that need to be addressed. Glycosylation, a common post-translational modification of proteins, plays a significant role in tumor transformation. Numerous studies have demonstrated a close relationship between glycosylation modifications and tumor progression. However, the biological function of glycosylation-related genes in LGG remains largely unexplored. Their potential roles within the LGG microenvironment are also not well understood. Methods We collected RNA-seq data and scRNA-seq data from patients with LGG from TCGA and GEO databases. The glycosylation pathway activity scores of each cluster and each patient were calculated by irGSEA and GSVA algorithms, and the differential genes between the high and low glycosylation pathway activity score groups were identified. Prognostic risk profiles of glycosylation-related genes were constructed using univariate Cox and LASSO regression analyses and validated in the CGGA database. Results An 8 genes risk score signature including ASPM, CHI3L1, LILRA4, MSN, OCIAD2, PTGER4, SERPING1 and TNFRSF12A was constructed based on the analysis of glycosylation-related genes. Patients with LGG were divided into high risk and low risk groups according to the median risk score. Significant differences in immunological characteristics, TIDE scores, drug sensitivity, and immunotherapy response were observed between these groups. Additionally, survival analysis of clinical medication information in the TCGA cohort indicated that high risk and low risk groups have different sensitivities to drug therapy. The risk score characteristics can thus guide clinical medication decisions for LGG patients. Conclusion Our study established glycosylation-related gene risk score signatures, providing new perspectives and approaches for prognostic prediction and treatment of LGG.
Collapse
Affiliation(s)
- Binbin Zou
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Mingtai Li
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jiachen Zhang
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yingzhen Gao
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoya Huo
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinhu Li
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yimin Fan
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanlin Guo
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaodong Liu
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
17
|
Zhang L, He J, Zhao W, Zhou Y, Li J, Li S, Zhao W, Zhang L, Tang Z, Tan G, Chen S, Zhang B, Zhang YW, Wang Z. CD2AP promotes the progression of glioblastoma multiforme via TRIM5-mediated NF-kB signaling. Cell Death Dis 2024; 15:722. [PMID: 39353894 PMCID: PMC11445578 DOI: 10.1038/s41419-024-07094-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024]
Abstract
CD2-associated protein (CD2AP) is a scaffolding/adaptive protein that regulates intercellular adhesion and multiple signaling pathways. Although emerging evidence suggests that CD2AP is associated with several malignant tumors, there is no study investigating the expression and biological significance of CD2AP in glioblastoma multiforme (GBM). Here by studying public datasets, we found that CD2AP expression was significantly elevated in GBM and that glioma patients with increased CD2AP expression had a worse prognosis. We also confirmed the increase of CD2AP expression in clinical GBM samples and GBM cell lines. CD2AP overexpression in GBM cells promoted their proliferation, colony formation, migration, and invasion in vitro and their tumorigenesis in vivo, and reduced cell apoptosis both at basal levels and in response to temozolomide. While CD2AP knockdown had the opposite effects. Mechanistically, we revealed that CD2AP interacted with TRIM5, an NF-κB modulator. CD2AP overexpression and knockdown increased and decreased TRIM5 levels as well as the NF-κB activity, respectively. Moreover, downregulation of TRIM5 reversed elevated NF-κB activity in GBM cells with CD2AP overexpression; and inhibition of the NF-κB activity attenuated malignant features of GBM cells with CD2AP overexpression. Our findings demonstrate that CD2AP promotes GBM progression through activating TRIM5-mediated NF-κB signaling and that downregulation of CD2AP can attenuate GBM malignancy, suggesting that CD2AP may become a biomarker and the CD2AP-TRIM5-NF-κB axis may become a therapeutic target for GBM.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Neurosurgery and Department of Neuroscience, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, China
- Xiamen Neurosurgical Quality Control Center, Xiamen, China
| | - Jiawei He
- Department of Neurosurgery and Department of Neuroscience, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wentao Zhao
- Department of Neurosurgery and Department of Neuroscience, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yuhang Zhou
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jin Li
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Shaobo Li
- Department of Neurosurgery, Xiamen Humanity Hospital Fujian Medical University, Xiamen, China
| | - Wenpeng Zhao
- Department of Neurosurgery and Department of Neuroscience, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Lingliang Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Ziqian Tang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Guowei Tan
- Department of Neurosurgery and Department of Neuroscience, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Neurosurgical Quality Control Center, Xiamen, China
| | - Sifang Chen
- Department of Neurosurgery and Department of Neuroscience, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Neurosurgical Quality Control Center, Xiamen, China
| | - Bingchang Zhang
- Department of Neurosurgery and Department of Neuroscience, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yun-Wu Zhang
- Department of Neurosurgery and Department of Neuroscience, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| | - Zhanxiang Wang
- Department of Neurosurgery and Department of Neuroscience, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
- Xiamen Neurosurgical Quality Control Center, Xiamen, China.
| |
Collapse
|
18
|
Yamanaka K, Koma YI, Urakami S, Takahashi R, Nagamata S, Omori M, Torigoe R, Yokoo H, Nakanishi T, Ishihara N, Tsukamoto S, Kodama T, Nishio M, Shigeoka M, Yokozaki H, Terai Y. YKL40/Integrin β4 Axis Induced by the Interaction between Cancer Cells and Tumor-Associated Macrophages Is Involved in the Progression of High-Grade Serous Ovarian Carcinoma. Int J Mol Sci 2024; 25:10598. [PMID: 39408927 PMCID: PMC11477481 DOI: 10.3390/ijms251910598] [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: 09/05/2024] [Revised: 09/24/2024] [Accepted: 09/28/2024] [Indexed: 10/20/2024] Open
Abstract
Macrophages in the tumor microenvironment, termed tumor-associated macrophages (TAMs), promote the progression of various cancer types. However, many mechanisms related to tumor-stromal interactions in epithelial ovarian cancer (EOC) progression remain unclear. High-grade serous ovarian carcinoma (HGSOC) is the most malignant EOC subtype. Herein, immunohistochemistry was performed on 65 HGSOC tissue samples, revealing that patients with a higher infiltration of CD68+, CD163+, and CD204+ macrophages had a poorer prognosis. We subsequently established an indirect co-culture system between macrophages and EOC cells, including HGSOC cells. The co-cultured macrophages showed increased expression of the TAM markers CD163 and CD204, and the co-cultured EOC cells exhibited enhanced proliferation, migration, and invasion. Cytokine array analysis revealed higher YKL40 secretion in the indirect co-culture system. The addition of YKL40 increased proliferation, migration, and invasion via extracellular signal-regulated kinase (Erk) signaling in EOC cells. The knockdown of integrin β4, one of the YKL40 receptors, suppressed YKL40-induced proliferation, migration, and invasion, as well as Erk phosphorylation in some EOC cells. Database analysis showed that high-level expression of YKL40 and integrin β4 correlated with a poor prognosis in patients with serous ovarian carcinoma. Therefore, the YKL40/integrin β4 axis may play a role in ovarian cancer progression.
Collapse
Affiliation(s)
- Keitaro Yamanaka
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Obstetrics and Gynecology, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.T.); (S.N.); (Y.T.)
| | - Yu-ichiro Koma
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Satoshi Urakami
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Gastroenterology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Ryosuke Takahashi
- Division of Obstetrics and Gynecology, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.T.); (S.N.); (Y.T.)
| | - Satoshi Nagamata
- Division of Obstetrics and Gynecology, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.T.); (S.N.); (Y.T.)
| | - Masaki Omori
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Rikuya Torigoe
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Gastrointestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Hiroki Yokoo
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Gastrointestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Takashi Nakanishi
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Gastrointestinal Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Nobuaki Ishihara
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
- Division of Hepato-Biliary-Pancreatic Surgery, Department of Surgery, Kobe University Graduate School of Medicine, Kobe, 650-0017, Japan
| | - Shuichi Tsukamoto
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Takayuki Kodama
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Mari Nishio
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Manabu Shigeoka
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Hiroshi Yokozaki
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (K.Y.); (S.U.); (M.O.); (R.T.); (H.Y.); (T.N.); (N.I.); (S.T.); (T.K.); (M.N.); (M.S.); (H.Y.)
| | - Yoshito Terai
- Division of Obstetrics and Gynecology, Department of Surgery Related, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan; (R.T.); (S.N.); (Y.T.)
| |
Collapse
|
19
|
Chen Y, Zhou Q, Jia Z, Cheng N, Zhang S, Chen W, Wang L. Enhancing cancer immunotherapy: Nanotechnology-mediated immunotherapy overcoming immunosuppression. Acta Pharm Sin B 2024; 14:3834-3854. [PMID: 39309502 PMCID: PMC11413684 DOI: 10.1016/j.apsb.2024.05.032] [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: 03/22/2024] [Revised: 05/12/2024] [Accepted: 05/24/2024] [Indexed: 09/25/2024] Open
Abstract
Immunotherapy is an important cancer treatment method that offers hope for curing cancer patients. While immunotherapy has achieved initial success, a major obstacle to its widespread adoption is the inability to benefit the majority of patients. The success or failure of immunotherapy is closely linked to the tumor's immune microenvironment. Recently, there has been significant attention on strategies to regulate the tumor immune microenvironment in order to stimulate anti-tumor immune responses in cancer immunotherapy. The distinctive physical properties and design flexibility of nanomedicines have been extensively utilized to target immune cells (including tumor-associated macrophages (TAMs), T cells, myeloid-derived suppressor cells (MDSCs), and tumor-associated fibroblasts (TAFs)), offering promising advancements in cancer immunotherapy. In this article, we have reviewed treatment strategies aimed at targeting various immune cells to regulate the tumor immune microenvironment. The focus is on cancer immunotherapy models that are based on nanomedicines, with the goal of inducing or enhancing anti-tumor immune responses to improve immunotherapy. It is worth noting that combining cancer immunotherapy with other treatments, such as chemotherapy, radiotherapy, and photodynamic therapy, can maximize the therapeutic effects. Finally, we have identified the challenges that nanotechnology-mediated immunotherapy needs to overcome in order to design more effective nanosystems.
Collapse
Affiliation(s)
- Yunna Chen
- Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Qianqian Zhou
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Zongfang Jia
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Nuo Cheng
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Sheng Zhang
- Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Weidong Chen
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| | - Lei Wang
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei 230012, China
| |
Collapse
|
20
|
Chen J, Zhang A, Nie A, Zuo X, Zhang L, Jiao Y, Wang L, Yang Y, Liu K, Xue X, Zhuang Y, Meng Y, Yang JH. Multi-omics analysis to reveal the synergistic mechanism underlying the multiple ingredients of Stephania tetrandra extract on rheumatoid arthritis through the PI3K/Akt signaling pathway. Front Pharmacol 2024; 15:1447283. [PMID: 39221139 PMCID: PMC11361992 DOI: 10.3389/fphar.2024.1447283] [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: 06/11/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
Background: Stephania tetrandra has been used for treating rheumatic diseases for thousands of years in rural areas of China. Several studies have found that tetrandrine and fangchinoline can inactivate the PI3K/Akt signaling pathway by reducing the expression and phosphorylation of AKT. However, the mechanism underlying the therapeutic actions of S. tetrandra on RA is not well known. Methods: In this study, we determined the molecular mechanism of the therapeutic effects of the multiple ingredients of S. tetrandra extract (STE) on collagen-induced arthritic (CIA) rats by integrating pharmacometabolomics, proteomics, and PTMomics. Results: In the multi-omics joint analysis, first, the expression signatures of proteins, PTMs, metabolites, and STE ingredients were profiled in CIA rats PBMCs that underwent STE treatment. Bioinformatics analysis were subsequently probed that STE mainly regulated tryptophan metabolism, inflammatory response, and cell adhesion pathways in CIA rats. The interrelated pathways were further constructed, and the findings revealed that STE attenuated the inflammatory response and proliferation of PBMCs in CIA rats by mediating the key targets of the PI3K/Akt pathway, including Hint1, ACP1, FGR, HSP90@157W + dioxidation, and Prkca@220N + 845.4540 Da. The rheumatic functions of Hint1 and ACP1 were further confirmed by applying a transcriptomic data of RA patients who clinically received abatacept therapy. Furthermore, a cross-ome correlation analysis was performed and major in vivo ingredients of STE, including coclaurine-N-glucuronide, Me,coclaurine-O-glc, N-gluA-schefferine, corydamine, corypamine, tetrandrine, and fangchiniline, were found to act on these targerts to inactivate the PI3K/Akt pathway. Conclusion: These results elucidated the molecular mechanism by which the ingredients of STE mediate the expression of the key targets in the PI3K/Akt pathway, leading to anti-rheumatic functions. The findings of this study provided new insights into the synergistic effect of STE against arthritis in rats.
Collapse
Affiliation(s)
- Jinfeng Chen
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - An Zhang
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Anzheng Nie
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoxiao Zuo
- Radiotherapy Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lei Zhang
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Yuxue Jiao
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Lulu Wang
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Yang Yang
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Kun Liu
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Xinli Xue
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| | - Yuanyuan Zhuang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yansha Meng
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jing-Hua Yang
- Research Center for Clinical Systems Biology, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou, Henan, China
| |
Collapse
|
21
|
Fan Y, Meng Y, Hu X, Liu J, Qin X. Uncovering novel mechanisms of chitinase-3-like protein 1 in driving inflammation-associated cancers. Cancer Cell Int 2024; 24:268. [PMID: 39068486 PMCID: PMC11282867 DOI: 10.1186/s12935-024-03425-y] [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: 02/25/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Chitinase-3-like protein 1 (CHI3L1) is a secreted glycoprotein that is induced and regulated by multiple factors during inflammation in enteritis, pneumonia, asthma, arthritis, and other diseases. It is associated with the deterioration of the inflammatory environment in tissues with chronic inflammation caused by microbial infection or autoimmune diseases. The expression of CHI3L1 expression is upregulated in several malignant tumors, underscoring the crucial role of chronic inflammation in the initiation and progression of cancer. While the precise mechanism connecting inflammation and cancer is unclear, the involvement of CHI3L1 is involved in chronic inflammation, suggesting its role as a contributing factor to in the link between inflammation and cancer. CHI3L1 can aggravate DNA oxidative damage, induce the cancerous phenotype, promote the development of a tumor inflammatory environment and angiogenesis, inhibit immune cells, and promote cancer cell growth, invasion, and migration. Furthermore, it participates in the initiation of cancer progression and metastasis by binding with transmembrane receptors to mediate intracellular signal transduction. Based on the current research on CHI3L1, we explore introduce the receptors that interact with CHI3L1 along with the signaling pathways that may be triggered during chronic inflammation to enhance tumorigenesis and progression. In the last section of the article, we provide a brief overview of anti-inflammatory therapies that target CHI3L1.
Collapse
Affiliation(s)
- Yan Fan
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Yuan Meng
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Xingwei Hu
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Jianhua Liu
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China
| | - Xiaosong Qin
- Department of Laboratory Medicine, Liaoning Clinical Research Center for Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110122, China.
- Liaoning Clinical Research Center for Laboratory Medicine, Shenyang, Liaoning Province, China.
| |
Collapse
|
22
|
Su PC, Chen CY, Yu MH, Kuo IY, Yang PS, Hsu CH, Hou YC, Hsieh HT, Chang CP, Shan YS, Wang YC. Fully human chitinase-3 like-1 monoclonal antibody inhibits tumor growth, fibrosis, angiogenesis, and immune cell remodeling in lung, pancreatic, and colorectal cancers. Biomed Pharmacother 2024; 176:116825. [PMID: 38820971 DOI: 10.1016/j.biopha.2024.116825] [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: 03/29/2024] [Revised: 05/23/2024] [Accepted: 05/26/2024] [Indexed: 06/02/2024] Open
Abstract
Considering the limited efficacy of current therapies in lung, colorectal, and pancreatic cancers, innovative combination treatments with diverse mechanisms of action are needed to improve patients' outcomes. Chitinase-3 like-1 protein (CHI3L1) emerges as a versatile factor with significant implications in various diseases, particularly cancers, fostering an immunosuppressive tumor microenvironment for cancer progression. Therefore, pre-clinical validation is imperative to fully realize its potential in cancer treatment. We developed phage display-derived fully human monoclonal CHI3L1 neutralizing antibodies (nAbs) and verified the nAbs-antigen binding affinity and specificity in lung, pancreatic and colorectal cancer cell lines. Tumor growth signals, proliferation and migration ability were all reduced by CHI3L1 nAbs in vitro. Orthotopic or subcutaneous tumor mice model and humanized mouse model were established for characterizing the anti-tumor properties of two CHI3L1 nAb leads. Importantly, CHI3L1 nAbs not only inhibited tumor growth but also mitigated fibrosis, angiogenesis, and restored immunostimulatory functions of immune cells in pancreatic, lung, and colorectal tumor mice models. Mechanistically, CHI3L1 nAbs directly suppressed the activation of pancreatic stellate cells and the transformation of macrophages into myofibroblasts, thereby attenuating fibrosis. These findings strongly support the therapeutic potential of CHI3L1 nAbs in overcoming clinical challenges, including the failure of gemcitabine in pancreatic cancer.
Collapse
Affiliation(s)
- Pei-Chia Su
- Department of Basic Medical Sciences, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan
| | - Ching-Yu Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan
| | - Min-Hua Yu
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan
| | - I-Ying Kuo
- Department of Biotechnology, College of Life Science, Kaohsiung Medical University, No.100, Shih-Chuan 1st Road, Kaohsiung 80708, Taiwan
| | - Pei-Shan Yang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan
| | - Ching-Hsuan Hsu
- AP Biosciences, Inc, No. 508, Sec. 7, Zhongxiao E. Rd, Taipei 115011, Taiwan
| | - Ya-Chin Hou
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, No.138, Sheng-Li Road, Tainan 70403, Taiwan; Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, No.138, Sheng-Li Road, Tainan 70403, Taiwan
| | - Hsin-Ta Hsieh
- AP Biosciences, Inc, No. 508, Sec. 7, Zhongxiao E. Rd, Taipei 115011, Taiwan
| | - Chih-Peng Chang
- Department of Basic Medical Sciences, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan; Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan
| | - Yan-Shen Shan
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, No.138, Sheng-Li Road, Tainan 70403, Taiwan; Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, No.138, Sheng-Li Road, Tainan 70403, Taiwan.
| | - Yi-Ching Wang
- Department of Basic Medical Sciences, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan; Department of Pharmacology, College of Medicine, National Cheng Kung University, No.1, Ta-Hsueh Road, Tainan 70101, Taiwan.
| |
Collapse
|
23
|
Li D, Fan G, Zhou Y. Chitinase 3 like-1 activates the Akt pathway, inducing NF-κB-dependent release of pro-inflammatory cytokines and promoting the proliferative ability in nasopharyngeal carcinoma cells. Cytokine 2024; 179:156631. [PMID: 38710115 DOI: 10.1016/j.cyto.2024.156631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND Chitinase 3 like-1 (CHI3L1) has been reported to function as an oncogene in many types of cancer. However, the biological function of CHI3L1 in nasopharyngeal carcinoma (NPC) remains unknown. METHODS Differentially expressed genes (DEGs) in NPC tissues in GSE64634 and GSE12452 were downloaded from Gene Expression Omnibus (GEO). CHI3L1, interleukin 6 (IL-6), and tumor necrosis factor α (TNF-α) mRNA expression was examined by qRT-PCR. Cell proliferation was evaluated by CCK-8 and EdU incorporation assays. Western blot analysis was used to measure the changes of CHI3L1, nuclear factor-κappaB (NF-κB), and protein kinase B (Akt) pathways. Gene ontology (GO) enrichment and Kyoto Encyclopedia of Gene and Genome (KEGG) pathway analyses were performed using DAVID database. RESULTS We identified 3 overlapping DEGs using Draw Venn diagram, among which CHI3L1 was chosen for the following analyses. CHI3L1 was upregulated in NPC tissues and cells. CHI3L1 silencing suppressed inflammatory response by inactivating the NF-κB pathway and inhibited cell proliferation in NPC cells. On the contrary, CHI3L1 overexpression induced inflammatory response by activating the NF-κB pathway and promoted cell proliferation in NPC cells. According to GO and KEGG analyses, CHI3L1 positive regulates Akt signaling and is enriched in the PI3K-Akt pathway. CHI3L1 knockdown inhibited the Akt pathway, and CHI3L1 overexpression activated the Akt pathway in NPC cells. Akt overexpression abolished the effects of CHI3L1 knockdown on inflammatory response, NF-κB pathway, and proliferation in NPC cells. On the contrary, Akt knockdown abolished the effects of CHI3L1 overexpression on inflammatory response, NF-κB pathway, and proliferation in NPC cells. CONCLUSION CHI3L1 knockdown inhibited NF-κB-dependent inflammatory response and promoting proliferation in NPC cells by inactivating the Akt pathway.
Collapse
Affiliation(s)
- Dajun Li
- Department of ENT, Nanyang First People's Hospital, Nanyang, China.
| | - Gai Fan
- Department of ENT, Nanyang First People's Hospital, Nanyang, China
| | - Yeqi Zhou
- Department of Radiotherapy, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an Second People's Hospital, Huai'an, China
| |
Collapse
|
24
|
Yu H, Wang Z, Zhu B, Jia Z, Luo J, Han X, Chen H, Shao R. A humanized Anti-YKL-40 antibody inhibits tumor development. Biochem Pharmacol 2024; 225:116335. [PMID: 38824968 DOI: 10.1016/j.bcp.2024.116335] [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: 03/12/2024] [Revised: 04/29/2024] [Accepted: 05/30/2024] [Indexed: 06/04/2024]
Abstract
Drugs specifically targeting YKL-40, an over-expressed gene (CHI3L1) in various diseases remain developed. The current study is to create a humanized anti-YKL-40 neutralizing antibody and characterize its potentially therapeutic signature. We utilized in silico CDR-grafting bioinformatics to replace the complementarity determining regions (CDRs) of human IgG1 with mouse CDRs of our previously established anti-YKL-40 antibody (mAY). In fifteen candidates (VL1-3/VH1-5) of heavy and light chain variable region combination, one antibody L3H4 named Rosazumab demonstrated strong binding affinity with YKL-40 (KD = 4.645 × 10-8 M) and high homology with human IgG (80 %). In addition, we established different overlapping amino acid peptides of YKL-40 and found that Rosazumab specifically bound to residues K337, K342, and R344, the KR-rich functional domain of YKL-40. Rosazumab inhibited migration and tube formation of YKL-40-expressing tumor cells and induced tumor cell apoptosis. Mechanistically, Rosazumab induced interaction of N-cadherin with β-catenin and activation of downstream MST1/RASSF1/Histone H2B axis, leading to chromosomal DNA breakage and cell apoptosis. Treatment of xenografted tumor mice with Rosazumab twice a week for 4 weeks inhibited tumor growth and angiogenesis, but induced tumor apoptosis. Rosazumab injected in mice distributed to blood, tumor, and other multiple organs, but did not impact in function or structure of liver and kidney, indicating non-detectable toxicity in vivo. Collectively, the study is the first one to demonstrate that a humanized YKL-40 neutralizing antibody offers a valuable means to block tumor development.
Collapse
Affiliation(s)
- Haihui Yu
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ziyi Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Bowen Zhu
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ziheng Jia
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jing Luo
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiao Han
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hui Chen
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Rong Shao
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
25
|
Wang Y, Zhong F, Xiao F, Li J, Liu X, Ni G, Wang T, Zhang W. Host-defence caerin 1.1 and 1.9 peptides suppress glioblastoma U87 and U118 cell proliferation through the modulation of mitochondrial respiration and induce the downregulation of CHI3L1. PLoS One 2024; 19:e0304149. [PMID: 38848430 PMCID: PMC11161062 DOI: 10.1371/journal.pone.0304149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 05/08/2024] [Indexed: 06/09/2024] Open
Abstract
Glioblastoma, the most aggressive form of brain cancer, poses a significant global health challenge with a considerable mortality rate. With the predicted increase in glioblastoma incidence, there is an urgent need for more effective treatment strategies. In this study, we explore the potential of caerin 1.1 and 1.9, host defence peptides derived from an Australian tree frog, in inhibiting glioblastoma U87 and U118 cell growth. Our findings demonstrate the inhibitory impact of caerin 1.1 and 1.9 on cell growth through CCK8 assays. Additionally, these peptides effectively curtail the migration of glioblastoma cells in a cell scratch assay, exhibiting varying inhibitory effects among different cell lines. Notably, the peptides hinder the G0/S phase replication in both U87 and U118 cells, pointing to their impact on the cell cycle. Furthermore, caerin 1.1 and 1.9 show the ability to enter the cytoplasm of glioblastoma cells, influencing the morphology of mitochondria. Proteomics experiments reveal intriguing insights, with a decrease in CHI3L1 expression and an increase in PZP and JUNB expression after peptide treatment. These proteins play roles in cell energy metabolism and inflammatory response, suggesting a multifaceted impact on glioblastoma cells. In conclusion, our study underscores the substantial anticancer potential of caerin 1.1 and 1.9 against glioblastoma cells. These findings propose the peptides as promising candidates for further exploration in the realm of glioblastoma management, offering new avenues for developing effective treatment strategies.
Collapse
Affiliation(s)
- Yichen Wang
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, China
| | - Furong Zhong
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, China
- Zhongˈao Biomedical Technology (Guangdong) Co., Ltd, Zhongshan, Guangdong, China
| | - Fengyun Xiao
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, China
| | - Junjie Li
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, China
- Zhongˈao Biomedical Technology (Guangdong) Co., Ltd, Zhongshan, Guangdong, China
| | - Xiaosong Liu
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, China
- Zhongˈao Biomedical Technology (Guangdong) Co., Ltd, Zhongshan, Guangdong, China
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Guoying Ni
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, China
- Zhongˈao Biomedical Technology (Guangdong) Co., Ltd, Zhongshan, Guangdong, China
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan, Guangdong, China
| | - Tianfang Wang
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore BC, QLD, Australia
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore BC, QLD, Australia
| | - Wei Zhang
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, China
- Cancer Research Institute, First People’s Hospital of Foshan, Foshan, Guangdong, China
- Guangdong Provincial Engineering and Technology Research Center of Stem Cell Therapy for Pituitary Disease, Guangzhou, China
| |
Collapse
|
26
|
Dankner M, Maritan SM, Priego N, Kruck G, Nkili-Meyong A, Nadaf J, Zhuang R, Annis MG, Zuo D, Nowakowski A, Biondini M, Kiepas A, Mourcos C, Le P, Charron F, Inglebert Y, Savage P, Théret L, Guiot MC, McKinney RA, Muller WJ, Park M, Valiente M, Petrecca K, Siegel PM. Invasive growth of brain metastases is linked to CHI3L1 release from pSTAT3-positive astrocytes. Neuro Oncol 2024; 26:1052-1066. [PMID: 38271182 PMCID: PMC11145453 DOI: 10.1093/neuonc/noae013] [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: 04/19/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Compared to minimally invasive brain metastases (MI BrM), highly invasive (HI) lesions form abundant contacts with cells in the peritumoral brain parenchyma and are associated with poor prognosis. Reactive astrocytes (RAs) labeled by phosphorylated STAT3 (pSTAT3) have recently emerged as a promising therapeutic target for BrM. Here, we explore whether the BrM invasion pattern is influenced by pSTAT3+ RAs and may serve as a predictive biomarker for STAT3 inhibition. METHODS We used immunohistochemistry to identify pSTAT3+ RAs in HI and MI human and patient-derived xenograft (PDX) BrM. Using PDX, syngeneic, and transgenic mouse models of HI and MI BrM, we assessed how pharmacological STAT3 inhibition or RA-specific STAT3 genetic ablation affected BrM growth in vivo. Cancer cell invasion was modeled in vitro using a brain slice-tumor co-culture assay. We performed single-cell RNA sequencing of human BrM and adjacent brain tissue. RESULTS RAs expressing pSTAT3 are situated at the brain-tumor interface and drive BrM invasive growth. HI BrM invasion pattern was associated with delayed growth in the context of STAT3 inhibition or genetic ablation. We demonstrate that pSTAT3+ RAs secrete Chitinase 3-like-1 (CHI3L1), which is a known STAT3 transcriptional target. Furthermore, single-cell RNA sequencing identified CHI3L1-expressing RAs in human HI BrM. STAT3 activation, or recombinant CHI3L1 alone, induced cancer cell invasion into the brain parenchyma using a brain slice-tumor plug co-culture assay. CONCLUSIONS Together, these data reveal that pSTAT3+ RA-derived CHI3L1 is associated with BrM invasion, implicating STAT3 and CHI3L1 as clinically relevant therapeutic targets for the treatment of HI BrM.
Collapse
Affiliation(s)
- Matthew Dankner
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Sarah M Maritan
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Neibla Priego
- Brain Metastasis Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Georgia Kruck
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - Andriniaina Nkili-Meyong
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute-Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - Javad Nadaf
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute-Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - Rebecca Zhuang
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Matthew G Annis
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - Dongmei Zuo
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - Alexander Nowakowski
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Marco Biondini
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - Alexander Kiepas
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Caitlyn Mourcos
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Phuong Le
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute-Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - François Charron
- Department of Pharmacology, McGill University, Montreal, Quebec, Canada
| | - Yanis Inglebert
- Department of Neurosciences, University of Montreal, Montreal, Quebec, Canada
| | - Paul Savage
- Department of Surgery, University of Toronto, Toronto, Ontario, Canada
| | - Louis Théret
- Research Institute of the University of Montreal (IRIC), Montreal, Quebec, Canada
| | - Marie-Christine Guiot
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute-Hospital, McGill University Health Centre, Montreal, Quebec, Canada
- Department of Pathology, McGill University, Montreal, Quebec, Canada
| | - R Anne McKinney
- Department of Pharmacology, McGill University, Montreal, Quebec, Canada
| | - William J Muller
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Morag Park
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Pathology, McGill University, Montreal, Quebec, Canada
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Manuel Valiente
- Brain Metastasis Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Kevin Petrecca
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
- Montreal Neurological Institute-Hospital, McGill University Health Centre, Montreal, Quebec, Canada
| | - Peter M Siegel
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
27
|
Wang K, Zhu W, Huang W, Huang K, Luo H, Long L, Yi B. TRIM Expression in HNSCC: Exploring the Link Between Ubiquitination, Immune Infiltration, and Signaling Pathways Through Bioinformatics. Int J Gen Med 2024; 17:2389-2405. [PMID: 38808201 PMCID: PMC11132118 DOI: 10.2147/ijgm.s463286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024] Open
Abstract
Objective Ubiquitination is an important post-translational modification. However, the significance of the TRIM family of E3 ubiquitin ligases in head and neck squamous cell carcinoma (HNSCC) has not been determined. In this study, the roles of TRIM E3 ubiquitin ligases in lymphovascular invasion in head and neck squamous cell carcinoma (HNSCC) were evaluated. Materials and Methods TRIM expression and related parameters were obtained from UbiBrowser2.0, UALCAN, TIMER, TISIDB, LinkedOmics, STRING, and GeneMANIA databases. Immunohistochemistry was used to confirm their expression. Results TRIM2, TRIM11, TRIM28, and TRIM56 were upregulated in HNSCC with lymphovascular invasion. TRIM expression was strongly associated with immune infiltration, including key treatment targets, like PD-1 and CTL4. Co-expressed genes and possible ubiquitination substrates included tumor-related factors. The TRIMs had predicted roles in ubiquitination-related pathways and vital signaling pathways, eg, MAPK, PI3K-Akt, and JAK-STAT signaling pathways. Conclusion Ubiquitination mediated by four TRIMs might be involved in the regulation of tumor immunity, laying the foundation for future studies of the roles of the TRIM family on the prediction and personalized medicine in HNSCC. The four TRIMs might exert oncogenic effects by promoting lymphovascular invasion in HNSCC.
Collapse
Affiliation(s)
- Kun Wang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
| | - Wei Zhu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
| | - Wei Huang
- Research Center of Carcinogenesis and Targeted Therapy, Xiangya Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
| | - Kangkang Huang
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
| | - Huidan Luo
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
| | - Lu Long
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
| | - Bin Yi
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan Province, People’s Republic of China
| |
Collapse
|
28
|
Rezaee A, Rahmanian P, Nemati A, Sohrabifard F, Karimi F, Elahinia A, Ranjbarpazuki A, Lashkarbolouki R, Dezfulian S, Zandieh MA, Salimimoghadam S, Nabavi N, Rashidi M, Taheriazam A, Hashemi M, Hushmandi K. NF-ĸB axis in diabetic neuropathy, cardiomyopathy and nephropathy: A roadmap from molecular intervention to therapeutic strategies. Heliyon 2024; 10:e29871. [PMID: 38707342 PMCID: PMC11066643 DOI: 10.1016/j.heliyon.2024.e29871] [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: 12/03/2023] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 05/07/2024] Open
Abstract
Diabetes mellitus (DM) is a metabolic illness defined by elevated blood glucose levels, mediating various tissue alterations, including the dysfunction of vital organs. Diabetes mellitus (DM) can lead to many consequences that specifically affect the brain, heart, and kidneys. These issues are known as neuropathy, cardiomyopathy, and nephropathy, respectively. Inflammation is acknowledged as a pivotal biological mechanism that contributes to the development of various diabetes consequences. NF-κB modulates inflammation and the immune system at the cellular level. Its abnormal regulation has been identified in several clinical situations, including cancer, inflammatory bowel illnesses, cardiovascular diseases, and Diabetes Mellitus (DM). The purpose of this review is to evaluate the potential impact of NF-κB on complications associated with DM. Enhanced NF-κB activity promotes inflammation, resulting in cellular harm and compromised organ performance. Phytochemicals, which are therapeutic molecules, can potentially decline the NF-κB level, therefore alleviating inflammation and the progression of problems correlated with DM. More importantly, the regulation of NF-κB can be influenced by various factors, such as TLR4 in DM. Highlighting these factors can facilitate the development of novel therapies in the future.
Collapse
Affiliation(s)
- Aryan Rezaee
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parham Rahmanian
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Amirreza Nemati
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farima Sohrabifard
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Fatemeh Karimi
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Ali Elahinia
- Department of Clinical Science, Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Ali Ranjbarpazuki
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rozhin Lashkarbolouki
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Sadaf Dezfulian
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Afshin Taheriazam
- Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| |
Collapse
|
29
|
Fu H, Wu S, Shen H, Luo K, Huang Z, Lu N, Li Y, Lan Q, Xian Y. Glutamine Metabolism Heterogeneity in Glioblastoma Unveils an Innovative Combination Therapy Strategy. J Mol Neurosci 2024; 74:52. [PMID: 38724832 DOI: 10.1007/s12031-024-02201-x] [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: 08/07/2023] [Accepted: 02/12/2024] [Indexed: 07/20/2024]
Abstract
Treatment of glioblastoma multiforme (GBM) remains challenging. Unraveling the orchestration of glutamine metabolism may provide a novel viewpoint on GBM therapy. The study presented a full and comprehensive comprehending of the glutamine metabolism atlas and heterogeneity in GBM for facilitating the development of a more effective therapeutic choice. Transcriptome data from large GBM cohorts were integrated in this study. A glutamine metabolism-based classification was established through consensus clustering approach, and a classifier by LASSO analysis was defined for differentiating the classification. Prognosis, signaling pathway activity, tumor microenvironment, and responses to immune checkpoint blockade (ICB) and small molecular drugs were characterized in each cluster. A combinational therapy of glutaminase inhibitor CB839 with dihydroartemisinin (DHA) was proposed, and the influence on glutamine metabolism, apoptosis, reactive oxygen species (ROS), and migration was measured in U251 and U373 cells. We discovered that GBM presented heterogeneous glutamine metabolism-based clusters, with unique survival outcomes, activity of signaling pathways, tumor microenvironment, and responses to ICB and small molecular compounds. In addition, the classifier could accurately differentiate the two clusters. Strikingly, the combinational therapy of CB839 with DHA synergistically attenuated glutamine metabolism, triggered apoptosis and ROS accumulation, and impaired migrative capacity in GBM cells, demonstrating the excellent preclinical efficacy. Altogether, our findings unveil the glutamine metabolism heterogeneity in GBM and propose an innovative combination therapy of CB839 with DHA for this malignant disease.
Collapse
Affiliation(s)
- Huangde Fu
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China.
- Department of Neurosurgery, The Third Nanning People's Hospital, Nanning, Guangxi, 530005, China.
| | - Shengtian Wu
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Hechun Shen
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Kai Luo
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Zhongxiang Huang
- Department of Pathology, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Nankun Lu
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Yaolin Li
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Qian Lan
- Department of Laboratory, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| | - Yishun Xian
- Department of Neurosurgery, The Second Nanning People's Hospital, Nanning, Guangxi, 530031, China
| |
Collapse
|
30
|
Fu XP, Ji CY, Tang WQ, Yu TT, Luo L. Long non-coding RNA LOXL1-AS1: a potential biomarker and therapeutic target in human malignant tumors. Clin Exp Med 2024; 24:93. [PMID: 38693424 PMCID: PMC11062969 DOI: 10.1007/s10238-024-01355-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 05/03/2024]
Abstract
Long non-coding RNAs (lncRNAs) are transcripts that contain more than 200 nucleotides. Despite their inability to code proteins, multiple studies have identified their important role in human cancer through different mechanisms. LncRNA lysyl oxidase like 1 antisense RNA 1 (LOXL1-AS1), a newly discovered lncRNA located on human chromosome 15q24.1, has recently been shown to be involved in the occurrence and progression of various malignancies, such as colorectal cancer, gastric cancer, hepatocellular carcinoma, prostate cancer, non-small cell lung cancer, ovarian cancer, cervical cancer, breast cancer, glioma, thymic carcinoma, pancreatic carcinoma. LOXL1-AS1 acts as competitive endogenous RNA (ceRNA) and via sponging various miRNAs, including miR-374b-5p, miR-21, miR-423-5p, miR-589-5p, miR-28-5p, miR-324-3p, miR-708-5p, miR-143-3p, miR-18b-5p, miR-761, miR-525-5p, miR-541-3p, miR-let-7a-5p, miR-3128, miR-3614-5p, miR-377-3p and miR-1224-5p to promote tumor cell proliferation, invasion, migration, apoptosis, cell cycle, and epithelial-mesenchymal transformation (EMT). In addition, LOXL1-AS1 is involved in the regulation of P13K/AKT and MAPK signaling pathways. This article reviews the current understanding of the biological function and clinical significance of LOXL1-AS1 in human cancers. These findings suggest that LOXL1-AS1 may be both a reliable biomarker and a potential therapeutic target for cancers.
Collapse
Affiliation(s)
- Xiao-Ping Fu
- Department of Health Management Center, Hubei Provincial Hospital of Traditional Chinese Medicine, Hongshan District, 856 Luoyu Road, Wuhan, 430070, People's Republic of China
| | - Chun-Yan Ji
- Department of Gastroenterology, Hubei Provincial Hospital of Traditional Chinese and Western Medicine, Wuhan, 430015, People's Republic of China
| | - Wen-Qian Tang
- Department of Health Management Center, Hubei Provincial Hospital of Traditional Chinese Medicine, Hongshan District, 856 Luoyu Road, Wuhan, 430070, People's Republic of China
| | - Ting-Ting Yu
- School of Clinical Medical, Hubei University of Chinese Medicine, Wuhan, 443000, People's Republic of China
| | - Lei Luo
- Department of Health Management Center, Hubei Provincial Hospital of Traditional Chinese Medicine, Hongshan District, 856 Luoyu Road, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
31
|
Tang W, Du J, Li L, Hu S, Ma S, Xue M, Zhu L. Hypoxia-related THBD + macrophages as a prognostic factor in glioma: Construction of a powerful risk model. J Cell Mol Med 2024; 28:e18393. [PMID: 38809929 PMCID: PMC11135907 DOI: 10.1111/jcmm.18393] [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: 01/19/2024] [Revised: 04/10/2024] [Accepted: 04/30/2024] [Indexed: 05/31/2024] Open
Abstract
Glioma is a prevalent malignant tumour characterized by hypoxia as a pivotal factor in its progression. This study aims to investigate the impact of the most severely hypoxic cell subpopulation in glioma. Our findings reveal that the THBD+ macrophage subpopulation is closely associated with hypoxia in glioma, exhibiting significantly higher infiltration in tumours compared to non-tumour tissues. Moreover, a high proportion of THBD+ cells correlates with poor prognosis in glioblastoma (GBM) patients. Notably, THBD+ macrophages exhibit hypoxic characteristics and epithelial-mesenchymal transition features. Silencing THBD expression leads to a notable reduction in the proliferation and metastasis of glioma cells. Furthermore, we developed a THBD+ macrophage-related risk signature (THBDMRS) through machine learning techniques. THBDMRS emerges as an independent prognostic factor for GBM patients with a substantial prognostic impact. By comparing THBDMRS with 119 established prognostic features, we demonstrate the superior prognostic performance of THBDMRS. Additionally, THBDMRS is associated with glioma metastasis and extracellular matrix remodelling. In conclusion, hypoxia-related THBD+ macrophages play a pivotal role in glioma pathogenesis, and THBDMRS emerges as a potent and promising prognostic tool for GBM, contributing to enhanced patient survival outcomes.
Collapse
Affiliation(s)
- Weichun Tang
- Blood Transfusion DepartmentThe Third People's Hospital of BengbuBengbuChina
| | - Juntao Du
- Department of Rehabilitation MedicineThe First Affiliated Hospital of Bengbu Medical CollegeBengbuChina
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
| | - Lin Li
- Department of Rehabilitation MedicineThe First Affiliated Hospital of Bengbu Medical CollegeBengbuChina
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
| | | | - Shuo Ma
- Medical School of Southeast UniversityNanjingChina
| | - Mengtong Xue
- Department of Rehabilitation MedicineThe First Affiliated Hospital of Bengbu Medical CollegeBengbuChina
- Anhui Key Laboratory of Tissue TransplantationBengbu Medical CollegeBengbuChina
| | - Linlin Zhu
- School of Medical TechnologyXinxiang Medical UniversityXinxiangChina
| |
Collapse
|
32
|
Mizoguchi E, Sadanaga T, Nanni L, Wang S, Mizoguchi A. Recently Updated Role of Chitinase 3-like 1 on Various Cell Types as a Major Influencer of Chronic Inflammation. Cells 2024; 13:678. [PMID: 38667293 PMCID: PMC11049018 DOI: 10.3390/cells13080678] [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: 02/27/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Chitinase 3-like 1 (also known as CHI3L1 or YKL-40) is a mammalian chitinase that has no enzymatic activity, but has the ability to bind to chitin, the polymer of N-acetylglucosamine (GlcNAc). Chitin is a component of fungi, crustaceans, arthropods including insects and mites, and parasites, but it is completely absent from mammals, including humans and mice. In general, chitin-containing organisms produce mammalian chitinases, such as CHI3L1, to protect the body from exogenous pathogens as well as hostile environments, and it was thought that it had a similar effect in mammals. However, recent studies have revealed that CHI3L1 plays a pathophysiological role by inducing anti-apoptotic activity in epithelial cells and macrophages. Under chronic inflammatory conditions such as inflammatory bowel disease and chronic obstructive pulmonary disease, many groups already confirmed that the expression of CHI3L1 is significantly induced on the apical side of epithelial cells, and activates many downstream pathways involved in inflammation and carcinogenesis. In this review article, we summarize the expression of CHI3L1 under chronic inflammatory conditions in various disorders and discuss the potential roles of CHI3L1 in those disorders on various cell types.
Collapse
Affiliation(s)
- Emiko Mizoguchi
- Department of Immunology, Kurume University School of Medicine, Kurume 830-0011, Japan; (T.S.); (S.W.); (A.M.)
- Department of Molecular Microbiology and Immunology, Brown University Alpert Medical School, Providence, RI 02912, USA
| | - Takayuki Sadanaga
- Department of Immunology, Kurume University School of Medicine, Kurume 830-0011, Japan; (T.S.); (S.W.); (A.M.)
- Department of Molecular Microbiology and Immunology, Brown University Alpert Medical School, Providence, RI 02912, USA
| | - Linda Nanni
- Catholic University of the Sacred Heart, 00168 Rome, Italy;
| | - Siyuan Wang
- Department of Immunology, Kurume University School of Medicine, Kurume 830-0011, Japan; (T.S.); (S.W.); (A.M.)
| | - Atsushi Mizoguchi
- Department of Immunology, Kurume University School of Medicine, Kurume 830-0011, Japan; (T.S.); (S.W.); (A.M.)
| |
Collapse
|
33
|
曹 越, 高 帅, 罗 刚, 赵 水, 唐 雅, 杜 占, 泮 思. [Role and mechanisms of CHI3L1 in coronary artery lesions in a mouse model of Kawasaki disease-like vasculitis]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:1227-1233. [PMID: 38112139 PMCID: PMC10731963 DOI: 10.7499/j.issn.1008-8830.2309080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 11/09/2023] [Indexed: 12/20/2023]
Abstract
OBJECTIVES To explore the role and potential mechanisms of chitinase-3-like protein 1 (CHI3L1) in coronary artery lesions in a mouse model of Kawasaki disease (KD)-like vasculitis. METHODS Four-week-old male SPF-grade C57BL/6 mice were randomly divided into a control group and a model group, with 10 mice in each group. The model group mice were intraperitoneally injected with 0.5 mL of lactobacillus casei cell wall extract (LCWE) to establish a mouse model of KD-like vasculitis, while the control group mice were injected with an equal volume of normal saline. The general conditions of the mice were observed on the 3rd, 7th, and 14th day after injection. Changes in coronary artery tissue pathology were observed using hematoxylin-eosin staining. The level of CHI3L1 in mouse serum was measured by enzyme-linked immunosorbent assay. Immunofluorescence staining was used to detect the expression and localization of CHI3L1, von Willebrand factor (vWF), and α-smooth muscle actin (α-SMA) in coronary artery tissue. Western blot analysis was used to detect the expression of CHI3L1, vWF, vascular endothelial cadherin (VE cadherin), Caspase-3, B cell lymphoma-2 (Bcl-2), Bcl-2 associated X protein (Bax), nuclear factor κB (NF-κB), and phosphorylated NF-κB (p-NF-κB) in coronary artery tissue. RESULTS The serum level of CHI3L1 in the model group was significantly higher than that in the control group (P<0.05). Compared to the control group, the expression of CHI3L1 in the coronary artery tissue was higher, while the expression of vWF was lower in the model group. The relative expression levels of CHI3L1, Bax, Caspase-3, NF-κB, and p-NF-κB were significantly higher in the model group than in the control group (P<0.05). The relative expression levels of vWF, VE cadherin, and Bcl-2 were lower in the model group than in the control group (P<0.05). CONCLUSIONS In the LCWE-induced mouse model of KD-like vasculitis, the expression levels of CHI3L1 in serum and coronary arteries increase, and it may play a role in coronary artery lesions through endothelial cell apoptosis mediated by inflammatory reactions.
Collapse
|
34
|
Zhao H, Huang M, Jiang L. Potential Roles and Future Perspectives of Chitinase 3-like 1 in Macrophage Polarization and the Development of Diseases. Int J Mol Sci 2023; 24:16149. [PMID: 38003338 PMCID: PMC10671302 DOI: 10.3390/ijms242216149] [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: 10/16/2023] [Revised: 11/03/2023] [Accepted: 11/05/2023] [Indexed: 11/26/2023] Open
Abstract
Chitinase-3-like protein 1 (CHI3L1), a chitinase-like protein family member, is a secreted glycoprotein that mediates macrophage polarization, inflammation, apoptosis, angiogenesis, and carcinogenesis. Abnormal CHI3L1 expression has been associated with multiple metabolic and neurological disorders, including diabetes, atherosclerosis, and Alzheimer's disease. Aberrant CHI3L1 expression is also reportedly associated with tumor migration and metastasis, as well as contributions to immune escape, playing important roles in tumor progression. However, the physiological and pathophysiological roles of CHI3L1 in the development of metabolic and neurodegenerative diseases and cancer remain unclear. Understanding the polarization relationship between CHI3L1 and macrophages is crucial for disease progression. Recent research has uncovered the complex mechanisms of CHI3L1 in different diseases, highlighting its close association with macrophage functional polarization. In this article, we review recent findings regarding the various disease types and summarize the relationship between macrophages and CHI3L1. Furthermore, this article also provides a brief overview of the various mechanisms and inhibitors employed to inhibit CHI3L1 and disrupt its interaction with receptors. These endeavors highlight the pivotal roles of CHI3L1 and suggest therapeutic approaches targeting CHI3L1 in the development of metabolic diseases, neurodegenerative diseases, and cancers.
Collapse
Affiliation(s)
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China;
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou 350116, China;
| |
Collapse
|
35
|
Song M, Zhang G, Shi H, Zhu E, Deng L, Shen H. Serum YKL-40 in coronary heart disease: linkage with inflammatory cytokines, artery stenosis, and optimal cut-off value for estimating major adverse cardiovascular events. Front Cardiovasc Med 2023; 10:1242339. [PMID: 38028459 PMCID: PMC10644235 DOI: 10.3389/fcvm.2023.1242339] [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: 06/19/2023] [Accepted: 09/22/2023] [Indexed: 12/01/2023] Open
Abstract
Objective YKL-40, previously known as chitinase-3-like protein 1 (CHI3L1), is an inflammation-related glycoprotein that promotes atherosclerosis, but its application and optimal cut-off value as a prognostic biomarker in coronary heart disease (CHD) require more clinical evidence. Thus, this prospective study aimed to evaluate the linkage of serum YKL-40 with disease features, inflammatory cytokines, and major adverse cardiovascular events (MACEs) in CHD patients. Methods A total of 410 CHD patients were enrolled for serum YKL-40 determination via enzyme-linked immunosorbent assay. Meanwhile, serum YKL-40 levels in 100 healthy controls (HCs) were also quantified. Results YKL-40 level was higher in CHD patients compared with that in HCs (P < 0.001). YKL-40 was positively linked with hyperlipidemia (P = 0.014), diabetes mellitus (P = 0.001), fasting blood glucose (P = 0.045), C-reactive protein (P < 0.001), the Gensini score (P < 0.001), and stenosis degree (graded by the Gensini score) (P < 0.001) in CHD patients. In addition, an elevated YKL-40 level was associated with increased levels of tumor necrosis factor alpha (P = 0.001), interleukin (IL)-1β (P = 0.001), IL-6 (P < 0.001), and IL-17A (P = 0.002) in CHD patients. The 1-/2-/3-year cumulative MACE rates of CHD patients were 5.5%, 14.4%, and 25.0%, respectively. Regarding the prognostic capability, YKL-40 ≥100 ng/ml (the median cut-off value) (P = 0.003) and YKL-40 ≥150 ng/ml (the third interquartile cut-off value) (P = 0.021) reflected an elevated accumulating MACE rate, whereas accumulating MACE was not different between CHD patients with YKL-40 ≥80 and <80 ng/ml (the first interquartile cut-off value) (P = 0.083). Conclusion Serum YKL-40 is positively linked with inflammatory cytokines and the Gensini score, whose high expression cut-off by 100 and 150 ng/ml estimates a higher MACE risk in CHD patients.
Collapse
Affiliation(s)
- Mowei Song
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Guofu Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Hang Shi
- Department of Cardiovascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Erjun Zhu
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Li Deng
- Department of Extracorporeal Life Support, The People’s Hospital of Gaozhou, Gaozhou, China
| | - Hongtao Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| |
Collapse
|
36
|
Lin P, He L, Tian N, Qi X. The evaluation of six genes combined value in glioma diagnosis and prognosis. J Cancer Res Clin Oncol 2023; 149:12413-12433. [PMID: 37439825 DOI: 10.1007/s00432-023-05082-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 06/29/2023] [Indexed: 07/14/2023]
Abstract
PURPOSE Glioma is the most common and fatal type of brain tumour. Owing to its aggressiveness and lethality, early diagnosis and prediction of patient survival are very important. This study aimed to identify key genes and biomarkers for glioma that can guide clinicians in making rapid diagnosis and prognostication. METHODS Data mining of The Cancer Genome Atlas (TCGA), Chinese Glioma Genome Atlas (CGGA), Repository of Molecular Brain Neoplasia Data, and Genotype-Tissue Expression Project brain expression data revealed significantly differentially expressed genes (DEGs), and the risk scores of individual patients were calculated. WGCNA was utilized to screen for genes most related to clinical diagnosis. Prognostic genes associated with glioma were selected via combining the LASSO regression with univariate and multivariate Cox regression and protein-protein interaction network analyses. Then, a nomogram was constructed. And CGGA dataset was utilized to validated. The protein expression levels of the signature were detected using the human protein atlas. Drug response prediction was carried out using the package "pRRophetic". RESULTS A six-gene signature (KLF6, CHI3L1, SERPINE1, ANGPT2, TGFBR1, and PTX3) was identified and used to stratify patients into low- and high-risk groups. Survival, ROC curve, and Cox analyses clarified that the six hub genes were a favourable independent prognostic factor for patients with glioma. A nomogram was set up by integrating clinical parameters with risk signatures, showing high precision for predicting 2-, 3-, 4-, 5-years survival. In addition, the expression of most genes was consistent with protein expression. Furthermore, the sensitivity to the top ten drugs in the GDSC database of the high-risk group was significantly higher than the low-risk group. CONCLUSION Based on genetic profiles and clinicopathological features, including age, grade, isocitrate dehydrogenase mutation status, we constructed a comprehensive prognostic model for patients with glioma. These signatures can be regarded as biomarkers to predict the prognosis of gliomas, possibly providing more therapeutic strategies for future clinical research.
Collapse
Affiliation(s)
- Ping Lin
- Department of Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Lingyan He
- Department of Traditional Chinese Medicine, Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Nan Tian
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Xuchen Qi
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing, Zhejiang, China.
| |
Collapse
|
37
|
Ren J, Xu B, Ren J, Liu Z, Cai L, Zhang X, Wang W, Li S, Jin L, Ding L. The Importance of M1-and M2-Polarized Macrophages in Glioma and as Potential Treatment Targets. Brain Sci 2023; 13:1269. [PMID: 37759870 PMCID: PMC10526262 DOI: 10.3390/brainsci13091269] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Glioma is the most common and malignant tumor of the central nervous system. Glioblastoma (GBM) is the most aggressive glioma, with a poor prognosis and no effective treatment because of its high invasiveness, metabolic rate, and heterogeneity. The tumor microenvironment (TME) contains many tumor-associated macrophages (TAMs), which play a critical role in tumor proliferation, invasion, metastasis, and angiogenesis and indirectly promote an immunosuppressive microenvironment. TAM is divided into tumor-suppressive M1-like (classic activation of macrophages) and tumor-supportive M2-like (alternatively activated macrophages) polarized cells. TAMs exhibit an M1-like phenotype in the initial stages of tumor progression, and along with the promotion of lysing tumors and the functions of T cells and NK cells, tumor growth is suppressed, and they rapidly transform into M2-like polarized macrophages, which promote tumor progression. In this review, we discuss the mechanism by which M1- and M2-polarized macrophages promote or inhibit the growth of glioblastoma and indicate the future directions for treatment.
Collapse
Affiliation(s)
- Jiangbin Ren
- Department of neurosurgery, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Nanjing Medical University, Huai’an 223000, China; (J.R.); (B.X.); (Z.L.); (L.C.); (X.Z.); (W.W.); (S.L.); (L.J.)
| | - Bangjie Xu
- Department of neurosurgery, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Nanjing Medical University, Huai’an 223000, China; (J.R.); (B.X.); (Z.L.); (L.C.); (X.Z.); (W.W.); (S.L.); (L.J.)
| | - Jianghao Ren
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai 200030, China;
| | - Zhichao Liu
- Department of neurosurgery, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Nanjing Medical University, Huai’an 223000, China; (J.R.); (B.X.); (Z.L.); (L.C.); (X.Z.); (W.W.); (S.L.); (L.J.)
| | - Lingyu Cai
- Department of neurosurgery, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Nanjing Medical University, Huai’an 223000, China; (J.R.); (B.X.); (Z.L.); (L.C.); (X.Z.); (W.W.); (S.L.); (L.J.)
| | - Xiaotian Zhang
- Department of neurosurgery, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Nanjing Medical University, Huai’an 223000, China; (J.R.); (B.X.); (Z.L.); (L.C.); (X.Z.); (W.W.); (S.L.); (L.J.)
| | - Weijie Wang
- Department of neurosurgery, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Nanjing Medical University, Huai’an 223000, China; (J.R.); (B.X.); (Z.L.); (L.C.); (X.Z.); (W.W.); (S.L.); (L.J.)
| | - Shaoxun Li
- Department of neurosurgery, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Nanjing Medical University, Huai’an 223000, China; (J.R.); (B.X.); (Z.L.); (L.C.); (X.Z.); (W.W.); (S.L.); (L.J.)
| | - Luhao Jin
- Department of neurosurgery, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Nanjing Medical University, Huai’an 223000, China; (J.R.); (B.X.); (Z.L.); (L.C.); (X.Z.); (W.W.); (S.L.); (L.J.)
| | - Lianshu Ding
- Department of neurosurgery, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Nanjing Medical University, Huai’an 223000, China; (J.R.); (B.X.); (Z.L.); (L.C.); (X.Z.); (W.W.); (S.L.); (L.J.)
| |
Collapse
|
38
|
Zhou H, Chen B, Zhang L, Li C. Machine learning-based identification of lower grade glioma stemness subtypes discriminates patient prognosis and drug response. Comput Struct Biotechnol J 2023; 21:3827-3840. [PMID: 37560125 PMCID: PMC10407594 DOI: 10.1016/j.csbj.2023.07.029] [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: 03/06/2023] [Revised: 07/06/2023] [Accepted: 07/19/2023] [Indexed: 08/11/2023] Open
Abstract
Glioma stem cells (GSCs) remodel their tumor microenvironment to sustain a supportive niche. Identification and stratification of stemness related characteristics in patients with glioma might aid in the diagnosis and treatment of the disease. In this study, we calculated the mRNA stemness index in bulk and single-cell RNA-sequencing datasets using machine learning methods and investigated the correlation between stemness and clinicopathological characteristics. A glioma stemness-associated score (GSScore) was constructed using multivariate Cox regression analysis. We also generated a GSC cell line derived from a patient diagnosed with glioma and used glioma cell lines to validate the performance of the GSScore in predicting chemotherapeutic responses. Differentially expressed genes (DEGs) between GSCs with high and low GSScores were used to cluster lower-grade glioma (LGG) samples into three stemness subtypes. Differences in clinicopathological characteristics, including survival, copy number variations, mutations, tumor microenvironment, and immune and chemotherapeutic responses, among the three LGG stemness-associated subtypes were identified. Using machine learning methods, we further identified genes as subtype predictors and validated their performance using the CGGA datasets. In the current study, we identified a GSScore that correlated with LGG chemotherapeutic response. Through the score, we also identified a novel classification of the LGG subtype and associated subtype predictors, which might facilitate the development of precision therapy.
Collapse
Affiliation(s)
- Hongshu Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- Hypothalamic-pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Bo Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- Hypothalamic-pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- Department of Surgery, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong SAR, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- Hypothalamic-pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- Clinical Diagnosis and Therapy Center for Glioma, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| | - Chuntao Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- Hypothalamic-pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- Clinical Diagnosis and Therapy Center for Glioma, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, PR China
| |
Collapse
|
39
|
Baeza-Kallee N, Bergès R, Hein V, Cabaret S, Garcia J, Gros A, Tabouret E, Tchoghandjian A, Colin C, Figarella-Branger D. Deciphering the Action of Neuraminidase in Glioblastoma Models. Int J Mol Sci 2023; 24:11645. [PMID: 37511403 PMCID: PMC10380381 DOI: 10.3390/ijms241411645] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/11/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Glioblastoma (GBM) contains cancer stem cells (CSC) that are resistant to treatment. GBM CSC expresses glycolipids recognized by the A2B5 antibody. A2B5, induced by the enzyme ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyl transferase 3 (ST8Sia3), plays a crucial role in the proliferation, migration, clonogenicity and tumorigenesis of GBM CSC. Our aim was to characterize the resulting effects of neuraminidase that removes A2B5 in order to target GBM CSC. To this end, we set up a GBM organotypic slice model; quantified A2B5 expression by flow cytometry in U87-MG, U87-ST8Sia3 and GBM CSC lines, treated or not by neuraminidase; performed RNAseq and DNA methylation profiling; and analyzed the ganglioside expression by liquid chromatography-mass spectrometry in these cell lines, treated or not with neuraminidase. Results demonstrated that neuraminidase decreased A2B5 expression, tumor size and regrowth after surgical removal in the organotypic slice model but did not induce a distinct transcriptomic or epigenetic signature in GBM CSC lines. RNAseq analysis revealed that OLIG2, CHI3L1, TIMP3, TNFAIP2, and TNFAIP6 transcripts were significantly overexpressed in U87-ST8Sia3 compared to U87-MG. RT-qPCR confirmed these results and demonstrated that neuraminidase decreased gene expression in GBM CSC lines. Moreover, neuraminidase drastically reduced ganglioside expression in GBM CSC lines. Neuraminidase, by its pleiotropic action, is an attractive local treatment against GBM.
Collapse
Affiliation(s)
| | - Raphaël Bergès
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
| | - Victoria Hein
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
| | - Stéphanie Cabaret
- ChemoSens Platform, Centre des Sciences du Goût et de l'Alimentation, InstitutAgro, CNRS, INRAE, Université de Bourgogne Franche-Comté, 21000 Dijon, France
| | - Jeremy Garcia
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, 13005 Marseille, France
| | - Abigaëlle Gros
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, 13005 Marseille, France
| | - Emeline Tabouret
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
- APHM, CHU Timone, Service de Neurooncologie, 13005 Marseille, France
| | | | - Carole Colin
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
| | | |
Collapse
|
40
|
Shi P, Xu J, Cui H. The Recent Research Progress of NF-κB Signaling on the Proliferation, Migration, Invasion, Immune Escape and Drug Resistance of Glioblastoma. Int J Mol Sci 2023; 24:10337. [PMID: 37373484 PMCID: PMC10298967 DOI: 10.3390/ijms241210337] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/09/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and invasive primary central nervous system tumor in humans, accounting for approximately 45-50% of all primary brain tumors. How to conduct early diagnosis, targeted intervention, and prognostic evaluation of GBM, in order to improve the survival rate of glioblastoma patients, has always been an urgent clinical problem to be solved. Therefore, a deeper understanding of the molecular mechanisms underlying the occurrence and development of GBM is also needed. Like many other cancers, NF-κB signaling plays a crucial role in tumor growth and therapeutic resistance in GBM. However, the molecular mechanism underlying the high activity of NF-κB in GBM remains to be elucidated. This review aims to identify and summarize the NF-κB signaling involved in the recent pathogenesis of GBM, as well as basic therapy for GBM via NF-κB signaling.
Collapse
Affiliation(s)
- Pengfei Shi
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China; (P.S.); (J.X.)
- Jinfeng Laboratory, Chongqing 401329, China
| | - Jie Xu
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China; (P.S.); (J.X.)
- Jinfeng Laboratory, Chongqing 401329, China
| | - Hongjuan Cui
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China; (P.S.); (J.X.)
- Jinfeng Laboratory, Chongqing 401329, China
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
| |
Collapse
|
41
|
Wang Z, Wang S, Jia Z, Hu Y, Cao D, Yang M, Liu L, Gao L, Qiu S, Yan W, Li Y, Luo J, Geng Y, Zhang J, Li Z, Wang X, Li M, Shao R, Liu Y. YKL-40 derived from infiltrating macrophages cooperates with GDF15 to establish an immune suppressive microenvironment in gallbladder cancer. Cancer Lett 2023; 563:216184. [PMID: 37088328 DOI: 10.1016/j.canlet.2023.216184] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 04/06/2023] [Accepted: 04/11/2023] [Indexed: 04/25/2023]
Abstract
Despite of the high lethality of gallbladder cancer (GBC), little is known regarding molecular regulation of the tumor immunosuppressive microenvironment. Here, we determined tumor expression levels of YKL-40 and the molecular mechanisms by which YKL-40 regulates escape of anti-tumor immune surveillance. We found that elevated expression levels of YKL-40 in plasma and tissue were correlated with tumor size, stage IV and lymph node metastasis. Single cell transcriptome analysis revealed that YKL-40 was predominantly derived from M2-like subtype of infiltrating macrophages. Blockade of M2-like macrophage differentiation of THP-1 cells with YKL-40 shRNA resulted in reprogramming to M1-like macrophages and restricting tumor development. YKL-40 induced tumor cell expression and secretion of growth differentiation factor 15 (GDF15), thus coordinating to promote PD-L1 expression mediated by PI3K, AKT and/or Erk activation. Interestingly, extracellular GDF15 inhibited intracellular expression of GDF15 that suppressed PD-L1 expression. Thus, YKL-40 disrupted the balance of pro- and anti-PD-L1 regulation to enhance expression of PD-L1 and inhibition of T cell cytotoxicity, leading to tumor immune evasion. The data suggest that YKL-40 and GDF15 could serve as diagnostic biomarkers and immunotherapeutic targets for GBC.
Collapse
Affiliation(s)
- Ziyi Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Shijia Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Ziheng Jia
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Yunping Hu
- Institute of Pathology and Southwest Cancer Center, The First Affiliated Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dongyan Cao
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Mingjie Yang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Liguo Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Li Gao
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shimei Qiu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Weikang Yan
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Yiming Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Jing Luo
- Department of Biliary Tract Surgery I, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Yajun Geng
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Jingyun Zhang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China
| | - Zhizhen Li
- Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuan Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Maolan Li
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Rong Shao
- Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China; Department of Pharmacology and Biochemistry, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai key lab for gallbladder - related gastroenterological diseases, Xinhua hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yingbin Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Cancer Institute, State Key Laboratory of Oncogenes and Related Genes, Shanghai, China.
| |
Collapse
|
42
|
Anderson G. Amyotrophic Lateral Sclerosis Pathoetiology and Pathophysiology: Roles of Astrocytes, Gut Microbiome, and Muscle Interactions via the Mitochondrial Melatonergic Pathway, with Disruption by Glyphosate-Based Herbicides. Int J Mol Sci 2022; 24:ijms24010587. [PMID: 36614029 PMCID: PMC9820185 DOI: 10.3390/ijms24010587] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/23/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
The pathoetiology and pathophysiology of motor neuron loss in amyotrophic lateral sclerosis (ALS) are still to be determined, with only a small percentage of ALS patients having a known genetic risk factor. The article looks to integrate wider bodies of data on the biological underpinnings of ALS, highlighting the integrative role of alterations in the mitochondrial melatonergic pathways and systemic factors regulating this pathway across a number of crucial hubs in ALS pathophysiology, namely glia, gut, and the muscle/neuromuscular junction. It is proposed that suppression of the mitochondrial melatonergic pathway underpins changes in muscle brain-derived neurotrophic factor, and its melatonergic pathway mimic, N-acetylserotonin, leading to a lack of metabolic trophic support at the neuromuscular junction. The attenuation of the melatonergic pathway in astrocytes prevents activation of toll-like receptor agonists-induced pro-inflammatory transcription factors, NF-kB, and yin yang 1, from having a built-in limitation on inflammatory induction that arises from their synchronized induction of melatonin release. Such maintained astrocyte activation, coupled with heightened microglia reactivity, is an important driver of motor neuron susceptibility in ALS. Two important systemic factors, gut dysbiosis/permeability and pineal melatonin mediate many of their beneficial effects via their capacity to upregulate the mitochondrial melatonergic pathway in central and systemic cells. The mitochondrial melatonergic pathway may be seen as a core aspect of cellular function, with its suppression increasing reactive oxygen species (ROS), leading to ROS-induced microRNAs, thereby altering the patterning of genes induced. It is proposed that the increased occupational risk of ALS in farmers, gardeners, and sportsmen and women is intimately linked to exposure, whilst being physically active, to the widely used glyphosate-based herbicides. This has numerous research and treatment implications.
Collapse
Affiliation(s)
- George Anderson
- CRC Scotland & London, Eccleston Square, London SW1V 1PG, UK
| |
Collapse
|