1
|
Xiang Z, Yu S, Xu Y, Xiong H, Hu D, Li Q, Wu Z. CH25H Promotes Autophagy and Regulates the Malignant Progression of Laryngeal Squamous Cell Carcinoma Through the PI3K-AKT Pathway. Cancer Med 2024; 13:e70312. [PMID: 39428922 PMCID: PMC11491687 DOI: 10.1002/cam4.70312] [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: 01/09/2024] [Revised: 09/24/2024] [Accepted: 09/29/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND Laryngeal squamous cell carcinoma (LSCC) is a type of cancer of the respiratory tract that often presents with subtle symptoms at the early stage and is susceptible to recurrence and metastasis. MATERIALS AND METHODS To find out key regulatory genes involved in LSCC development, we downloaded LSCC-related sequencing datasets for bioinformatics analysis. WGCNA was performed on GSE142083 and differential analysis was conducted on GSE51985 and TCGA-HNSC. Intersection genes were taken from the above three datasets. To confirm the function of genes, we overexpressed and knocked down genes in cells and treated them with autophagy agonist Rapamycin and PI3K-AKT pathway inhibitor. At the cellular level, the expression of CH25H, autophagy-related proteins (LC3 I, LC3 II, p62, and Beclin 1), and PI3K-AKT pathway-related proteins (PI3K, AKT, and p-AKT) were assessed via Western blot; the mRNA level of CH25H was evaluated through qRT-PCR; the cell activity was examined by CCK8; the apoptosis was assessed through flow cytometry; and the cell migration and invasion were assessed through wound healing and Transwell assays. RESULTS Through bioinformatics analysis, we screened 7 genes (CH25H, NELL2, STC2, TMEM158, ZIC2, HOXD11, and HOXD10). Ultimately, CH25H was selected for follow-up experiments. By detecting CH25H expression in human immortalized keratinocytes (HaCaT) and LSCC cells (Tu-686, SNU899, and AMC-HN-8), it was found out that CH25H expression was higher in HaCaT cells than in LSCC cells. To elucidate the role of CH25H in LSCC development, we overexpressed CH25H in Tu-686 cells and downregulated its expression in AMC-HN-8 cells. CH25H was revealed to reduce the proliferation, activity, invasion, and migration of LSCC cells while increasing their apoptosis levels. Significant changes were also observed in the expressions of autophagy- and PI3K-AKT pathway-related proteins. To further investigate the roles of autophagy and the PI3K-AKT pathway in LSCC development, we respectively employed autophagy agonists and inhibitors targeting the PI3K-AKT pathway to intervene the cells, and found that CH25H regulated the PI3K-AKT pathway to promote autophagy, thus enhancing the apoptosis of LSCC cells. We further investigated CH25H's impact on tumor growth, autophagy, and the PI3K-AKT pathway at the animal level and found that CH25H promoted autophagy of LSCC cells and inhibited the PI3K-AKT pathway, and ultimately inhibiting the progression of LSCC. CONCLUSIONS In summary, CH25H promotes autophagy and affects the malignant progression of LSCC through the PI3K-AKT pathway.
Collapse
Affiliation(s)
- Zhenfei Xiang
- Department of Radiation OncologyNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| | - Senquan Yu
- Department of OncologyThe Second Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Yuxin Xu
- Department of Otolaryngology, Head and Neck SurgeryNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| | - Huacai Xiong
- Department of Radiation OncologyNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| | - Danfei Hu
- Department of Radiation OncologyNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| | - Qun Li
- Department of Otolaryngology, Head and Neck SurgeryNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| | - Zhenhua Wu
- Department of Otolaryngology, Head and Neck SurgeryNingbo Medical Center Lihuili Hospital, Ningbo UniversityNingboZhejiangChina
| |
Collapse
|
2
|
Ye Y, Wan L, Hu J, Li X, Zhang K. Combined single-cell RNA sequencing and mendelian randomization to identify biomarkers associated with necrotic apoptosis in intervertebral disc degeneration. Spine J 2024:S1529-9430(24)01030-1. [PMID: 39332686 DOI: 10.1016/j.spinee.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 09/02/2024] [Accepted: 09/14/2024] [Indexed: 09/29/2024]
Abstract
BACKGROUND Intervertebral disc degeneration (IDD) is associated with back pain; back pain is a world-wide contributor to poor quality of life, while necroptosis has the characteristics of necroptosis and apoptosis, however, its role in IDD is still unclear. Therefore, the aim of this study was to identify biomarkers associated with necroptosis in IDD. PURPOSE To explore biomarkers associated with necroptosis in IDD, reveal the pathogenesis of IDD, as well as provide new directions for the diagnosis and treatment of this disease. STUDY DESIGN/SETTINGS Retrospective cohort study. Our study employs scRNA-seq coupled with MR analysis to investigate the causal relationship between necroptosis and IDD, laying a foundational groundwork for unveiling the intricate pathogenic mechanisms of this condition. METHODS Data quality control and normalisation was executed in single-cell dataset, GSE205535. Then, different cell types were obtained by cell annotation through marker genes. Subsequently, chi-square test was employed to assess the distribution difference of different cell types between IDD and control to screen key cells. AUCell was applied to calculate necroptosis-related genes (NRGs) scores of all cell types, further key cells were divided into high and low NRGs groups according to the median AUC scores of different cell types. Afterwards, the differentially expressed genes (DEGs) within the 2 score groups were screened. Then, the genes that had causal relationship with IDD were selected as biomarkers by univariate and multivariate Mendelian randomization (MR) analysis. Finally, the expression of biomarkers in different cell types and pseudo-time analysis was analyzed separately. RESULTS In GSE205535, 16 different cell populations identified by UMAP cluster analysis were further annotated to 8 cell types using maker genes. Afterwards, 53 DEGs were screened between the high and low NRGs groups. In addition, 9 genes with causal relationship with IDD were obtained by univariate MR analysis, further multivariate MR analysis proved that NT5E and TMEM158 had a direct causal relationship with IDD, which were used as biomarkers in this study. This study not only found that the expression levels of NT5E and TMEM158 were higher in IDD group, but also found that fibrochondrocytes and inflammatory chondrocytes were the key cells of NT5E and TMEM158, respectively. In the end, the biomarkers had the same expression trend in the quasi-time series, and both of them from high to low and then increased. CONCLUSION NT5E and TMEM158, as biomarkers of necroptotic apoptotic IDD, were causally associated with IDD. CLINICAL SIGNIFICANCE The understanding of chondrocytes as key cells provides new perspectives for deeper elucidation of the pathogenesis of IDD, improved diagnostic methods, and the development of more effective treatments. These findings are expected to provide a more accurate and personalised approach to clinical diagnosis and treatment, thereby improving the prognosis and quality of life of patients with IDD.
Collapse
Affiliation(s)
- Yi Ye
- Orthopaedic Department, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 32# W. Sec 2, 1st Ring Rd. Sichuan, 610072, China
| | - Lun Wan
- Orthopaedic Department, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 32# W. Sec 2, 1st Ring Rd. Sichuan, 610072, China
| | - Jiang Hu
- Orthopaedic Department, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 32# W. Sec 2, 1st Ring Rd. Sichuan, 610072, China
| | - Xiaoxue Li
- Orthopaedic Department, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 32# W. Sec 2, 1st Ring Rd. Sichuan, 610072, China
| | - Kun Zhang
- Orthopaedic Department, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, 32# W. Sec 2, 1st Ring Rd. Sichuan, 610072, China.
| |
Collapse
|
3
|
Quach H, Farrell S, Wu MJM, Kanagarajah K, Leung JWH, Xu X, Kallurkar P, Turinsky AL, Bear CE, Ratjen F, Kalish B, Goyal S, Moraes TJ, Wong AP. Early human fetal lung atlas reveals the temporal dynamics of epithelial cell plasticity. Nat Commun 2024; 15:5898. [PMID: 39003323 PMCID: PMC11246468 DOI: 10.1038/s41467-024-50281-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 07/05/2024] [Indexed: 07/15/2024] Open
Abstract
Studying human fetal lungs can inform how developmental defects and disease states alter the function of the lungs. Here, we sequenced >150,000 single cells from 19 healthy human pseudoglandular fetal lung tissues ranging between gestational weeks 10-19. We capture dynamic developmental trajectories from progenitor cells that express abundant levels of the cystic fibrosis conductance transmembrane regulator (CFTR). These cells give rise to multiple specialized epithelial cell types. Combined with spatial transcriptomics, we show temporal regulation of key signalling pathways that may drive the temporal and spatial emergence of specialized epithelial cells including ciliated and pulmonary neuroendocrine cells. Finally, we show that human pluripotent stem cell-derived fetal lung models contain CFTR-expressing progenitor cells that capture similar lineage developmental trajectories as identified in the native tissue. Overall, this study provides a comprehensive single-cell atlas of the developing human lung, outlining the temporal and spatial complexities of cell lineage development and benchmarks fetal lung cultures from human pluripotent stem cell differentiations to similar developmental window.
Collapse
Affiliation(s)
- Henry Quach
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Spencer Farrell
- Department of Physics, University of Toronto, Toronto, Ontario, Canada
| | - Ming Jia Michael Wu
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kayshani Kanagarajah
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Joseph Wai-Hin Leung
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Xiaoqiao Xu
- Centre for Computational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Prajkta Kallurkar
- Centre for Computational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Andrei L Turinsky
- Centre for Computational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Christine E Bear
- Program in Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Felix Ratjen
- Program in Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Brian Kalish
- Program in Neurosciences and Mental Health, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
- Division of Neonatology, Department of Paediatrics, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sidhartha Goyal
- Department of Physics, University of Toronto, Toronto, Ontario, Canada
| | - Theo J Moraes
- Program in Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Amy P Wong
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Ontario, Canada.
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
4
|
Wang XP, Guo W, Chen YF, Hong C, Ji J, Zhang XY, Dong YF, Sun XL. PD-1/PD-L1 axis is involved in the interaction between microglial polarization and glioma. Int Immunopharmacol 2024; 133:112074. [PMID: 38615383 DOI: 10.1016/j.intimp.2024.112074] [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: 02/28/2024] [Revised: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
The tumor microenvironment plays a vital role in glioblastoma growth and invasion. PD-1 and PD-L1 modulate the immunity in the brain tumor microenvironment. However, the underlying mechanisms remain unclear. In the present study, in vivo and in vitro experiments were conducted to reveal the effects of PD-1/PD-L1 on the crosstalk between microglia and glioma. Results showed that glioma cells secreted PD-L1 to the peritumoral areas, particularly microglia containing highly expressed PD-1. In the early stages of glioma, microglia mainly polarized into the pro-inflammatory subtype (M1). Subsequently, the secreted PD-L1 accumulated and bound to PD-1 on microglia, facilitating their polarization toward the microglial anti-inflammatory (M2) subtype primarily via the STAT3 signaling pathway. The role of PD-1/PD-L1 in M2 polarization of microglia was partially due to PD-1/PD-L1 depletion or application of BMS-1166, a novel inhibitor of PD-1/PD-L1. Consistently, co-culturing with microglia promoted glioma cell growth and invasion, and blocking PD-1/PD-L1 significantly suppressed these processes. Our findings reveal that the PD-1/PD-L1 axis engages in the microglial M2 polarization in the glioma microenvironment and promotes tumor growth and invasion.
Collapse
Affiliation(s)
- Xi-Peng Wang
- Nanjing University of Chinese Medicine, Nanjing, China; Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Wei Guo
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Ye-Fan Chen
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Chen Hong
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Juan Ji
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Xi-Yue Zhang
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Yin-Feng Dong
- Nanjing University of Chinese Medicine, Nanjing, China.
| | - Xiu-Lan Sun
- Nanjing University of Chinese Medicine, Nanjing, China; Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
5
|
Zhang C, Du FH, Wang RX, Han WB, Lv X, Zeng LH, Chen GQ. TSPAN6 reinforces the malignant progression of glioblastoma via interacting with CDK5RAP3 and regulating STAT3 signaling pathway. Int J Biol Sci 2024; 20:2440-2453. [PMID: 38725860 PMCID: PMC11077372 DOI: 10.7150/ijbs.85984] [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: 05/09/2023] [Accepted: 11/20/2023] [Indexed: 05/12/2024] Open
Abstract
Glioblastoma is the prevailing and highly malignant form of primary brain neoplasm with poor prognosis. Exosomes derived from glioblastoma cells act a vital role in malignant progression via regulating tumor microenvironment (TME), exosomal tetraspanin protein family members (TSPANs) are important actors of cell communication in TME. Among all the TSPANs, TSPAN6 exhibited predominantly higher expression levels in comparison to normal tissues. Meanwhile, glioblastoma patients with high level of TSPAN6 had shorter overall survival compared with low level of TSPAN6. Furthermore, TSPAN6 promoted the malignant progression of glioblastoma via promoting the proliferation and metastatic potential of glioblastoma cells. More interestingly, TSPAN6 overexpression in glioblastoma cells promoted the migration of vascular endothelial cell, and exosome secretion inhibitor reversed the migrative ability of vascular endothelial cells enhanced by TSPAN6 overexpressing glioblastoma cells, indicating that TSPAN6 might reinforce angiogenesis via exosomes in TME. Mechanistically, TSPAN6 enhanced the malignant progression of glioblastoma by interacting with CDK5RAP3 and regulating STAT3 signaling pathway. In addition, TSPAN6 overexpression in glioblastoma cells enhanced angiogenesis via regulating TME and STAT3 signaling pathway. Collectively, TSPAN6 has the potential to serve as both a therapeutic target and a prognostic biomarker for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Chong Zhang
- Affiliated Luqiao Hospital, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China, 310015
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China, 310015
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China, 310015
| | - Fei-hua Du
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China, 310015
- Department of Pharmacology, Zhejiang University, Hangzhou, Zhejiang, China, 310058
| | - Rou-xin Wang
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China, 310015
- Department of Pharmacology, Zhejiang University, Hangzhou, Zhejiang, China, 310058
| | - Wen-bo Han
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China, 310015
| | - Xing Lv
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China, 310015
| | - Ling-hui Zeng
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China, 310015
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China, 310015
| | - Guo-qing Chen
- Affiliated Luqiao Hospital, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China, 310015
| |
Collapse
|
6
|
Liang J, Yun D, Jin W, Fan J, Wang X, Wang X, Li Y, Yu S, Zhang C, Li T, Yang X. NCAPH serves as a prognostic factor and promotes the tumor progression in glioma through PI3K/AKT signaling pathway. Mol Cell Biochem 2024:10.1007/s11010-024-04976-4. [PMID: 38587786 DOI: 10.1007/s11010-024-04976-4] [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: 09/26/2023] [Accepted: 02/24/2024] [Indexed: 04/09/2024]
Abstract
Non-SMC (Structural Maintenance of Chromosomes) condensin I complex subunit H (NCAPH) has been shown to facilitate progression and predict adverse prognostic outcome in many cancer types. However, the function of NCAPH in gliomas is still unclear. Series of experiments were taken to uncover the function of NCAPH in glioma. The expression of NCAPH and potential mechanism regulating progression of glioma was verified by bioinformatics analysis. Lentiviral transfection was used for establishment of loss-of-function and gain-of-function cell lines. CCK-8 assay and Colony-formation assay were used to evaluate proliferation. Transwell assay and Cell wound healing assay were used to assess migration and invasion. Cell cycle and apoptosis were measured by flow cytometry. Protein and RNA were quantified by WB and RT-PCR, respectively. The nude mice model of glioma was used to evaluate the effect of NCAPH in vivo. The expression of NCAPH increased significantly in glioma tissues and correlated with WHO grade, IDH wild-type and non-1p/19q codeletion. Glioma patients with high expression of NCAPH had an undesirable prognosis. Functionally, upregulated NCAPH promotes the malignant hallmarks of glioma cells in vivo and in vitro. NCAPH correlated with DNA damage repair ability of glioma cells and facilitated the proliferation, invasion, and migration of glioma cells by promoting the PI3K/AKT signaling pathway. This study identifies the important pro-tumor role of NCAPH in glioma and suggests that NCAPH is a potential therapeutic target.
Collapse
Affiliation(s)
- Jianshen Liang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300000, China
| | - Debo Yun
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300000, China
- Department of Neurosurgery, Nanchong Central Hospital, Nanchong, 637000, Sichuan, China
| | - Wenzhe Jin
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300000, China
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, 071000, Hebei, China
| | - Jikang Fan
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300000, China
| | - Xuya Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300000, China
| | - Xisen Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300000, China
| | - Yiming Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300000, China
| | - Shengping Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300000, China
| | - Chen Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300000, China
| | - Tao Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China.
| | - Xuejun Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China.
- Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, 300000, China.
- Department of Neurosurgery, Tsinghua University Beijing Tsinghua Changgung Hospital, Beijing, 102218, China.
| |
Collapse
|
7
|
Xu T, Yin F, Shi K. TMEM158 functions as an oncogene and promotes lung adenocarcinoma progression through the PI3K/AKT pathway via interaction with TWIST1. Exp Cell Res 2024; 437:114010. [PMID: 38508329 DOI: 10.1016/j.yexcr.2024.114010] [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: 01/06/2024] [Revised: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 03/22/2024]
Abstract
Lung adenocarcinoma (LUAD) is a common and deadly form of lung cancer, with high rates of metastasis and unsatisfactory clinical outcomes. Herein, we examined the influence of TMEM158 on the LUAD progression. A combination of bioinformatic analyses was used to assess the TMEM158 expression pattern, prognostic implications, and potential function in LUAD. The levels of TMEM158 and TWIST1 were evaluated in clinical samples from LUAD patients using Western blot analysis and qRT-PCR. To discover the function and underlying molecular pathways of TMEM158 in LUAD, we employed a combination of experimental approaches in vitro, such as flow cytometry analysis and colony formation, Co-IP, CCK-8, Transwell, and wound-healing assays. Elevated expression of TMEM158 in LUAD is associated with increased cancer aggressiveness and a poor prognosis. In vitro experiments demonstrated that high levels of TMEM158 promote cell proliferation, progression through the cell cycle, migration, and invasion while suppressing apoptosis. Knockdown of TMEM158 produced opposite effects. The underlying mechanism involves TMEM158 and TWIST1 directly interacting, stimulating the PI3K/AKT signaling pathway in LUAD cells. This investigation emphasizes the molecular functions of TMEM158 in LUAD progression and proposes targeting it as a promising treatment approach for managing LUAD.
Collapse
Affiliation(s)
- Tao Xu
- Anhui Medical University, Hefei, 230000, China; Department of Cardiothoracic Surgery, The First People's Hospital of Wuhu, Wuhu, 241000, China.
| | - Fang Yin
- Department of Cardiothoracic Surgery, The First People's Hospital of Wuhu, Wuhu, 241000, China.
| | - Kaihu Shi
- Anhui Medical University, Hefei, 230000, China; Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing, 210028, China.
| |
Collapse
|
8
|
Wang Y, Wang B, Cao W, Xu X. TGF-β-activated circRYK drives glioblastoma progression by increasing VLDLR mRNA expression and stability in a ceRNA- and RBP-dependent manner. J Exp Clin Cancer Res 2024; 43:73. [PMID: 38454465 PMCID: PMC10921701 DOI: 10.1186/s13046-024-03000-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 03/01/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND The TGF-β signalling pathway is intricately associated with the progression of glioblastoma (GBM). The objective of this study was to examine the role of circRNAs in the TGF-β signalling pathway. METHODS In our research, we used transcriptome analysis to search for circRNAs that were activated by TGF-β. After confirming the expression pattern of the selected circRYK, we carried out in vitro and in vivo cell function assays. The underlying mechanisms were analysed via RNA pull-down, luciferase reporter, and RNA immunoprecipitation assays. RESULTS CircRYK expression was markedly elevated in GBM, and this phenotype was strongly associated with a poor prognosis. Functionally, circRYK promotes epithelial-mesenchymal transition and GSC maintenance in GBM. Mechanistically, circRYK sponges miR-330-5p and promotes the expression of the oncogene VLDLR. In addition, circRYK could enhance the stability of VLDLR mRNA via the RNA-binding protein HuR. CONCLUSION Our findings show that TGF-β promotes epithelial-mesenchymal transition and GSC maintenance in GBM through the circRYK-VLDLR axis, which may provide a new therapeutic target for the treatment of GBM.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210000, China
| | - Binbin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210000, China
| | - Wenping Cao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210000, China.
| | - Xiupeng Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, 210000, China.
| |
Collapse
|
9
|
Tang N, Zhu Y, Yu J. Xihuang pill facilitates glioma cell pyroptosis via the POU4F1/STAT3 axis. Funct Integr Genomics 2023; 23:334. [PMID: 37962640 DOI: 10.1007/s10142-023-01263-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/20/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023]
Abstract
Glioma is the most common malignancy in the central nervous system. This study aims to disclose the impacts of Xihuang pill (XHP), a traditional Chinese formula, on glioma cell pyroptosis and relevant molecular mechanism. U251 and SHG-44 cells were treated with XHP alone or together with oe-POU4F1 and sh-STAT3. CCK8 assay detected the viability, flow cytometry evaluated pyroptosis, and microscopy observed cell morphology. LDH release was determined by the LDH kit and the levels of IL-1β and IL-18 were detected by ELISA. Immunofluorescence showed NLRP3 expression in glioma cells and western blotting measured the levels of POU4F1, STAT3, NLRP3, ASC, cleaved caspase-1, and IL-1β. The binding of POU4F1 to STAT3 was verified. Primary glioma model was established to observe tumor change by in vivo imaging, determine the levels of Ki67 and NLRP3 by immunochemistry, and detect relevant protein levels by western blotting. XHP treatment alone downregulated POU4F1 and STAT3 levels, aroused pyroptotic appearance in glioma cells such as ballooning swelling, reduced cell viability and number of pyroptotic cells, increased LDH release and IL-1β and IL-18 levels, formed NLRP3 sports in cells, and elevated the levels of pyroptosis-related proteins. However, POU4F1 overexpression or STAT3 silencing suppressed XHP-promoted pyroptosis. Mechanistically, POU4F1 acted as a transcription factor of STAT3 and regulated its transcription. In primary glioma models, XHP enhanced glioma cell pyroptosis and blocked glioma growth. XHP facilitates glioma cell pyroptosis via the POU4F1/STAT3 axis.
Collapse
Affiliation(s)
- Ning Tang
- Department of Neurosurgery, The First Affiliated Hospital of Hunan University of Chinese Medicine, No. 95 Shaoshan Middle Road, Changsha, Hunan, 410007, People's Republic of China
| | - Yuanyuan Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Hunan University of Chinese Medicine, No. 95 Shaoshan Middle Road, Changsha, Hunan, 410007, People's Republic of China
| | - Jianbai Yu
- Department of Neurosurgery, The First Affiliated Hospital of Hunan University of Chinese Medicine, No. 95 Shaoshan Middle Road, Changsha, Hunan, 410007, People's Republic of China.
| |
Collapse
|
10
|
Cui X, Lu J, Zhao C, Duan Y. Oncogenic transmembrane protein 158 drives the PI3K/Akt signaling pathway to accelerate gastric cancer cell growth. Braz J Med Biol Res 2023; 56:e12943. [PMID: 37970923 PMCID: PMC10644964 DOI: 10.1590/1414-431x2023e12943] [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: 07/27/2023] [Accepted: 10/14/2023] [Indexed: 11/19/2023] Open
Abstract
Gastric cancer (GC) is a serious threat to human health and an important cause of cancer-related death. Herein, we evaluated the influence of transmembrane protein 158 (TMEM158) on GC cell growth. According to Genomic Spatial Event (GSE) and The Cancer Genome Atlas (TCGA) databases, TMEM158 content is amplified in GC tissues. The diagnostic value of TMEM158 expression in GC is huge. GC sufferers with high expression of TMEM158 were associated with poor overall survival. In addition, TMEM158 content was increased in GC cells. TMEM158 promoted GC cell proliferation by modulating the PI3K/Akt signaling pathway. Lack of TMEM158 reduced GC tumor growth. Collectively, TMEM158 accelerated GC cell proliferation by modulating the PI3K/Akt signaling pathway, making it a prospective biomarker for survival in GC patients.
Collapse
Affiliation(s)
- Xiaoting Cui
- Department of Gastroenterology, The Third Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Baogang Hospital, Baotou, China
| | - Jie Lu
- Department of Gastroenterology, The Fourth Affiliated Hospital of Baotou Medical College, Baotou Eighth Hospital, Baotou, China
| | - Cuijuan Zhao
- Department of Gastroenterology, The Third Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Baogang Hospital, Baotou, China
| | - Yu Duan
- Department of Gastroenterology, The Third Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia Baogang Hospital, Baotou, China
| |
Collapse
|
11
|
Herrera-Quiterio GA, Encarnación-Guevara S. The transmembrane proteins (TMEM) and their role in cell proliferation, migration, invasion, and epithelial-mesenchymal transition in cancer. Front Oncol 2023; 13:1244740. [PMID: 37936608 PMCID: PMC10627164 DOI: 10.3389/fonc.2023.1244740] [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: 06/23/2023] [Accepted: 09/11/2023] [Indexed: 11/09/2023] Open
Abstract
Transmembrane proteins (TMEM) are located in the different biological membranes of the cell and have at least one passage through these cellular compartments. TMEM proteins carry out a wide variety of functions necessary to maintain cell homeostasis TMEM165 participates in glycosylation protein, TMEM88 in the development of cardiomyocytes, TMEM45A in epidermal keratinization, and TMEM74 regulating autophagy. However, for many TMEM proteins, their physiological function remains unknown. The role of these proteins is being recently investigated in cancer since transcriptomic and proteomic studies have revealed that exits differential expression of TMEM proteins in different neoplasms concerning cancer-free tissues. Among the cellular processes in which TMEM proteins have been involved in cancer are the promotion or suppression of cell proliferation, epithelial-mesenchymal transition, invasion, migration, intravasation/extravasation, metastasis, modulation of the immune response, and response to antineoplastic drugs. Inclusive data suggests that the participation of TMEM proteins in these cellular events could be carried out through involvement in different cell signaling pathways. However, the exact mechanisms not clear. This review shows a description of the involvement of TMEM proteins that promote or decrease cell proliferation, migration, and invasion in cancer cells, describes those TMEM proteins for which both a tumor suppressor and a tumor promoter role have been identified, depending on the type of cancer in which the protein is expressed. As well as some TMEM proteins involved in chemoresistance. A better characterization of these proteins is required to improve the understanding of the tumors in which their expression and function are altered; in addition to improving the understanding of the role of these proteins in cancer will show those TMEM proteins be potential candidates as biomarkers of response to chemotherapy or prognostic biomarkers or as potential therapeutic targets in cancer.
Collapse
Affiliation(s)
| | - Sergio Encarnación-Guevara
- Laboratorio de Proteómica, Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| |
Collapse
|
12
|
Tamai S, Ichinose T, Jiapaer S, Hirai N, Sabit H, Tanaka S, Kinoshita M, Kobayashi M, Hirao A, Nakada M. Therapeutic potential of pentamidine for glioma-initiating cells and glioma cells through multimodal antitumor effects. Cancer Sci 2023. [PMID: 37142416 DOI: 10.1111/cas.15827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/07/2023] [Accepted: 04/12/2023] [Indexed: 05/06/2023] Open
Abstract
Glioma-initiating cells, which comprise a heterogeneous population of glioblastomas, contribute to resistance against aggressive chemoradiotherapy. Using drug reposition, we investigated a therapeutic drug for glioma-initiating cells. Drug screening was undertaken to select candidate agents that inhibit proliferation of two different glioma-initiating cells lines. The alteration of proliferation and stemness of the two glioma-initiating cell lines, and proliferation, migration, cell cycle, and survival of these two differentiated glioma-initiating cell lines and three different glioblastoma cell lines treated with the candidate agent were evaluated. We also used a xenograft glioma mouse model to evaluate anticancer effects of treated glioma cell lines. Among the 1301 agents, pentamidine-an antibiotic for Pneumocystis jirovecii-emerged as a successful antiglioma agent. Pentamidine treatment suppressed proliferation and stemness in glioma-initiating cell lines. Proliferation and migration were inhibited in all differentiated glioma-initiating cells and glioblastoma cell lines, with cell cycle arrest and caspase-dependent apoptosis induction. The in vivo study reproduced the same findings as the in vitro studies. Pentamidine showed a stronger antiproliferative effect on glioma-initiating cells than on differentiated cells. Western blot analysis revealed pentamidine inhibited phosphorylation of signal transducer and activator of transcription 3 in all cell lines, whereas Akt expression was suppressed in glioma-initiating cells but not in differentiated lines. In the present study, we identified pentamidine as a potential therapeutic drug for glioma. Pentamidine could be promising for the treatment of glioblastomas by targeting both glioma-initiating cells and differentiated cells through its multifaceted antiglioma effects.
Collapse
Affiliation(s)
- Sho Tamai
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - Toshiya Ichinose
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - Shabierjiang Jiapaer
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - Nozomi Hirai
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
- Department of Neurosurgery, Toho University Ohashi Medical Center, Tokyo, Japan
| | - Hemragul Sabit
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - Shingo Tanaka
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - Masashi Kinoshita
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| | - Masahiko Kobayashi
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Ishikawa, Japan
| | - Atsushi Hirao
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Ishikawa, Japan
| | - Mitsutoshi Nakada
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Ishikawa, Japan
| |
Collapse
|
13
|
Li J, Hou H, Sun J, Ding Z, Xu Y, Li G. Systematic pan-cancer analysis identifies transmembrane protein 158 as a potential therapeutic, prognostic and immunological biomarker. Funct Integr Genomics 2023; 23:105. [PMID: 36977915 DOI: 10.1007/s10142-023-01032-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/12/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023]
Abstract
The purpose of this study was to investigate the expression significance, predictive value, immunologic function, and biological role of transmembrane protein 158 (TMEM158) in the development of pan-cancer. To achieve this, we utilized data from multiple databases, including TCGA, GTEx, GEPIA, and TIMER, to collect gene transcriptome, patient prognosis, and tumor immune data. We evaluated the association of TMEM158 with patient prognosis, tumor mutational burden (TMB), and microsatellite instability (MSI) in pan-cancer samples. We performed immune checkpoint gene co-expression analysis and gene set enrichment analysis (GSEA) to better understand the immunologic function of TMEM158. Our findings revealed that TMEM158 was significantly differentially expressed between most types of cancer tissues and their adjacent normal tissues and was associated with prognosis. Moreover, TMEM158 was significantly correlated with TMB, MSI, and tumor immune cell infiltration in multiple cancers. Co-expression analysis of immune checkpoint genes showed that TMEM158 was related to the expression of several common immune checkpoint genes, especially CTLA4 and LAG3. Gene enrichment analysis further revealed that TMEM158 was involved in multiple immune-related biological pathways in pan-cancer. Overall, this systematic pan-cancer analysis suggests that TMEM158 is generally highly expressed in various cancer tissues and is closely related to patient prognosis and survival across multiple cancer types. TMEM158 may serve as a significant predictor of cancer prognosis and modulate immune responses to various types of cancer.
Collapse
Affiliation(s)
- Jiayi Li
- School of Management, Shandong University, Jinan, 250100, Shandong, China
- School of Graduate, Hanyang University, Seoul, 04763, South Korea
| | - Haiguang Hou
- Department of Anatomy, School of Basic Medical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Jinhao Sun
- Department of Anatomy, School of Basic Medical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Zhaoxi Ding
- Department of Anatomy, School of Basic Medical Sciences, Shandong University, Jinan, 250012, Shandong, China
| | - Yingkun Xu
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Guibao Li
- Department of Anatomy, School of Basic Medical Sciences, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
14
|
PDPN contributes to constructing immunosuppressive microenvironment in IDH wildtype glioma. Cancer Gene Ther 2023; 30:345-357. [PMID: 36434176 PMCID: PMC9935394 DOI: 10.1038/s41417-022-00550-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 09/05/2022] [Accepted: 10/17/2022] [Indexed: 11/26/2022]
Abstract
The tumor immunosuppressive microenvironment (IME) significantly affects tumor occurrence, progression, and prognosis, but the underlying molecular mechanisms remain to make known. We investigated the prognostic significance of PDPN and its role in IME in glioma. Weighted gene co-expression network analysis (WGCNA) found PDPN closely related to IDH wildtype status and higher immune score. Correlation analysis suggested PDPN was highly positively relevant to immune checkpoints expression and immune checkpoints block responding status. Correlation analysis together with verification in vitro suggested PDPN highly positively relevant tumor-associated neutrophils (TANs) and tumor-associated macrophages (TAMs). Least absolute shrinkage and selection operator (LASSO) regression employed to develop the prediction model with TANs and TAMs markers showed that high risk scores predicted worse prognosis. We highlight that PDPN overexpression is an independent prognostic indicator, and promotes macrophage M2 polarization and neutrophil degranulation, ultimately devotes to the formation of an immunosuppressive tumor microenvironment. Our findings contribute to re-recognizing the role of PDPN in IDH wildtype gliomas and implicate promising target therapy combined with immunotherapy for this highly malignant tumor.
Collapse
|
15
|
Feng R, Lu M, Yin C, Xu K, Liu L, Xu P. Identification of candidate genes and pathways associated with juvenile idiopathic arthritis by integrative transcriptome-wide association studies and mRNA expression profiles. Arthritis Res Ther 2023; 25:19. [PMID: 36755318 PMCID: PMC9906884 DOI: 10.1186/s13075-023-03003-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 01/31/2023] [Indexed: 02/10/2023] Open
Abstract
AIM Juvenile idiopathic arthritis (JIA) is the most common chronic rheumatic disease of childhood, with genetic susceptibility and pathological processes such as autoimmunity and autoinflammation, but its pathogenesis is unclear. We conducted a transcriptome-wide association study (TWAS) using expression interpolation from a large-scale genome-wide association study (GWAS) dataset to identify genes, biological pathways, and environmental chemicals associated with JIA. METHODS We obtained published GWAS data on JIA for TWAS and used mRNA expression profiling to validate the genes identified by TWAS. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed. A protein-protein interaction (PPI) network was generated, and central genes were obtained using Molecular Complex Detection (MCODE). Finally, chemical gene expression datasets were obtained from the Comparative Toxicogenomics database for chemical genome enrichment analysis. RESULTS TWAS identified 1481 genes associated with JIA, and 154 differentially expressed genes were identified based on mRNA expression profiles. After comparing the results of TWAS and mRNA expression profiles, we obtained eight overlapping genes. GO and KEGG enrichment analyses of the genes identified by TWAS yielded 163 pathways, and PPI network analysis as well as MCODE resolution identified a total of eight clusters. Through chemical gene set enrichment analysis, 287 environmental chemicals associated with JIA were identified. CONCLUSION By integrating TWAS and mRNA expression profiles, genes, biological pathways, and environmental chemicals associated with JIA were identified. Our findings provide new insights into the pathogenesis of JIA, including candidate genetic and environmental factors contributing to its onset and progression.
Collapse
Affiliation(s)
- Ruoyang Feng
- grid.452452.00000 0004 1757 9282Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xi’an, 710054 Shanxi China
| | - Mengnan Lu
- grid.452672.00000 0004 1757 5804Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi China
| | - Chunyan Yin
- grid.452672.00000 0004 1757 5804Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi China
| | - Ke Xu
- grid.452452.00000 0004 1757 9282Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xi’an, 710054 Shanxi China
| | - Lin Liu
- grid.452452.00000 0004 1757 9282Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xi’an, 710054 Shanxi China
| | - Peng Xu
- Department of Joint Surgery, HongHui Hospital, Xian Jiaotong University, Xi'an, 710054, Shanxi, China.
| |
Collapse
|
16
|
Bou Zerdan M, Atoui A, Hijazi A, Basbous L, Abou Zeidane R, Alame SM, Assi HI. Latest updates on cellular and molecular biomarkers of gliomas. Front Oncol 2022; 12:1030366. [PMID: 36425564 PMCID: PMC9678906 DOI: 10.3389/fonc.2022.1030366] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Accepted: 10/05/2022] [Indexed: 03/05/2024] Open
Abstract
Gliomas are the most common central nervous system malignancies, compromising almost 80% of all brain tumors and is associated with significant mortality. The classification of gliomas has shifted from basic histological perspective to one that is based on molecular biomarkers. Treatment of this type of tumors consists currently of surgery, chemotherapy and radiation therapy. During the past years, there was a limited development of effective glioma diagnostics and therapeutics due to multiple factors including the presence of blood-brain barrier and the heterogeneity of this type of tumors. Currently, it is necessary to highlight the advantage of molecular diagnosis of gliomas to develop patient targeted therapies based on multiple oncogenic pathway. In this review, we will evaluate the development of cellular and molecular biomarkers for the diagnosis of gliomas and the impact of these diagnostic tools for better tailored and targeted therapies.
Collapse
Affiliation(s)
- Maroun Bou Zerdan
- Department of Internal Medicine, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Ali Atoui
- Hematology-Oncology Division, Internal Medicine Department, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ali Hijazi
- Hematology-Oncology Division, Internal Medicine Department, American University of Beirut Medical Center, Beirut, Lebanon
| | - Lynn Basbous
- Hematology-Oncology Division, Internal Medicine Department, American University of Beirut Medical Center, Beirut, Lebanon
| | - Reine Abou Zeidane
- Hematology-Oncology Division, Internal Medicine Department, American University of Beirut Medical Center, Beirut, Lebanon
| | - Saada M Alame
- Department of Pediatrics, Faculty of Medicine, Lebanese University, Beirut, Lebanon
| | - Hazem I Assi
- Hematology-Oncology Division, Internal Medicine Department, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
17
|
Huang J, Liu W, Zhang D, Lin B, Li B. TMEM158 expression is negatively regulated by AR signaling and associated with favorite survival outcomes in prostate cancers. Front Oncol 2022; 12:1023455. [PMID: 36387246 PMCID: PMC9663988 DOI: 10.3389/fonc.2022.1023455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/18/2022] [Indexed: 08/30/2023] Open
Abstract
BACKGROUND Membrane protein TMEM158 was initially reported as a Ras-induced gene during senescence and has been implicated as either an oncogenic factor or tumor suppressor, depending on tumor types. It is unknown if TMEM158 expression is altered in prostate cancers. METHODS Multiple public gene expression datasets from RNA-seq and cDNA microarray assays were utilized to analyze candidate gene expression profiles. TMEM158 protein expression was assessed using an immunohistochemistry approach on a tissue section array from benign and malignant prostate tissues. Comparisons of gene expression profiles were conducted using the bioinformatics software R package. RESULTS COX regression-based screening identified the membrane protein TMEM158 gene as negatively associated with disease-specific and progression-free survival in prostate cancer patients. Gene expression at the mRNA and protein levels revealed that TMEM158 expression was significantly reduced in malignant tissues compared to benign compartments. Meanwhile, TMEM158 downregulation was strongly correlated with advanced clinicopathological features, including late-stage diseases, lymph node invasion, higher PSA levels, residual tumors after surgery, and adverse Gleason scores. In castration-resistant prostate cancers, TMEM158 expression was negatively correlated with AR signaling activity but positively correlated with neuroendocrinal progression index. Consistently, in cell culture models, androgen treatment reduced TMEM158 expression, while androgen deprivation led to upregulation of TMEM158 expression. Correlation analysis showed a tight correlation of TMEM158 expression with the level of R-Ras gene expression, which was also significantly downregulated in prostate cancers. Tumor immune infiltration profiling analysis discovered a strong association of TMEM158 expression with NK cell and Mast cell enrichment. CONCLUSION The membrane protein TMEM158 is significantly downregulated in prostate cancer and is tightly associated with disease progression, anti-tumor immune infiltration, and patient survival outcome.
Collapse
Affiliation(s)
- Jian Huang
- Center for Pathological Diagnosis and Research, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Wang Liu
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS, United States
| | - Da Zhang
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS, United States
| | - Biyun Lin
- Center for Pathological Diagnosis and Research, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Benyi Li
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS, United States
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
18
|
Allen DE, Donohue KC, Cadwell CR, Shin D, Keefe MG, Sohal VS, Nowakowski TJ. Fate mapping of neural stem cell niches reveals distinct origins of human cortical astrocytes. Science 2022; 376:1441-1446. [PMID: 35587512 PMCID: PMC9233096 DOI: 10.1126/science.abm5224] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Progenitors of the developing human neocortex reside in the ventricular and outer subventricular zones (VZ and OSVZ, respectively). However, whether cells derived from these niches have similar developmental fates is unknown. By performing fate mapping in primary human tissue, we demonstrate that astrocytes derived from these niches populate anatomically distinct layers. Cortical plate astrocytes emerge from VZ progenitors and proliferate locally, while putative white matter astrocytes are morphologically heterogeneous and emerge from both VZ and OSVZ progenitors. Furthermore, via single-cell sequencing of morphologically defined astrocyte subtypes using Patch-seq, we identify molecular distinctions between VZ-derived cortical plate astrocytes and OSVZ-derived white matter astrocytes that persist into adulthood. Together, our study highlights a complex role for cell lineage in the diversification of human neocortical astrocytes.
Collapse
Affiliation(s)
- Denise E Allen
- Department of Anatomy, The University of California San Francisco, San Francisco, USA,Department of Psychiatry and Behavioral Sciences, The University of California San Francisco, San Francisco, USA,Department of Neurological Surgery, The University of California San Francisco, San Francisco, USA,Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, The University of California San Francisco, San Francisco, USA
| | - Kevin C Donohue
- Department of Psychiatry and Behavioral Sciences, The University of California San Francisco, San Francisco, USA,School of Medicine, The University of California San Francisco, San Francisco, USA,Center for Integrative Neuroscience, The University of California San Francisco; San Francisco, USA,Weill Institute for Neurosciences, The University of California San Francisco; San Francisco, USA,Kavli Institute for Fundamental Neuroscience, The University of California San Francisco, San Francisco, USA
| | - Cathryn R Cadwell
- Department of Pathology, The University of California San Francisco, San Francisco, USA
| | - David Shin
- Department of Anatomy, The University of California San Francisco, San Francisco, USA,Department of Psychiatry and Behavioral Sciences, The University of California San Francisco, San Francisco, USA,Department of Neurological Surgery, The University of California San Francisco, San Francisco, USA,Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, The University of California San Francisco, San Francisco, USA
| | - Matthew G Keefe
- Department of Anatomy, The University of California San Francisco, San Francisco, USA,Department of Psychiatry and Behavioral Sciences, The University of California San Francisco, San Francisco, USA,Department of Neurological Surgery, The University of California San Francisco, San Francisco, USA,Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, The University of California San Francisco, San Francisco, USA
| | - Vikaas S Sohal
- Department of Psychiatry and Behavioral Sciences, The University of California San Francisco, San Francisco, USA,Weill Institute for Neurosciences, The University of California San Francisco; San Francisco, USA,Kavli Institute for Fundamental Neuroscience, The University of California San Francisco, San Francisco, USA
| | - Tomasz J Nowakowski
- Department of Anatomy, The University of California San Francisco, San Francisco, USA,Department of Psychiatry and Behavioral Sciences, The University of California San Francisco, San Francisco, USA,Department of Neurological Surgery, The University of California San Francisco, San Francisco, USA,Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, The University of California San Francisco, San Francisco, USA,Weill Institute for Neurosciences, The University of California San Francisco; San Francisco, USA,Corresponding author.
| |
Collapse
|