1
|
Bagci H, Winkler M, Grädel B, Uliana F, Boulais J, Mohamed WI, Park SL, Côté JF, Pertz O, Peter M. The hGID GID4 E3 ubiquitin ligase complex targets ARHGAP11A to regulate cell migration. Life Sci Alliance 2024; 7:e202403046. [PMID: 39389782 PMCID: PMC11467045 DOI: 10.26508/lsa.202403046] [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: 09/15/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
The human CTLH/GID (hGID) complex emerged as an important E3 ligase regulating multiple cellular processes, including cell cycle progression and metabolism. However, the range of biological functions controlled by hGID remains unexplored. Here, we used proximity-dependent biotinylation (BioID2) to identify proteins interacting with the hGID complex, among them, substrate candidates that bind GID4 in a pocket-dependent manner. Biochemical and cellular assays revealed that the hGIDGID4 E3 ligase binds and ubiquitinates ARHGAP11A, thereby targeting this RhoGAP for proteasomal degradation. Indeed, GID4 depletion or impeding the GID4 substrate binding pocket with the PFI-7 inhibitor stabilizes ARHGAP11A protein amounts, although it carries no functional N-terminal degron. Interestingly, GID4 inactivation impairs cell motility and directed cell movement by increasing ARHGAP11A levels at the cell periphery, where it inactivates RhoA. Together, we identified a wide range of hGIDGID4 E3 ligase substrates and uncovered a unique function of the hGIDGID4 E3 ligase regulating cell migration by targeting ARHGAP11A.
Collapse
Affiliation(s)
- Halil Bagci
- Institute of Biochemistry, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Martin Winkler
- Institute of Biochemistry, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Benjamin Grädel
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Federico Uliana
- Institute of Biochemistry, Department of Biology, ETH Zürich, Zürich, Switzerland
| | | | - Weaam I Mohamed
- Institute of Biochemistry, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Sophia L Park
- Institute of Biochemistry, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Jean-François Côté
- Montreal Clinical Research Institute (IRCM), Montréal, Canada
- Molecular Biology Programs, Université de Montréal, Montréal, Canada
| | - Olivier Pertz
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Matthias Peter
- Institute of Biochemistry, Department of Biology, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
2
|
Zhang W, Bai X, Liu T, Mao Y, Zhang L, Wang W, Yu H. Rho GTPase activating protein 11A promotes tongue squamous cell carcinoma proliferation and is a transcriptional target of forkhead box M1. J Dent Sci 2024; 19:2268-2277. [PMID: 39347089 PMCID: PMC11437295 DOI: 10.1016/j.jds.2024.02.015] [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: 01/07/2024] [Revised: 02/16/2024] [Indexed: 10/01/2024] Open
Abstract
Background/purpose Rho GTPase activating protein 11A (ARHGAP11A) can facilitate GTP hydrolysis in RhoA. The functions of ARHGAP11A in oral squamous cell carcinoma (OSCC) have not yet been explored. This study aimed to investigate the expression profile of ARHGAP11A in OSCC, its correlation with patient prognosis, its effect on cell-cycle progression, and the mechanisms by which it is dysregulated. Materials and methods Bioinformatics analysis was conducted using data from The Cancer Genome Atlas-Head and Neck Squamous Cell Carcinoma (TCGA-HNSC). Lentiviruses carrying ARHGAP11A shRNAs were employed to determine the effects of ARHGAP11A knockdown on tumor cell proliferation and cell-cycle progression. Dual-luciferase reporter assays were utilized to examine how FOXM1 transcriptionally regulates ARHGAP11A expression. Results ARHGAP11A upregulation was associated with unfavorable overall survival (OS) in patients with TSCC (HR: 2.142, 95%CI: 1.224-3.749, P = 0.007), but not in patients with OSCC of sites other than the tongue. ARHGAP11A knockdown inhibited the proliferation of TSCC cells in vitro and in vivo, and induced G1 phase arrest. ARHGAP11A knockdown increased GTP-RhoA but decreased p-RB levels, while it did not affect the total expression of RhoA and RB. ARHGAP11A knockdown increased p27 and decreased cyclin E1 expression. ARHGAP11A is transcriptionally activated by FOXM1 via multiple FOXM1 binding sites in the promoter regions in TSCC cells. Conclusion This study revealed the oncogenic role of ARHGAP11A in TSCC, highlighting its impact on cell-cycle control and tumor proliferation. Furthermore, the regulatory relationship between FOXM1 and ARHGAP11A provides new insights into the transcriptional networks in TSCC.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Orthodontics, Binzhou Medical University Hospital, Binzhou, China
| | - Xueyan Bai
- Department of Orthodontics, Binzhou Medical University Hospital, Binzhou, China
| | - Tingting Liu
- Department of Oral Medicine, Binzhou Medical University Hospital, Binzhou, China
| | - Yulong Mao
- Department of Oral and Maxillofacial Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Lingnan Zhang
- Department of Orthodontics, Binzhou Medical University Hospital, Binzhou, China
| | - Wenlong Wang
- Department of Oral and Maxillofacial Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Huanying Yu
- Department of Prosthodontics, Binzhou Medical University Hospital, Binzhou, China
| |
Collapse
|
3
|
Huang J, Wang G, Zhang J, Liu Y, Shen Y, Chen G, Ji W, Shao J. A novel ARHGAP family gene signature for survival prediction in glioma patients. J Cell Mol Med 2024; 28:e18555. [PMID: 39075640 PMCID: PMC11286547 DOI: 10.1111/jcmm.18555] [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: 05/02/2024] [Revised: 06/15/2024] [Accepted: 07/13/2024] [Indexed: 07/31/2024] Open
Abstract
ARHGAP family genes are often used as glioma oncogenic factors, and their mechanism of action remains unexplained. Our research entailed a thorough examination of the immune microenvironment and enrichment pathways across various glioma subtypes. A distinctive 6-gene signature was developed employing the CGGA cohort, leading to insights into the disparities in clinical characteristics, mutation patterns, and immune cell infiltration among distinct risk categories. Additionally, a unique nomogram was established, grounded on ARHGAPs, with DCA curves illustrating the model's prospective clinical utility in guiding therapeutic strategies. Emphasizing the role of ARHGAP30, integral to our model, its impact on glioma severity and the credibility of our risk assessment model were substantiated through RT-qPCR, Western blot analysis, and cellular functional assays. We identified 6 ARHGAP family genes associated with glioma prognosis. Analysis using the Kaplan-Meier method indicated a correlation between elevated risk levels and adverse outcomes in glioma patients. The risk score, linked with tumour staging and IDH mutation status, emerged as an independent factor predicting prognosis. Patients in the high-risk category exhibited increased immune cell infiltration, enhanced tumour mutational burden, more pronounced expression of immune checkpoint genes, and a better response to ICB therapy. A nomogram, integrating the risk score with the pathological features of glioma patients, was developed. DCA analysis and cellular studies confirmed the model's potential to improve clinical treatment outcomes for patients. A novel ARHGAP family gene signature reveals the prognosis of glioma.
Collapse
Affiliation(s)
- Jin Huang
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuChina
| | - Gaosong Wang
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuChina
| | - Jiahao Zhang
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuChina
| | - Yuankun Liu
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuChina
| | - Yifan Shen
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuChina
| | - Gengjing Chen
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuChina
| | - Wei Ji
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuChina
| | - Junfei Shao
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical UniversityWuxiJiangsuChina
| |
Collapse
|
4
|
Zhang Y, Ma D, Gong Y, Wang F, Wu J, Wu C. IL1R2 is a Novel Prognostic Biomarker for Lung Adenocarcinoma. Curr Mol Med 2024; 24:620-629. [PMID: 37078353 DOI: 10.2174/1566524023666230420092142] [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: 10/03/2022] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 04/21/2023]
Abstract
AIMS The aim of this study is to figure out the role of IL1R2 in LUAD (lung adenocarcinoma). BACKGROUND IL1R2, a special member of IL-1 receptor family, binds to IL-1 and plays an important role in inhibiting IL-1 pathway, which seems to be involved in tumorigenesis. Emerging studies demonstrated higher IL1R2 expression levels in several malignancies. OBJECTIVE In the present study, we assessed the expression of IL1R2 in LUAD tissues with immunohistochemistry and explored various databases to determine whether it could be a potential prognostic biomarker and therapeutic target. METHODS The expression level of IL1R2 in lung adenocarcinoma was analyzed by Immunohistochemistry and UALCAN database. The correlation between IL1R2 expression and the patient prognosis was identified by Kaplan-Meier plotter. The correlation of IL1R2 expression with immune infiltrates was clarified by TIMER database. The protein-protein interaction network and gene functional enrichment analysis were constructed and performed by STRING and Metascape database. RESULTS Immunohistochemistry showed that the expression of IL1R2 was higher in tumor tissues of LUAD patients and that patients with lower IL1R2 level have a better prognosis than their counterparts. We validated our findings in several online databases and found that IL1R2 gene was also positively correlated with B cells and neutrophils and biomarkers of CD8+T cells and exhausted T cells. PPI network and gene enrichment analyses showed that expression of IL1R2 was also associated with complex functionspecific networks involving IL-1 signal, NF-KappaB transcription factors. CONCLUSION According to these findings, we demonstrated that IL1R2 was involved in the progression and prognosis of LUAD and the underlying mechanism needs further investigation.
Collapse
Affiliation(s)
- Ying Zhang
- Departments of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, P.R. China
- Departments of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, 214000, P.R. China
| | - Danyu Ma
- Departments of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, P.R. China
| | - Yile Gong
- Department of General Internal Medicine, People's hospital of Ziyang, Ankang, Shanxi, 725399, P.R. China
| | - Fan Wang
- Department of General Internal Medicine, People's hospital of Ziyang, Ankang, Shanxi, 725399, P.R. China
| | - Jingping Wu
- Department of General Internal Medicine, People's hospital of Ziyang, Ankang, Shanxi, 725399, P.R. China
| | - Chen Wu
- Departments of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, 213003, P.R. China
- Department of General Internal Medicine, People's hospital of Ziyang, Ankang, Shanxi, 725399, P.R. China
| |
Collapse
|
5
|
Quadri R, Rotondo G, Sertic S, Pozzi S, dell’Oca MC, Guerrini L, Muzi-Falconi M. A Haspin-ARHGAP11A axis regulates epithelial morphogenesis through Rho-ROCK dependent modulation of LIMK1-Cofilin. iScience 2023; 26:108011. [PMID: 37841592 PMCID: PMC10570125 DOI: 10.1016/j.isci.2023.108011] [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: 05/02/2023] [Revised: 07/20/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
Throughout mitosis, a plethora of processes must be efficiently concerted to ensure cell proliferation and tissue functionality. The mitotic spindle does not only mediate chromosome segregation, but also defines the axis of cellular division, thus determining tissue morphology. Functional spindle orientation relies on precise actin dynamics, shaped in mitosis by the LIMK1-Cofilin axis. The kinase Haspin acts as a guardian of faithful chromosome segregation that ensures amphitelic chromosome attachment and prevents unscheduled cohesin cleavage. Here, we report an unprecedented role for Haspin in the determination of spindle orientation in mitosis. We show that, during mitosis, Haspin regulates Rho-ROCK activity through ARHGAP11A, a poorly characterized GAP, and that ROCK is in turn responsible for the mitotic activation of LIMK1 and stabilization of the actin cytoskeleton, thus supporting a functional spindle orientation. By exploiting 3D cell cultures, we show that this pathway is pivotal for the establishment of a morphologically functional tissue.
Collapse
Affiliation(s)
- Roberto Quadri
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | - Giuseppe Rotondo
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | - Sarah Sertic
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | - Sara Pozzi
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | | | - Luisa Guerrini
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| | - Marco Muzi-Falconi
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy
| |
Collapse
|
6
|
Yang H, Zhang H, Zhang L, Tusuphan P, Zheng J. ARHGAP11A Is a Novel Prognostic and Predictive Biomarker Correlated with Immunosuppressive Microenvironment in Clear Cell Renal Cell Carcinoma. Int J Mol Sci 2023; 24:ijms24097755. [PMID: 37175461 PMCID: PMC10178328 DOI: 10.3390/ijms24097755] [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: 02/24/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a highly immunogenic tumor and immune dysfunction is associated with ccRCC poor prognosis. The RhoGTPase-activating proteins (RhoGAPs) family was reported to affect ccRCC development, but its role in immunity and prognosis prediction for ccRCC remain unknown. In the current study, we found ARHGAP11A was the only independent risk factor among 33 RhoGAPs (hazard ratio [HR] 1.949, 95% confidence interval [CI] 1.364-2.785). High ARHGAP11A level was associated with shorter overall survival (OS, HR 2.040, 95% CI 1.646-3.417) and ARHGAP11A is a prognostic biomarker for ccRCC. ARHGAP11A knockdown suppressed renal cell carcinoma (RCC) cell proliferation, colony formation, and migration, suggesting the promoting role of ARHGAP11A on RCC development. Mechanistically, ARHGAP11A might contribute to the suppressive tumor immune microenvironment (TIME). High ARHGAP11A level was correlated with infiltration of immunosuppressive cells (including T helper 2 (Th2) cells, regulatory T (Treg) cells, myeloid derived suppressor cells (MDSC), and M2 macrophage cells), activation of immunosuppressive pathways (IL6-JAK-STAT3 signaling and IFNγ response), and expression of inhibitory immune checkpoints (ICs). ARHGAP11A could promote T cell exhaustion and induce immune escape. ccRCC patients with low ARHGAP11A level were more suitable for immune checkpoint inhibitors (ICIs) therapy, while those with high ARHGAP11A level might benefit from a combination of ARHGAP11A blockade and ICIs. In all, ARHGAP11A might serve as a novel prognostic marker, therapeutic target, and predictor in the clinical response to ICIs therapy for ccRCC.
Collapse
Affiliation(s)
- Huihui Yang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing100069, China
| | - Hongning Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing100069, China
| | - Liuxu Zhang
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing100069, China
| | - Paizigul Tusuphan
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing100069, China
| | - Junfang Zheng
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing100069, China
| |
Collapse
|
7
|
Kavousi N, Tonge DP, Mourtada-Maarabouni M. New insights into the functional role of protein phosphatase 4 regulatory subunit PP4R3A/SMEK1 in the regulation of leukemic cell fate. Int J Biol Macromol 2023; 233:123467. [PMID: 36731689 DOI: 10.1016/j.ijbiomac.2023.123467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 01/08/2023] [Accepted: 01/25/2023] [Indexed: 02/01/2023]
Abstract
The serine/threonine protein phosphatase 4 holoenzyme consists of a PP4 catalytic subunit (PP4c), which interacts with four different regulatory subunits. Previous studies have shown that PP4c acts as a tumour suppressor. Emerging evidence suggests that the protein phosphatase 4 regulatory subunits might regulate cell fate independently of PP4c. To this end, we investigated the role of PP4R3A (SMEK1) in Jurkat and CEM-C7 leukemic cell lines. SMEK1 overexpression decreased cell growth, increased spontaneous apoptosis, and reduced the colony forming ability of leukemic cells. Conversely, siRNA-mediated silencing of SMEK1 led to increased short and long-term survival in these cells. Phospho-protein arrays revealed that increased expression of SMEK1 affected the phosphorylation of key proteins involved in MAPK3, AKT, JAK/STAT, NFκB and TGFβ signalling pathways. These proteins include transcription factors such as NFκB, STAT3, c-JUN, SMAD1, and SMAD5, suggesting a role for SMEK1 in the regulation of gene expression. RNA sequencing confirmed the role of SMEK1 in the regulation of gene expression. RNA sequencing also confirmed the tumour suppressor role of SMEK1. Taken together, this study shows that SMEK1 regulates leukemic T cell survival, indicating that SMEK1 dysfunction may be important in the development and progression of leukemia.
Collapse
Affiliation(s)
- Nadieh Kavousi
- School of Life Sciences, Faculty of Natural Sciences, Keele University, Newcastle-under-Lyme ST5 5BG, UK
| | - Daniel P Tonge
- School of Life Sciences, Faculty of Natural Sciences, Keele University, Newcastle-under-Lyme ST5 5BG, UK
| | - Mirna Mourtada-Maarabouni
- School of Life Sciences, Faculty of Natural Sciences, Keele University, Newcastle-under-Lyme ST5 5BG, UK.
| |
Collapse
|
8
|
Peng J, He C, Yan H, Zhou W. Prognostic value of genes related to cancer-associated fibroblasts in lung adenocarcinoma. Technol Health Care 2023; 31:2339-2354. [PMID: 37661904 DOI: 10.3233/thc-230453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
BACKGROUND Although it has been established that cancer-associated fibroblasts (CAFs) facilitate tumor development, the relationship between CAFs and the prognosis of patients with lung adenocarcinoma (LUAD) has not been extensively explored. OBJECTIVE This study was formulated to investigate the prognostic value of CAF-related genes in LUAD. METHODS Differential analysis was carried out with TCGA-LUAD dataset as the training set. By overlapping differentially expressed genes (DEGs) with genes associated with CAF, CAF-related DEGs specific to LUAD were obtained. A prognostic risk model was constructed by Lasso and Cox regression analysis, and samples were grouped according to median risk score. The efficacy of the model was accessed through survival curve and receiver operating characteristic curve (ROC) analyses, with the validation set for verification. Risk score combined with clinical factors was utilized for Cox analysis to verify the independence of the model, and a nomogram was drawn. GSEA was performed on different risk groups. Immunologic infiltration and tumor mutational burden were assessed in different risk groups. RESULTS Eleven feature genes including DLGAP5, KCNE2, UPK2, NPAS2, ARHGAP11A, ANGPTL4, ANLN, DKK1, SMUG1, C16orf74, and ACAD8 were identified, based on which a prognostic model was constructed. Risk score could predict the prognosis of LUAD patients and could be an independent prognostic factor for LUAD patients. GSEA outcomes displayed significant enrichment of genes in the high-risk group in the P53 SIGNALING PATHWAY. In comparison to the low-risk group, the high-risk group exhibited a decreased degree of immune infiltration and an elevated level of tumor mutational burden. CONCLUSION An 11-gene model was constructed based on CAF-related genes to predict LUAD prognosis. This model represented an independent prognostic factor for LUAD.
Collapse
Affiliation(s)
- Jigui Peng
- Department of Cardiothoracic Surgery, Ningde Municipal Hospital Affiliated to Ningde Normal University, Ningde, Fujian, China
| | - Changjin He
- Department of Cardiothoracic Surgery, School of Clinical Medicine, Fujian Medical University, Ningde Municipal Hospital, Ningde, Fujian, China
| | - Haiqiang Yan
- Department of Cardiothoracic Surgery, Ningde Municipal Hospital Affiliated to Ningde Normal University, Ningde, Fujian, China
| | - Wang Zhou
- Department of Cardiothoracic Surgery, Ningde Municipal Hospital Affiliated to Ningde Normal University, Ningde, Fujian, China
| |
Collapse
|
9
|
Lin Q, Chen Z, Shen ZL, Xue F, Qin JJ, Kang XP, Chen ZR, Xia ZY, Gao L, Chen XZ. TRAF3IP3 promotes glioma progression through the ERK signaling pathway. Front Oncol 2022; 12:776834. [PMID: 36185204 PMCID: PMC9523251 DOI: 10.3389/fonc.2022.776834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 08/19/2022] [Indexed: 12/02/2022] Open
Abstract
TRAF3IP3 was reportedly associated with poor prognosis in patients with melanoma; however, its role in glioma is unknown. We aimed to demonstrate the relationship between TRAF3IP3 and glioma and to investigate the potential role of TRAF3IP3 in glioma. Datasets were collected from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. We used the Wilcoxon rank-sum test to compared TRAF3IP3 expression in normal and glioma tissues. Kaplan–Meier analysis was performed to evaluate the correlation between TRAF3IP3 and patient survival rate. Gene set enrichment analysis (GSEA) was used to annotate the biological function of TRAF3IP3 in glioma. We also examined the effects of TRAF3IP3 on glioma progression, including characteristics such as cell proliferation, migration, and invasion, using cell proliferation, wound healing, and Transwell assays, respectively, paired with in vitro glioma cell lines and in vivo mouse xenograft models to determine the molecular mechanisms underlying these effects. High TRAF3IP3 expression in glioma tissues was associated with patients with neoplasm cancer tissue source site, and poorer overall survival (OS) (p = 0.03), which was validated using TCGA. GSEA revealed the enrichment of neuroactive ligand–receptor interactions, the olfactory pathway, proteasome pathway, cytokine–cytokine receptor interactions, and calcium signaling pathway in the TRAF3IP3 high-expression phenotype. TRAF3IP3 knockdown markedly suppressed the proliferation, migration, and invasion abilities of U251 glioma cells, whereas TRAF3IP3 overexpression notably promoted the progression of U118 cell tumors. Mechanistic studies revealed that TRAF3IP3 upregulated p-ERK expression in glioma cells. Notably, the ERK signaling pathway inhibitor U0126 drastically attenuated the effects of TRAF3IP3 on p-ERK and markedly blocked its tumor-promoting activity. TRAF3IP3 overexpression also promoted in vivo tumor growth in a nude mouse xenograft model. Collectively, TRAF3IP3 stimulates glioma cell proliferation, migration, and invasion, at least partly by activating the ERK signaling pathway. We hypothesize that TRAF3IP3 may participate in glioma development via the ERK signaling pathway and that elevated TRAF3IP3 expression may serve as a potential biomarker for glioma prognosis.
Collapse
Affiliation(s)
- Qi Lin
- Department of Neurosurgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhen Chen
- Department of Neurosurgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhao-Li Shen
- Department of Neurosurgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fei Xue
- Department of Neurosurgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jia-Jun Qin
- Tongji University School of Medicine, Shanghai, China
| | - Xi-Peng Kang
- Department of Neurosurgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhong-Rong Chen
- Department of Neurosurgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhong -Yuan Xia
- Department of Neurosurgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liang Gao
- Department of Neurosurgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Liang Gao, ; Xian-Zhen Chen,
| | - Xian-Zhen Chen
- Department of Neurosurgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Liang Gao, ; Xian-Zhen Chen,
| |
Collapse
|
10
|
Zheng L, Cai X, Song J, Shi H, Zhang J, Ke X, Li H, Chen Y. MicroRNA-30c-2-3p represses malignant progression of gastric adenocarcinoma cells via targeting ARHGAP11A. Bioengineered 2022; 13:14534-14544. [PMID: 35754342 PMCID: PMC9342190 DOI: 10.1080/21655979.2022.2090222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
MicroRNAs are crucial tumor regulators to tumor development and progression. MiR-30c-2-3p can suppress malignant progression of tumor cells, but no study has reported the modulatory process of miR-30c-2-3p in gastric adenocarcinoma (GA). We herein investigated role of miR-30c-2-3p in GA cells. Here, we evaluated gene level in cancer cells by qRT-PCR. CCK-8, colony formation, flow cytometry, and transwell assays revealed biological functions of miR-30c-2-3p and ARHGAP11A. Genes downstream of miR-30c-2-3p were acquired through bioinformatics analysis. Our results suggested a low level of miR-30c-2-3p in GA tissue and cells, while its high expression could repress the malignant progression and promote cell cycle arrest and apoptosis of GA cells. Besides, ARHGAP11A was downstream of miR-30c-2-3p, with up-regulated ARHGAP11A facilitating malignant progression and repressing cell cycle arrest and apoptosis of GA cells. In addition, changes in GA cell functions caused by high ARHGAP11A expression could be partially offset by enhancing miR-30c-2-3p. Thus, our observations indicated that miR-30c-2-3p was a tumor repressor that could inhibit GA progression via modulating ARHGAP11A.
Collapse
Affiliation(s)
- Liang Zheng
- Department of Abdominal Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Xiongchao Cai
- Department of Abdominal Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Jintian Song
- Department of Abdominal Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Huaijing Shi
- Department of Gynecology Surgery, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Jiulong Zhang
- Department of Thoracic Surgical Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Xi Ke
- Department of Abdominal Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Hui Li
- Department of Abdominal Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Yigui Chen
- Department of Abdominal Oncology, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, China
| |
Collapse
|
11
|
Ju W, Zhang G, Zhang X, Wang J, Wu T, Li H. Involvement of MiRNA-211-5p and Arhgap11a Interaction During Osteogenic Differentiation of MC3T3-E1 Cells. Front Surg 2022; 9:857170. [PMID: 35495761 PMCID: PMC9051074 DOI: 10.3389/fsurg.2022.857170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/17/2022] [Indexed: 11/17/2022] Open
Abstract
Objective MicroRNAs (miRNAs) are well-recognized for their abilities to regulate gene expression post-transcriptionally in plants and animals. Recently, miRNA-messenger RNA (mRNA) regulatory relationships have been confirmed during biological processes, including osteogenic differentiation. This study aimed to find out more candidate miRNA-mRNA pairs involved in the osteogenic differentiation of MC3T3-E1 cells. Methods An MC3T3-E1-based microarray dataset (accessioned as GSE46400) downloaded from the Gene Expression Omnibus included MC3T3-E1 cells with or without 14-day osteoblast differentiation osteoblast induction. Multiple miRNA-mRNA prediction databases were searched by differentially expressed genes (DEGs) to obtain pairs of a miRNA-DEG regulatory network. The MC3T3-E1 cells were cultured and incubated in the osteogenic differentiation medium for 14 days. The expressions of candidate miRNAs and mRNAs were determined by real-time quantitative PCR(RT-qPCR) in MC3T3-E1 cells. The miRNA-mRNA interactions were verified by dual-luciferase reporter gene assays and experiments using mimics miRNA or their inhibitors. Results We identified 715 upregulated DEGs and 603 downregulated DEGs between MC3T3-E1 cells with and without osteoblast induction by analyzing the raw data of the GSE46400 dataset. There were 7 overlapped miRNA-mRNA pairs identified during osteogenic differentiation of MC3T3-E1 cells, including mmu-miR-204-5p-Arhgap11a, mmu-miR-211-5p-Arhgap11a, mmu-miR-24-3p-H2afx, mmu-miR-3470b-Chek2, mmu-miR-3470b-Dlgap5, mmu-miR-466b-3p-Chek1, and mmu-miR-466c-3p-Chek1. The Arhgap11a, H2afx, Chek2, Dlgap5, and Chek1 were hub genes downregulated in MC3T3-E1 cells after osteogenic differentiation, verified by RT-qPCR results. The RT-qPCR also determined declined expressions of miR-204-5p and miR-24-3p concomitant with elevated expressions of miR-211-5p, miR-3470b, miR-466b-3p, and miR-466c-3p in the MC3T3-E1 cells, with osteoblast induction compared with undifferentiated MC3T3-E1 cells. Dual-luciferase reporter gene assays demonstrated Arhgap11a as the target of miR-211-5p. MiR-211-5p upregulation by its mimic increased Arhgap11a expression in MC3T3-E1 cells. Conclusion Our study characterizes miR-211-5p targeting Arhgap11a promotes the osteogenic differentiation of MC3T3-E1 cells, which provides novel targets to promote the osteogenesis process during bone repair.
Collapse
Affiliation(s)
- Wenwen Ju
- Department of Endocrinology (I), The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Guangfeng Zhang
- Departments of Magnetic Resonance Imaging (MRI), The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Xu Zhang
- Department of Endocrinology, Zhongshan City People's Hospital, Zhongshan, China
| | - Jingting Wang
- Department of Endocrinology (I), The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
| | - Tong Wu
- Mental & Health College, Qiqihar Medical University, Qiqihar, China
| | - Huafeng Li
- Department of Endocrinology (I), The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, China
- *Correspondence: Huafeng Li
| |
Collapse
|
12
|
Fixing the GAP: the role of RhoGAPs in cancer. Eur J Cell Biol 2022; 101:151209. [DOI: 10.1016/j.ejcb.2022.151209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/29/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
|
13
|
ARHGAP11A Promotes the Malignant Progression of Gastric Cancer by Regulating the Stability of Actin Filaments through TPM1. JOURNAL OF ONCOLOGY 2021; 2021:4146910. [PMID: 34912455 PMCID: PMC8668285 DOI: 10.1155/2021/4146910] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/07/2021] [Accepted: 11/12/2021] [Indexed: 12/29/2022]
Abstract
The mechanism underlying the poor prognosis of gastric cancer, including its high degree of malignancy, invasion, and metastasis, is extremely complicated. Rho GTPases are involved in the occurrence and development of a variety of malignant tumors. ARHGAP11A, in the Rho GTPase activating protein family, is highly expressed in gastric cancer, but its function and mechanism have not yet been explored. In this study, the effect of ARHGAP11A on the occurrence and development of gastric cancer and the mechanism related to this effect were studied. The expression of ARHGAP11A was increased in gastric cancer cells and tissues, and high ARHGAP11A expression in tissues was related to the degree of tumor differentiation and poor prognosis. Moreover, ARHGAP11A knockout significantly inhibited cell proliferation, cell migration, and invasion in vitro and significantly inhibited the tumorigenic ability of gastric cancer cells in nude mice in vivo. Further studies revealed that ARHGAP11A promotes the malignant progression of gastric cancer cells by interacting with TPM1 to affect cell migration and invasion and the stability of actin filaments. These results suggest that ARHGAP11A plays an important role in gastric cancer and may become a useful prognostic biomarker and therapeutic target for gastric cancer patients.
Collapse
|