1
|
Wang GC, Gan X, Zeng YQ, Chen X, Kang H, Huang SW, Hu WH. The Role of NCS1 in Immunotherapy and Prognosis of Human Cancer. Biomedicines 2023; 11:2765. [PMID: 37893139 PMCID: PMC10604305 DOI: 10.3390/biomedicines11102765] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/01/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
The Neural Calcium Sensor1 (NCS1) is a crucial protein that binds to Ca2+ and is believed to play a role in regulating tumor invasion and cell proliferation. However, the role of NCS1 in immune infiltration and cancer prognosis is still unknown. Our study aimed to explore the expression profile, immune infiltration pattern, prognostic value, biological function, and potential compounds targeting NCS1 using public databases. High expression of NCS1 was detected by immune histochemical staining in LIHC (Liver hepatocellular carcinoma), BRCA (Breast invasive carcinoma), KIRC (Kidney renal clear cell carcinoma), and SKCM (Skin Cutaneous Melanoma). The expression of NCS1 in cancer was determined by TCGA (The Cancer Genome Atlas Program), GTEx (The Genotype-Tissue Expression), the Kaplan-Meier plotter, GEO (Gene Expression Omnibus), GEPIA2.0 (Gene Expression Profiling Interactive Analysis 2.0), HPA (The Human Protein Atlas), UALCAN, TIMER2.0, TISIDB, Metascape, Drugbank, chEMBL, and ICSDB databases. NCS1 has genomic mutations as well as aberrant DNA methylation in multiple cancers compared to normal tissues. Also, NCS1 was significantly different in the immune microenvironment, tumor mutational burden (TMB), microsatellite instability (MSI), and immune infiltrate-associated cells in different cancers, which could be used for the typing of immune and molecular subtypes of cancer and the presence of immune checkpoint resistance in several cancers. Univariate regression analysis, multivariate regression analysis, and gene enrichment analysis to construct prognostic models revealed that NCS1 is involved in immune regulation and can be used as a prognostic biomarker for SKCM, LIHC, BRCA, COAD, and KIRC. These results provide clues from a bioinformatic perspective and highlight the importance of NCS1 in a variety of cancers.
Collapse
Affiliation(s)
- Gen-Chun Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xin Gan
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yun-Qian Zeng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xin Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hao Kang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shuai-Wen Huang
- Department of General Practice, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wei-Hua Hu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
2
|
Li W, Wang M, Ma W, Liu P, Zhang M, He J, Cui Y. Temozolomide protects against the progression of glioblastoma via SOX4 downregulation by inhibiting the LINC00470-mediated transcription factor EGR2. CNS Neurosci Ther 2023; 29:2292-2307. [PMID: 36987665 PMCID: PMC10352878 DOI: 10.1111/cns.14181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 02/17/2023] [Accepted: 03/05/2023] [Indexed: 03/30/2023] Open
Abstract
OBJECTIVE Temozolomide is extensively applied in chemotherapy for glioblastoma with unclear exact action mechanisms. This article seeks to address the potential molecular mechanisms in temozolomide therapy for glioblastoma involving LINC00470. METHODS Bioinformatics analysis was conducted to predict the potential mechanism of LINC00470 in glioblastoma, which was validated by dual-luciferase reporter, RIP, ChIP, and RNA pull-down assays. LINC00470 expression and the predicted downstream transcription factor early growth response 2 (EGR2) were detected in the collected brain tissues from glioblastoma patients. Following temozolomide treatment and/or gain- and loss-of-function approaches in glioblastoma cells, cell viability, invasion, migration, cycle distribution, angiogenesis, autophagy, and apoptosis were measured. In addition, the expression of mesenchymal surface marker proteins was assessed by western blot. Tumor xenograft in nude mice was conducted for in vivo validation. RESULTS Mechanistic analysis and bioinformatics analysis revealed that LINC00470 transcriptionally activated SRY-related high-mobility-group box 4 (SOX4) through the transcription factor EGR2. LINC00470 and EGR2 were highly expressed in brain tissues of glioblastoma patients. LINC00470 and EGR2 mRNA expression gradually decreased with increasing concentrations of temozolomide in glioblastoma cells, and SOX4 expression was reduced in cells by temozolomide and LINC00470 knockdown. Temozolomide treatment induced cell cycle arrest, diminished cell viability, migration, invasion, and angiogenesis, and increased apoptosis and autophagy in glioblastoma, which was counteracted by overexpressing LINC00470 or SOX4 but was further promoted by LINC00470 knockdown. Temozolomide restrained glioblastoma growth and angiogenesis in vivo, while LINC00470 or SOX4 overexpression nullified but LINC00470 knockdown further facilitated these trends. CONCLUSION Conclusively, temozolomide repressed glioblastoma progression by repressing the LINC00470/EGR2/SOX4 axis.
Collapse
Affiliation(s)
- Wenyang Li
- Department of NeurosurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Ming Wang
- Department of NeurosurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Wenjia Ma
- Department of NeurosurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Ping Liu
- Department of OncologyThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Mingming Zhang
- Department of NeurosurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Jiarong He
- Department of NeurosurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Yan Cui
- Department of NeurosurgeryThe Second Xiangya Hospital of Central South UniversityChangshaChina
| |
Collapse
|
3
|
Li P, Chen Y, Peng Y, Zhang Y, Zhou H, Chen X, Li T, Li S, Yang H, Wu C, Zheng C, Zhu J, You F, Li L, Qin X, Liu Y. Notch-1 signaling promotes reattachment of suspended cancer cells by cdc42-dependent microtentacles formation. Cancer Sci 2021; 112:4894-4908. [PMID: 34582616 PMCID: PMC8645759 DOI: 10.1111/cas.15146] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/15/2021] [Accepted: 09/23/2021] [Indexed: 01/14/2023] Open
Abstract
Circulating tumor cells (CTCs) are associated with a higher risk of metastasis in tumor patients. The adhesion and arrest of CTCs at a secondary site is an essential prerequisite for the occurrence of tumor metastasis. CTC reattachment has shown to be dependent on microtentacle (McTN) formation in vivo. However, the specific molecular mechanism of McTN formation in suspended cancer cells remains largely unclear. Here, we demonstrated that the activation of Notch-1 signaling triggers McTN formation to facilitate cell reattachment in suspended cell culture conditions. Moreover, molecular mechanistic studies revealed that McTN formation is governed by the balance between microtubule-driven outgrowth and actomyosin-driven cell contractility. The activation of Notch-1 downregulates the acetylation level of microtubules via the Cdc42/HDAC6 pathway, which contributes to microtubule polymerization. Simultaneously, Notch-1 signaling-induced Cdc42 activation also reduced phosphorylation of myosin regulatory light chain, leading to cell contractility attenuation. Altogether, these results defined a novel mechanism by which Notch-1 signaling disturbs the balance between the expansion of microtubules and contraction of the cortical actin, which promotes McTN formation and cell reattachment. Our findings provide a new perspective on the effective therapeutic target to prevent CTC reattachment.
Collapse
Affiliation(s)
- Ping Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Yu Chen
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Yueting Peng
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Yixi Zhang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Hanying Zhou
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiangyan Chen
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Tingting Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Shun Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Hong Yang
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Chunhui Wu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Chuan Zheng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Zhu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fengming You
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Li
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiang Qin
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China
| | - Yiyao Liu
- Department of Biophysics, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, China.,TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
4
|
Zimu Z, Jia Z, Xian F, Rui M, Yuting R, Yuan W, Tianhong W, Mian M, Yinlong L, Enfang S. Decreased Expression of PACSIN1 in Brain Glioma Samples Predicts Poor Prognosis. Front Mol Biosci 2021; 8:696072. [PMID: 34422904 PMCID: PMC8375027 DOI: 10.3389/fmolb.2021.696072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 07/27/2021] [Indexed: 12/03/2022] Open
Abstract
Gliomas are the most severe brain tumours with a poor prognosis. Although surgery, postoperative radiotherapy and chemotherapy can improve the survival rate of glioma patients, the prognosis of most glioma patients is still poor. In recent years, the influence of gene-targeted therapy on gliomas has been gradually discovered, and intervening the occurrence and development of brain gliomas from the perspective of the gene will significantly improve treatment prognosis. Protein Kinase C and Casein Kinase Substrate in Neurons 1 (PACSIN1) is a member of the conserved peripheral membrane protein family in eukaryotes. Improper expression of PACSIN1 can lead to neurological diseases such as Huntington’s disease and schizophrenia. However, its relationship with tumours or even gliomas has not been explored. The study aims to explore PACSIN1 as a prognostic factor that can predict overall survival (OS) for gliomas. We collected the data from CGGA, TCGA, GEO databases and the pathological glioma tissue specimens from 15 clinical glioma patients surgically resected. The differential expression of PACSIN1 in various clinical indicators, the genes related to PACSIN1 expression, the prognostic value of PACSIN1 and the functional annotations and pathway analysis of differently expressed genes (DEGs) were analysed. The results revealed that PACSIN1 had low expression levels in grade IV, IDH1 wild-type and 1p/19q non-codel group gliomas, and PACSIN1 was considered a mesenchymal molecular subtype marker. PACSIN1 expression is positively correlated with OS in all gliomas and it was found that PACSIN1 influenced the occurrence and development of gliomas through synaptic transmission. The PACSIN1 expression is negatively correlated with the malignant degree of gliomas and positively associated with the OS, indicating that PACSIN1 would play an essential role in the occurrence and development of gliomas and might be a potential new biomarker and targeted therapy site for gliomas.
Collapse
Affiliation(s)
- Zhou Zimu
- School of Nursing, Nanjing Medical University, Nanjing, China.,Cancer Nursing Research Branch, Nursing Research Center, Nanjing Medical University, Nanjing, China
| | - Zhang Jia
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Fu Xian
- Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Ma Rui
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Ren Yuting
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Wei Yuan
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Wen Tianhong
- School of Nursing, Nanjing Medical University, Nanjing, China
| | - Ma Mian
- Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Liu Yinlong
- Department of Neurosurgery, The Affiliated Huashan Hospital, Fudan University, Shanghai, China.,Department of Neurosurgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Shan Enfang
- School of Nursing, Nanjing Medical University, Nanjing, China.,Cancer Nursing Research Branch, Nursing Research Center, Nanjing Medical University, Nanjing, China
| |
Collapse
|
5
|
Li C, Guo H, Wang C, Zhan W, Tan Q, Xie C, Sharma A, Sharma HS, Chen L, Zhang Z. Network pharmacological mechanism of Cinobufotalin against glioma. PROGRESS IN BRAIN RESEARCH 2021; 265:119-137. [PMID: 34560920 DOI: 10.1016/bs.pbr.2021.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Cinobufotalin was extracted from the skin of Chinese giant salamander or black sable with good clinical effect against tumor. This study aims to explore the mechanism of Cinobufotalin components and predict the target of action of Cinobufotalin on glioma. METHODS The active components of Cinobufotalin were screened by the Chinese medicine pharmacology database and analysis platform (TCMSP), PubChem database, etc. The potential molecular components and targets were identified and enrichment analysis was conducted through the construction of related networks and analysis of their characteristics. Relevant targets of glioma were searched through TTD, DRUGBANK, and other databases, and the intersection was found and the key targets were found too. RESULTS A total of 21 active components and 184 target genes of Cinobufotalin were found. According to the enrichment analysis results, the pharmacological mechanism of Cinobufotalin mainly includes inhibition of the cell cycle, promotion of cell apoptosis, and regulation of immunity. On this basis, RAC1, FOS, and NOS3 can be preliminarily predicted as potential targets of Cinobufotalin in the treatment of glioma. CONCLUSIONS The screening of active ingredients and target prediction based on network pharmacology can provide a new research idea for the multi-target treatment of glioma with Cinobufotalin.
Collapse
Affiliation(s)
- Cong Li
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province Hospital of Chinese Medical, Guangzhou, China
| | - Hanyu Guo
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province Hospital of Chinese Medical, Guangzhou, China
| | - Chao Wang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province Hospital of Chinese Medical, Guangzhou, China
| | - Wengang Zhan
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province Hospital of Chinese Medical, Guangzhou, China
| | - Qijia Tan
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province Hospital of Chinese Medical, Guangzhou, China
| | - Caijun Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province Hospital of Chinese Medical, Guangzhou, China
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Lin Chen
- Department of Neurosurgery, Dongzhimen Hospital of Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Zhiqiang Zhang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Province Hospital of Chinese Medical, Guangzhou, China.
| |
Collapse
|
6
|
Margarido AS, Bornes L, Vennin C, van Rheenen J. Cellular Plasticity during Metastasis: New Insights Provided by Intravital Microscopy. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a037267. [PMID: 31615867 DOI: 10.1101/cshperspect.a037267] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Metastasis is a highly dynamic process during which cancer and microenvironmental cells undergo a cascade of events required for efficient dissemination throughout the body. During the metastatic cascade, tumor cells can change their state and behavior, a phenomenon commonly defined as cellular plasticity. To monitor cellular plasticity during metastasis, high-resolution intravital microscopy (IVM) techniques have been developed and allow us to visualize individual cells by repeated imaging in animal models. In this review, we summarize the latest technological advancements in the field of IVM and how they have been applied to monitor metastatic events. In particular, we highlight how longitudinal imaging in native tissues can provide new insights into the plastic physiological and developmental processes that are hijacked by cancer cells during metastasis.
Collapse
Affiliation(s)
- Andreia S Margarido
- Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - Laura Bornes
- Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - Claire Vennin
- Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - Jacco van Rheenen
- Molecular Pathology, Oncode Institute, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| |
Collapse
|
7
|
Matsuda M, Terai K. Experimental pathology by intravital microscopy and genetically encoded fluorescent biosensors. Pathol Int 2020; 70:379-390. [PMID: 32270554 PMCID: PMC7383902 DOI: 10.1111/pin.12925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/13/2020] [Accepted: 03/16/2020] [Indexed: 01/03/2023]
Abstract
The invention of two‐photon excitation microscopes widens the potential application of intravital microscopy (IVM) to the broad field of experimental pathology. Moreover, the recent development of fluorescent protein‐based, genetically encoded biosensors provides an ideal tool to visualize the cell function in live animals. We start from a brief review of IVM with two‐photon excitation microscopes and genetically encoded biosensors based on the principle of Förster resonance energy transfer (FRET). Then, we describe how IVM using biosensors has revealed the pathogenesis of several disease models.
Collapse
Affiliation(s)
- Michiyuki Matsuda
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Kenta Terai
- Research Center for Dynamic Living Systems, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
8
|
Ueyama T. Rho-Family Small GTPases: From Highly Polarized Sensory Neurons to Cancer Cells. Cells 2019; 8:cells8020092. [PMID: 30696065 PMCID: PMC6406560 DOI: 10.3390/cells8020092] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/19/2019] [Accepted: 01/23/2019] [Indexed: 12/22/2022] Open
Abstract
The small GTPases of the Rho-family (Rho-family GTPases) have various physiological functions, including cytoskeletal regulation, cell polarity establishment, cell proliferation and motility, transcription, reactive oxygen species (ROS) production, and tumorigenesis. A relatively large number of downstream targets of Rho-family GTPases have been reported for in vitro studies. However, only a small number of signal pathways have been established at the in vivo level. Cumulative evidence for the functions of Rho-family GTPases has been reported for in vivo studies using genetically engineered mouse models. It was based on different cell- and tissue-specific conditional genes targeting mice. In this review, we introduce recent advances in in vivo studies, including human patient trials on Rho-family GTPases, focusing on highly polarized sensory organs, such as the cochlea, which is the primary hearing organ, host defenses involving reactive oxygen species (ROS) production, and tumorigenesis (especially associated with RAC, novel RAC1-GSPT1 signaling, RHOA, and RHOBTB2).
Collapse
Affiliation(s)
- Takehiko Ueyama
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe 657-8501, Japan.
| |
Collapse
|
9
|
High expression of GALNT7 promotes invasion and proliferation of glioma cells. Oncol Lett 2018; 16:6307-6314. [PMID: 30405766 PMCID: PMC6202485 DOI: 10.3892/ol.2018.9498] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 07/20/2018] [Indexed: 01/06/2023] Open
Abstract
Polypeptide-N-acetyl-galactosaminlytransferase 7 (GALNT7), a member of the GalNAc-transferase family, has not been previously evaluated as a prognostic factor of glioblastoma (GBM) or low-grade glioma (LGG). Based on The Cancer Genome Atlas database and bioinformatics analyses, the expression of GALNT7 was demosntrated to be higher in GBM and LGG tissues than in normal brain tissue. The expression levels of GANLT7 were associated with age, tumor grade, survival rate, disease-free survival time and overall survival time. Gene correlation and gene-set enrichment analyses suggested that GALNT7 may affect the proliferative and invasive abilities of glioma cells through multiple signaling pathways, including regulation of the actin cytoskeleton, natural killer cell-mediated cytotoxicity, the janus kinase-signal transducer and activator of transcription (STAT) signaling pathway, cell adhesion molecules and extracellular matrix receptor interaction pathways. Furthermore, 5 target genes of GALNT7 involved in these signaling pathways were identified, including Crk, Rac family small GTPase 1, STAT3, poliovirus receptor and Tenascin C. In summary, high expression of GALNT7 was associated with poor prognosis of glioma, and may be used as an effective biomarker of glioma.
Collapse
|
10
|
Dali R, Verginelli F, Pramatarova A, Sladek R, Stifani S. Characterization of a FOXG1:TLE1 transcriptional network in glioblastoma-initiating cells. Mol Oncol 2018; 12:775-787. [PMID: 29316219 PMCID: PMC5983107 DOI: 10.1002/1878-0261.12168] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 12/12/2017] [Accepted: 12/21/2017] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) is the most common and deadly malignant brain cancer of glial cell origin, with a median patient survival of less than 20 months. Transcription factors FOXG1 and TLE1 promote GBM propagation by supporting maintenance of brain tumour-initiating cells (BTICs) with stem-like properties. Here, we characterize FOXG1 and TLE1 target genes in GBM patient-derived BTICs using ChIP-Seq and RNA-Seq approaches. These studies identify 150 direct FOXG1 targets, several of which are also TLE1 targets, involved in cell proliferation, differentiation, survival, chemotaxis and angiogenesis. Negative regulators of NOTCH signalling, including CHAC1, are among the transcriptional repression targets of FOXG1:TLE1 complexes, suggesting a crosstalk between FOXG1:TLE1 and NOTCH-mediated pathways in GBM. These results provide previously unavailable insight into the transcriptional programs underlying the tumour-promoting functions of FOXG1:TLE1 in GBM.
Collapse
Affiliation(s)
- Rola Dali
- Department of Neurology and NeurosurgeryMontreal Neurological InstituteMcGill UniversityMontrealCanada
- McGill Center for BioinformaticsMcGill UniversityMontrealCanada
| | - Federica Verginelli
- Department of Neurology and NeurosurgeryMontreal Neurological InstituteMcGill UniversityMontrealCanada
- Present address:
Laboratory of Cancer Stem Cell ResearchCandiolo Cancer InstituteFPO‐IRCCSCandioloItaly
| | - Albena Pramatarova
- Departments of Human Genetics and MedicineMcGill UniversityMontrealCanada
| | - Robert Sladek
- Departments of Human Genetics and MedicineMcGill UniversityMontrealCanada
| | - Stefano Stifani
- Department of Neurology and NeurosurgeryMontreal Neurological InstituteMcGill UniversityMontrealCanada
| |
Collapse
|
11
|
Bright MD, Clarke PA, Workman P, Davies FE. Oncogenic RAC1 and NRAS drive resistance to endoplasmic reticulum stress through MEK/ERK signalling. Cell Signal 2018; 44:127-137. [PMID: 29329780 PMCID: PMC6562199 DOI: 10.1016/j.cellsig.2018.01.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/18/2017] [Accepted: 01/07/2018] [Indexed: 12/16/2022]
Abstract
Cancer cells are able to survive under conditions that cause endoplasmic reticulum stress (ER-stress), and can adapt to this stress by upregulating cell-survival signalling pathways and down-regulating apoptotic pathways. The cellular response to ER-stress is controlled by the unfolded protein response (UPR). Small Rho family GTPases are linked to many cell responses including cell growth and apoptosis. In this study, we investigate the function of small GTPases in cell survival under ER-stress. Using siRNA screening we identify that RAC1 promotes cell survival under ER-stress in cells with an oncogenic N92I RAC1 mutation. We uncover a novel connection between the UPR and N92I RAC1, whereby RAC1 attenuates phosphorylation of EIF2S1 under ER-stress and drives over-expression of ATF4 in basal conditions. Interestingly, the UPR connection does not drive resistance to ER-stress, as knockdown of ATF4 did not affect this. We further investigate cancer-associated kinase signalling pathways and show that RAC1 knockdown reduces the activity of AKT and ERK, and using a panel of clinically important kinase inhibitors, we uncover a role for MEK/ERK, but not AKT, in cell viability under ER-stress. A known major activator of ERK phosphorylation in cancer is oncogenic NRAS and we show that knockdown of NRAS in cells, which bear a Q61 NRAS mutation, sensitises to ER-stress. These findings highlight a novel mechanism for resistance to ER-stress through oncogenic activation of MEK/ERK signalling by small GTPases.
Collapse
Affiliation(s)
- Michael D Bright
- The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK.
| | - Paul A Clarke
- The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK
| | - Paul Workman
- The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK
| | - Faith E Davies
- The Institute of Cancer Research, 15 Cotswold Road, Sutton SM2 5NG, UK
| |
Collapse
|
12
|
Luo W, Song Z, Sun H, Liang J, Zhao S. Bergamottin, a natural furanocoumarin abundantly present in grapefruit juice, suppresses the invasiveness of human glioma cells via inactivation of Rac1 signaling. Oncol Lett 2017; 15:3259-3266. [PMID: 29435067 DOI: 10.3892/ol.2017.7641] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 02/14/2017] [Indexed: 12/11/2022] Open
Abstract
The aim of the present study was to explore the effect of bergamottin, a natural furanocoumarin obtained from grapefruit juice, on the invasiveness of human glioma cells. The results revealed that treatment with bergamottin for 48 h significantly inhibited wound-healing migration and Matrigel invasion of human glioma cells, compared with untreated cells (P<0.05). Bergamottin treatment caused a significant decrease in the expression and secretion of matrix metalloproteinase (MMP)-9 in glioma cells compared with untreated cells (P<0.05). A Rac1-GTP pull-down assay demonstrated that bergamottin-treated glioma cells had a significantly decreased level of active Rac1-GTP compared with untreated cells (P<0.05). However, bergamottin had no significant effect on cell division cycle 42 activity. Expression of constitutively activated Rac1 almost completely restored the migration and invasion of bergamottin-treated glioma cells. In addition, bergamottin-induced downregulation of MMP-9 was prevented by exogenous activated Rac1. The results of the present study demonstrated that bergamottin exhibits anti-invasive activity in human glioma cells through the inactivation of Rac1 and downregulation of MMP-9.
Collapse
Affiliation(s)
- Wenzheng Luo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhenyu Song
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Hongwei Sun
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Junxin Liang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shanshan Zhao
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
13
|
Ishii T, Ueyama T, Shigyo M, Kohta M, Kondoh T, Kuboyama T, Uebi T, Hamada T, Gutmann DH, Aiba A, Kohmura E, Tohda C, Saito N. A Novel Rac1-GSPT1 Signaling Pathway Controls Astrogliosis Following Central Nervous System Injury. J Biol Chem 2016; 292:1240-1250. [PMID: 27941025 DOI: 10.1074/jbc.m116.748871] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 11/29/2016] [Indexed: 01/31/2023] Open
Abstract
Astrogliosis (i.e. glial scar), which is comprised primarily of proliferated astrocytes at the lesion site and migrated astrocytes from neighboring regions, is one of the key reactions in determining outcomes after CNS injury. In an effort to identify potential molecules/pathways that regulate astrogliosis, we sought to determine whether Rac/Rac-mediated signaling in astrocytes represents a novel candidate for therapeutic intervention following CNS injury. For these studies, we generated mice with Rac1 deletion under the control of the GFAP (glial fibrillary acidic protein) promoter (GFAP-Cre;Rac1flox/flox). GFAP-Cre;Rac1flox/flox (Rac1-KO) mice exhibited better recovery after spinal cord injury and exhibited reduced astrogliosis at the lesion site relative to control. Reduced astrogliosis was also observed in Rac1-KO mice following microbeam irradiation-induced injury. Moreover, knockdown (KD) or KO of Rac1 in astrocytes (LN229 cells, primary astrocytes, or primary astrocytes from Rac1-KO mice) led to delayed cell cycle progression and reduced cell migration. Rac1-KD or Rac1-KO astrocytes additionally had decreased levels of GSPT1 (G1 to S phase transition 1) expression and reduced responses of IL-1β and GSPT1 to LPS treatment, indicating that IL-1β and GSPT1 are downstream molecules of Rac1 associated with inflammatory condition. Furthermore, GSPT1-KD astrocytes had cell cycle delay, with no effect on cell migration. The cell cycle delay induced by Rac1-KD was rescued by overexpression of GSPT1. Based on these results, we propose that Rac1-GSPT1 represents a novel signaling axis in astrocytes that accelerates proliferation in response to inflammation, which is one important factor in the development of astrogliosis/glial scar following CNS injury.
Collapse
Affiliation(s)
- Taiji Ishii
- From the Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe 657-8501, Japan
| | - Takehiko Ueyama
- From the Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe 657-8501, Japan,
| | - Michiko Shigyo
- the Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Masaaki Kohta
- the Department of Neurosurgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Takeshi Kondoh
- the Department of Neurosurgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Tomoharu Kuboyama
- the Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Tatsuya Uebi
- From the Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe 657-8501, Japan
| | - Takeshi Hamada
- From the Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe 657-8501, Japan
| | - David H Gutmann
- the Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, and
| | - Atsu Aiba
- the Laboratory of Animal Resources, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Eiji Kohmura
- the Department of Neurosurgery, Kobe University Graduate School of Medicine, Kobe 650-0017, Japan
| | - Chihiro Tohda
- the Division of Neuromedical Science, Department of Bioscience, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan
| | - Naoaki Saito
- From the Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe 657-8501, Japan,
| |
Collapse
|
14
|
Hirata E, Kiyokawa E. Future Perspective of Single-Molecule FRET Biosensors and Intravital FRET Microscopy. Biophys J 2016; 111:1103-1111. [PMID: 27475975 DOI: 10.1016/j.bpj.2016.01.037] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/23/2015] [Accepted: 01/11/2016] [Indexed: 01/03/2023] Open
Abstract
Förster (or fluorescence) resonance energy transfer (FRET) is a nonradiative energy transfer process between two fluorophores located in close proximity to each other. To date, a variety of biosensors based on the principle of FRET have been developed to monitor the activity of kinases, proteases, GTPases or lipid concentration in living cells. In addition, generation of biosensors that can monitor physical stresses such as mechanical power, heat, or electric/magnetic fields is also expected based on recent discoveries on the effects of these stressors on cell behavior. These biosensors can now be stably expressed in cells and mice by transposon technologies. In addition, two-photon excitation microscopy can be used to detect the activities or concentrations of bioactive molecules in vivo. In the future, more sophisticated techniques for image acquisition and quantitative analysis will be needed to obtain more precise FRET signals in spatiotemporal dimensions. Improvement of tissue/organ position fixation methods for mouse imaging is the first step toward effective image acquisition. Progress in the development of fluorescent proteins that can be excited with longer wavelength should be applied to FRET biosensors to obtain deeper structures. The development of computational programs that can separately quantify signals from single cells embedded in complicated three-dimensional environments is also expected. Along with the progress in these methodologies, two-photon excitation intravital FRET microscopy will be a powerful and valuable tool for the comprehensive understanding of biomedical phenomena.
Collapse
Affiliation(s)
- Eishu Hirata
- Department of Oncologic Pathology, Kanazawa Medical University, Ishikawa, Japan.
| | - Etsuko Kiyokawa
- Department of Oncologic Pathology, Kanazawa Medical University, Ishikawa, Japan.
| |
Collapse
|
15
|
Photo-enhancement of macrophage phagocytic activity via Rac1-mediated signaling pathway: Implications for bacterial infection. Int J Biochem Cell Biol 2016; 78:206-216. [PMID: 27345261 DOI: 10.1016/j.biocel.2016.06.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 05/12/2016] [Accepted: 06/19/2016] [Indexed: 11/20/2022]
Abstract
Phagocytosis and the subsequent destruction of invading pathogens by macrophages are indispensable steps in host immune responses to microbial infections. Low-power laser irradiation (LPLI) has been found to exert photobiological effects on immune responses, but the signaling mechanisms underlying this photobiomodulation of phagocytosis remains largely unknown. Here, we demonstrated for the first time that LPLI enhanced the phagocytic activity of macrophages by stimulating the activation of Rac1. The overexpression of constitutively activated Rac1 clearly enhanced LPLI-induced phagocytosis, whereas the overexpression of dominant negative Rac1 exerted the opposite effect. The phosphorylation of cofilin was involved in the effects of LPLI on phagocytosis, which was regulated by the membrane translocation and activation of Rac1. Furthermore, the photoactivation of Rac1 was dependent on the Src/PI3K/Vav1 pathway. The inhibition of the Src/PI3K pathway significantly suppressed LPLI-induced actin polymerization and phagocytosis enhancement. Additionally, LPLI-treated mice exhibited increased survival and a decreased organ bacterial load when challenged with Listeria monocytogenes, indicating that LPLI enhanced macrophage phagocytosis in vivo. These findings highlight the important roles of the Src/PI3K/Vav1/Rac1/cofilin pathway in regulating macrophage phagocytosis and provide a potential strategy for treating phagocytic deficiency via LPLI.
Collapse
|
16
|
Takaoka S, Kamioka Y, Takakura K, Baba A, Shime H, Seya T, Matsuda M. Live imaging of transforming growth factor-β activated kinase 1 activation in Lewis lung carcinoma 3LL cells implanted into syngeneic mice and treated with polyinosinic:polycytidylic acid. Cancer Sci 2016; 107:644-52. [PMID: 26931406 PMCID: PMC4970831 DOI: 10.1111/cas.12923] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 02/01/2016] [Accepted: 02/28/2016] [Indexed: 12/15/2022] Open
Abstract
Transforming growth factor‐β activated kinase 1 (TAK1) has been shown to play a crucial role in cell death, differentiation, and inflammation. Here, we live‐imaged robust TAK1 activation in Lewis lung carcinoma 3LL cells implanted into the s.c. tissue of syngeneic C57BL/6 mice and treated with polyinosinic:polycytidylic acid (PolyI:C). First, we developed and characterized a Förster resonance energy transfer‐based biosensor for TAK1 activity. The TAK1 biosensor, named Eevee‐TAK1, responded to stress‐inducing reagents such as anisomycin, tumor necrosis factor‐α, and interleukin1‐β. The anisomycin‐induced increase in Förster resonance energy transfer was abolished by the TAK1 inhibitor (5z)‐7‐oxozeaenol. Activity of TAK1 in 3LL cells was markedly increased by PolyI:C in the presence of macrophages. 3LL cells expressing Eevee‐TAK1 were implanted into mice and observed through imaging window by two‐photon excitation microscopy. During the growth of tumor, the 3LL cells at the periphery of the tumor showed higher TAK1 activity than the 3LL cells located at the center of the tumor, suggesting that cells at the periphery of the tumor mass were under stronger stress. Injection of PolyI:C, which is known to induce regression of the implanted tumors, induced marked and homogenous TAK1 activation within the tumor tissues. The effect of PolyI:C faded within 4 days. These observations suggest that Eevee‐TAK1 is a versatile tool to monitor cellular stress in cancer tissues.
Collapse
Affiliation(s)
- Saori Takaoka
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuji Kamioka
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Innovative Techno-Hub for Integrated Medical Bio-Imaging, Kyoto University, Kyoto, Japan
| | - Kanako Takakura
- Imaging Platform for Spatio-Temporal Regulation, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ai Baba
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Hiroaki Shime
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tsukasa Seya
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Michiyuki Matsuda
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
17
|
Komatsubara AT, Matsuda M, Aoki K. Quantitative analysis of recombination between YFP and CFP genes of FRET biosensors introduced by lentiviral or retroviral gene transfer. Sci Rep 2015; 5:13283. [PMID: 26290434 PMCID: PMC4542544 DOI: 10.1038/srep13283] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 07/22/2015] [Indexed: 11/25/2022] Open
Abstract
Biosensors based on the principle of Förster (or fluorescence) resonance energy transfer (FRET) have been developed to visualize spatio-temporal dynamics of signalling molecules in living cells. Many of them adopt a backbone of intramolecular FRET biosensor with a cyan fluorescent protein (CFP) and yellow fluorescent protein (YFP) as donor and acceptor, respectively. However, there remains the difficulty of establishing cells stably expressing FRET biosensors with a YFP and CFP pair by lentiviral or retroviral gene transfer, due to the high incidence of recombination between YFP and CFP genes. To address this, we examined the effects of codon-diversification of YFP on the recombination of FRET biosensors introduced by lentivirus or retrovirus. The YFP gene that was fully codon-optimized to E.coli evaded the recombination in lentiviral or retroviral gene transfer, but the partially codon-diversified YFP did not. Further, the length of spacer between YFP and CFP genes clearly affected recombination efficiency, suggesting that the intramolecular template switching occurred in the reverse-transcription process. The simple mathematical model reproduced the experimental data sufficiently, yielding a recombination rate of 0.002–0.005 per base. Together, these results show that the codon-diversified YFP is a useful tool for expressing FRET biosensors by lentiviral or retroviral gene transfer.
Collapse
Affiliation(s)
- Akira T Komatsubara
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Michiyuki Matsuda
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan.,Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Kazuhiro Aoki
- Imaging Platform for Spatio-Temporal Information, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
18
|
ZHENG ZHONGHUI, DING MULIANG, NI JIANGDONG, SONG DEYE, HUANG JUN, WANG JUNJIE. miR-142 acts as a tumor suppressor in osteosarcoma cell lines by targeting Rac1. Oncol Rep 2014; 33:1291-9. [DOI: 10.3892/or.2014.3687] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 11/12/2014] [Indexed: 11/05/2022] Open
|