1
|
Tian Z, Yu S, Cai R, Zhang Y, Liu Q, Zhu Y. SH3GL2 and MMP17 as lung adenocarcinoma biomarkers: a machine-learning based approach. Biochem Biophys Rep 2024; 38:101693. [PMID: 38571554 PMCID: PMC10987888 DOI: 10.1016/j.bbrep.2024.101693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/19/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
Objective Using bioinformatics machine learning methods, our research aims to identify the potential key genes associated with Lung adenocarcinoma (LUAD). Methods We obtained two gene expression profiling microarrays (GSE68571 and GSE74706) from the public Gene Expression Omnibus (GEO) database at the National Centre for Biotechnology Information (NCBI). The purpose was to identify Differentially Expressed Genes (DEGs) between the lung adenocarcinoma group and the healthy control group. The limma R package in R was utilized for this analysis. For the differential gene diagnosis of lung adenocarcinoma, we employed the least absolute shrinkage and selection operator (LASSO) regression and SVM-RFE screening crossover. To evaluate the performance, ROC curves were plotted. We performed immuno-infiltration analysis using CIBERSORT. Finally, we validated the key genes through qRT-PCR and Western-blot verification, then downregulated MMP17 gene expression, upregulated SH3GL2 gene expression, and performed CCK8 experiments. Results A total of 32 Differentially Expressed Genes (DEGs) were identified. Two diagnostic marker genes, SH3GL2 and MMP17, were selected by employing LASSO and SVM-RFE machine learning methods. In Lung adenocarcinoma cells, the expression of MMP17 was observed to be elevated compared to normal lung epithelial cells in the control group (P < 0.05). In contrast, a down-regulation of SH3GL2 was found in Lung adenocarcinoma cells (P < 0.05). Finally, we downregulated MMP17 and upregulated SH3GL2 gene expression, then the CCK8 showed that the proliferation of both lung cancer cells was inhibited. Conclusion SH3GL2 and MMP17 are expected to be potential biomarkers for Lung adenocarcinoma.
Collapse
Affiliation(s)
- Zengjian Tian
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Shilong Yu
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Ruizhi Cai
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Yinghui Zhang
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Qilun Liu
- General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| | - Yongzhao Zhu
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, 750004, China
| |
Collapse
|
2
|
Yang LQ, Huang AF, Xu WD. Biology of endophilin and it's role in disease. Front Immunol 2023; 14:1297506. [PMID: 38116012 PMCID: PMC10728279 DOI: 10.3389/fimmu.2023.1297506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/22/2023] [Indexed: 12/21/2023] Open
Abstract
Endophilin is an evolutionarily conserved family of protein that involves in a range of intracellular membrane dynamics. This family consists of five isoforms, which are distributed in various tissues. Recent studies have shown that Endophilin regulates diseases pathogenesis, including neurodegenerative diseases, tumors, cardiovascular diseases, and autoimmune diseases. In vivo, it regulates different biological functions such as vesicle endocytosis, mitochondrial morphological changes, apoptosis and autophagosome formation. Functional studies confirmed the role of Endophilin in development and progression of these diseases. In this study, we have comprehensively discussed the complex function of Endophilin and how the family contributes to diseases development. It is hoped that this study will provide new ideas for targeting Endophilin in diseases.
Collapse
Affiliation(s)
- Lu-Qi Yang
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - An-Fang Huang
- Department of Rheumatology and Immunology, Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Wang-Dong Xu
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
3
|
Molecular characterization of early breast cancer onset to understand disease phenotypes in African patients. Med Oncol 2023; 40:13. [PMID: 36352274 PMCID: PMC9646617 DOI: 10.1007/s12032-022-01877-8] [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: 08/15/2022] [Accepted: 10/18/2022] [Indexed: 11/11/2022]
Abstract
Female breast cancer (BC) is the leading cause of cancer-related deaths worldwide with higher mortality rates and early onset in developing countries. The molecular basis of early disease onset is still elusive. We recruited 472 female breast cancer from two sub-Saharan African countries (Cameroon and Congo) between 2007 and 2018 and collected clinical data from these patients. To investigate the molecular drivers of early disease onset, we analyzed publicly available breast cancer molecular data from the cancer genome atlas (TCGA) and the gene expression omnibus (GEO) for copy number alteration, mutation and gene expression. Early BC onset (EOBRCA) (diagnosis before 45 years) was higher in African women compared with the TCGA cohort (51.7% vs 15.6%). The tumor grade, mitotic index, HER2 + phenotype, basal-like phenotype and ki67 were higher in EOBRCA for all cohorts. BC risk factors such as parity, breastfeeding early onset of menarche and use of hormonal contraceptives were significantly associated with EOBRCA (p < 0.05). EOBRCA was equally associated with copy number alterations in several oncogenes including CDH6 and FOXM1 and tumor suppressor including TGM3 and DMBT1 as well as higher TP53 mutation rates (OR: 2.93, p < 0.01). There was a significant enrichment of TGFß signaling in EOBRCA with TGM3 deletions, which was associated with high expression of all SMAD transcription factors as well as WNT ligands. The Frizzled receptors FZD1, FZD4 and FZD6 were significantly upregulated in EOBRCA, suggesting activation of non-canonical WNT signaling. Our data, suggest the implication of TGM3 deletion in early breast cancer onset. Further molecular investigations are warranted in African patients.
Collapse
|
4
|
Chen S, Chen X, Zhang P, Chen S, Wang X, Luo Q, Cui Z, Huang Y, Wan L, Hou X, Yao H, Liu X, He A, Jiang Z, Qiu J, Li Y, Yu K, Zhuang J. Bioinformatics Analysis and Experimental Identification of Immune-Related Genes and Immune Cells in the Progression of Retinoblastoma. Invest Ophthalmol Vis Sci 2022; 63:28. [PMID: 36315123 PMCID: PMC9631497 DOI: 10.1167/iovs.63.11.28] [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] [Indexed: 11/16/2022] Open
Abstract
Purpose Retinoblastoma (RB) is the most common type of aggressive intraocular malignancy in children. The alteration of immunity during RB progression and invasion has not yet been well defined. This study investigated significantly altered immune-associated genes and cells related to RB invasion. Methods The differentially expressed immune-related genes (IRGs) in noninvasive RB and invasive RB were identified by analysis of two microarray datasets (GSE97508 and GSE110811). Hub IRGs were further identified by real time PCR. The single-sample gene set enrichment analysis algorithm and Pearson correlation analysis were used to define immune cell infiltration and the relationships between hub IRGs and immune cells. Cell viability and migration were evaluated by CCK-8 and Transwell assays. A xenograft mouse model was used to verify the relationship between Src homology 3 (SH3) domain GRB2-like 2 (SH3GL2) expression and myeloid-derived suppressor cells (MDSCs). Results Eight upregulated genes and six downregulated IRGs were identified in invasive RB. Seven IRGs were confirmed by real-time PCR. Moreover, the proportions of MDSCs were higher in invasive RB tissues than in noninvasive RB tissues. Furthermore, correlation analysis of altered immune genes and cells suggested that SH3GL2, Langerhans cell protein 1 (LCP1) and transmembrane immune signaling adaptor TYROBP have strong connections with MDSCs. Specifically, decreased SH3GL2 expression promoted the migration of RB cells in vitro, increased the tumor size and weight, and increased the numbers of MDSCs in the tumor and spleen in vivo. Conclusions This study indicated that SH3GL2 and MDSCs play a critical role in RB progression and invasion and provide candidate targets for the treatment of RB.
Collapse
Affiliation(s)
- Shuilian Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Xi Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Ping Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Shuxia Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Xiao Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Qian Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Zedu Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Yuke Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Linxi Wan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Xiangtao Hou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Huan Yao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Xuan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Anqi He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Zihua Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Jin Qiu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Yan Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Keming Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| | - Jing Zhuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou City, China
| |
Collapse
|
5
|
Capdeville C, Russo L, Penton D, Migliavacca J, Zecevic M, Gries A, Neuhauss SC, Grotzer MA, Baumgartner M. Spatial proteomics finds CD155 and Endophilin-A1 as mediators of growth and invasion in medulloblastoma. Life Sci Alliance 2022; 5:5/6/e202201380. [PMID: 35296518 PMCID: PMC8926928 DOI: 10.26508/lsa.202201380] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/18/2022] [Accepted: 02/24/2022] [Indexed: 11/24/2022] Open
Abstract
The composition of the plasma membrane (PM)-associated proteome of tumor cells determines cell-cell and cell-matrix interactions and the response to environmental cues. Whether the PM-associated proteome impacts the phenotype of Medulloblastoma (MB) tumor cells and how it adapts in response to growth factor cues is poorly understood. Using a spatial proteomics approach, we observed that hepatocyte growth factor (HGF)-induced activation of the receptor tyrosine kinase c-MET in MB cells changes the abundance of transmembrane and membrane-associated proteins. The depletion of MAP4K4, a pro-migratory effector kinase downstream of c-MET, leads to a specific decrease of the adhesion and immunomodulatory receptor CD155 and of components of the fast-endophilin-mediated endocytosis (FEME) machinery in the PM-associated proteome of HGF-activated MB cells. The decreased surface expression of CD155 or of the fast-endophilin-mediated endocytosis effector endophilin-A1 reduces growth and invasiveness of MB tumor cells in the tissue context. These data thus describe a novel function of MAP4K4 in the control of the PM-associated proteome of tumor cells and identified two downstream effector mechanisms controlling proliferation and invasiveness of MB cells.
Collapse
Affiliation(s)
- Charles Capdeville
- Pediatric Molecular Neuro-Oncology Lab, Children's Research Center, University Children's Hospital Zürich, Zürich, Switzerland
| | - Linda Russo
- Pediatric Molecular Neuro-Oncology Lab, Children's Research Center, University Children's Hospital Zürich, Zürich, Switzerland
| | - David Penton
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Jessica Migliavacca
- Pediatric Molecular Neuro-Oncology Lab, Children's Research Center, University Children's Hospital Zürich, Zürich, Switzerland
| | - Milica Zecevic
- Pediatric Molecular Neuro-Oncology Lab, Children's Research Center, University Children's Hospital Zürich, Zürich, Switzerland
| | - Alexandre Gries
- Pediatric Molecular Neuro-Oncology Lab, Children's Research Center, University Children's Hospital Zürich, Zürich, Switzerland
| | - Stephan Cf Neuhauss
- Department of Molecular Life Sciences, University of Zurich, Zürich, Switzerland
| | - Michael A Grotzer
- Department of Oncology, University Children's Hospital Zürich, Zürich, Switzerland
| | - Martin Baumgartner
- Pediatric Molecular Neuro-Oncology Lab, Children's Research Center, University Children's Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
6
|
Casamento A, Boucrot E. Molecular mechanism of Fast Endophilin-Mediated Endocytosis. Biochem J 2020; 477:2327-2345. [PMID: 32589750 PMCID: PMC7319585 DOI: 10.1042/bcj20190342] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/11/2020] [Accepted: 05/18/2020] [Indexed: 12/13/2022]
Abstract
Endocytosis mediates the cellular uptake of micronutrients and cell surface proteins. Clathrin-mediated endocytosis (CME) is the housekeeping pathway in resting cells but additional Clathrin-independent endocytic (CIE) routes, including Fast Endophilin-Mediated Endocytosis (FEME), internalize specific cargoes and support diverse cellular functions. FEME is part of the Dynamin-dependent subgroup of CIE pathways. Here, we review our current understanding of the molecular mechanism of FEME. Key steps are: (i) priming, (ii) cargo selection, (iii) membrane curvature and carrier formation, (iv) membrane scission and (v) cytosolic transport. All steps are controlled by regulatory mechanisms mediated by phosphoinositides and by kinases such as Src, LRRK2, Cdk5 and GSK3β. A key feature of FEME is that it is not constitutively active but triggered upon the stimulation of selected cell surface receptors by their ligands. In resting cells, there is a priming cycle that concentrates Endophilin into clusters on discrete locations of the plasma membrane. In the absence of receptor activation, the patches quickly abort and new cycles are initiated nearby, constantly priming the plasma membrane for FEME. Upon activation, receptors are swiftly sorted into pre-existing Endophilin clusters, which then bud to form FEME carriers within 10 s. We summarize the hallmarks of FEME and the techniques and assays required to identify it. Next, we review similarities and differences with other CIE pathways and proposed cargoes that may use FEME to enter cells. Finally, we submit pending questions and future milestones and discuss the exciting perspectives that targeting FEME may boost treatments against cancer and neurodegenerative diseases.
Collapse
Affiliation(s)
- Alessandra Casamento
- Institute of Structural and Molecular Biology, University College London, Gower Street, London WC1E 6BT, U.K
| | - Emmanuel Boucrot
- Institute of Structural and Molecular Biology, University College London, Gower Street, London WC1E 6BT, U.K
- Institute of Structural and Molecular Biology, Birkbeck College, Malet Street, London WC1E 7HX, U.K
| |
Collapse
|
7
|
Repression of transcription factor AP-2 alpha by PPARγ reveals a novel transcriptional circuit in basal-squamous bladder cancer. Oncogenesis 2019; 8:69. [PMID: 31772149 PMCID: PMC6879593 DOI: 10.1038/s41389-019-0178-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 11/05/2019] [Accepted: 11/06/2019] [Indexed: 11/13/2022] Open
Abstract
The discovery of bladder cancer transcriptional subtypes provides an opportunity to identify high risk patients, and tailor disease management. Recent studies suggest tumor heterogeneity contributes to regional differences in molecular subtype within the tumor, as well as during progression and following treatment. Nonetheless, the transcriptional drivers of the aggressive basal-squamous subtype remain unidentified. As PPARɣ has been repeatedly implicated in the luminal subtype of bladder cancer, we hypothesized inactivation of this transcriptional master regulator during progression results in increased expression of basal-squamous specific transcription factors (TFs) which act to drive aggressive behavior. We initiated a pharmacologic and RNA-seq-based screen to identify PPARɣ-repressed, basal-squamous specific TFs. Hierarchical clustering of RNA-seq data following treatment of three human bladder cancer cells with a PPARɣ agonist identified a number of TFs regulated by PPARɣ activation, several of which are implicated in urothelial and squamous differentiation. One PPARɣ-repressed TF implicated in squamous differentiation identified is Transcription Factor Activating Protein 2 alpha (TFAP2A). We show TFAP2A and its paralog TFAP2C are overexpressed in basal-squamous bladder cancer and in squamous areas of cystectomy samples, and that overexpression is associated with increased lymph node metastasis and distant recurrence, respectively. Biochemical analysis confirmed the ability of PPARɣ activation to repress TFAP2A, while PPARɣ antagonist and PPARɣ siRNA knockdown studies indicate the requirement of a functional receptor. In vivo tissue recombination studies show TFAP2A and TFAP2C promote tumor growth in line with the aggressive nature of basal-squamous bladder cancer. Our findings suggest PPARɣ inactivation, as well as TFAP2A and TFAP2C overexpression cooperate with other TFs to promote the basal-squamous transition during tumor progression.
Collapse
|
8
|
Kannan A, Philley JV, Hertweck KL, Ndetan H, Singh KP, Sivakumar S, Wells RB, Vadlamudi RK, Dasgupta S. Cancer Testis Antigen Promotes Triple Negative Breast Cancer Metastasis and is Traceable in the Circulating Extracellular Vesicles. Sci Rep 2019; 9:11632. [PMID: 31406142 PMCID: PMC6690992 DOI: 10.1038/s41598-019-48064-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 07/16/2019] [Indexed: 12/15/2022] Open
Abstract
Triple negative breast cancer (TNBC) has poor survival, exhibits rapid metastases, lacks targeted therapies and reliable prognostic markers. Here, we examined metastasis promoting role of cancer testis antigen SPANXB1 in TNBC and its utility as a therapeutic target and prognostic biomarker. Expression pattern of SPANXB1 was determined using matched primary cancer, lymph node metastatic tissues and circulating small extracellular vesicles (sEVs). cDNA microarray analysis of TNBC cells stably integrated with a metastasis suppressor SH3GL2 identified SPANXB1 as a potential target gene. TNBC cells overexpressing SH3GL2 exhibited decreased levels of both SPANXB1 mRNA and protein. Silencing of SPANXB1 reduced migration, invasion and reactive oxygen species production of TNBC cells. SPANXB1 depletion augmented SH3GL2 expression and decreased RAC-1, FAK, A-Actinin and Vinculin expression. Phenotypic and molecular changes were reversed upon SPANXB1 re-expression. SPANXB1 overexpressing breast cancer cells with an enhanced SPANXB1:SH3GL2 ratio achieved pulmonary metastasis within 5 weeks, whereas controls cells failed to do so. Altered expression of SPANXB1 was detected in the sEVs of SPANXB1 transduced cells. Exclusive expression of SPANXB1 was traceable in circulating sEVs, which was associated with TNBC progression. SPANXB1 represents a novel and ideal therapeutic target for blocking TNBC metastases due to its unique expression pattern and may function as an EV based prognostic marker to improve TNBC survival. Uniquely restricted expression of SPANXB1 in TNBCs, makes it an ideal candidate for targeted therapeutics and prognostication.
Collapse
Affiliation(s)
- Anbarasu Kannan
- Departments of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Julie V Philley
- Departments of Medicine, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Kate L Hertweck
- Departments of Biology, The University of Texas at Tyler, Tyler, Texas, USA.,Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Harrison Ndetan
- Departments of Epidemiology and Biostatistics, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Karan P Singh
- Departments of Epidemiology and Biostatistics, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Subramaniam Sivakumar
- Departments of Biochemistry, Sri Sankara Arts and Science College, Kanchipuram, India
| | - Robert B Wells
- Departments of Pathology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA
| | - Ratna K Vadlamudi
- Departments of Obstetrics and Gynecology, CDP program, Mays Cancer Center, University of Texas Health at San Antonio, San Antonio, Texas, USA
| | - Santanu Dasgupta
- Departments of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA. .,Departments of Medicine, The University of Texas Health Science Center at Tyler, Tyler, Texas, USA.
| |
Collapse
|
9
|
Quantitative proteomics reveals reduction of endocytic machinery components in gliomas. EBioMedicine 2019; 46:32-41. [PMID: 31331834 PMCID: PMC6711119 DOI: 10.1016/j.ebiom.2019.07.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 02/04/2023] Open
Abstract
Background Gliomas are the most frequent and aggressive malignancies of the central nervous system. Decades of molecular analyses have demonstrated that gliomas accumulate genetic alterations that culminate in enhanced activity of receptor tyrosine kinases and downstream mediators. While the genetic alterations, like gene amplification or loss, have been well characterized, little information exists about changes in the proteome of gliomas of different grades. Methods We performed unbiased quantitative proteomics of human glioma biopsies by mass spectrometry followed by bioinformatic analysis. Findings Various pathways were found to be up- or downregulated. In particular, endocytosis as pathway was affected by a vast and concomitant reduction of multiple machinery components involved in initiation, formation, and scission of endocytic carriers. Both clathrin-dependent and -independent endocytosis were changed, since not only clathrin, AP-2 adaptins, and endophilins were downregulated, but also dynamin that is shared by both pathways. The reduction of endocytic machinery components caused increased receptor cell surface levels, a prominent phenotype of defective endocytosis. Analysis of additional biopsies revealed that depletion of endocytic machinery components was a common trait of various glioma grades and subclasses. Interpretation We propose that impaired endocytosis creates a selective advantage in glioma tumor progression due to prolonged receptor tyrosine kinase signaling from the cell surface. Fund This work was supported by Grants 316030-164105 (to P. Jenö), 31003A-162643 (to M. Spiess) and PP00P3-176974 (to G. Hutter) from the Swiss National Science Foundation. Further funding was received by the Department of Surgery from the University Hospital Basel.
Collapse
|
10
|
Yu X, Xu T, Ou S, Yuan J, Deng J, Li R, Yang J, Liu X, Li Q, Chen Y. Endophilin A1 mediates seizure activity via regulation of AMPARs in a PTZ-kindled epileptic mouse model. Exp Neurol 2018; 304:41-57. [PMID: 29481784 DOI: 10.1016/j.expneurol.2018.02.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 02/08/2018] [Accepted: 02/21/2018] [Indexed: 11/24/2022]
Abstract
Endophilin A1 is a member of the endophilin A family and is primarily expressed in the central nervous system. Endophilin A1 can mediate neuronal excitability by regulating neuronal synaptic plasticity, which indicates that the protein may be involved in epilepsy. However, to date, its role in epilepsy remains unclear. To explore the role of endophilin A1 in epilepsy, we aimed to investigate the expression patterns of endophilin A1 in patients with temporal lobe epilepsy (TLE) and in a pentylenetetrazole (PTZ)-kindled epileptic mouse model and to conduct behavioral and electrophysiological analyses after lentivirus-mediated knockdown of endophilin A1 in the hippocampus of epileptic mice. This study found that the expression of endophilin A1 was significantly up-regulated in the temporal neocortex of TLE patients and in the hippocampus and adjacent temporal cortex of the PTZ-kindled epileptic mouse model. Behavioral analyses indicated that knockdown of endophilin A1 in the mouse hippocampus increased the latency of the first seizure and reduced the frequency and duration of seizure activity. Whole-cell patch-clamp recordings of pyramidal neurons in the hippocampal CA3 area indicated that knockdown of endophilin A1 in the mouse hippocampus resulted in a reduced frequency of action potentials and decreased amplitudes of miniature excitatory postsynaptic currents (mEPSCs) and evoked AMPA-dependent EPSCs. Moreover, western blotting analysis showed that the surface expression of the AMPAR GluR2 subunit was also decreased after endophilin A1 knockdown, and co-immunoprecipitation indicated an association between endophilin A1 and AMPAR GluR2 in the mouse hippocampus. Further, when AMPARs were activated by CX546, the antiepileptic function of endophilin A1 knockdown was decreased. Based on these results, endophilin A1 plays a critical role in epilepsy, and its suppression in the mouse hippocampus can restrain neuronal excitability and seizure activity via regulating AMPARs.
Collapse
Affiliation(s)
- Xinyuan Yu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tao Xu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shu Ou
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinxian Yuan
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Deng
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Rong Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Juan Yang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xi Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qi Li
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yangmei Chen
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
11
|
Identification of genes and critical control proteins associated with inflammatory breast cancer using network controllability. PLoS One 2017; 12:e0186353. [PMID: 29108005 PMCID: PMC5673205 DOI: 10.1371/journal.pone.0186353] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 10/01/2017] [Indexed: 12/29/2022] Open
Abstract
One of the most aggressive forms of breast cancer is inflammatory breast cancer (IBC), whose lack of tumour mass also makes a prompt diagnosis difficult. Moreover, genomic differences between common breast cancers and IBC have not been completely assessed, thus substantially limiting the identification of biomarkers unique to IBC. Here, we developed a novel statistical analysis of gene expression profiles corresponding to microdissected IBC, non-IBC (nIBC) and normal samples that enabled us to identify a set of genes significantly associated with a specific disease state. Second, by using advanced methods based on controllability network theory, we identified a set of critical control proteins that uniquely and structurally control the entire proteome. By mapping high change variance genes in protein interaction networks, we found that a large statistically significant fraction of genes whose variance changed significantly between normal and IBC and nIBC disease states were among the set of critical control proteins. Moreover, this analysis identified the overlapping genes with the highest statistical significance; these genes may assist in developing future biomarkers and determining drug targets to disrupt the molecular pathways driving carcinogenesis in IBC.
Collapse
|
12
|
John BA, Said N. Insights from animal models of bladder cancer: recent advances, challenges, and opportunities. Oncotarget 2017; 8:57766-57781. [PMID: 28915710 PMCID: PMC5593682 DOI: 10.18632/oncotarget.17714] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 04/18/2017] [Indexed: 12/16/2022] Open
Abstract
Bladder cancer (urothelial cancer of the bladder) is the most common malignancy affecting the urinary system with increasing incidence and mortality. Treatment of bladder cancer has not advanced in the past 30 years. Therefore, there is a crucial unmet need for novel therapies, especially for high grade/stage disease that can only be achieved by preclinical model systems that faithfully recapitulate the human disease. Animal models are essential elements in bladder cancer research to comprehensively study the multistep cascades of carcinogenesis, progression and metastasis. They allow for the investigation of premalignant phases of the disease that are not clinically encountered. They can be useful for identification of diagnostic and prognostic biomarkers for disease progression and for preclinical identification and validation of therapeutic targets/candidates, advancing translation of basic research to clinic. This review summarizes the latest advances in the currently available bladder cancer animal models, their translational potential, merits and demerits, and the prevalent tumor evaluation modalities. Thereby, findings from these model systems would provide valuable information that can help researchers and clinicians utilize the model that best answers their research questions.
Collapse
Affiliation(s)
- Bincy Anu John
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Neveen Said
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA.,Department of Urology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
13
|
Zhu Y, Zhang X, Wang L, Ji Z, Xie M, Zhou X, Liu Z, Shi H, Yu R. Loss of SH3GL2 promotes the migration and invasion behaviours of glioblastoma cells through activating the STAT3/MMP2 signalling. J Cell Mol Med 2017; 21:2685-2694. [PMID: 28470949 PMCID: PMC5661104 DOI: 10.1111/jcmm.13184] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/06/2017] [Indexed: 12/22/2022] Open
Abstract
SH3GL2 (Src homology 3 (SH3) domain GRB2‐like 2) is mainly expressed in the central nervous system and regarded as a tumour suppressor in human glioma. However, the molecular mechanism of the SH3GL2 protein involved in malignant behaviours of human glioma has not been elucidated. In this study, we tried to investigate the role of SH3GL2 in glioma cell migration and invasion and explore its underlined molecular mechanism. Firstly, we discovered that the protein level of SH3GL2 was widely decreased in the human glioma patients, especially in high‐grade glioma tissues. Then, we determined the role of SH3GL2 in migration and invasion of glioma cells upon SH3GL2 knocking down and overexpressing. It was showed that knocking down of SH3GL2 promoted the migration and invasion of glioma cells, whereas overexpression of SH3GL2 inhibited them. Further study on molecular mechanism disclosed that silencing of SH3GL2 obviously activated the STAT3 (signal transducer and activator of transcription 3) signalling thereby promoting the expression and secretion of MMP2. On the contrary, overexpression of SH3GL2 had opposite effect. Taken together, the above results suggest that SH3GL2 suppresses migration and invasion behaviours of glioma cells through negatively regulating STAT3/MMP2 signalling and that loss of SH3GL2 may intensify the STAT3/MMP2 signalling thereby contributing to the migration and invasion of glioma cells.
Collapse
Affiliation(s)
- Yufu Zhu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiang Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,The Graduate School, Xuzhou Medical University, Xuzhou, China
| | - Lei Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhe Ji
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,The Graduate School, Xuzhou Medical University, Xuzhou, China
| | - Manyi Xie
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xinyu Zhou
- The Graduate School, Xuzhou Medical University, Xuzhou, China.,Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhiyi Liu
- The Graduate School, Xuzhou Medical University, Xuzhou, China.,Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Hengliang Shi
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, China.,Brain Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
14
|
Mitochondrial Reprogramming Regulates Breast Cancer Progression. Clin Cancer Res 2016; 22:3348-60. [DOI: 10.1158/1078-0432.ccr-15-2456] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 02/06/2016] [Indexed: 11/16/2022]
|
15
|
Kiselyov A, Bunimovich-Mendrazitsky S, Startsev V. Key signaling pathways in the muscle-invasive bladder carcinoma: Clinical markers for disease modeling and optimized treatment. Int J Cancer 2015; 138:2562-9. [PMID: 26547270 DOI: 10.1002/ijc.29918] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 10/03/2015] [Accepted: 11/04/2015] [Indexed: 02/01/2023]
Abstract
In this review, we evaluate key molecular pathways and markers of muscle-invasive bladder cancer (MIBC). Overexpression and activation of EGFR, p63, and EMT genes are suggestive of basal MIBC subtype generally responsive to chemotherapy. Alterations in PPARγ, ERBB2/3, and FGFR3 gene products and their signaling along with deregulated p53, cytokeratins KRT5/6/14 in combination with the cellular proliferation (Ki-67), and cell cycle markers (p16) indicate the need for more radical treatment protocols. Similarly, the "bell-shape" dynamics of Shh expression levels may suggest aggressive MIBC. A panel of diverse biological markers may be suitable for simulation studies of MIBC and development of an optimized treatment protocol. We conducted a critical evaluation of PubMed/Medline and SciFinder databases related to MIBC covering the period 2009-2015. The free-text search was extended by adding the following keywords and phrases: bladder cancer, metastatic, muscle-invasive, basal, luminal, epithelial-to-mesenchymal transition, cancer stem cell, mutations, immune response, signaling, biological markers, molecular markers, mathematical models, simulation, epigenetics, transmembrane, transcription factor, kinase, predictor, prognosis. The resulting selection of ca 500 abstracts was further analyzed in order to select the latest publications relevant to MIBC molecular markers of immediate clinical significance.
Collapse
Affiliation(s)
- Alex Kiselyov
- NBIC, Moscow Institute of Physics and Technology (MIPT), 9 Institutsky per, Dolgoprudny, Moscow Region, 141700, Russia
| | | | - Vladimir Startsev
- Department of Oncology, State Pediatric Medical University, St.-Petersburg, 194100, Russia
| |
Collapse
|
16
|
Distinct Functions of Endophilin Isoforms in Synaptic Vesicle Endocytosis. Neural Plast 2015; 2015:371496. [PMID: 26682072 PMCID: PMC4670672 DOI: 10.1155/2015/371496] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/15/2015] [Accepted: 06/23/2015] [Indexed: 11/23/2022] Open
Abstract
Endophilin isoforms perform distinct characteristics in their interactions with N-type Ca2+ channels and dynamin. However, precise functional differences for the endophilin isoforms on synaptic vesicle (SV) endocytosis remain unknown. By coupling RNA interference and electrophysiological recording techniques in cultured rat hippocampal neurons, we investigated the functional differences of three isoforms of endophilin in SV endocytosis. The results showed that the amplitude of normalized evoked excitatory postsynaptic currents in endophilin1 knockdown neurons decreased significantly for both single train and multiple train stimulations. Similar results were found using endophilin2 knockdown neurons, whereas endophilin3 siRNA exhibited no change compared with control neurons. Endophilin1 and endophilin2 affected SV endocytosis, but the effect of endophilin1 and endophilin2 double knockdown was not different from that of either knockdown alone. This result suggested that endophilin1 and endophilin2 functioned together but not independently during SV endocytosis. Taken together, our results indicate that SV endocytosis is sustained by endophilin1 and endophilin2 isoforms, but not by endophilin3, in primary cultured hippocampal neurons.
Collapse
|
17
|
Li B, He Y, Han X, Zhang S, Xu Y, Zhou Y, Song Z, Ouyang L. Aberrant promoter methylation of SH3GL2 gene in vulvar squamous cell carcinoma correlates with clinicopathological characteristics and HPV infection status. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:15442-15447. [PMID: 26823912 PMCID: PMC4713698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 10/23/2015] [Indexed: 06/05/2023]
Abstract
OBJECTIVE This study attempted to examine the methylation status of SH3GL2 gene in different types of human vulvar lesions and its correlation with clinicopathological parameters. METHODS Immunohistochemical analysis was used to identify the expression status of SH3GL2 in vulvar squamous cell carcinoma (VSCC), vulvar intraepithelial neoplasia (VIN) and benign vulvar squamous epithelium tissues. Bisulfite genomic sequencing method was used to detect methylation status of the SH3GL2 gene. Clinicopathological correlation of the alterations was analysed by the chi-square tests. RESULTS Immunohistochemical analysis showed expression of SH3GL2 in VSCC was significantly downregulated than that in VIN and normal vulvar tissues. In accordance with higher frequency of methylation status in SH3GL2, statistical analysis showed methylation status of SH3GL2 was closely related to tumor TNM stage (P=0.003), but not related to age, tumor volume, tumor differentiation, lymph node metastasis and VIN grade. High-methylation status of SH3GL2 showed significant association with HPV infection status. CONCLUSIONS Our results indicated that the methylation status of SH3GL2 gene was associated with the TNM staging and HPV infection status of VSCC, suggesting that it might play a synergistic role in the development of VSCC.
Collapse
Affiliation(s)
- Bo Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University Shenyang 110004, China
| | - Yinghui He
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University Shenyang 110004, China
| | - Xue Han
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University Shenyang 110004, China
| | - Shitai Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University Shenyang 110004, China
| | - Yan Xu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University Shenyang 110004, China
| | - Yang Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University Shenyang 110004, China
| | - Zixuan Song
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University Shenyang 110004, China
| | - Ling Ouyang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University Shenyang 110004, China
| |
Collapse
|
18
|
Adam RM, DeGraff DJ. Molecular mechanisms of squamous differentiation in urothelial cell carcinoma: a paradigm for molecular subtyping of urothelial cell carcinoma of the bladder. Urol Oncol 2015; 33:444-50. [PMID: 26254697 DOI: 10.1016/j.urolonc.2015.06.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 05/26/2015] [Accepted: 06/15/2015] [Indexed: 12/21/2022]
Abstract
BACKGROUND Recent molecular characterization studies focusing on bladder cancer have provided a wealth of information, including the identification of specific molecular subtypes of this disease. Interestingly, a particular molecular subtype identified by several different groups is characterized, at least in part, by the presence of squamous differentiation (SqD) in a significant fraction of primary tumors. Tumors that exhibit SqD are extremely aggressive. Moreover, conflicting reports exist relative to the sensitivity of bladder tumors exhibiting SqD to multimodal treatment. Bladder cancers that exhibit SqD appear to be distinct clinical entities and are often associated with a specific molecular subtype; therefore, it is important to understand the molecular drivers of this process. PURPOSE Because presence of SqD is closely associated with a basal molecular phenotype, we review the evidence for specific pathways in SqD. In addition, we pose key areas for future exploration.
Collapse
Affiliation(s)
- Rosalyn M Adam
- Department of Urology, Boston Children׳s Hospital, Boston, MA; Department of Surgery, Harvard Medical School, Boston, MA
| | - David J DeGraff
- Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA; Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA; Department of Surgery, Division of Urology, Pennsylvania State University College of Medicine, Hershey, PA.
| |
Collapse
|
19
|
Progress made in the use of animal models for the study of high-risk, nonmuscle invasive bladder cancer. Curr Opin Urol 2015; 24:512-6. [PMID: 24921908 DOI: 10.1097/mou.0000000000000087] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
PURPOSE OF REVIEW High-risk, nonmuscle invasive bladder cancer (HR-NMIBC) represents a costly and difficult-to-treat disease, the molecular pathogenesis of which has a limited understanding. Most preclinical models for the study of bladder cancer are more appropriate for the study of advanced disease. However, recent key advances in preclinical animal models places us at an opportune position to better understand HR-NMIBC. RECENT FINDINGS Discoveries in the basic sciences allow us to better understand tumor biology when building models of bladder cancer. Of note, a key study on urothelial progenitor cells recently highlighted an important role for Sonic hedgehog-positive cells and retinoid signaling that is essential for urothelial development and regeneration. In the translational realm, transgenic mouse models continue to be used, with a recent interest in the role of Wnt/beta-catenin in urothelial carcinomas. Tissue recombination models are also being increasingly utilized to better recreate the tissue microenvironment and better understand stromal-epithelial interactions and the impact of genetic alterations on tissue differentiation. Lastly, the avatar mouse systems, which involve direct xenotransplantation of human tumor specimens into immunocompromised mice, represent an additional approach to study cancer characteristics in a preserved tissue context. SUMMARY With molecular alterations remaining an unclear area of our understanding of HR-NMIBC, preclinical models of bladder cancer serve as essential tools to discover specific genetic compromises in disease pathogenesis and the therapeutics to treat them.
Collapse
|
20
|
Chen L, Liu W, Wang P, Xue Y, Su Q, Zeng C, Shang X. Endophilin-1 regulates blood-brain barrier permeability via EGFR-JNK signaling pathway. Brain Res 2015; 1606:44-53. [PMID: 25721793 DOI: 10.1016/j.brainres.2015.02.032] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 02/08/2015] [Accepted: 02/10/2015] [Indexed: 10/23/2022]
Abstract
Endophilin-1 (Endo1), a multifunctional protein, is essential for synaptic vesicle endocytosis. However, the role and mechanism of endophilin-1 in blood-brain barrier (BBB) function are still unclear. This study was performed to determine whether endophilin-1 regulated BBB permeability via the EGFR-JNK signaling pathway. In the present study, we found that endophilin-1 over-expression in human cerebral microvascular endothelial cell (hCMEC/D3) increased BBB permeability and meanwhile reduced the expression levels of epidermal growth factor receptor (EGFR), phosphorylated c-Jun N-terminal kinase (p-JNK). While endophilin-1 knockdown led to the contrary results. After JNK inhibitor SP600125 was administered to the endophilin-1 silenced hCMEC/D3 cells, the transendothelial electrical resistance (TEER) value was decreased and the permeability coefficient values to 4kDa and 40kDa FITC-dextran were increased. Results observed by Transmission electron microscopy (TEM) showed that tight junctions (TJs) were opened. Moreover, immunofluorescence and Western blot assays revealed the discontinuous distribution of TJ-associated proteins ZO-1, occludin on cell-cell boundaries and a significant decrease in protein expressing levels. Therefore, these results indicated that endophilin-1 positively regulated BBB permeability via the EGFR-JNK signaling pathway in hCMEC/D3 cells, which would provide an experimental basis for further research on endophilin-1 mediated the opening of BBB.
Collapse
Affiliation(s)
- Lin Chen
- Department of Neurology, First Affiliated Hospital, China Medical University, Shenyang 110001, People׳s Republic of China; Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110001, People׳s Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110001, People׳s Republic of China; Department of Neurology, Hainan Provincial Nongken General Hospital, Haikou 570311, People׳s Republic of China
| | - Wenjing Liu
- Department of Neurology, First Affiliated Hospital, China Medical University, Shenyang 110001, People׳s Republic of China; Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110001, People׳s Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110001, People׳s Republic of China
| | - Ping Wang
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110001, People׳s Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110001, People׳s Republic of China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang 110001, People׳s Republic of China; Institute of Pathology and Pathophysiology, China Medical University, Shenyang 110001, People׳s Republic of China
| | - Qingjie Su
- Department of Neurology, Hainan Provincial Nongken General Hospital, Haikou 570311, People׳s Republic of China
| | - Chaosheng Zeng
- Department of Neurology, Hainan Provincial Nongken General Hospital, Haikou 570311, People׳s Republic of China
| | - Xiuli Shang
- Department of Neurology, First Affiliated Hospital, China Medical University, Shenyang 110001, People׳s Republic of China.
| |
Collapse
|
21
|
Cancer subclonal genetic architecture as a key to personalized medicine. Neoplasia 2014; 15:1410-20. [PMID: 24403863 DOI: 10.1593/neo.131972] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 12/03/2013] [Accepted: 12/03/2013] [Indexed: 02/08/2023] Open
Abstract
The future of personalized oncological therapy will likely rely on evidence-based medicine to integrate all of the available evidence to delineate the most efficacious treatment option for the patient. To undertake evidence-based medicine through use of targeted therapy regimens, identification of the specific underlying causative mutation(s) driving growth and progression of a patient's tumor is imperative. Although molecular subtyping is important for planning and treatment, intraclonal genetic diversity has been recently highlighted as having significant implications for biopsy-based prognosis. Overall, delineation of the clonal architecture of a patient's cancer and how this will impact on the selection of the most efficacious therapy remain a topic of intense interest.
Collapse
|
22
|
Mooso BA, Vinall RL, Mudryj M, Yap SA, deVere White RW, Ghosh PM. The role of EGFR family inhibitors in muscle invasive bladder cancer: a review of clinical data and molecular evidence. J Urol 2014; 193:19-29. [PMID: 25158272 DOI: 10.1016/j.juro.2014.07.121] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE Conventional platinum based chemotherapy for advanced urothelial carcinoma is plagued by common resistance to this regimen. Several studies implicate the EGFR family of RTKs in urothelial carcinoma progression and chemoresistance. Many groups have investigated the effects of inhibitors of this family in patients with urothelial carcinoma. This review focuses on the underlying molecular pathways that lead to urothelial carcinoma resistance to EGFR family inhibitors. MATERIALS AND METHODS We performed a PubMed® search for peer reviewed literature on bladder cancer development, EGFR family expression, clinical trials of EGFR family inhibitors and molecular bypass pathways. Research articles deemed to be relevant were examined and a summary of original data was created. Meta-analysis of expression profiles was also performed for each EGFR family member based on data sets accessible via Oncomine®. RESULTS Many clinical trials using inhibitors of EGFR family RTKs have been done or are under way. Those that have concluded with results published to date do not show an added benefit over standard of care chemotherapy in an adjuvant or second line setting. However, a neoadjuvant study using erlotinib before radical cystectomy demonstrated promising results. CONCLUSIONS Clinical and preclinical studies show that for reasons not currently clear prior treatment with chemotherapeutic agents rendered patients with urothelial carcinoma with muscle invasive bladder cancer resistant to EGFR family inhibitors as well. However, EGFR family inhibitors may be of use in patients with no prior chemotherapy in whom EGFR or ERBB2 is over expressed.
Collapse
Affiliation(s)
- Benjamin A Mooso
- Research Service, Veterans Affairs Northern California Health Care System, Sacramento, California
| | - Ruth L Vinall
- California Northstate College of Pharmacy, Rancho Cordova, California
| | - Maria Mudryj
- Research Service, Veterans Affairs Northern California Health Care System, Sacramento, California; Department of Medical Microbiology and Immunology, University of California-Davis, Sacramento, California
| | - Stanley A Yap
- Research Service, Veterans Affairs Northern California Health Care System, Sacramento, California; Department of Urology, University of California-Davis, Sacramento, California
| | | | - Paramita M Ghosh
- Research Service, Veterans Affairs Northern California Health Care System, Sacramento, California; Department of Biochemistry and Molecular Medicine, University of California-Davis, Sacramento, California; Department of Urology, University of California-Davis, Sacramento, California
| |
Collapse
|
23
|
Liu W, Wang P, Shang C, Chen L, Cai H, Ma J, Yao Y, Shang X, Xue Y. Endophilin-1 regulates blood–brain barrier permeability by controlling ZO-1 and occludin expression via the EGFR–ERK1/2 pathway. Brain Res 2014; 1573:17-26. [DOI: 10.1016/j.brainres.2014.05.022] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 05/07/2014] [Accepted: 05/13/2014] [Indexed: 01/07/2023]
|
24
|
Yao Y, Xue Y, Ma J, Shang C, Wang P, Liu L, Liu W, Li Z, Qu S, Li Z, Liu Y. MiR-330-mediated regulation of SH3GL2 expression enhances malignant behaviors of glioblastoma stem cells by activating ERK and PI3K/AKT signaling pathways. PLoS One 2014; 9:e95060. [PMID: 24736727 PMCID: PMC3988141 DOI: 10.1371/journal.pone.0095060] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 03/23/2014] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs are currently considered as an active and rapidly evolving area for the treatment of tumors. In this study, we elucidated the biological significance of miR-330 in glioblastoma stem cells (GSCs) as well as the possible molecular mechanisms. SH3GL2 is mainly distributed in the central nervous system and considered to be a tumor suppressor in many tumors. In the present study, we identified miR-330 as a potential regulator of SH3GL2 and we found that it was to be inversely correlated with SH3GL2 expression in GSCs which were isolated from U87 cell lines. The expression of miR-330 enhanced cellular proliferation, promoted cell migration and invasion, and dampened cell apoptosis. When the GSCs were co-transfected with the plasmid containing short hairpin RNA directed against human SH3GL2 gene and miR-330 mimic, we found that miR-330 promoted the malignant behavior of GSCs by down-regulating the expression of SH3GL2. Meanwhile, the ERK and PI3K/AKT signaling pathways were significantly activated, leading to the decreased expression of apoptotic protein and increased expression of anti-apoptotic protein. Furthermore, in orthotopic mouse xenografts, the mice given stable over-expressed SH3GL2 cells co-transfected with miR-330 knockdown plasmid had the smallest tumor sizes and longest survival. In conclusion, these results suggested that miR-330 negatively regulated the expression of SH3GL2 in GSCs, which promoted the oncogenic progression of GSCs through activating ERK and PI3K/AKT signaling pathways. The elucidation of these mechanisms will provide potential therapeutic approaches for human glioblastoma.
Collapse
Affiliation(s)
- Yilong Yao
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, People’s Republic of China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
- Institute of Pathology and Pathophysiology, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
| | - Jun Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
- Institute of Pathology and Pathophysiology, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
| | - Chao Shang
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
- Institute of Pathology and Pathophysiology, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
| | - Ping Wang
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
- Institute of Pathology and Pathophysiology, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
| | - Libo Liu
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
- Institute of Pathology and Pathophysiology, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
| | - Wenjing Liu
- Department of Neurology, The First Affiliated Hospital, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, People’s Republic of China
| | - Shengtao Qu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, People’s Republic of China
| | - Zhiqing Li
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, Liaoning Province, People’s Republic of China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning Province, People’s Republic of China
- * E-mail:
| |
Collapse
|