1
|
Wang CX, Liu F, Wang Y. RBM5 suppresses proliferation, metastasis and glycolysis of colorectal cancer cells via stabilizing phosphatase and tensin homolog mRNA. World J Gastrointest Oncol 2024; 16:3241-3255. [PMID: 39072172 PMCID: PMC11271791 DOI: 10.4251/wjgo.v16.i7.3241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/07/2024] [Accepted: 05/24/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND RNA binding motif 5 (RBM5) has emerged as crucial regulators in many cancers. AIM To explore more functional and mechanistic exploration of RBM5 since the lack of research on RBM5 in colorectal cancer (CRC) dictates that is essential. METHODS Through Gene Expression Profiling Interactive Analysis, we analyzed RBM5 expression in colon adenocarcinoma and rectum adenocarcinoma tissues. For detecting the mRNA expression of RBM5, quantitative real time-polymerase chain reaction was performed. Protein expression levels of RBM5, hexokinase 2, lactate dehydrogenase A, phosphatase and tensin homolog (PTEN), phosphoinositide 3-kinase (PI3K), phosphorylated-protein kinase B (p-AKT), and AKT were determined via Western blot. Functionally, cell counting kit-8 and 5-ethynyl-2'-deoxyuridine (EDU) assay were performed to evaluate proliferation of CRC cells. Invasiveness and migration of CRC cells were evaluated through conducting transwell assays. Glucose consumption, lactate production and adenosine-triphosphate (ATP) production were measured through a glucose assay kit, a lactate assay kit and an ATP production assay kit, respectively. Besides, RNA immunoprecipitation assay, half-life RT-PCR and dual-luciferase reporter assay were applied to detect interaction between RBM5 and PTEN. To establish a xenotypic tumor mice, CRC cells were subcutaneously injected into the right flank of each mouse. Protein expression of RBM5, Ki67, and PTEN in tumor tissues was examined using immunohistochemistry staining. Haematoxylin and eosin staining was used to evaluate tumor liver metastasis in mice. RESULTS We discovered down-regulation of RBM5 expression in CRC tissues and cells. RBM5 overexpression repressed proliferation, migration and invasion of CRC cells. Meantime, RBM5 impaired glycolysis in CRC cells, presenting as decreased glucose consumption, decreased lactate production and decreased ATP production. Besides, RBM5 bound to PTEN mRNA to stabilize its expression. PTEN expression was positively regulated by RBM5 in CRC cells. The protein levels of PI3K and p-AKT were significantly decreased after RBM5 overexpression. The suppressive influences of RBM5 on glycolysis, proliferation and metastasis of CRC cells were partially counteracted by PTEN knockdown. RBM5 suppressed tumor growth and liver metastasis in vivo. CONCLUSION This investigation provided new evidence that RBM5 was involved in CRC by binding to PTEN, expanding the importance of RBM5 in the treatment of CRC.
Collapse
Affiliation(s)
- Chu-Xiang Wang
- Department of Anorectal Surgery, The Affiliated Hospital of Dalian Medical University, Dalian 116000, Liaoning Province, China
| | - Feng Liu
- Department of Scientific Research, Beijing Fresta Technologies Co, Ltd., Beijing 100176, China
| | - Yi Wang
- Department of VIP Medical Services, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
2
|
Zhou X, Cao Y, Li R, Di X, Wang Y, Wang K. PEI, a new transfection method, augments the inhibitory effect of RBM5 on prostate cancer. Biochem Biophys Res Commun 2024; 704:149703. [PMID: 38402723 DOI: 10.1016/j.bbrc.2024.149703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 02/27/2024]
Abstract
PEI is a cationic polymer, serving as a non-viral transfection carrier grounded in nanotechnology that enhances transfection efficiency via the proton sponge effect. RBM5 is an RNA-binding protein that can inhibit tumor development. This study involved the transfection of RBM5 in prostate cancer cells with PEI, Lipo2000, and their combination. Transwell and wound healing assays were used to observe invasion and migration of prostate cancer cells and flow cytometry was used to observe the apoptosis. Detect the expression of invasion and migration-related protein MMP9 through western blotting experiment. An activity detection kit was used to detect the activity of apoptotic protein caspase-3. We found that there was no significant difference in transfection efficiency when PEI and Lipo2000 were used alone but it significantly improved when they are combined. RBM5 reduced invasion, migration, and proliferation of prostate cancer and enhanced apoptosis. MMP9 expression was reduced, and the activity of caspase-3 was increased. PEI transfection could improve the inhibition of RBM5 on tumors more than Lipo2000. The inhibitory effect is more obvious when the two are used together. RBM5 transfected with PEI can amplify its inhibitory effect on prostate cancer, and this effect is more evident when combined with Lipo2000.
Collapse
Affiliation(s)
- Xijia Zhou
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Yingshu Cao
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Ranwei Li
- Department of Urinary Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xin Di
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Yanqiao Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Ke Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
3
|
Silva VAR, Lavinsky J, Pauna HF, Vianna MF, Santos VM, Ikino CMY, Sampaio ALL, Tardim Lopes P, Lamounier P, Maranhão ASDA, Soares VYR, Polanski JF, Denaro MMDC, Chone CT, Bento RF, Castilho AM. Brazilian Society of Otology task force - Vestibular Schwannoma ‒ evaluation and treatment. Braz J Otorhinolaryngol 2023; 89:101313. [PMID: 37813009 PMCID: PMC10563065 DOI: 10.1016/j.bjorl.2023.101313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 08/17/2023] [Indexed: 10/11/2023] Open
Abstract
OBJECTIVE To review the literature on the diagnosis and treatment of vestibular schwannoma. METHODS Task force members were educated on knowledge synthesis methods, including electronic database search, review and selection of relevant citations, and critical appraisal of selected studies. Articles written in English or Portuguese on vestibular schwannoma were eligible for inclusion. The American College of Physicians' guideline grading system and the American Thyroid Association's guideline criteria were used for critical appraisal of evidence and recommendations for therapeutic interventions. RESULTS The topics were divided into 2 parts: (1) Diagnosis - audiologic, electrophysiologic tests, and imaging; (2) Treatment - wait and scan protocols, surgery, radiosurgery/radiotherapy, and systemic therapy. CONCLUSIONS Decision making in VS treatment has become more challenging. MRI can diagnose increasingly smaller tumors, which has disastrous consequences for the patients and their families. It is important to develop an individualized approach for each case, which highly depends on the experience of each surgical team.
Collapse
Affiliation(s)
- Vagner Antonio Rodrigues Silva
- Universidade Estadual de Campinas (Unicamp), Faculdade de Ciências Médicas (FCM), Departamento de Otorrinolaringologia e Cirurgia de Cabeça e Pescoço, Campinas, SP, Brazil; Sociedade Brasileira de Otologia - SBO
| | - Joel Lavinsky
- Sociedade Brasileira de Otologia - SBO; Universidade Federal do Rio Grande do Sul (UFRGS), Departamento de Ciências Morfológicas, Porto Alegre, RS, Brazil
| | - Henrique Furlan Pauna
- Hospital Universitário Cajuru, Departamento de Otorrinolaringologia, Curitiba, PR, Brazil
| | - Melissa Ferreira Vianna
- Sociedade Brasileira de Otologia - SBO; Irmandade Santa Casa de Misericórdia de São Paulo, Departamento de Otorrinolaringologia, São Paulo, SP, Brazil
| | - Vanessa Mazanek Santos
- Universidade Federal do Paraná, Hospital de Clínicas, Departamento de Otorrinolaringologia e Cirurgia de Cabeça e Pescoço, Curitiba, PR, Brazil
| | - Cláudio Márcio Yudi Ikino
- Universidade Federal de Santa Catarina, Hospital Universitário, Departamento de Cirurgia, Florianópolis, SC, Brazil
| | - André Luiz Lopes Sampaio
- Sociedade Brasileira de Otologia - SBO; Universidade de Brasília (UnB), Faculdade de Medicina, Laboratório de Ensino e Pesquisa em Otorrinolaringologia, Brasília, DF, Brazil
| | - Paula Tardim Lopes
- Faculdade de Medicina da Universidade de São Paulo (FMUSP), Departamento de Otorrinolaringologia, São Paulo, SP, Brazil
| | - Pauliana Lamounier
- Centro de Reabilitação e Readaptação Dr. Henrique Santillo (CRER), Departamento de Otorrinolaringologia, Goiânia, GO, Brazil
| | - André Souza de Albuquerque Maranhão
- Universidade Federal de São Paulo (UNIFESP), Escola Paulista de Medicina, Departamento de Otorrinolaringologia e Cirurgia de Cabeça e Pescoço, São Paulo, SP, Brazil
| | - Vitor Yamashiro Rocha Soares
- Hospital Flavio Santos e Hospital Getúlio Vargas, Grupo de Otologia e Base Lateral do Crânio, Teresina, PI, Brazil
| | - José Fernando Polanski
- Universidade Federal do Paraná, Hospital de Clínicas, Departamento de Otorrinolaringologia e Cirurgia de Cabeça e Pescoço, Curitiba, PR, Brazil; Faculdade Evangélica Mackenzie do Paraná, Faculdade de Medicina, Curitiba, PR, Brazil
| | | | - Carlos Takahiro Chone
- Universidade Estadual de Campinas (Unicamp), Faculdade de Ciências Médicas (FCM), Departamento de Otorrinolaringologia e Cirurgia de Cabeça e Pescoço, Campinas, SP, Brazil
| | - Ricardo Ferreira Bento
- Faculdade de Medicina da Universidade de São Paulo (FMUSP), Departamento de Otorrinolaringologia, São Paulo, SP, Brazil
| | - Arthur Menino Castilho
- Universidade Estadual de Campinas (Unicamp), Faculdade de Ciências Médicas (FCM), Departamento de Otorrinolaringologia e Cirurgia de Cabeça e Pescoço, Campinas, SP, Brazil; Sociedade Brasileira de Otologia - SBO.
| |
Collapse
|
4
|
Soni K, Jagtap PKA, Martínez-Lumbreras S, Bonnal S, Geerlof A, Stehle R, Simon B, Valcárcel J, Sattler M. Structural basis for specific RNA recognition by the alternative splicing factor RBM5. Nat Commun 2023; 14:4233. [PMID: 37454201 PMCID: PMC10349855 DOI: 10.1038/s41467-023-39961-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 07/05/2023] [Indexed: 07/18/2023] Open
Abstract
The RNA-binding motif protein RBM5 belongs to a family of multi-domain RNA binding proteins that regulate alternative splicing of genes important for apoptosis and cell proliferation and have been implicated in cancer. RBM5 harbors structural modules for RNA recognition, such as RRM domains and a Zn finger, and protein-protein interactions such as an OCRE domain. Here, we characterize binding of the RBM5 RRM1-ZnF1-RRM2 domains to cis-regulatory RNA elements. A structure of the RRM1-ZnF1 region in complex with RNA shows how the tandem domains cooperate to sandwich target RNA and specifically recognize a GG dinucleotide in a non-canonical fashion. While the RRM1-ZnF1 domains act as a single structural module, RRM2 is connected by a flexible linker and tumbles independently. However, all three domains participate in RNA binding and adopt a closed architecture upon RNA binding. Our data highlight how cooperativity and conformational modularity of multiple RNA binding domains enable the recognition of distinct RNA motifs, thereby contributing to the regulation of alternative splicing. Remarkably, we observe surprising differences in coupling of the RNA binding domains between the closely related homologs RBM5 and RBM10.
Collapse
Affiliation(s)
- Komal Soni
- Helmholtz Munich, Molecular Targets and Therapeutics Center, Institute of Structural Biology, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Bavarian NMR Center, Lichtenbergstrasse 4, 85748, Garching, Germany
| | - Pravin Kumar Ankush Jagtap
- Helmholtz Munich, Molecular Targets and Therapeutics Center, Institute of Structural Biology, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Bavarian NMR Center, Lichtenbergstrasse 4, 85748, Garching, Germany
| | - Santiago Martínez-Lumbreras
- Helmholtz Munich, Molecular Targets and Therapeutics Center, Institute of Structural Biology, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Bavarian NMR Center, Lichtenbergstrasse 4, 85748, Garching, Germany
| | - Sophie Bonnal
- Centre de Regulació Genòmica, Barcelona Institute of Science and Technology and Universitat Pompeu Fabra, Barcelona, Spain
| | - Arie Geerlof
- Helmholtz Munich, Molecular Targets and Therapeutics Center, Institute of Structural Biology, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Ralf Stehle
- Helmholtz Munich, Molecular Targets and Therapeutics Center, Institute of Structural Biology, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Bavarian NMR Center, Lichtenbergstrasse 4, 85748, Garching, Germany
| | - Bernd Simon
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, 69117, Heidelberg, Germany
| | - Juan Valcárcel
- Centre de Regulació Genòmica, Barcelona Institute of Science and Technology and Universitat Pompeu Fabra, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Michael Sattler
- Helmholtz Munich, Molecular Targets and Therapeutics Center, Institute of Structural Biology, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany.
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Bavarian NMR Center, Lichtenbergstrasse 4, 85748, Garching, Germany.
| |
Collapse
|
5
|
Tumor Biology and Microenvironment of Vestibular Schwannoma-Relation to Tumor Growth and Hearing Loss. Biomedicines 2022; 11:biomedicines11010032. [PMID: 36672540 PMCID: PMC9856152 DOI: 10.3390/biomedicines11010032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022] Open
Abstract
Vestibular schwannoma is the most common benign neoplasm of the cerebellopontine angle. It arises from Schwann cells of the vestibular nerve. The first symptoms of vestibular schwannoma include hearing loss, tinnitus, and vestibular symptoms. In the event of further growth, cerebellar and brainstem symptoms, along with palsy of the adjacent cranial nerves, may be present. Although hearing impairment is present in 95% of patients diagnosed with vestibular schwannoma, most tumors do not progress in size or have low growth rates. However, the clinical picture has unpredictable dynamics, and there are currently no reliable predictors of the tumor's behavior. The etiology of the hearing loss in patients with vestibular schwannoma is unclear. Given the presence of hearing loss in patients with non-growing tumors, a purely mechanistic approach is insufficient. A possible explanation for this may be that the function of the auditory system may be affected by the paracrine activity of the tumor. Moreover, initiation of the development and growth progression of vestibular schwannomas is not yet clearly understood. Biallelic loss of the NF2 gene does not explain the occurrence in all patients; therefore, detection of gene expression abnormalities in cases of progressive growth is required. As in other areas of cancer research, the tumor microenvironment is coming to the forefront, also in vestibular schwannomas. In the paradigm of the tumor microenvironment, the stroma of the tumor actively influences the tumor's behavior. However, research in the area of vestibular schwannomas is at an early stage. Thus, knowledge of the molecular mechanisms of tumorigenesis and interactions between cells present within the tumor is crucial for the diagnosis, prediction of tumor behavior, and targeted therapeutic interventions. In this review, we provide an overview of the current knowledge in the field of molecular biology and tumor microenvironment of vestibular schwannomas, as well as their relationship to tumor growth and hearing loss.
Collapse
|
6
|
Tamura R, Toda M. A Critical Overview of Targeted Therapies for Vestibular Schwannoma. Int J Mol Sci 2022; 23:5462. [PMID: 35628268 PMCID: PMC9143502 DOI: 10.3390/ijms23105462] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 12/10/2022] Open
Abstract
Vestibular schwannoma (VS) is a benign tumor that originates from Schwann cells in the vestibular component. Surgical treatment for VS has gradually declined over the past few decades, especially for small tumors. Gamma knife radiosurgery has become an accepted treatment for VS, with a high rate of tumor control. For neurofibromatosis type 2 (NF2)-associated VS resistant to radiotherapy, vascular endothelial growth factor (VEGF)-A/VEGF receptor (VEGFR)-targeted therapy (e.g., bevacizumab) may become the first-line therapy. Recently, a clinical trial using a VEGFR1/2 peptide vaccine was also conducted in patients with progressive NF2-associated schwannomas, which was the first immunotherapeutic approach for NF2 patients. Targeted therapies for the gene product of SH3PXD2A-HTRA1 fusion may be effective for sporadic VS. Several protein kinase inhibitors could be supportive to prevent tumor progression because merlin inhibits signaling by tyrosine receptor kinases and the activation of downstream pathways, including the Ras/Raf/MEK/ERK and PI3K/Akt/mTORC1 pathways. Tumor-microenvironment-targeted therapy may be supportive for the mainstays of management. The tumor-associated macrophage is the major component of immunosuppressive cells in schwannomas. Here, we present a critical overview of targeted therapies for VS. Multimodal therapy is required to manage patients with refractory VS.
Collapse
Affiliation(s)
- Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan;
| | | |
Collapse
|
7
|
Nagururu NV, Akbar A, Ward BK. Using magnetic resonance imaging to improve diagnosis of peripheral vestibular disorders. J Neurol Sci 2022; 439:120300. [DOI: 10.1016/j.jns.2022.120300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/18/2022] [Accepted: 05/23/2022] [Indexed: 12/01/2022]
|
8
|
Welling DB, Collier KA, Burns SS, Oblinger JL, Shu E, Miles‐Markley BA, Hofmeister CC, Makary MS, Slone HW, Blakeley JO, Mansouri SA, Neff BA, Jackler RK, Mortazavi A, Chang L. Early phase clinical studies of AR-42, a histone deacetylase inhibitor, for neurofibromatosis type 2-associated vestibular schwannomas and meningiomas. Laryngoscope Investig Otolaryngol 2021; 6:1008-1019. [PMID: 34667843 PMCID: PMC8513424 DOI: 10.1002/lio2.643] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/16/2021] [Accepted: 08/10/2021] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVES Two pilot studies of AR-42, a pan-histone deacetylase inhibitor, in human neurofibromatosis type 2 (NF2), vestibular schwannomas (VS), and meningiomas are presented. Primary endpoints included safety, and intra-tumoral pharmacokinetics (PK) and pharmacodynamics (PD). METHODS Pilot 1 is a subset analysis of a phase 1 study of AR-42 in solid tumors, which included NF2 or sporadic meningiomas. Tumor volumes and treatment-related adverse events (TRAEs) are reported (NCT01129193).Pilot 2 is a phase 0 surgical study of AR-42 assessing intra-tumoral PK and PD. AR-42 was administered for 3 weeks pre-operatively. Plasma and tumor drug concentrations and p-AKT expression were measured (NCT02282917). RESULTS Pilot 1: Five patients with NF2 and two with sporadic meningiomas experienced a similar incidence of TRAEs to the overall phase I trial. The six evaluable patients had 15 tumors (8 VS, 7 meningiomas). On AR-42, tumor volume increased in six, remained stable in eight, and decreased in one tumor. The annual percent growth rate decreased in eight, remained stable in three, and increased in four tumors. Pilot 2: Four patients with sporadic VS and one patient with meningioma experienced no grade 3/4 toxicities. Expression of p-AKT decreased in three of four VS. All tumors had higher AR-42 concentrations than plasma. CONCLUSIONS AR-42 is safe. Tumor volumes showed a mixed response, but most slowed growth. On a 40-mg regimen, drug concentrated in tumors and growth pathways were suppressed in most tumors, suggesting this may be a well-tolerated and effective dose. A phase 2 study of AR-42 for NF2-associated tumors appears warranted. LEVEL OF EVIDENCE 1b, 4.
Collapse
Affiliation(s)
- D. Bradley Welling
- Department of Otolaryngology Head and Neck SurgeryHarvard Medical School, Massachusetts Eye and Ear Infirmary, Massachusetts General HospitalBostonMassachusettsUSA
| | - Katharine A. Collier
- Division of Medical Oncology, Department of Internal MedicineThe Ohio State University College of Medicine and the Comprehensive Cancer CenterColumbusOhioUSA
| | - Sarah S. Burns
- Center for Childhood Cancer and Blood diseasesAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOhioUSA
| | - Janet L. Oblinger
- Center for Childhood Cancer and Blood diseasesAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOhioUSA
| | - Edina Shu
- Department of Otolaryngology Head and Neck SurgeryHarvard Medical School, Massachusetts Eye and Ear Infirmary, Massachusetts General HospitalBostonMassachusettsUSA
| | - Beth A. Miles‐Markley
- Department of Otolaryngology‐Head and Neck SurgeryThe Ohio State University College of MedicineColumbusOhioUSA
| | - Craig C. Hofmeister
- Department of Hematology & OncologyWinship Cancer Institute of Emory UniversityAtlantaGeorgiaUSA
| | - Mina S. Makary
- Department of RadiologyThe Ohio State University College of MedicineColumbusOhioUSA
| | - H. Wayne Slone
- Department of RadiologyThe Ohio State University College of MedicineColumbusOhioUSA
| | - Jaishri O. Blakeley
- Departments of Neurology, Neurosurgery, & OncologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - S. Alireza Mansouri
- Departments of Neurology, Neurosurgery, & OncologyJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Brian A. Neff
- Department of Otolaryngology Head and Neck SurgeryMayo ClinicRochesterMinnesotaUSA
| | - Robert K. Jackler
- Department of Otolaryngology Head and Neck SurgeryStanford UniversityPalo AltoCaliforniaUSA
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal MedicineThe Ohio State University College of Medicine and the Comprehensive Cancer CenterColumbusOhioUSA
| | - Long‐Sheng Chang
- Center for Childhood Cancer and Blood diseasesAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State University College of MedicineColumbusOhioUSA
- Department of Otolaryngology‐Head and Neck SurgeryThe Ohio State University College of MedicineColumbusOhioUSA
| |
Collapse
|
9
|
Integrated Analysis of Transcriptome and Differential Methylation of Neurofibromatosis Type 2 Vestibular Schwannomas. World Neurosurg 2021; 157:e66-e76. [PMID: 34587518 DOI: 10.1016/j.wneu.2021.09.094] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/19/2021] [Accepted: 09/20/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Vestibular schwannoma is the third most common benign intracranial tumor that can occur sporadically or be associated with neurofibromatosis type 2 (neurofibromatosis type 2 vestibular schwannoma [NF2-VS]). The aim of this study is to provide a comprehensive bioinformatic analysis of methylated-differentially expressed genes (MDEGs) in NF2-VS. METHODS Transcriptional sequencing datasets (GSE141801 and GSE108524) and gene methylation microarrays (GSE56598) from the Gene Expression Omnibus database were used to identify and analyze MDEGs in NF2-VS. A protein-protein interaction (PPI) network was built, and the hub genes and modules were identified. Finally, potential pharmacotherapy targeting MDEGs were extracted for NF2-VS. RESULTS A total of 57 hypermethylation-low expression genes and 88 hypomethylation-high expression genes were identified. Pathways associated with aberrantly MDEGs included P13K-AKT, MAPK, and Ras, which were also involved in NF2-VS. Six hub genes (EGFR, CCND1, CD53, CSF1R, PLAU, and FGFR1) were identified from the PPI network. Modification of the aforementioned genes altered cell-to-cell communication, response to stimulus, cellular regulation, and membrane and protein bindings. Thirty drugs targeting these pathways were selected based on the hub genes. CONCLUSIONS Analysis of MDEGs may enrich the understanding of the molecular mechanisms of NF2-VS pathogenesis and lay the groundwork for potential biomarkers and therapeutic targets for NF2-VS.
Collapse
|
10
|
Cao Y, Di X, Zhang Q, Li R, Wang K. RBM10 Regulates Tumor Apoptosis, Proliferation, and Metastasis. Front Oncol 2021; 11:603932. [PMID: 33718153 PMCID: PMC7943715 DOI: 10.3389/fonc.2021.603932] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/12/2021] [Indexed: 12/15/2022] Open
Abstract
The RNA-binding motif protein 10 (RBM10) is involved in alternative splicing and modifies mRNA post-transcriptionally. RBM10 is abnormally expressed in the lung, breast, and colorectal cancer, female genital tumors, osteosarcoma, and other malignant tumors. It can inhibit proliferation, promote apoptosis, and inhibit invasion and metastasis. RBM10 has long been considered a tumor suppressor because it promotes apoptosis through the regulation of the MDM2-p53 negative feedback loop, Bcl-2, Bax, and other apoptotic proteins and inhibits proliferation through the Notch signaling and rap1a/Akt/CREB pathways. However, it has been recently demonstrated that RBM10 can also promote cancer. Given these different views, it is necessary to summarize the research progress of RBM10 in various fields to reasonably analyze the underlying molecular mechanisms, and provide new ideas and directions for the clinical research of RBM10 in various cancer types. In this review, we provide a new perspective on the reasons for these opposing effects on cancer biology, molecular mechanisms, research progress, and clinical value of RBM10.
Collapse
Affiliation(s)
- Yingshu Cao
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Xin Di
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Qinghua Zhang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Ranwei Li
- Department of Urinary Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Ke Wang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Neurofibromatosis Type 2 (NF2) and the Implications for Vestibular Schwannoma and Meningioma Pathogenesis. Int J Mol Sci 2021; 22:ijms22020690. [PMID: 33445724 PMCID: PMC7828193 DOI: 10.3390/ijms22020690] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/06/2021] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
Patients diagnosed with neurofibromatosis type 2 (NF2) are extremely likely to develop meningiomas, in addition to vestibular schwannomas. Meningiomas are a common primary brain tumor; many NF2 patients suffer from multiple meningiomas. In NF2, patients have mutations in the NF2 gene, specifically with loss of function in a tumor-suppressor protein that has a number of synonymous names, including: Merlin, Neurofibromin 2, and schwannomin. Merlin is a 70 kDa protein that has 10 different isoforms. The Hippo Tumor Suppressor pathway is regulated upstream by Merlin. This pathway is critical in regulating cell proliferation and apoptosis, characteristics that are important for tumor progression. Mutations of the NF2 gene are strongly associated with NF2 diagnosis, leading to benign proliferative conditions such as vestibular schwannomas and meningiomas. Unfortunately, even though these tumors are benign, they are associated with significant morbidity and the potential for early mortality. In this review, we aim to encompass meningiomas and vestibular schwannomas as they pertain to NF2 by assessing molecular genetics, common tumor types, and tumor pathogenesis.
Collapse
|
12
|
Long J, Zhang Y, Huang X, Ren J, Zhong P, Wang B. A Review of Drug Therapy in Vestibular Schwannoma. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:75-85. [PMID: 33447015 PMCID: PMC7802892 DOI: 10.2147/dddt.s280069] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022]
Abstract
Vestibular schwannomas (VSs, also known as acoustic neuromas) are benign intracranial tumors commonly managed with observation, surgery, and radiotherapy. There is currently no approved pharmacotherapy for VS patients, which is why we conducted a detailed search of relevant literature from PubMed and Web of Science to explore recent advances and experiences in drug therapy. VSs feature a long course of disease that requires treatment to have minimal long-term side effects. Conventional chemotherapeutic agents are characterized by neurotoxicity or ototoxicity, poor effect on slow-growing tumors, and may induce new mutations in patients who have lost tumor suppressor function, and therefore are unsuitable for treating VSs. Along with the well-investigated molecular pathophysiology of VS and the increasingly accessible technology such as drug repositioning platform, many molecular targeted inhibitors have been identified and shown certain therapeutic effects in preclinical experiments or clinical trials.
Collapse
Affiliation(s)
- Jianfei Long
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Yu Zhang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Xiang Huang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Junwei Ren
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Ping Zhong
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Bin Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, People's Republic of China
| |
Collapse
|
13
|
Gugel I, Ebner FH, Grimm F, Czemmel S, Paulsen F, Hagel C, Tatagiba M, Nahnsen S, Tabatabai G. Contribution of mTOR and PTEN to Radioresistance in Sporadic and NF2-Associated Vestibular Schwannomas: A Microarray and Pathway Analysis. Cancers (Basel) 2020; 12:cancers12010177. [PMID: 31936793 PMCID: PMC7016954 DOI: 10.3390/cancers12010177] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 01/07/2020] [Indexed: 01/29/2023] Open
Abstract
The use of radiation treatment has increased for both sporadic and neurofibromatosis type 2 (NF2)-associated vestibular schwannoma (VS). However, there are a subset of radioresistant tumors and systemic treatments that are seldom used in these patients. We investigated molecular alterations after radiation in three NF2-associated and five sporadically operated recurrent VS after primary irradiation. We compared these findings with 49 non-irradiated (36 sporadic and 13 NF2-associated) VS through gene-expression profiling and pathway analysis. Furthermore, we stained the key molecules of the distinct pathway by immunohistochemistry. A total of 195 differentially expressed genes in sporadic and NF2-related comparisons showed significant differences based on the criteria of p value < 0.05 and a two-fold change. These genes were involved in pathways that are known to be altered upon irradiation (e.g., mammalian target of rapamycin (mTOR), phosphatase and tensin homolog (PTEN) and vascular endothelial growth factor (VEGF) signaling). We observed a combined downregulation of PTEN signaling and an upregulation of mTOR signaling in progressive NF2-associated VS after irradiation. Immunostainings with mTOR and PTEN antibodies confirmed the respective molecular alterations. Taken together, mTOR inhibition might be a promising therapeutic strategy in NF2-associated VS progress after irradiation.
Collapse
Affiliation(s)
- Isabel Gugel
- Center for Neuro-Oncol., Comprehensive Cancer Center Tübingen Stuttgart, 72076 Tübingen, Germany
- Department of Neurosurgery, University Hospital Tübingen, 72076 Tübingen, Germany
- Centre of Neurofibromatosis and Rare Diseases, University Hospital Tübingen, 72076 Tübingen, Germany
- Interdisciplinary Division of Neuro-Oncol., University Hospital Tübingen, 72076 Tübingen, Germany
- Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
- Correspondence: ; Tel.: +49-7071-2980325; Fax: +49-07071-295245
| | - Florian H. Ebner
- Department of Neurosurgery, Alfried Krupp Hospital, 45131 Essen, Germany
| | - Florian Grimm
- Center for Neuro-Oncol., Comprehensive Cancer Center Tübingen Stuttgart, 72076 Tübingen, Germany
- Department of Neurosurgery, University Hospital Tübingen, 72076 Tübingen, Germany
- Interdisciplinary Division of Neuro-Oncol., University Hospital Tübingen, 72076 Tübingen, Germany
| | - Stefan Czemmel
- Quantitative Biology Center (QBiC), University of Tübingen, 72076 Tübingen, Germany
| | - Frank Paulsen
- Center for Neuro-Oncol., Comprehensive Cancer Center Tübingen Stuttgart, 72076 Tübingen, Germany
- Interdisciplinary Division of Neuro-Oncol., University Hospital Tübingen, 72076 Tübingen, Germany
- Department of Radiation Oncology, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Christian Hagel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Marcos Tatagiba
- Center for Neuro-Oncol., Comprehensive Cancer Center Tübingen Stuttgart, 72076 Tübingen, Germany
- Department of Neurosurgery, University Hospital Tübingen, 72076 Tübingen, Germany
- Centre of Neurofibromatosis and Rare Diseases, University Hospital Tübingen, 72076 Tübingen, Germany
- Interdisciplinary Division of Neuro-Oncol., University Hospital Tübingen, 72076 Tübingen, Germany
- Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Sven Nahnsen
- Quantitative Biology Center (QBiC), University of Tübingen, 72076 Tübingen, Germany
| | - Ghazaleh Tabatabai
- Center for Neuro-Oncol., Comprehensive Cancer Center Tübingen Stuttgart, 72076 Tübingen, Germany
- Department of Neurosurgery, University Hospital Tübingen, 72076 Tübingen, Germany
- Interdisciplinary Division of Neuro-Oncol., University Hospital Tübingen, 72076 Tübingen, Germany
- Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
14
|
Sass H, Cayé-Thomasen P. Contemporary Molecular Biology of Sporadic Vestibular Schwannomas: A Systematic Review and Clinical Implications. J Int Adv Otol 2019; 14:322-329. [PMID: 30100540 DOI: 10.5152/iao.2018.4929] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In light of missing systematic reviews in the literature, the objective of this paper is to present the contemporary knowledge on the molecular biology of vestibular schwannomas (VS), based on a systematic literature search. In addition, current and prospected medical therapy based on molecular biology is addressed. A systematic literature search was conducted using the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. The systematic search was performed in the Pubmed and Embase databases. The following were the words searched: acoustic neuroma/vestibular schwannoma, molecular biology, gene, and microRNA. Specific inclusion and exclusion criteria were determined prior to search. The systematic search rendered 486 articles, ultimately yielding 69 included articles, whereas 35 were from relevant references. The occurrence of at least one mutation in the merlin gene was reported to range between 54% and 76%, whereas the loss of heterozygosity (LOH) corresponding to chromosome 22 occurs in 25% to 83% of sporadic VS. Global gene expression studies indicate that a number of genes other than merlin are at play. No high-level methylation of the merlin gene has been found. Several miRNAs are deregulated in tumor tissue, among others let-7d, miR-221, and miR-21. The acquired knowledge on molecular biology has led to several clinical implementations. Lack of the tumor suppressor merlin plays a principal role in the development of VS. Existing knowledge on the molecular biology has led to the first attempts of targeted medical treatment to prevent tumor growth. Future research is likely to introduce potential imaging markers with prognostic value and new targets for medical therapy.
Collapse
Affiliation(s)
- Hjalte Sass
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Copenhagen University Hospital, Copenhagen, Denmark; University of Copenhagen, School of Health and Medical Sciences, Copenhagen, Denmark
| | - Per Cayé-Thomasen
- Department of Otorhinolaryngology, Head and Neck Surgery and Audiology, Copenhagen University Hospital, Copenhagen, Denmark; University of Copenhagen, School of Health and Medical Sciences, Copenhagen, Denmark
| |
Collapse
|
15
|
Zhang YP, Liu KL, Wang YX, Yang Z, Han ZW, Lu BS, Qi JC, Yin YW, Teng ZH, Chang XL, Li JD, Xin H, Li W. Down-regulated RBM5 inhibits bladder cancer cell apoptosis by initiating an miR-432-5p/β-catenin feedback loop. FASEB J 2019; 33:10973-10985. [PMID: 31318608 DOI: 10.1096/fj.201900537r] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
RNA-binding motif protein 5 (RBM5) acts as a tumor suppressor in various human cancers and presents with several important characteristics, such as the potentiation of apoptosis, inhibition of the cell cycle, and alternative splicing of Fas and caspase-2 precursor mRNA. However, its role in bladder urothelial carcinoma (BUC) remains unknown. In this study, we found that RBM5 expression was significantly down-regulated in BUC tissues when compared with the adjacent nontumor tissues. The down-regulation of RBM5 activates β-catenin, which binds to the T-cell factor/lymphocyte enhancer factor element of the miR-432-5p promoter and elevates the expression of miR-432-5p in bladder cancer cells. The up-regulated miR-432-5p directly targets 3'-UTR and depresses RBM5 expression. Thus, RBM5-miR-432-5p-β-catenin forms a feedback loop in regulating bladder cancer cell apoptosis. Our findings provide evidence that the regulatory feedback loop among RBM5, miR-432-5p, and Wnt-β-catenin is responsible for the progress of bladder cancer cells.-Zhang, Y.-P., Liu, K.-L., Wang, Y.-X., Yang, Z., Han, Z.-W., Lu, B.-S., Qi, J.-C., Yin, Y.-W., Teng, Z.-H., Chang, X.-L., Li, J.-D., Xin, H., Li, W. Down-regulated RBM5 inhibits bladder cancer cell apoptosis by initiating an miR-432-5p/β-catenin feedback loop.
Collapse
Affiliation(s)
- Yan-Ping Zhang
- Department of Obstetrics, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Kai-Long Liu
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ya-Xuan Wang
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhan Yang
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhen-Wei Han
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bao-Sai Lu
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jin-Chun Qi
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yue-Wei Yin
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhi-Hai Teng
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xue-Liang Chang
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing-Dong Li
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hong Xin
- Department of Obstetrics, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wei Li
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
16
|
Xu J, Zhang Y, Shi Y, Yin D, Dai P, Zhao W, Zhang T. Identification of Predictive Proteins and Biological Pathways for the Tumorigenicity of Vestibular Schwannoma by Proteomic Profiling. Proteomics Clin Appl 2019; 13:e1800175. [PMID: 31120176 DOI: 10.1002/prca.201800175] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 04/17/2019] [Indexed: 12/30/2022]
Abstract
PURPOSE Vestibular schwannomas (VSs) are benign tumors that account for 8-10% of all intracranial tumors. So far, the tumorigenesis of VS has not been fully elucidated. This study is designed to identify differently expressed proteins involved in VS tumorigenesis. EXPERIMENTAL DESIGN An isobaric tag is used for relative and absolute quantification (iTRAQ) approach to characterize the protein expression profiles from pooled VS tissues (n = 12) and pooled matched normal vestibular tissues (n = 12). RESULTS A total of 933 differentially expressed proteins are identified between VS and the matched normal vestibular tissues, with 489 being upregulated and 444 being downregulated. Bioinformatics analyses are performed according to Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses. Several of the differentially expressed proteins are validated by western blotting analyses, and upregulation of LGALS1, ANXA1, GRB2, and STAT1 is validated in VS tissue by immunohistochemistry. CONCLUSIONS AND CLINICAL RELEVANCE The study represents the successful application of iTRAQ technology to an investigation of VS. Many of the differentially expressed proteins identified here have not been linked to VS before, and these dysregulated proteins may provide potential biomarkers for human VS diagnosis.
Collapse
Affiliation(s)
- Jianhui Xu
- ENT Institute and Otorhinolaryngology Department , Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| | - Yang Zhang
- ENT Institute and Otorhinolaryngology Department , Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| | - Yuxuan Shi
- ENT Institute and Otorhinolaryngology Department , Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| | - Dongming Yin
- ENT Institute and Otorhinolaryngology Department , Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| | - Peidong Dai
- ENT Institute and Otorhinolaryngology Department , Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| | - Weidong Zhao
- ENT Institute and Otorhinolaryngology Department , Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Department of Otology and Skull Base Surgery, Eye and Ear Nose Throat Hospital of Fudan University, Shanghai, 200031, China
| | - Tianyu Zhang
- ENT Institute and Otorhinolaryngology Department , Affiliated Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200031, China.,Key Laboratory of Hearing Medicine of NHFPC, Shanghai, 200031, China
| |
Collapse
|
17
|
Lei Y, Guo P, Li X, Zhang Y, Du T. Identification of Differentially Expressed miRNAs and mRNAs in Vestibular Schwannoma by Integrated Analysis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7267816. [PMID: 31309113 PMCID: PMC6594327 DOI: 10.1155/2019/7267816] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/22/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Vestibular schwannoma (VS) is benign, slow-growing brain tumor that negatively impacts patient quality of life, which may cause even death. This study aimed to explore key genes and microRNAs (miRNAs) associated with VS. METHODS The mRNA and miRNA expression profiles of VS downloaded from Gene Expression Omnibus (GEO) database were included in this study to perform an integrated analysis. The differentially expressed mRNAs (DEmRNAs) and miRNAs (DEmiRNAs) were identified. Then, functional annotation and protein-protein interaction networks (PPI) of DEmRNAs were constructed. DEmiRNA-target DEmRNAs analysis and functional annotation of DEmiRNA-target DEmRNAs were performed. RESULTS A total of 2627 DEmRNAs (1194 upregulated and 1433 downregulated DEmRNAs) and 21 DEmiRNAs (12 upregulated and 9 downregulated DEmiRNAs) were identified. ISG15, TLE1, and XPC were three hub proteins of VS-specific PPI network. A total of 2970 DEmiRNAs-DEmRNAs pairs were obtained. Among which, hsa-miR-181a-5p, hsa-miR-106-5p, and hsa-miR-34a-5p were the top three DEmiRNAs that covered most DEmRNAs. The functional annotation of DEmiRNA-target DEmRNAs revealed that the DEmiRNA-target DEmRNAs were significantly enriched in cGMP-PKG signaling pathway, adrenergic signaling in cardiomyocytes, and pathways in cancer. CONCLUSION The results of this present study may provide a comprehensive understanding for the roles of DEmRNAs and DEmiRNAs in the pathogenesis of VS and developing potential biomarkers of VS.
Collapse
Affiliation(s)
- Yanhua Lei
- Department of Otolaryngology, Jining No. 1 People's Hospital, China
| | - Ping Guo
- Department of Otolaryngology, Jining No. 1 People's Hospital, China
| | - Xiuguo Li
- Department of Otolaryngology, Jining No. 1 People's Hospital, China
| | - Yuanyuan Zhang
- Department of Otolaryngology, Jining No. 1 People's Hospital, China
| | - Ting Du
- Department of Otolaryngology, Jining No. 1 People's Hospital, China
| |
Collapse
|
18
|
Eser Ocak P, Ocak U, Tang J, Zhang JH. The role of caveolin-1 in tumors of the brain - functional and clinical implications. Cell Oncol (Dordr) 2019; 42:423-447. [PMID: 30993541 DOI: 10.1007/s13402-019-00447-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Caveolin-1 (cav-1) is the major structural protein of caveolae, the flask-shaped invaginations of the plasma membrane mainly involved in cell signaling. Today, cav-1 is believed to play a role in a variety of disease processes including cancer, owing to the variations of its expression in association with tumor progression, invasive behavior, metastasis and therapy resistance. Since first detected in the brain, a number of studies has particularly focused on the role of cav-1 in the various steps of brain tumorigenesis. In this review, we discuss the different roles of cav-1 and its contributions to the molecular mechanisms underlying the pathobiology and natural behavior of brain tumors including glial, non-glial and metastatic subtypes. These contributions could be attributed to its co-localization with important players in tumorigenesis within the lipid-enriched domains of the plasma membrane. In that regard, the ability of cav-1 to interact with various cell signaling molecules as well as the impact of caveolae depletion on important pathways acting in brain tumor pathogenesis are noteworthy. We also discuss conversant causes hampering the treatment of malignant glial tumors such as limited transport of chemotherapeutics across the blood tumor barrier and resistance to chemoradiotherapy, by focusing on the molecular fundamentals involving cav-1 participation. CONCLUSIONS Cav-1 has the potential to pivot the molecular basis underlying the pathobiology of brain tumors, particularly the malignant glial subtype. In addition, the regulatory effect of cav-1-dependent and caveola-mediated transcellular transport on the permeability of the blood tumor barrier could be of benefit to overcome the restricted transport across brain barriers when applying chemotherapeutics. The association of cav-1 with tumors of the brain other than malignant gliomas deserves to be underlined, as well given the evidence suggesting its potential in predicting tumor grade and recurrence rates together with determining patient prognosis in oligodendrogliomas, ependymomas, meningiomas, vestibular schwannomas and brain metastases.
Collapse
Affiliation(s)
- Pinar Eser Ocak
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Umut Ocak
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA. .,Department of Anesthesiology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA. .,Department of Neurology, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA. .,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92354, USA.
| |
Collapse
|
19
|
Rusheen AE, Smadbeck JB, Schimmenti LA, Klee EW, Link MJ, Vasmatzis G, Carlson ML. Proposal for Modification of Cahan's Criteria Utilizing Molecular Genetic Analyses for Cases without Baseline Histopathology: A Unique Method Applicable to Primary Radiosurgery. J Neurol Surg B Skull Base 2019; 80:10-17. [PMID: 30733895 PMCID: PMC6365249 DOI: 10.1055/s-0038-1655759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
Background Cahan's criteria have been utilized since 1948 to establish causality between prior radiation treatment and the development of secondary malignancy. One major criterion specifies that histological and radiographic evidence collected before and after radiation treatment must confirm separate tumor types; however, pretreatment biopsy is rarely obtained prior to radiosurgery for vestibular schwannoma and many other skull base and cranial lesions. Therefore, in these cases Cahan's criteria cannot be validly applied. Objective This article proposes an update to Cahan's criteria using modern molecular genetic analysis for cases lacking baseline histopathology. Methods Mate-pair sequencing and whole exome sequencing of a cerebellopontine angle undifferentiated high-grade pleomorphic sarcoma (UHGPS) that developed after stereotactic radiosurgery of a presumed benign vestibular schwannoma. Results Mate-pair sequencing and whole exome sequencing of the sarcoma revealed complex chromosomal aberrations. Notably, the tumor contained a deletion in the NF2 gene at 22q12 and an in-frame deletion on exon 5 of the remaining copy of NF2 . Biallelic events impacting NF2 are atypical for UHGPS but are characteristic for vestibular schwannoma. These findings help support the conclusion that the UHGPS arose from a benign vestibular schwannoma all along. Conclusions Next-generation sequencing can be successfully applied to a radiation-induced sarcoma when both the original and malignant tumors harbor separate signature genetic markers. As our understanding of the genetic profile of various tumors expand, we believe that next-generation sequencing and other genomic tools will play an increasingly important role in establishing causality between radiation and the development of secondary malignancy.
Collapse
Affiliation(s)
- Aaron E. Rusheen
- Medical Scientist Training Program, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States
| | - James B. Smadbeck
- Biomarker Discovery Program, Center of Individualized Medicine, Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Lisa A. Schimmenti
- Department of Otolaryngology-Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota, United States
| | - Eric W. Klee
- Biomarker Discovery Program, Center of Individualized Medicine, Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Michael J. Link
- Department of Otolaryngology-Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota, United States
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, United States
| | - George Vasmatzis
- Biomarker Discovery Program, Center of Individualized Medicine, Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States
| | - Matthew L. Carlson
- Department of Otolaryngology-Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota, United States
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, United States
| |
Collapse
|
20
|
Lovato A, García ibañez E, García ibañez L, Filippis C. Tumor growth rate: A new prognostic indicator of hearing preservation in vestibular schwannoma surgery. Laryngoscope 2019; 129:2378-2383. [DOI: 10.1002/lary.27805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 10/12/2018] [Accepted: 12/19/2018] [Indexed: 11/11/2022]
Affiliation(s)
- Andrea Lovato
- Department of NeurosciencesUniversity of Padova, Audiology Unit at Treviso Hospital Treviso Italy
| | | | | | - Cosimo Filippis
- Department of NeurosciencesUniversity of Padova, Audiology Unit at Treviso Hospital Treviso Italy
| |
Collapse
|
21
|
Xu Y, Su Z, Li J, Wang Q, Meng G, Zhang Y, Yang W, Zhang J, Gao P. Role of RNA-binding protein 5 in the diagnosis and chemotherapeutic response of lung cancer. Oncol Lett 2018; 17:2013-2019. [PMID: 30675268 DOI: 10.3892/ol.2018.9818] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 10/04/2018] [Indexed: 01/16/2023] Open
Abstract
Lung cancer remains one of the leading causes of cancer-associated mortality in the world. Lung carcinogenesis is frequently associated with deletions or the loss of heterozygosity at the critical chromosomal region 3p21.3, where RNA-binding protein 5 (RBM5) is localized. RBM5 regulates cell growth, cell cycle progression and apoptosis in cell homeostasis. In the lungs, altered RBM5 protein expression leads to alterations in cell growth and apoptosis, with subsequent lung pathogenesis and varied responses to treatment in patients with lung cancer. Detection of RBM5 expression may be a tumor marker for diagnosis, prediction and treatment response in lung cancer, and may be developed as a potential therapeutic target for drug resistant lung cancer. This review discusses the most recent progress on the role of RBM5 in lung cancer.
Collapse
Affiliation(s)
- Yanling Xu
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China.,Department of Geriatrics and General Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Zhenzhong Su
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Junyao Li
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Qi Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Guangping Meng
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yu Zhang
- Department of Geriatrics and General Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Wen Yang
- Department of Geriatrics and General Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Jie Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Peng Gao
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
22
|
Zhou C, Gao X, Hu S, Gan W, Xu J, Ma YC, Ma L. RBM-5 modulates U2AF large subunit-dependent alternative splicing in C. elegans. RNA Biol 2018; 15:1295-1308. [PMID: 30295127 PMCID: PMC6284560 DOI: 10.1080/15476286.2018.1526540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/23/2018] [Accepted: 09/11/2018] [Indexed: 01/06/2023] Open
Abstract
A key step in pre-mRNA splicing is the recognition of 3' splicing sites by the U2AF large and small subunits, a process regulated by numerous trans-acting splicing factors. How these trans-acting factors interact with U2AF in vivo is unclear. From a screen for suppressors of the temperature-sensitive (ts) lethality of the C. elegans U2AF large subunit gene uaf-1(n4588) mutants, we identified mutations in the RNA binding motif gene rbm-5, a homolog of the tumor suppressor gene RBM5. rbm-5 mutations can suppress uaf-1(n4588) ts-lethality by loss of function and neuronal expression of rbm-5 was sufficient to rescue the suppression. Transcriptome analyses indicate that uaf-1(n4588) affected the expression of numerous genes and rbm-5 mutations can partially reverse the abnormal gene expression to levels similar to that of wild type. Though rbm-5 mutations did not obviously affect alternative splicing per se, they can suppress or enhance, in a gene-specific manner, the altered splicing of genes in uaf-1(n4588) mutants. Specifically, the recognition of a weak 3' splice site was more susceptible to the effect of rbm-5. Our findings provide novel in vivo evidence that RBM-5 can modulate UAF-1-dependent RNA splicing and suggest that RBM5 might interact with U2AF large subunit to affect tumor formation.
Collapse
Affiliation(s)
- Chuanman Zhou
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Xiaoyang Gao
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Surong Hu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Wenjing Gan
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Jing Xu
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Yongchao C. Ma
- Departments of Pediatrics, Neurology and Physiology, Northwestern University Feinberg School of Medicine, Anne & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| | - Long Ma
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| |
Collapse
|
23
|
Gettelfinger JD, Dahl JP. Syndromic Hearing Loss: A Brief Review of Common Presentations and Genetics. J Pediatr Genet 2018; 7:1-8. [PMID: 29441214 PMCID: PMC5809162 DOI: 10.1055/s-0037-1617454] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023]
Abstract
Congenital hearing loss is one of the most common birth defects worldwide, with around 1 in 500 people experiencing some form of severe hearing loss. While over 400 different syndromes involving hearing loss have been described, it is important to be familiar with a wide range of syndromes involving hearing loss so an early diagnosis can be made and early intervention can be pursued to maximize functional hearing and speech-language development in the setting of verbal communication. This review aims to describe the presentation and genetics for some of the most frequently occurring syndromes involving hearing loss, including neurofibromatosis type 2, branchio-oto-renal syndrome, Treacher Collins syndrome, Stickler syndrome, Waardenburg syndrome, Pendred syndrome, Jervell and Lange-Nielsen syndrome, Usher syndromes, Refsum disease, Alport syndrome, MELAS, and MERRF.
Collapse
Affiliation(s)
- John D. Gettelfinger
- Department of Otolaryngology – Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - John P. Dahl
- Department of Otolaryngology – Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, Indiana, United States
| |
Collapse
|
24
|
Jamsai D, Watkins DN, O'Connor AE, Merriner DJ, Gursoy S, Bird AD, Kumar B, Miller A, Cole TJ, Jenkins BJ, O'Bryan MK. In vivo evidence that RBM5 is a tumour suppressor in the lung. Sci Rep 2017; 7:16323. [PMID: 29176597 PMCID: PMC5701194 DOI: 10.1038/s41598-017-15874-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 11/03/2017] [Indexed: 01/04/2023] Open
Abstract
Cigarette smoking is undoubtedly a risk factor for lung cancer. Moreover, smokers with genetic mutations on chromosome 3p21.3, a region frequently deleted in cancer and notably in lung cancer, have a dramatically higher risk of aggressive lung cancer. The RNA binding motif 5 (RBM5) is one of the component genes in the 3p21.3 tumour suppressor region. Studies using human cancer specimens and cell lines suggest a role for RBM5 as a tumour suppressor. Here we demonstrate, for the first time, an in vivo role for RBM5 as a tumour suppressor in the mouse lung. We generated Rbm5 loss-of-function mice and exposed them to a tobacco carcinogen NNK. Upon exposure to NNK, Rbm5 loss-of-function mice developed lung cancer at similar rates to wild type mice. As tumourigenesis progressed, however, reduced Rbm5 expression lead to significantly more aggressive lung cancer i.e. increased adenocarcinoma nodule numbers and tumour size. Our data provide in vivo evidence that reduced RBM5 function, as occurs in a large number of patients, coupled with exposure to tobacco carcinogens is a risk factor for an aggressive lung cancer phenotype. These data suggest that RBM5 loss-of-function likely underpins at least part of the pro-tumourigenic consequences of 3p21.3 deletion in humans.
Collapse
Affiliation(s)
- Duangporn Jamsai
- The School of Biological Sciences, Monash University, 25 Rainforest Walk, Clayton, Victoria, 3800, Australia.,The Development and Stem Cells Program of Monash Biomedicine Discovery Institute, 19 Innovation Walk, Clayton, Victoria, 3800, Australia
| | - D Neil Watkins
- Cancer Developmental Biology Group, The Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, Sydney, NSW 2010, Australia
| | - Anne E O'Connor
- The School of Biological Sciences, Monash University, 25 Rainforest Walk, Clayton, Victoria, 3800, Australia.,The Development and Stem Cells Program of Monash Biomedicine Discovery Institute, 19 Innovation Walk, Clayton, Victoria, 3800, Australia
| | - D Jo Merriner
- The School of Biological Sciences, Monash University, 25 Rainforest Walk, Clayton, Victoria, 3800, Australia.,The Development and Stem Cells Program of Monash Biomedicine Discovery Institute, 19 Innovation Walk, Clayton, Victoria, 3800, Australia
| | - Selen Gursoy
- The School of Biological Sciences, Monash University, 25 Rainforest Walk, Clayton, Victoria, 3800, Australia.,The Development and Stem Cells Program of Monash Biomedicine Discovery Institute, 19 Innovation Walk, Clayton, Victoria, 3800, Australia
| | - Anthony D Bird
- The Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia
| | - Beena Kumar
- Department of Anatomical Pathology, Monash Medical Centre, Monash Health, 246 Clayton Rd, Clayton, Victoria 3168, Australia
| | - Alistair Miller
- General and Respiratory Medicine, Monash Medical Centre, Monash Health, 246 Clayton Rd, Clayton, Victoria 3168, Australia
| | - Timothy J Cole
- The Development and Stem Cells Program of Monash Biomedicine Discovery Institute, 19 Innovation Walk, Clayton, Victoria, 3800, Australia.,The Department of Biochemistry and Molecular Biology, Monash University, 19 Innovation Walk, Clayton, Victoria 3800, Australia
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, 27-31 Wright St, Clayton, Victoria 3168, Australia
| | - Moira K O'Bryan
- The School of Biological Sciences, Monash University, 25 Rainforest Walk, Clayton, Victoria, 3800, Australia. .,The Development and Stem Cells Program of Monash Biomedicine Discovery Institute, 19 Innovation Walk, Clayton, Victoria, 3800, Australia.
| |
Collapse
|
25
|
Loiselle JJ, Roy JG, Sutherland LC. RBM10 promotes transformation-associated processes in small cell lung cancer and is directly regulated by RBM5. PLoS One 2017; 12:e0180258. [PMID: 28662214 PMCID: PMC5491171 DOI: 10.1371/journal.pone.0180258] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 06/13/2017] [Indexed: 11/19/2022] Open
Abstract
Lung cancers are the leading cause of cancer-related deaths worldwide, with small cell lung cancer (SCLC) being the most aggressive type. At the time of diagnosis, SCLC has usually already metastasized, and an astonishing 95% of patients eventually succumb to the disease. This highlights the need for more effective SCLC screening and treatment options. Interestingly, the earliest and most frequent genetic alteration associated with lung cancers involves a lesion in the region to which the RNA binding protein RBM5 maps. We have recently shown that a decrease in RBM5 expression may be a key step in SCLC development, as RBM5 regulated many transformation-associated processes in SCLC cells. RBM5 is structurally and functionally similar to another RNA binding protein, RBM10. Both proteins have tumor-suppressor properties in a variety of cancer cell lines, and it has been suggested that RBM5 expression can influence RBM10. Due to their similarities, and the recent evidence that RBM10 is mutated in up to 21% of lung cancers, we hypothesized that RBM10 would share RBM5's tumor-suppressor properties in SCLC. Using transcriptome analysis and functional assays, we show, however, that RBM10's function was opposite to what we hypothesized; in the endogenously RBM5-null GLC20 SCLC cell line, RBM10 actually promoted cell proliferation and other transformation-associated processes. Using RNA immunoprecipitation followed by next generation sequencing (RIP-Seq) and Western blotting, we demonstrate that RBM5 post-transcriptionally regulated RBM10 expression via direct interaction with specific RBM10 splice variants. We propose a working model describing the impact of this interaction on cellular processes. Our results provide evidence that RBM10 expression, in RBM5-null tumors, may contribute to tumor growth and metastasis. Measurement of both RBM10 and RBM5 expression in clinical samples may therefore hold prognostic and/or potentially predictive value.
Collapse
Affiliation(s)
- Julie J. Loiselle
- Biomolecular Sciences Program, Laurentian University, Sudbury, Ontario, Canada
| | - Justin G. Roy
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
| | - Leslie C. Sutherland
- Biomolecular Sciences Program, Laurentian University, Sudbury, Ontario, Canada
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Ontario, Canada
- Health Sciences North Research Institute (HSNRI), Sudbury, Ontario, Canada
| |
Collapse
|
26
|
Håvik AL, Bruland O, Myrseth E, Miletic H, Aarhus M, Knappskog PM, Lund-Johansen M. Genetic landscape of sporadic vestibular schwannoma. J Neurosurg 2017; 128:911-922. [PMID: 28409725 DOI: 10.3171/2016.10.jns161384] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Vestibular schwannoma (VS) is a benign tumor with associated morbidities and reduced quality of life. Except for mutations in NF2, the genetic landscape of VS remains to be elucidated. Little is known about the effect of Gamma Knife radiosurgery (GKRS) on the VS genome. The aim of this study was to characterize mutations occurring in this tumor to identify new genes and signaling pathways important for the development of VS. In addition, the authors sought to evaluate whether GKRS resulted in an increase in the number of mutations. METHODS Forty-six sporadic VSs, including 8 GKRS-treated tumors and corresponding blood samples, were subjected to whole-exome sequencing and tumor-specific DNA variants were called. Pathway analysis was performed using the Ingenuity Pathway Analysis software. In addition, multiplex ligation-dependent probe amplification was performed to characterize copy number variations in the NF2 gene, and microsatellite instability testing was done to investigate for DNA replication error. RESULTS With the exception of a single sample with an aggressive phenotype that harbored a large number of mutations, most samples showed a relatively low number of mutations. A median of 14 tumor-specific mutations in each sample were identified. The GKRS-treated tumors harbored no more mutations than the rest of the group. A clustering of mutations in the cancer-related axonal guidance pathway was identified (25 patients), as well as mutations in the CDC27 (5 patients) and USP8 (3 patients) genes. Thirty-five tumors harbored mutations in NF2 and 16 tumors had 2 mutational hits. The samples without detectable NF2 mutations harbored mutations in genes that could be linked to NF2 or to NF2-related functions. None of the tumors showed microsatellite instability. CONCLUSIONS The genetic landscape of VS seems to be quite heterogeneous; however, most samples had mutations in NF2 or in genes that could be linked to NF2. The results of this study do not link GKRS to an increased number of mutations.
Collapse
Affiliation(s)
- Aril Løge Håvik
- Departments of1Clinical Medicine.,2Center for Medical Genetics and Molecular Medicine, and.,3Clinical Science, and
| | - Ove Bruland
- 2Center for Medical Genetics and Molecular Medicine, and
| | | | - Hrvoje Miletic
- 5Pathology, Haukeland University Hospital, Bergen; and.,6K.G. Jebsen Brain Tumor Research Center, University of Bergen.,7Biomedicine, and
| | - Mads Aarhus
- 8Department of Neurosurgery, Oslo University Hospitals, Ullevål Sykehus, Oslo,Norway
| | - Per-Morten Knappskog
- 2Center for Medical Genetics and Molecular Medicine, and.,3Clinical Science, and
| | - Morten Lund-Johansen
- Departments of1Clinical Medicine.,Departments of4Neurosurgery and.,6K.G. Jebsen Brain Tumor Research Center, University of Bergen
| |
Collapse
|
27
|
Overexpression of RBM5 induces autophagy in human lung adenocarcinoma cells. World J Surg Oncol 2016; 14:57. [PMID: 26923134 PMCID: PMC4770605 DOI: 10.1186/s12957-016-0815-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/17/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Dysfunctions in autophagy and apoptosis are closely interacted and play an important role in cancer development. RNA binding motif 5 (RBM5) is a tumor suppressor gene, which inhibits tumor cells' growth and enhances chemosensitivity through inducing apoptosis in our previous studies. In this study, we investigated the relationship between RBM5 overexpression and autophagy in human lung adenocarcinoma cells. METHODS Human lung adenocarcinoma cancer (A549) cells were cultured in vitro and were transiently transfected with a RBM5 expressing plasmid (GV287-RBM5) or plasmid with scrambled control sequence. RBM5 expression was determined by semi-quantitative reverse transcription polymerase chain reaction (RT-PCR) and Western blot. Intracellular LC-3 I/II, Beclin-1, lysosome associated membrane protein-1 (LAMP1), Bcl-2, and NF-κB/p65 protein levels were detected by Western blot. Chemical staining with monodansylcadaverine (MDC) and acridine orange (AO) was applied to detect acidic vesicular organelles (AVOs). The ultrastructure changes were observed under transmission electron microscope (TEM). Then, transplanted tumor models of A549 cells on BALB/c nude mice were established and treated with the recombinant plasmids carried by attenuated Salmonella to induce RBM5 overexpression in tumor tissues. RBM5, LC-3, LAMP1, and Beclin1 expression was determined by immunohistochemistry staining in plasmids-treated A549 xenografts. RESULTS Our study demonstrated that overexpression of RBM5 caused an increase in the autophagy-related proteins including LC3-I, LC3-II, LC3-II/LC3-I ratio, Beclin1, and LAMP1 in A549 cells. A large number of autophagosomes with double-membrane structure and AVOs were detected in the cytoplasm of A549 cells transfected with GV287-RBM5 at 24 h. We observed that the protein level of NF-κB/P65 was increased and the protein level of Bcl-2 decreased by RBM5 overexpression. Furthermore, treatment with an autophagy inhibitor, 3-MA, enhanced RBM5-induced cell death and chemosensitivity in A549 cells. Furthermore, we successfully established the lung adenocarcinoma animal model using A549 cells. Overexpression of RBM5 enhanced the LC-3, LAMP1, and Beclin1 expression in the A549 xenografts. CONCLUSIONS Our findings showed for the first time that RBM5 overexpression induced autophagy in human lung adenocarcinoma cells, which might be driven by upregulation of Beclin1, NF-κB/P65, and downregulation of Bcl-2. RBM5-enhanced autophagy acts in a cytoprotective way and inhibition of autophagy may improve the anti-tumor efficacy of RBM5 in lung cancer.
Collapse
|
28
|
Ozuemba B, Masilamani TJ, Loiselle JJ, Koenderink B, Vanderbeck KA, Knee J, Larivière C, Sutherland LC. Co- and post-transcriptional regulation of Rbm5 and Rbm10 in mouse cells as evidenced by tissue-specific, developmental and disease-associated variation of splice variant and protein expression levels. Gene 2016; 580:26-36. [PMID: 26784654 DOI: 10.1016/j.gene.2015.12.070] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/31/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND Expression and function of the two RNA binding proteins and regulators of alternative splicing, RBM5 and RBM10, have largely been studied in human tissue and cell lines. The objective of the study described herein was to examine their expression in mouse tissue, in order to lay the framework for comprehensive functional studies using mouse models. METHODS All RNA variants of Rbm5 and Rbm10 were examined in a range of normal primary mouse tissues. RNA and protein were examined in differentiating C2C12 myoblasts and in denervated and dystonin-deficient mouse skeletal muscle. RESULTS All Rbm5 and Rbm10 variants examined were expressed in all mouse tissues and cell lines. In general, Rbm5 and Rbm10 RNA expression was higher in brain than in skin. RNA expression levels were more varied between cardiac and skeletal muscle, depending on the splice variant: for instance, Rbm10v1 RNA was higher in skeletal than cardiac muscle, whereas Rbm10v3 RNA was higher in cardiac than skeletal muscle. In mouse brain, cardiac and skeletal muscle, RNA encoding an approximately 17kDa potential paralogue of a small human RBM10 isoform was detected, and the protein observed in myoblasts and myotubes. Expression of Rbm5 and Rbm10 RNA remained constant during C2C12 myogenesis, but protein levels significantly decreased. In two muscle disease models, neither Rbm10 nor Rbm5 showed significant transcriptional changes, although significant specific alternative splicing changes of Rbm5 pre-mRNA were observed. Increased RBM10 protein levels were observed following denervation. CONCLUSIONS The varied co-transcriptional and post-transcriptional regulation aspects of Rbm5 and Rbm10 expression associated with mouse tissues, myogenesis and muscle disease states suggest that a mouse model would be an interesting and useful model in which to study comprehensive functional aspects of RBM5 and RBM10.
Collapse
Affiliation(s)
| | - Twinkle J Masilamani
- Biomolecular Sciences Program, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada
| | - Julie J Loiselle
- Biomolecular Sciences Program, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada
| | - Benjamin Koenderink
- AMRIC, Health Sciences North, 41 Ramsey Lake Road, Sudbury, ON, P3E 5J1, Canada
| | - Kaitlin A Vanderbeck
- School of Human Kinetics, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada
| | - Jose Knee
- AMRIC, Health Sciences North, 41 Ramsey Lake Road, Sudbury, ON, P3E 5J1, Canada
| | - Céline Larivière
- Biomolecular Sciences Program, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada; School of Human Kinetics, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada
| | - Leslie C Sutherland
- Biomolecular Sciences Program, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada; AMRIC, Health Sciences North, 41 Ramsey Lake Road, Sudbury, ON, P3E 5J1, Canada; Division of Medical Sciences, Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada; Department of Chemistry and Biochemistry, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada; Department of Medicine, Division of Medical Oncology, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
29
|
Lv XJ, Du YW, Hao YQ, Su ZZ, Zhang L, Zhao LJ, Zhang J. RNA-binding motif protein 5 inhibits the proliferation of cigarette smoke-transformed BEAS-2B cells through cell cycle arrest and apoptosis. Oncol Rep 2016; 35:2315-27. [PMID: 26782095 DOI: 10.3892/or.2016.4551] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/11/2015] [Indexed: 11/06/2022] Open
Abstract
Cigarette smoking has been shown to be the most significant risk factor for lung cancer. Recent studies have also indicated that RNA-binding motif protein 5 (RBM5) can modulate apoptosis and suppress tumor growth. The present study focused on the role of RBM5 in the regulation of cigarette smoke extract (CSE)-induced transformation of bronchial epithelial cells into the cancerous phenotype and its mechanism of action. Herein, we exposed normal BEAS-2B cells for 8 days to varying concentrations of CSE or dimethylsulfoxide (DMSO), followed by a recovery period of 2 weeks. Next, the RBM5 protein was overexpressed in these transformed BEAS-2B cells though lentiviral infection. Later, the morphological changes, cell proliferation, cell cycle, apoptosis, invasion and migration were assessed. In addition, we analyzed the role of RBM5 in xenograft growth. The expression of RBM5 along with the genes related to cell cycle regulation, apoptosis and invasion were also examined. Finally, our results revealed that BEAS-2B cells exposed to 100 µg/ml CSE acquired phenotypic changes and formed tumors in nude mice, indicative of their cancerous transformation and had reduced RBM5 expression. Subsequent overexpression of RBM5 in these cells significantly inhibited their proliferation, induced G1/S arrest, triggered apoptosis and inhibited their invasion and migration, including xenograft growth. Thus, we established an in vitro model of CSE-induced cancerous transformation and concluded that RBM5 overexpression inhibited the growth of these transformed cells through cell cycle arrest and induction of apoptosis. Therefore, our study suggests the importance of RBM5 in the pathogenesis of smoking-related cancer.
Collapse
Affiliation(s)
- Xue-Jiao Lv
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yan-Wei Du
- Department of Pathology, College of Basic Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Yu-Qiu Hao
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Zhen-Zhong Su
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Lin Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Li-Jing Zhao
- Department of Pathophysiology, College of Basic Medicine, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jie Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
30
|
Loiselle JJ, Tessier SJ, Sutherland LC. Post-transcriptional regulation of Rbm5 expression in undifferentiated H9c2 myoblasts. In Vitro Cell Dev Biol Anim 2015; 52:327-336. [PMID: 26659391 PMCID: PMC4833810 DOI: 10.1007/s11626-015-9976-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 11/04/2015] [Indexed: 12/20/2022]
Abstract
We previously examined the expression of Rbm5 during myoblast differentiation and found significantly more protein in the early stages of skeletal myoblast differentiation than during the later stages. We decided to determine if this elevated level was necessary for differentiation. Our hypothesis was that if high levels of Rbm5 protein expression were necessary for the initiation of skeletal myoblast differentiation, then inhibition of expression would prevent differentiation. Our long-term objective is to inhibit Rbm5 expression and examine the effect on H9c2 differentiation. Towards this end, stable knockdown clones and transient knockdown populations were generated. Expression analyses in H9c2 myoblasts demonstrated significant Rbm5 messenger RNA (mRNA) inhibition but, surprisingly, no effect on RBM5 protein levels. Expression of the Rbm5 paralogue Rbm10 was examined in order to (a) ensure no off-target knockdown effect, and (b) investigate any possible compensatory effects. RBM10 protein levels were found to be elevated, in both the clonal and transiently transfected populations. These results suggest that myoblast RBM5 expression is regulated by a process that includes RNA sequestration and/or controlled translation, and that (a) RBM5 function is compensated for by RBM10, and/or (b) RBM5 regulates RBM10 expression. We have developed a model to describe our findings, and suggest further experiments for testing its validity. Since upregulation of Rbm10 might compensate for downregulated Rbm5, and consequently might mask any potential knockdown effect, it could lead to incorrect conclusions regarding the importance of Rbm5 for differentiation. It is therefore imperative to determine how both RBM5 and RBM10 protein expression is regulated.
Collapse
Affiliation(s)
- Julie J Loiselle
- Biomolecular Sciences Program, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada. .,AMRIC, Health Sciences North, 41 Ramsey Lake Road, Sudbury, ON, P3E 5J1, Canada.
| | - Sarah J Tessier
- Department of Biology, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada
| | - Leslie C Sutherland
- Biomolecular Sciences Program, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada.,AMRIC, Health Sciences North, 41 Ramsey Lake Road, Sudbury, ON, P3E 5J1, Canada.,Department of Chemistry and Biochemistry, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada.,Division of Medical Sciences, Northern Ontario School of Medicine, Laurentian University, 935 Ramsey Lake Road, Sudbury, ON, P3E 2C6, Canada.,Department of Medicine, Division of Medical Oncology, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
31
|
Dilwali S, Briët MC, Kao SY, Fujita T, Landegger LD, Platt MP, Stankovic KM. Preclinical validation of anti-nuclear factor-kappa B therapy to inhibit human vestibular schwannoma growth. Mol Oncol 2015; 9:1359-70. [PMID: 25891780 DOI: 10.1016/j.molonc.2015.03.009] [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: 08/12/2014] [Revised: 02/22/2015] [Accepted: 03/23/2015] [Indexed: 01/25/2023] Open
Abstract
Vestibular schwannomas (VSs), the most common tumors of the cerebellopontine angle, arise from Schwann cells lining the vestibular nerve. Pharmacotherapies against VS are almost non-existent. Although the therapeutic inhibition of inflammatory modulators has been established for other neoplasms, it has not been explored in VS. A bioinformatic network analysis of all genes reported to be differentially expressed in human VS revealed a pro-inflammatory transcription factor nuclear factor-kappa B (NF-κB) as a central molecule in VS pathobiology. Assessed at the transcriptional and translational level, canonical NF-κB complex was aberrantly activated in human VS and derived VS cultures in comparison to control nerves and Schwann cells, respectively. Cultured primary VS cells and VS-derived human cell line HEI-193 were treated with specific NF-κB siRNAs, experimental NF-κB inhibitor BAY11-7082 (BAY11) and clinically relevant NF-κB inhibitor curcumin. Healthy human control Schwann cells from the great auricular nerve were also treated with BAY11 and curcumin to assess toxicity. All three treatments significantly reduced proliferation in primary VS cultures and HEI-193 cells, with siRNA, 5 μM BAY11 and 50 μM curcumin reducing average proliferation (±standard error of mean) to 62.33% ± 10.59%, 14.3 ± 9.7%, and 23.0 ± 20.9% of control primary VS cells, respectively. These treatments also induced substantial cell death. Curcumin, unlike BAY11, also affected primary Schwann cells. This work highlights NF-κB as a key modulator in VS cell proliferation and survival and demonstrates therapeutic efficacy of directly targeting NF-κB in VS.
Collapse
Affiliation(s)
- Sonam Dilwali
- Eaton Peabody Laboratories, Department of Otolaryngology, 243 Charles Street, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Harvard-MIT Program in Speech and Hearing Bioscience and Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | - Martijn C Briët
- Eaton Peabody Laboratories, Department of Otolaryngology, 243 Charles Street, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Otorhinolaryngology, Leiden University Medical Centre, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Shyan-Yuan Kao
- Eaton Peabody Laboratories, Department of Otolaryngology, 243 Charles Street, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA.
| | - Takeshi Fujita
- Eaton Peabody Laboratories, Department of Otolaryngology, 243 Charles Street, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Otology and Laryngology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA.
| | - Lukas D Landegger
- Eaton Peabody Laboratories, Department of Otolaryngology, 243 Charles Street, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Department of Otology and Laryngology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA.
| | - Michael P Platt
- Department of Otology and Laryngology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA; Department of Otolaryngology-Head and Neck Surgery, Boston University, 72 E Concord Street, Boston, MA 02118, USA.
| | - Konstantina M Stankovic
- Eaton Peabody Laboratories, Department of Otolaryngology, 243 Charles Street, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, USA; Harvard-MIT Program in Speech and Hearing Bioscience and Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Department of Otology and Laryngology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA.
| |
Collapse
|
32
|
Hao YQ, Su ZZ, Lv XJ, Li P, Gao P, Wang C, Bai Y, Zhang J. RNA-binding motif protein 5 negatively regulates the activity of Wnt/β-catenin signaling in cigarette smoke-induced alveolar epithelial injury. Oncol Rep 2015; 33:2438-44. [PMID: 25738917 DOI: 10.3892/or.2015.3828] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/17/2015] [Indexed: 11/06/2022] Open
Abstract
Cigarette smoking is closely associated with various respiratory diseases. Oxidants and carcinogens in cigarettes are reported to induce various airway epithelial injuries. However, the underlying mechanisms remain unclear. The aims of the present study were to determine the involvement of RNA-binding motif protein 5 (RBM5) and Wnt/β-catenin signaling in cigarette smoke-induced alveolar epithelial injury, as well as the interaction between both. A549 cells were treated with cigarette smoke extract (CSE). The MTT assay was used to assess the effects of CSE on cell viability. The levels of RBM5 and Wnt/β-catenin/GSK3β were detected by semi-quantitative reverse transcription-polymerase chain reaction (RT-PCR) and western blot analysis. A luciferase assay was used to assess the activity of β-catenin/T-cell factor (TCF) signaling. The results revealed that CSE inhibited A549 cell viability in both a dose- and time-dependent manner. Cytosolic and nuclear β-catenin levels were significantly increased following CSE treatment, compared with those in the control cells (P<0.05). The luciferase activity in CSE-exposed cells transfected with the TCF luciferase reporter wild-type plasmid (pGL3-OT) was significantly greater than that in cells without CSE exposure (33,167±3,085 vs. 19,978±1,916, respectively, P<0.05). Both the mRNA and protein levels of RBM5 in the CSE-treated cells were significantly reduced compared to the levels in the controls (all P<0.05). The overexpression of RBM5 inhibited Wnt/β-catenin signaling in the A549 cells, while silencing of RBM5 enhanced Wnt/β-catenin signaling. The β-catenin/TCF signaling inhibitor ICG-001 had no apparent effect on the RBM5 levels. Downregulation of RBM5 and activation of Wnt/β-catenin signaling are involved in CSE-induced alveolar epithelial injury. RBM5 acts as an upstream molecule that negatively regulates the activity of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Yu-Qiu Hao
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Chuangchun, Jilin 130041, P.R. China
| | - Zhen-Zhong Su
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Chuangchun, Jilin 130041, P.R. China
| | - Xue-Jiao Lv
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Chuangchun, Jilin 130041, P.R. China
| | - Ping Li
- Department of Paediatrics, The Second Affiliated Hospital of Jilin University, Chuangchun, Jilin 130041, P.R. China
| | - Peng Gao
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Chuangchun, Jilin 130041, P.R. China
| | - Chen Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Chuangchun, Jilin 130041, P.R. China
| | - Yue Bai
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Chuangchun, Jilin 130041, P.R. China
| | - Jie Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Chuangchun, Jilin 130041, P.R. China
| |
Collapse
|
33
|
|
34
|
Zhang L, Zhang Q, Yang Y, Wu C. The RNA recognition motif domains of RBM5 are required for RNA binding and cancer cell proliferation inhibition. Biochem Biophys Res Commun 2014; 444:445-50. [PMID: 24486491 DOI: 10.1016/j.bbrc.2014.01.102] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 01/21/2014] [Indexed: 01/08/2023]
Abstract
RBM5 is a known putative tumor suppressor gene that has been shown to function in cell growth inhibition by modulating apoptosis. RBM5 also plays a critical role in alternative splicing as an RNA binding protein. However, it is still unclear which domains of RBM5 are required for RNA binding and related functional activities. We hypothesized the two putative RNA recognition motif (RRM) domains of RBM5 spanning from amino acids 98-178 and 231-315 are essential for RBM5-mediated cell growth inhibition, apoptosis regulation, and RNA binding. To investigate this hypothesis, we evaluated the activities of the wide-type and mutant RBM5 gene transfer in low-RBM5 expressing A549 cells. We found that, unlike wild-type RBM5 (RBM5-wt), a RBM5 mutant lacking the two RRM domains (RBM5-ΔRRM), is unable to bind RNA, has compromised caspase-2 alternative splicing activity, lacks cell proliferation inhibition and apoptosis induction function in A549 cells. These data provide direct evidence that the two RRM domains of RBM5 are required for RNA binding and the RNA binding activity of RBM5 contributes to its function on apoptosis induction and cell growth inhibition.
Collapse
Affiliation(s)
- Lei Zhang
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, PR China
| | - Qing Zhang
- Jiangsu Key Laboratory of Biological Cancer Therapy, Xuzhou Medical College, Xuzhou 221002, PR China
| | - Yu Yang
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, PR China
| | - Chuanfang Wu
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, PR China.
| |
Collapse
|
35
|
Bechara EG, Sebestyén E, Bernardis I, Eyras E, Valcárcel J. RBM5, 6, and 10 differentially regulate NUMB alternative splicing to control cancer cell proliferation. Mol Cell 2013; 52:720-33. [PMID: 24332178 DOI: 10.1016/j.molcel.2013.11.010] [Citation(s) in RCA: 253] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 09/18/2013] [Accepted: 10/22/2013] [Indexed: 12/25/2022]
Abstract
RBM5, a regulator of alternative splicing of apoptotic genes, and its highly homologous RBM6 and RBM10 are RNA-binding proteins frequently deleted or mutated in lung cancer. We report that RBM5/6 and RBM10 antagonistically regulate the proliferative capacity of cancer cells and display distinct positional effects in alternative splicing regulation. We identify the Notch pathway regulator NUMB as a key target of these factors in the control of cell proliferation. NUMB alternative splicing, which is frequently altered in lung cancer, can regulate colony and xenograft tumor formation, and its modulation recapitulates or antagonizes the effects of RBM5, 6, and 10 in cell colony formation. RBM10 mutations identified in lung cancer cells disrupt NUMB splicing regulation to promote cell growth. Our results reveal a key genetic circuit in the control of cancer cell proliferation.
Collapse
Affiliation(s)
- Elias G Bechara
- Centre de Regulació Genòmica, Dr. Aiguader, 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra, Dr. Aiguader, 88, 08003 Barcelona, Spain
| | - Endre Sebestyén
- Universitat Pompeu Fabra, Dr. Aiguader, 88, 08003 Barcelona, Spain
| | | | - Eduardo Eyras
- Universitat Pompeu Fabra, Dr. Aiguader, 88, 08003 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Dr. Aiguader, 88, 08003 Barcelona, Spain
| | - Juan Valcárcel
- Centre de Regulació Genòmica, Dr. Aiguader, 88, 08003 Barcelona, Spain; Universitat Pompeu Fabra, Dr. Aiguader, 88, 08003 Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Dr. Aiguader, 88, 08003 Barcelona, Spain.
| |
Collapse
|
36
|
Luo W, Ren X, Chen S, Liu H, Sui D, Lin S. Intracranial intraparenchymal and intraventricular schwannomas: Report of 18 cases. Clin Neurol Neurosurg 2013. [DOI: 10.1016/j.clineuro.2012.10.029] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
37
|
Torres-Martin M, Lassaletta L, de Campos JM, Isla A, Gavilan J, Pinto GR, Burbano RR, Latif F, Melendez B, Castresana JS, Rey JA. Global profiling in vestibular schwannomas shows critical deregulation of microRNAs and upregulation in those included in chromosomal region 14q32. PLoS One 2013; 8:e65868. [PMID: 23776562 PMCID: PMC3679163 DOI: 10.1371/journal.pone.0065868] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 04/29/2013] [Indexed: 12/21/2022] Open
Abstract
Background Vestibular schwannomas are benign tumors that arise from Schwann cells in the VIII cranial pair and usually present NF2 gene mutations and/or loss of heterozygosity on chromosome 22q. Deregulation has also been found in several genes, such as ERBB2 and NRG1. MicroRNAs are non-coding RNAs approximately 21 to 23 nucleotides in length that regulate mRNAs, usually by degradation at the post-transcriptional level. Methods We used microarray technology to test the deregulation of miRNAs and other non-coding RNAs present in GeneChip miRNA 1.0 (Affymetrix) over 16 vestibular schwannomas and 3 control-nerves, validating 10 of them by qRT-PCR. Findings Our results showed the deregulation of 174 miRNAs, including miR-10b, miR-206, miR-183 and miR-204, and the upregulation of miR-431, miR-221, miR-21 and miR-720, among others. The results also showed an aberrant expression of other non-coding RNAs. We also found a general upregulation of the miRNA cluster located at chromosome 14q32. Conclusion Our results suggest that several miRNAs are involved in tumor formation and/or maintenance and that global upregulation of the 14q32 chromosomal site contains miRNAs that may represent a therapeutic target for this neoplasm.
Collapse
Affiliation(s)
- Miguel Torres-Martin
- Neuro-Oncology Laboratory, Research Unit, La Paz University Hospital, IdiPAZ, Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Torres-Martin M, Lassaletta L, San-Roman-Montero J, De Campos JM, Isla A, Gavilan J, Melendez B, Pinto GR, Burbano RR, Castresana JS, Rey JA. Microarray analysis of gene expression in vestibular schwannomas reveals SPP1/MET signaling pathway and androgen receptor deregulation. Int J Oncol 2013; 42:848-62. [PMID: 23354516 PMCID: PMC3597452 DOI: 10.3892/ijo.2013.1798] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 01/04/2013] [Indexed: 11/06/2022] Open
Abstract
Vestibular schwannomas are benign neoplasms that arise from the vestibular nerve. The hallmark of these tumors is the biallelic inactivation of neurofibromin 2 (NF2). Transcriptomic alterations, such as the neuregulin 1 (NRG1)/ErbB2 pathway, have been described in schwannomas. In this study, we performed a whole transcriptome analysis in 31 vestibular schwannomas and 9 control nerves in the Affymetrix Gene 1.0 ST platform, validated by quantitative real-time PCR (qRT-PCR) using TaqMan Low Density arrays. We performed a mutational analysis of NF2 by PCR/denaturing high-performance liquid chromatography (dHPLC) and multiplex ligation-dependent probe amplification (MLPA), as well as a microsatellite marker analysis of the loss of heterozygosity (LOH) of chromosome 22q. The microarray analysis demonstrated that 1,516 genes were deregulated and 48 of the genes were validated by qRT-PCR. At least 2 genetic hits (allelic loss and/or gene mutation) in NF2 were found in 16 tumors, seven cases showed 1 hit and 8 tumors showed no NF2 alteration. MET and associated genes, such as integrin, alpha 4 (ITGA4)/B6, PLEXNB3/SEMA5 and caveolin-1 (CAV1) showed a clear deregulation in vestibular schwannomas. In addition, androgen receptor (AR) downregulation may denote a hormonal effect or cause in this tumor. Furthermore, the osteopontin gene (SPP1), which is involved in merlin protein degradation, was upregulated, which suggests that this mechanism may also exert a pivotal role in schwannoma merlin depletion. Finally, no major differences were observed among tumors of different size, histological type or NF2 status, which suggests that, at the mRNA level, all schwannomas, regardless of their molecular and clinical characteristics, may share common features that can be used in their treatment.
Collapse
Affiliation(s)
- Miguel Torres-Martin
- Research Unit, La Paz University Hospital, Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Burns SS, Akhmametyeva EM, Oblinger JL, Bush ML, Huang J, Senner V, Chen CS, Jacob A, Welling DB, Chang LS. Histone deacetylase inhibitor AR-42 differentially affects cell-cycle transit in meningeal and meningioma cells, potently inhibiting NF2-deficient meningioma growth. Cancer Res 2013; 73:792-803. [PMID: 23151902 PMCID: PMC3549000 DOI: 10.1158/0008-5472.can-12-1888] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Meningiomas constitute about 34% of primary intracranial tumors and are associated with increased mortality in patients with neurofibromatosis type 2 (NF2). To evaluate potential medical therapies for these tumors, we have established a quantifiable orthotopic model for NF2-deficient meningiomas. We showed that telomerase-immortalized Ben-Men-1 benign meningioma cells harbored a single nucleotide deletion in NF2 exon 7 and did not express the NF2 protein, merlin. We also showed that AR-42, a pan-histone deacetylase inhibitor, inhibited proliferation of both Ben-Men-1 and normal meningeal cells by increasing expression of p16(INK4A), p21(CIP1/WAF1), and p27(KIP1). In addition, AR-42 increased proapoptotic Bim expression and decreased anti-apoptotic Bcl(XL) levels. However, AR-42 predominantly arrested Ben-Men-1 cells at G(2)-M whereas it induced cell-cycle arrest at G(1) in meningeal cells. Consistently, AR-42 substantially decreased the levels of cyclin D1, E, and A, and proliferating cell nuclear antigen in meningeal cells while significantly reducing the expression of cyclin B, important for progression through G(2), in Ben-Men-1 cells. In addition, AR-42 decreased Aurora A and B expression. To compare the in vivo efficacies of AR-42 and AR-12, a PDK1 inhibitor, we generated and used luciferase-expressing Ben-Men-1-LucB cells to establish intracranial xenografts that grew over time. While AR-12 treatment moderately slowed tumor growth, AR-42 caused regression of Ben-Men-1-LucB tumors. Importantly, AR-42-treated tumors showed minimal regrowth when xenograft-bearing mice were switched to normal diet. Together, these results suggest that AR-42 is a potential therapy for meningiomas. The differential effect of AR-42 on cell-cycle progression of normal meningeal and meningioma cells may have implications for why AR-42 is well-tolerated while it potently inhibits tumor growth.
Collapse
Affiliation(s)
- Sarah S. Burns
- Center for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- Department of Otolaryngology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Elena M. Akhmametyeva
- Center for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Janet L. Oblinger
- Center for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- Department of Otolaryngology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Matthew L. Bush
- Department of Otolaryngology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Jie Huang
- Center for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Volker Senner
- Institut für Neuropathologie, Universitätsklinikum Münster, Germany
| | - Ching-Shih Chen
- The Ohio State University College of Pharmacy, Columbus, Ohio, USA
| | - Abraham Jacob
- Department of Otolaryngology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - D. Bradley Welling
- Department of Otolaryngology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Long-Sheng Chang
- Center for Childhood Cancer, The Research Institute at Nationwide Children’s Hospital, Columbus, Ohio, USA
- Department of Otolaryngology, The Ohio State University College of Medicine, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
40
|
Peng J, Valeshabad AK, Li Q, Wang Y. Differential expression of RBM5 and KRAS in pancreatic ductal adenocarcinoma and their association with clinicopathological features. Oncol Lett 2012; 5:1000-1004. [PMID: 23425895 PMCID: PMC3576315 DOI: 10.3892/ol.2012.1080] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 12/11/2012] [Indexed: 12/13/2022] Open
Abstract
RNA binding motif 5 (RBM5) is a tumor suppressor gene that regulates cell proliferation, differentiation and apoptosis through pre-mRNA splicing of related genes. This study aimed to detect RBM5 and KRAS expression in pancreatic ductal adenocarcinoma and their association with clinicopathological features. Detection of RBM5 and KRAS expression by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and western blotting was performed at mRNA and protein levels, respectively, in pancreatic cancer and non-tumor tissues. In addition, the association of RBM5 and KRAS expression with clinicopathological parameters and tumor recurrence was analyzed. The expression of RBM5 was significantly downregulated in pancreatic cancer tissues compared to peritumoral tissues at the mRNA and protein levels. Contrastingly, KRAS was significantly overexpressed in pancreatic cancerous tissues compared to peritumoral tissues. Analysis revealed that RBM5 expression was negatively correlated with KRAS expression in pancreatic cancer. Furthermore, reduced RBM5 expression has a close association with lymph node metastasis, distant metastasis, Union for International Cancer Control (UICC) stage and nerve and venous invasion, while overexpression of KRAS proteins was significantly correlated with tumor size, lymph node metastasis, UICC stage and nerve and venous invasion of pancreatic cancer. Significant RBM5 underexpression and KRAS overexpression were observed in pancreatic cancer compared to non-tumor tissues. There is a close association of differential RBM5 and KRAS with poor clinicopathological features, suggesting their potential roles in the progression and metastasis of pancreatic cancer.
Collapse
Affiliation(s)
- Jie Peng
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P.R. China
| | | | | | | |
Collapse
|
41
|
Zhao L, Li R, Shao C, Li P, Liu J, Wang K. 3p21.3 tumor suppressor gene RBM5 inhibits growth of human prostate cancer PC-3 cells through apoptosis. World J Surg Oncol 2012; 10:247. [PMID: 23158838 PMCID: PMC3544648 DOI: 10.1186/1477-7819-10-247] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 10/24/2012] [Indexed: 01/12/2023] Open
Abstract
Background Recent studies have indicated that the nuclear RNA-binding protein RBM5 has the ability to modulate apoptosis and suppress tumor growth. The aim of this study is to investigate the expression of RBM5 in human prostate cancer and its mechanism of tumor suppression. Methods The expression of RBM5 protein in cancerous prostatic tissues and normal tissues was examined by IHC. PC-3 cell line was used to determine the apoptotic function of RBM5 in vitro. PC-3 cells were transiently transfected with pcDNA3.1-RBM5. Cell viability was determined by MTT assay. Rhodamine 123 staining and Annexin V analysis were performed to observe the apoptotic activity of PC-3 cells overexpressing RBM5. Expression of apoptosis-related genes was assessed by western blot. Results The expression of RBM5 protein was significantly decreased in cancerous prostatic tissues compared to the normal tissues. PC-3 cells overexpressing RBM5 showed not only significant growth inhibition compared with the vector controls, but also dysfunction of mitochondrial membrane potential and increased apoptotic activity. To further define RBM5 function in apoptotic pathways, we investigated differential expression profiles of various BH3-only proteins including Bid, Bad, and Bim, and apoptosis regulatory proteins include P53, cleaved caspase9, and cleaved caspase3. We found that the expression of both BH3-only proteins and apoptosis regulatory proteins was increased in RBM5 transfected cells. Conclusion The expression of RBM5 protein was significantly decreased in cancerous prostatic tissues, which suggests that RBM5 plays an important role in the pathogenesis of prostate cancer. RBM5 may induce the apoptosis of prostate cancer PC-3 cells by modulating the mitochondrial apoptotic pathway, and thus RBM5 might be a promising target for gene therapy on prostate cancer.
Collapse
Affiliation(s)
- Lijing Zhao
- Department of Pathophysiology, Norman Bethune College of Medicine of Jilin University, Changchun, Jilin 130021, China
| | | | | | | | | | | |
Collapse
|
42
|
Shao C, Zhao L, Wang K, Xu W, Zhang J, Yang B. The tumor suppressor gene RBM5 inhibits lung adenocarcinoma cell growth and induces apoptosis. World J Surg Oncol 2012; 10:160. [PMID: 22866867 PMCID: PMC3502321 DOI: 10.1186/1477-7819-10-160] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 07/15/2012] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND The loss of tumor suppressor gene (TSG) function is a critical step in the pathogenesis of human lung cancer. RBM5 (RNA-binding motif protein 5, also named H37/LUCA-15) gene from chromosome 3p21.3 demonstrated tumor suppressor activity. However, the role of RBM5 played in the occurrence and development of lung cancer is still not well understood. METHOD Paired non-tumor and tumor tissues were obtained from 30 adenocarcinomas. The expression of RBM5 mRNA and protein was examined by RT-PCR and Western blot. A549 cell line was used to determine the apoptotic function of RBM5 in vitro. A549 cells were transiently transfected with pcDNA3.1-RBM5. AnnexinV analysis was performed by flow cytometry. Expression of Bcl-2, cleaved caspase-3, caspase-9 and PAPP proteins in A549 lung cancer cells and the A549 xenograft BALB/c nude mice model was determined by Western blot. Tumor suppressor activity of RBM5 was also examined in the A549 xenograft model treated with pcDNA3.1-RBM5 plasmid carried by attenuated Salmonella typhi Ty21a. RESULT The expression of RBM5 mRNA and protein was decreased significantly in adenocarcinoma tissues compared to that in the non-tumor tissues. In addition, as compared to the vector control, a significant growth inhibition of A549 lung cancer cells was observed when transfected with pcDNA3.1-RBM5 as determined by cell proliferation assay. We also found that overexpression of RBM5 induced both early and late apoptosis in A549 cells using AnnexinV/PI staining as determined by flow cytometry. Furthermore, the expression of Bcl-2 protein was decreased, whereas the expression of cleaved caspase-3, caspase-9 and PARP proteins was significantly increased in the RBM5 transfected cells; similarly, expression of decreased Bcl-2 and increased cleaved caspase-3 proteins was also examined in the A549 xenograft model. More importantly, we showed that accumulative and stable overexpression of RBM5 in the A549 xenograft BALB/c nude mice model significantly inhibited the tumor growth rate in vivo as compared to that in the control. CONCLUSION Our study demonstrates that RBM5 can inhibit the growth of lung cancer cells and induce apoptosis both in vitro and in vivo, which suggests that RBM5 might be used as a potential biomarker or target for lung cancer diagnosis and chemotherapy. Moreover, we propose a novel animal model set up in BALB/c nude mice treated with attenuated Salmonella as a vector carrying plasmids to determine RBM5 function in vivo.
Collapse
Affiliation(s)
- Chen Shao
- Department of Pathophysiology, Norman Bethune College of Medicine of Jilin University, Changchun, Jilin 130021, China
| | | | | | | | | | | |
Collapse
|
43
|
Li P, Wang K, Zhang J, Zhao L, Liang H, Shao C, Sutherland LC. The 3p21.3 tumor suppressor RBM5 resensitizes cisplatin-resistant human non-small cell lung cancer cells to cisplatin. Cancer Epidemiol 2012; 36:481-9. [PMID: 22609235 DOI: 10.1016/j.canep.2012.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Revised: 04/06/2012] [Accepted: 04/18/2012] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Increasing RBM5 levels inhibit tumor cell growth and promote apoptosis. In this study, we investigated the role of RBM5 in the cisplatin resistance observed in human lung non-small cell lung cancer cells and evaluated the effect of RBM5 modulation on cell growth inhibition and apoptosis induced by cisplatin in the parental non-small cell lung cancer cells A549 and their cisplatin resistant counterparts, A549/DDP cells. METHODS RBM5 mRNA and protein expression in the A549 and A549/DDP cells was analyzed by semi-quantitative RT-PCR and western blot. The A549/DDP cells were then transfected with a pcDNA3-RBM5 plasmid, and an RBM5-specific siRNA was transfected into A549 cells, prior to treatment with cisplatin. Semi-quantitative RT-PCR and western blot analyses were performed to confirm the expression of RBM5 mRNA or protein, and knockdown of RBM5 mRNA or protein, respectively. MTT assays were used to evaluate chemosensitivity to cisplatin. Apoptosis was assessed by DAPI nuclear staining and flow cytometric analysis with an Annexin-V-FITC apoptosis kit. Cytosolic cytochrome c, cleaved caspase-3 and cleaved caspase-9 were detected by western blot. RESULTS The expression of RBM5 mRNA and protein was significantly reduced in the A549/DDP cells compared with the A549 cells. Exogenous expression of RBM5 by the pcDNA3-RBM5 resensitized the response of A549/DDP to cisplatin, resulting in a significant increase in tumor-suppressing activity induced by cisplatin. In contrast, downregulation of RBM5 with siRNA in the A549 cells inhibited cisplatin-induced apoptosis. We also found that the RBM5-enhanced chemosensitivity was associated with the release of cytochrome c into the cytosol, activation of caspase-9 and the downstream marker caspase-3. CONCLUSION Our results demonstrate that RBM5 may serve as a biomarker with the ability to predict a response to cisplatin. It may also act as a prognostic indicator in lung cancer patients. Our findings suggest that there may be clinical utility for ectopic RBM5 such as enhancing and resensitizing nonresponders to cisplatin.
Collapse
Affiliation(s)
- Ping Li
- Department of Respiratory Medicine, The Second Affiliated Hospital of Jilin University, Changchun, China.
| | | | | | | | | | | | | |
Collapse
|
44
|
Liang H, Zhang J, Shao C, Zhao L, Xu W, Sutherland LC, Wang K. Differential expression of RBM5, EGFR and KRAS mRNA and protein in non-small cell lung cancer tissues. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:36. [PMID: 22537942 PMCID: PMC3403968 DOI: 10.1186/1756-9966-31-36] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 04/26/2012] [Indexed: 01/22/2023]
Abstract
Background RNA binding motif 5 (RBM5) is a tumor suppressor gene that modulates apoptosis through the regulation of alternative splicing of apoptosis-related genes. This study aimed to detect RBM5 expression in non-small cell lung cancer (NSCLC) and to associate RBM5 expression with clinicopathological data from NSCLC patients and EGFR and KRAS expression to better understand the potential role of RBM5 in NSCLC. Method Semi-quantitative reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting were performed to detect expression of mRNA and protein, respectively, of RBM5, EGFR and KRAS in 120 paired non-tumor and tumor samples of NSCLC. Results The data showed that expression of RBM5 mRNA and protein was significantly reduced in NSCLC compared to normal tissues, whereas expression of both EGFR and KRAS genes was increased in NSCLC compared to normal tissues. Furthermore, the reduced RBM5 protein expression correlated with smoking status, tumor stage and lymph node metastasis of NSCLC, while overexpression of EGFR and KRAS proteins correlated with tumor stage and lymph node metastasis of NSCLC. Overexpression of KRAS protein was more frequent in smokers with NSCLC. In addition, expression of RBM5 mRNA and protein was negatively correlated with expression of EGFR and KRAS mRNA and protein in NSCLC tissues. Conclusion This study suggests further evaluation of RBM5 expression is warranted for use of RBM5 as a biomarker for NSCLC patients.
Collapse
Affiliation(s)
- Hong Liang
- Department of Respiratory Medicine, Second Affiliated Hospital of Jilin University, Changchun, Jilin 130041, China
| | | | | | | | | | | | | |
Collapse
|
45
|
Niu Z, Jin W, Zhang L, Li X. Tumor suppressor RBM5 directly interacts with the DExD/H-box protein DHX15 and stimulates its helicase activity. FEBS Lett 2012; 586:977-83. [DOI: 10.1016/j.febslet.2012.02.052] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 02/11/2012] [Accepted: 02/21/2012] [Indexed: 02/04/2023]
|
46
|
Celis-Aguilar E, Lassaletta L, Torres-Martín M, Rodrigues FY, Nistal M, Castresana JS, Gavilan J, Rey JA. The molecular biology of vestibular schwannomas and its association with hearing loss: a review. GENETICS RESEARCH INTERNATIONAL 2012; 2012:856157. [PMID: 22567403 PMCID: PMC3335540 DOI: 10.1155/2012/856157] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 10/08/2011] [Accepted: 11/03/2011] [Indexed: 01/16/2023]
Abstract
Hearing loss is the most common symptom in patients with vestibular schwannoma (VS). In the past, compressive mechanisms caused by the tumoral mass and its growth have been regarded as the most likely causes of the hearing loss associated with VS. Interestingly, new evidence proposes molecular mechanisms as an explanation for such hearing loss. Among the molecular mechanisms proposed are methylation of TP73, negative expression of cyclin D1, expression of B7-H1, increased expression of the platelet-derived growth factor A, underexpression of PEX5L, RAD54B, and PSMAL, and overexpression of CEA. Many molecular mechanisms are involved in vestibular schwannoma development; we review some of these mechanisms with special emphasis on hearing loss associated with vestibular schwannoma.
Collapse
Affiliation(s)
- Erika Celis-Aguilar
- Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, 14269 Ciudad de México, DF, Mexico
| | - Luis Lassaletta
- Department of Otolaryngology, “La Paz” University Hospital, 28046 Madrid, Spain
| | - Miguel Torres-Martín
- Unidad de Investigación, Laboratorio Oncogenetica Molecular, “La Paz” University Hospital, 28046 Madrid, Spain
| | - F. Yuri Rodrigues
- Department of Pathology, “La Paz” University Hospital, 28046 Madrid, Spain
| | - Manuel Nistal
- Department of Pathology, “La Paz” University Hospital, 28046 Madrid, Spain
| | - Javier S. Castresana
- Brain Tumor Biology Unit, CIFA, University of Navarra School of Sciences, 31009 Pamplona, Spain
| | - Javier Gavilan
- Department of Otolaryngology, “La Paz” University Hospital, 28046 Madrid, Spain
| | - Juan A. Rey
- Department of Pathology, “La Paz” University Hospital, 28046 Madrid, Spain
| |
Collapse
|
47
|
Lee JD, Kwon TJ, Kim UK, Lee WS. Genetic and epigenetic alterations of the NF2 gene in sporadic vestibular schwannomas. PLoS One 2012; 7:e30418. [PMID: 22295085 PMCID: PMC3266248 DOI: 10.1371/journal.pone.0030418] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Accepted: 12/15/2011] [Indexed: 12/11/2022] Open
Abstract
Background Mutations in the neurofibromatosis type 2 (NF2) tumor-suppressor gene have been identified in not only NF2-related tumors but also sporadic vestibular schwannomas (VS). This study investigated the genetic and epigenetic alterations in tumors and blood from 30 Korean patients with sporadic VS and correlated these alterations with tumor behavior. Methodology/Principal Findings NF2 gene mutations were detected using PCR and direct DNA sequencing and three highly polymorphic microsatellite DNA markers were used to assess the loss of heterozygosity (LOH) from chromosome 22. Aberrant hypermethylation of the CpG island of the NF2 gene was also analyzed. The tumor size, the clinical growth index, and the proliferative activity assessed using the Ki-67 labeling index were evaluated. We found 18 mutations in 16 cases of 30 schwannomas (53%). The mutations included eight frameshift mutations, seven nonsense mutations, one in-frame deletion, one splicing donor site, and one missense mutation. Nine patients (30%) showed allelic loss. No patient had aberrant hypermethylation of the NF2 gene and correlation between NF2 genetic alterations and tumor behavior was not observed in this study. Conclusions/Significance The molecular genetic changes in sporadic VS identified here included mutations and allelic loss, but no aberrant hypermethylation of the NF2 gene was detected. In addition, no clear genotype/phenotype correlation was identified. Therefore, it is likely that other factors contribute to tumor formation and growth.
Collapse
Affiliation(s)
- Jong Dae Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Tae Jun Kwon
- Department of Biology, College of Natural Sciences, Kyungpook National University, Daegu, Korea
| | - Un-Kyung Kim
- Department of Biology, College of Natural Sciences, Kyungpook National University, Daegu, Korea
- * E-mail:
| | - Won-Sang Lee
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
48
|
Singh A, Mishra AK, Ylaya K, Hewitt SM, Sharma KC, Saxena S. Wilms tumor-1, claudin-1 and ezrin are useful immunohistochemical markers that help to distinguish schwannoma from fibroblastic meningioma. Pathol Oncol Res 2011; 18:383-9. [PMID: 21909685 DOI: 10.1007/s12253-011-9456-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 08/31/2011] [Indexed: 10/17/2022]
Abstract
The aim of this study is to identify immunohistochemical (IHC) markers that can reliably separate schwannoma (SCHW) and fibroblastic meningioma (FM). We selected 106 cases of intracranial SCHW (n = 56) and FM (n = 50) and constructed a tissue microarray (TMA) of core diameter of 1.0 mm from archival formalin-fixed paraffin-embedded tissue. A TMA-IHC was performed using 14 antibodies. After IHC staining, 98 cores were found suitable for evaluation. The IHC staining was scored as 0-2+ (0, negative; 1+, weak and/or focal 2+ strong and/or diffuse positive). A discriminant analysis (DA) (Wilks'Lambda test) was performed to assess the relative importance of these biomarkers in classifying the two groups FM and SCHW. It showed that WT-1 (Wilks'λ 0.085, p < 0.001), EMA (Wilks'λ 0.253, p < 0.001), S100 (Wilks'λ 0.487, p < 0.001), Claudin-1 (Wilks' λ 0.57, p < 0.001) and Ezrin (Wilks'λ 0.656, p < 0.001), SPARC (Wilks'λ 0.751, p < 0.01), NP-Y (Wilks'λ, 0.819, p < 0.001) and EGFR (Wilks'λ 0.845, p = 0.026) were some of the statistically significant markers that discriminated SCHW and FM. For sensitivity and specificity for SCHW the significant markers [Area under the curve (95% CI), p-value] by ROC analysis were WT-1 [0.990(0.000, 1.000), <0.001], S100 [0.880(0.808, 0.951), <0.001] while for diagnosing FM the most sensitive and specific markers were EMA [0.957(0.914, 1.000), <. 001], Claudin-1 [0.857(0.782, 0.932), <0.001] and ezrin [0.792(0.700,0.884),<0.001]. WT-1, Claudin-1 and Ezrin may be potentially useful immunohistochemical adjuncts to EMA and S100 that differentiate SCHW from FM.
Collapse
Affiliation(s)
- Avninder Singh
- Safdarjung Hospital Campus, National Institute of Pathology (ICMR), Room 602, 6th floor, New Delhi, 110029, India.
| | | | | | | | | | | |
Collapse
|
49
|
Aarhus M, Bruland O, Sætran HA, Mork SJ, Lund-Johansen M, Knappskog PM. Global gene expression profiling and tissue microarray reveal novel candidate genes and down-regulation of the tumor suppressor gene CAV1 in sporadic vestibular schwannomas. Neurosurgery 2011; 67:998-1019; discussion 1019. [PMID: 20881564 DOI: 10.1227/neu.0b013e3181ec7b71] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The vestibular nerve is the predilection site for schwannomas. Few transcriptomic studies have been performed on solely sporadic vestibular schwannomas (VSs). OBJECTIVE To detect genes with altered expression levels in sporadic VSs. METHODS We studied 25 VSs and 3 tibial nerves (controls) with the ABI 1700 microarray platform. Significance analysis of microarrays was performed to explore differential gene expression. Selected genes were validated with quantitative reverse transcriptase polymerase chain reaction. A tissue microarray was constructed for immunohistochemistry. Neurofibromatosis type II cDNA was sequenced for mutations. RESULTS The VSs formed 2 clusters based on the total expression of 23,055 genes. Tumor size, previous Gamma Knife surgery, neurofibromatosis type II mutations, and cystic tumors were distributed equally in both. Significance analysis of microarrays detected 1650 differentially expressed genes. On the top 500 list, several cancer-related genes with an unrecognized role in VSs were down-regulated: CAV1, TGFB3, VCAM1, GLI1, GLI2, PRKAR2B, EPHA4, and FZD1. Immunohistochemistry showed no CAV1 expression in the VSs. The ERK pathway was the central core in the network linking the differentially expressed genes. The previously reported VS candidate genes SPARC, PLAT, and FGF1 were up-regulated. Nineteen of 25 VSs had NF2 mutations. CONCLUSION Using microarray technology, we identified novel genes and pathways with a putative role in VSs, confirmed previous candidate genes, and found cancer-related genes with no reported role in VSs. Among these, down-regulation of CAV1 at both the mRNA and protein levels is of particular interest because this tumor suppressor normally is expressed in Schwann cells.
Collapse
Affiliation(s)
- Mads Aarhus
- Centre for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway.
| | | | | | | | | | | |
Collapse
|
50
|
Kobayashi T, Ishida J, Musashi M, Ota S, Yoshida T, Shimizu Y, Chuma M, Kawakami H, Asaka M, Tanaka J, Imamura M, Kobayashi M, Itoh H, Edamatsu H, Sutherland LC, Brachmann RK. p53 transactivation is involved in the antiproliferative activity of the putative tumor suppressor RBM5. Int J Cancer 2010; 128:304-18. [PMID: 20309933 DOI: 10.1002/ijc.25345] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Accepted: 03/09/2010] [Indexed: 12/12/2022]
Abstract
RBM5 (RNA-binding motif protein 5) is a nuclear RNA binding protein containing 2 RNA recognition motifs. The RBM5 gene is located at the tumor suppressor locus 3p21.3. Deletion of this locus is the most frequent genetic alteration in lung cancer, but is also found in other human cancers. RBM5 is known to induce apoptosis and cell cycle arrest but the molecular mechanisms of RBM5 function are poorly understood. Here, we show that RBM5 is important for the activity of the tumor suppressor protein p53. Overexpression of RBM5 enhanced p53-mediated inhibition of cell growth and colony formation. Expression of RBM5 augmented p53 transcriptional activity in reporter gene assays and resulted in increased mRNA and protein levels for endogenous p53 target genes. In contrast, shRNA-mediated knockdown of endogenous RBM5 led to decreased p53 transcriptional activity and reduced levels of mRNA and protein for endogenous p53 target genes. RBM5 affected protein, but not mRNA, levels of endogenous p53 after DNA damage suggest that RBM5 contributes to p53 activity through post-transcriptional mechanisms. Our results show that RBM5 contributes to p53 transcriptional activity after DNA damage and that growth suppression and apoptosis mediated by RBM5 are linked to activity of the tumor suppressor protein p53.
Collapse
|