1
|
Xu D, Yin S, Shu Y. NF2: An underestimated player in cancer metabolic reprogramming and tumor immunity. NPJ Precis Oncol 2024; 8:133. [PMID: 38879686 PMCID: PMC11180135 DOI: 10.1038/s41698-024-00627-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/02/2024] [Indexed: 06/19/2024] Open
Abstract
Neurofibromatosis type 2 (NF2) is a tumor suppressor gene implicated in various tumors, including mesothelioma, schwannomas, and meningioma. As a member of the ezrin, radixin, and moesin (ERM) family of proteins, merlin, which is encoded by NF2, regulates diverse cellular events and signalling pathways, such as the Hippo, mTOR, RAS, and cGAS-STING pathways. However, the biological role of NF2 in tumorigenesis has not been fully elucidated. Furthermore, cross-cancer mutations may exert distinct biological effects on tumorigenesis and treatment response. In addition to the functional inactivation of NF2, the codeficiency of other genes, such as cyclin-dependent kinase inhibitor 2A/B (CDKN2A/B), BRCA1-associated protein-1 (BAP1), and large tumor suppressor 2 (LATS2), results in unique tumor characteristics that should be considered in clinical treatment decisions. Notably, several recent studies have explored the metabolic and immunological features associated with NF2, offering potential insights into tumor biology and the development of innovative therapeutic strategies. In this review, we consolidate the current knowledge on NF2 and examine the potential connection between cancer metabolism and tumor immunity in merlin-deficient malignancies. This review may provide a deeper understanding of the biological roles of NF2 and guide possible therapeutic avenues.
Collapse
Affiliation(s)
- Duo Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shiyuan Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
2
|
Diaz LR, Gil-Ranedo J, Jaworek KJ, Nsek N, Marques JP, Costa E, Hilton DA, Bieluczyk H, Warrington O, Hanemann CO, Futschik ME, Bossing T, Barros CS. Ribogenesis boosts controlled by HEATR1-MYC interplay promote transition into brain tumour growth. EMBO Rep 2024; 25:168-197. [PMID: 38225354 PMCID: PMC10897169 DOI: 10.1038/s44319-023-00017-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 01/17/2024] Open
Abstract
Cell commitment to tumourigenesis and the onset of uncontrolled growth are critical determinants in cancer development but the early events directing tumour initiating cell (TIC) fate remain unclear. We reveal a single-cell transcriptome profile of brain TICs transitioning into tumour growth using the brain tumour (brat) neural stem cell-based Drosophila model. Prominent changes in metabolic and proteostasis-associated processes including ribogenesis are identified. Increased ribogenesis is a known cell adaptation in established tumours. Here we propose that brain TICs boost ribogenesis prior to tumour growth. In brat-deficient TICs, we show that this dramatic change is mediated by upregulated HEAT-Repeat Containing 1 (HEATR1) to promote ribosomal RNA generation, TIC enlargement and onset of overgrowth. High HEATR1 expression correlates with poor glioma patient survival and patient-derived glioblastoma stem cells rely on HEATR1 for enhanced ribogenesis and tumourigenic potential. Finally, we show that HEATR1 binds the master growth regulator MYC, promotes its nucleolar localisation and appears required for MYC-driven ribogenesis, suggesting a mechanism co-opted in ribogenesis reprogramming during early brain TIC development.
Collapse
Affiliation(s)
- Laura R Diaz
- Peninsula Medical School, Faculty of Health, John Bull Building, University of Plymouth, PL6 8BU, Plymouth, UK
| | - Jon Gil-Ranedo
- Peninsula Medical School, Faculty of Health, John Bull Building, University of Plymouth, PL6 8BU, Plymouth, UK
| | - Karolina J Jaworek
- Peninsula Medical School, Faculty of Health, John Bull Building, University of Plymouth, PL6 8BU, Plymouth, UK
- School of Biological Sciences, Bangor University, LL57 2UW, Bangor, UK
| | - Nsikan Nsek
- Peninsula Medical School, Faculty of Health, John Bull Building, University of Plymouth, PL6 8BU, Plymouth, UK
| | - Joao Pinheiro Marques
- Peninsula Medical School, Faculty of Health, John Bull Building, University of Plymouth, PL6 8BU, Plymouth, UK
| | - Eleni Costa
- Peninsula Medical School, Faculty of Health, John Bull Building, University of Plymouth, PL6 8BU, Plymouth, UK
| | - David A Hilton
- Department of Cellular and Anatomical Pathology, University Hospitals Plymouth, PL6 8DH, Plymouth, UK
| | - Hubert Bieluczyk
- Peninsula Medical School, Faculty of Health, John Bull Building, University of Plymouth, PL6 8BU, Plymouth, UK
| | - Oliver Warrington
- Peninsula Medical School, Faculty of Health, John Bull Building, University of Plymouth, PL6 8BU, Plymouth, UK
- Wellcome Centre for Human Neuroimaging, UCL Queen Square Institute of Neurology, University College London, WC1N 3AR, London, UK
| | - C Oliver Hanemann
- Peninsula Medical School, Faculty of Health, John Bull Building, University of Plymouth, PL6 8BU, Plymouth, UK
| | - Matthias E Futschik
- School of Biomedical Sciences, Faculty of Health, Derriford Research Facility, University of Plymouth, PL6 8BU, Plymouth, UK
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-504, Coimbra, Portugal
| | - Torsten Bossing
- Peninsula Medical School, Faculty of Health, John Bull Building, University of Plymouth, PL6 8BU, Plymouth, UK
| | - Claudia S Barros
- Peninsula Medical School, Faculty of Health, John Bull Building, University of Plymouth, PL6 8BU, Plymouth, UK.
| |
Collapse
|
3
|
Dougherty MC, Shibata SB, Clark JJ, Canady FJ, Yates CW, Hansen MR. Reduction of sporadic and neurofibromatosis type 2-associated vestibular schwannoma growth in vitro and in vivo after treatment with the c-Jun N-terminal kinase inhibitor AS602801. J Neurosurg 2022; 138:962-971. [PMID: 36087315 DOI: 10.3171/2022.7.jns22934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 07/12/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Vestibular schwannomas (VSs) are benign nerve sheath tumors that result from mutation in the tumor suppressor gene NF2, with functional loss of the protein merlin. The authors have previously shown that c-Jun N-terminal kinase (JNK) is constitutively active in human VS cells and plays a central role in their survival by suppressing accumulation of mitochondrial superoxides, implicating JNK inhibitors as a potential systemic treatment for VS. Thus, the authors hypothesized that the adenosine 5'-triphosphate-competitive JNK inhibitor AS602801 would demonstrate antitumor activity in multiple VS models. METHODS Treatment with AS602801 was tested in primary human VS cultures, human VS xenografts, and a genetic mouse model of schwannoma (Postn-Cre;Nf2flox/flox). Primary human VS cell cultures were established from freshly obtained surgical tumor specimens; treatment group media was enriched with AS602801. VS xenograft tumors were established in male athymic nude mice from freshly collected human tumor. Four weeks postimplantation, a pretreatment MRI scan was obtained, followed by 65 days of AS602801 (n = 18) or vehicle control (n = 19) treatment. Posttreatment MRI scans were used to measure final tumor volume. Tumors were then harvested. Finally, Postn-Cre;Nf2flox/flox mice were treated with AS602801 (n = 10) or a vehicle (n = 13) for 65 days. Posttreatment auditory brainstem responses were obtained. Dorsal root ganglia from Postn-Cre;Nf2flox/flox mice were then harvested. In all models, schwannoma identity was confirmed with anti-S100 staining, cell proliferation was measured with the EdU assay, and cell death was measured with terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining. All protocols were approved by the local institutional review board and Institutional Animal Care and Use Committees. RESULTS Treatment with AS602801 decreased cell proliferation and increased apoptosis in primary human VS cultures. The systemic administration of AS602801 in mice with human VS xenografts reduced tumor volume and cell proliferation. Last, the AS602801-treated Postn-Cre;Nf2flox/flox mice demonstrated decreased cell proliferation in glial cells in the dorsal root ganglia. However, AS602801 did not significantly delay hearing loss in Postn-Cre;Nf2flox/flox mice up to 3 months posttreatment. CONCLUSIONS The data suggest that JNK inhibition with AS602801 suppresses growth of sporadic and neurofibromatosis type 2-associated VSs. As such, AS602801 is a potential systemic therapy for VS and warrants further investigation.
Collapse
Affiliation(s)
| | - Seiji B Shibata
- 2Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa; and
| | - J Jason Clark
- 2Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa; and
| | - Franklin J Canady
- 2Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa; and
| | - Charles W Yates
- 3Department of Otolaryngology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Marlan R Hansen
- 2Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa; and
| |
Collapse
|
4
|
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
|
5
|
Catasús N, Garcia B, Galván-Femenía I, Plana A, Negro A, Rosas I, Ros A, Amilibia E, Becerra JL, Hostalot C, Rocaribas F, Bielsa I, Lazaro Garcia C, de Cid R, Serra E, Blanco I, Castellanos E. Revisiting the UK Genetic Severity Score for NF2: a proposal for the addition of a functional genetic component. J Med Genet 2021; 59:678-686. [PMID: 34348961 DOI: 10.1136/jmedgenet-2020-107548] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 06/10/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND Neurofibromatosis type 2 (NF2) is an autosomal dominant disorder characterised by the development of multiple schwannomas, especially on vestibular nerves, and meningiomas. The UK NF2 Genetic Severity Score (GSS) is useful to predict the progression of the disease from germline NF2 pathogenic variants, which allows the clinical follow-up and the genetic counselling offered to affected families to be optimised. METHODS 52 Spanish patients were classified using the GSS, and patients' clinical severity was measured and compared between GSS groups. The GSS was reviewed with the addition of phenotype quantification, genetic variant classification and functional assays of Merlin and its downstream pathways. Principal component analysis and regression models were used to evaluate the differences between severity and the effect of NF2 germline variants. RESULTS The GSS was validated in the Spanish NF2 cohort. However, for 25% of mosaic patients and patients harbouring variants associated with mild and moderate phenotypes, it did not perform as well for predicting clinical outcomes as it did for pathogenic variants associated with severe phenotypes. We studied the possibility of modifying the mutation classification in the GSS by adding the impact of pathogenic variants on the function of Merlin in 27 cases. This revision helped to reduce variability within NF2 mutation classes and moderately enhanced the correlation between patient phenotype and the different prognosis parameters analysed (R2=0.38 vs R2=0.32, p>0001). CONCLUSIONS We validated the UK NF2 GSS in a Spanish NF2 cohort, despite the significant phenotypic variability identified within it. The revision of the GSS, named Functional Genetic Severity Score, could add value for the classification of mosaic patients and patients showing mild and moderate phenotypes once it has been validated in other cohorts.
Collapse
Affiliation(s)
- Núria Catasús
- Clinical Genomics Research Unit, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP-PMPPC), Badalona, Spain
| | - Belen Garcia
- Clinical Genomics Research Unit, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP-PMPPC), Badalona, Spain.,Genetic Counseling Unit, Clinical Genetics Service, Northern Metropolitan Clinical Laboratory, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Iván Galván-Femenía
- Genomes for Life-GCAT lab Group, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Adrià Plana
- Dermatology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Alejandro Negro
- Clinical Genomics Research Unit, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP-PMPPC), Badalona, Spain.,Genetic Counseling Unit, Clinical Genetics Service, Northern Metropolitan Clinical Laboratory, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Inma Rosas
- Clinical Genomics Research Unit, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP-PMPPC), Badalona, Spain.,Clinical Genomics Unit, Clinical Genetics Service, Northern Metropolitan Clinical Laboratory, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Andrea Ros
- Clinical Genomics Research Unit, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP-PMPPC), Badalona, Spain.,Genetic Counseling Unit, Clinical Genetics Service, Northern Metropolitan Clinical Laboratory, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Emilio Amilibia
- Otorhinolaryngology, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Juan Luis Becerra
- Neurology, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Cristina Hostalot
- Neurosurgery, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Francesc Rocaribas
- Otorhinolaryngology, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Isabel Bielsa
- Dermatology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Conxi Lazaro Garcia
- Hereditary Cancer Program, ICO-IDIBELL-CIBERONC, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Rafael de Cid
- Genomes for Life-GCAT lab Group, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Eduard Serra
- Hereditary Cancer Group, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP-PMPPC), Badalona, Spain
| | - Ignacio Blanco
- Clinical Genomics Research Unit, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP-PMPPC), Badalona, Spain.,Genetic Counseling Unit, Clinical Genetics Service, Northern Metropolitan Clinical Laboratory, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Elisabeth Castellanos
- Clinical Genomics Research Unit, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP-PMPPC), Badalona, Spain .,Clinical Genomics Unit, Clinical Genetics Service, Northern Metropolitan Clinical Laboratory, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | |
Collapse
|
6
|
Fibulin-2: A Novel Biomarker for Differentiating Grade II from Grade I Meningiomas. Int J Mol Sci 2021; 22:ijms22020560. [PMID: 33429944 PMCID: PMC7827565 DOI: 10.3390/ijms22020560] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 12/21/2022] Open
Abstract
There is an unmet need for the identification of biomarkers to aid in the diagnosis, clinical management, prognosis and follow-up of meningiomas. There is currently no consensus on the optimum management of WHO grade II meningiomas. In this study, we identified the calcium binding extracellular matrix glycoprotein, Fibulin-2, via mass-spectrometry-based proteomics, assessed its expression in grade I and II meningiomas and explored its potential as a grade II biomarker. A total of 87 grade I and 91 grade II different meningioma cells, tissue and plasma samples were used for the various experimental techniques employed to assess Fibulin-2 expression. The tumours were reviewed and classified according to the 2016 edition of the Classification of the Tumours of the central nervous system (CNS). Mass spectrometry proteomic analysis identified Fibulin-2 as a differentially expressed protein between grade I and II meningioma cell cultures. Fibulin-2 levels were further evaluated in meningioma cells using Western blotting and Real-time Quantitative Polymerase Chain Reaction (RT-qPCR); in meningioma tissues via immunohistochemistry and RT-qPCR; and in plasma via Enzyme-Linked Immunosorbent Assay (ELISA). Proteomic analyses (p < 0.05), Western blotting (p < 0.05) and RT-qPCR (p < 0.01) confirmed significantly higher Fibulin-2 (FBLN2) expression levels in grade II meningiomas compared to grade I. Fibulin-2 blood plasma levels were also significantly higher in grade II meningioma patients compared to grade I patients. This study suggests that elevated Fibulin-2 might be a novel grade II meningioma biomarker, when differentiating them from the grade I tumours. The trend of Fibulin-2 expression observed in plasma may serve as a useful non-invasive biomarker.
Collapse
|
7
|
Coy S, Rashid R, Stemmer-Rachamimov A, Santagata S. An update on the CNS manifestations of neurofibromatosis type 2. Acta Neuropathol 2020; 139:643-665. [PMID: 31161239 PMCID: PMC7038792 DOI: 10.1007/s00401-019-02029-5] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 05/23/2019] [Accepted: 05/25/2019] [Indexed: 12/21/2022]
Abstract
Neurofibromatosis type II (NF2) is a tumor predisposition syndrome characterized by the development of distinctive nervous system lesions. NF2 results from loss-of-function alterations in the NF2 gene on chromosome 22, with resultant dysfunction of its protein product merlin. NF2 is most commonly associated with the development of bilateral vestibular schwannomas; however, patients also have a predisposition to development of other tumors including meningiomas, ependymomas, and peripheral, spinal, and cranial nerve schwannomas. Patients may also develop other characteristic manifestations such as ocular lesions, neuropathies, meningioangiomatosis, and glial hamartia. NF2 has a highly variable clinical course, with some patients exhibiting a severe phenotype and development of multiple tumors at an early age, while others may be nearly asymptomatic throughout their lifetime. Despite the high morbidity associated with NF2 in severe cases, management of NF2-associated lesions primarily consists of surgical resection and treatment of symptoms, and there are currently no FDA-approved systemic therapies that address the underlying biology of the syndrome. Refinements to the diagnostic criteria of NF2 have been proposed over time due to increasing understanding of clinical and molecular data. Large-population studies have demonstrated that some features such as the development of gliomas and neurofibromas, currently included as diagnostic criteria, may require further clarification and modification. Meanwhile, burgeoning insights into the molecular biology of NF2 have shed light on the etiology and highly variable severity of the disease and suggested numerous putative molecular targets for therapeutic intervention. Here, we review the clinicopathologic features of NF2, current understanding of the molecular biology of NF2, particularly with regard to central nervous system lesions, ongoing therapeutic studies, and avenues for further research.
Collapse
Affiliation(s)
- Shannon Coy
- Division of Neuropathology, Department of Pathology, Brigham and Women's Hospital, Hale Building for Transformative Medicine, BTM8002P, 60 Fenwood Road, Boston, MA, 02115, USA
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Rumana Rashid
- Division of Neuropathology, Department of Pathology, Brigham and Women's Hospital, Hale Building for Transformative Medicine, BTM8002P, 60 Fenwood Road, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, USA
- Laboratory for Systems Pharmacology, Harvard Program in Therapeutic Science, Boston, MA, USA
| | - Anat Stemmer-Rachamimov
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Sandro Santagata
- Division of Neuropathology, Department of Pathology, Brigham and Women's Hospital, Hale Building for Transformative Medicine, BTM8002P, 60 Fenwood Road, Boston, MA, 02115, USA.
- Department of Pathology, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Laboratory for Systems Pharmacology, Harvard Program in Therapeutic Science, Boston, MA, USA.
- Ludwig Center at Harvard, Boston, MA, USA.
| |
Collapse
|
8
|
Ferluga S, Baiz D, Hilton DA, Adams CL, Ercolano E, Dunn J, Bassiri K, Kurian KM, Hanemann CO. Constitutive activation of the EGFR-STAT1 axis increases proliferation of meningioma tumor cells. Neurooncol Adv 2020; 2:vdaa008. [PMID: 32642677 PMCID: PMC7212880 DOI: 10.1093/noajnl/vdaa008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Background Meningiomas are the most frequent primary brain tumors of the central nervous system. The standard of treatment is surgery and radiotherapy, but effective pharmacological options are not available yet. The well-characterized genetic background stratifies these tumors in several subgroups, thus increasing diversification. We identified epidermal growth factor receptor–signal transducer and activator of transcription 1 (EGFR–STAT1) overexpression and activation as a common identifier of these tumors. Methods We analyzed STAT1 overexpression and phosphorylation in 131 meningiomas of different grades and locations by utilizing several techniques, including Western blots, qPCR, and immunocytochemistry. We also silenced and overexpressed wild-type and mutant forms of the gene to assess its biological function and its network. Results were further validated by drug testing. Results STAT1 was found widely overexpressed in meningioma but not in the corresponding healthy controls. The protein showed constitutive phosphorylation not dependent on the JAK–STAT pathway. STAT1 knockdown resulted in a significant reduction of cellular proliferation and deactivation of AKT and ERK1/2. STAT1 is known to be activated by EGFR, so we investigated the tyrosine kinase and found that EGFR was also constitutively phosphorylated in meningioma and was responsible for the aberrant phosphorylation of STAT1. The pharmaceutical inhibition of EGFR caused a significant reduction in cellular proliferation and of overall levels of cyclin D1, pAKT, and pERK1/2. Conclusions STAT1–EGFR-dependent constitutive phosphorylation is responsible for a positive feedback loop that causes its own overexpression and consequently an increased proliferation of the tumor cells. These findings provide the rationale for further studies aiming to identify effective therapeutic options in meningioma.
Collapse
Affiliation(s)
- Sara Ferluga
- Faculty of Health: Medicine, Dentistry and Human Sciences, Institute of Translational and Stratified Medicine, University of Plymouth, Plymouth, UK
| | - Daniele Baiz
- Faculty of Health: Medicine, Dentistry and Human Sciences, Institute of Translational and Stratified Medicine, University of Plymouth, Plymouth, UK
| | - David A Hilton
- Cellular and Anatomical Pathology, Plymouth Hospitals NHS Trust, Plymouth, UK
| | - Claire L Adams
- Faculty of Health: Medicine, Dentistry and Human Sciences, Institute of Translational and Stratified Medicine, University of Plymouth, Plymouth, UK
| | - Emanuela Ercolano
- Faculty of Health: Medicine, Dentistry and Human Sciences, Institute of Translational and Stratified Medicine, University of Plymouth, Plymouth, UK
| | - Jemma Dunn
- Faculty of Health: Medicine, Dentistry and Human Sciences, Institute of Translational and Stratified Medicine, University of Plymouth, Plymouth, UK
| | - Kayleigh Bassiri
- Faculty of Health: Medicine, Dentistry and Human Sciences, Institute of Translational and Stratified Medicine, University of Plymouth, Plymouth, UK
| | - Kathreena M Kurian
- Department of Neuropathology, Pathology Sciences, Southmead Hospital, Bristol, UK
| | - Clemens O Hanemann
- Faculty of Health: Medicine, Dentistry and Human Sciences, Institute of Translational and Stratified Medicine, University of Plymouth, Plymouth, UK
| |
Collapse
|
9
|
Fuse MA, Dinh CT, Vitte J, Kirkpatrick J, Mindos T, Plati SK, Young JI, Huang J, Carlstedt A, Franco MC, Brnjos K, Nagamoto J, Petrilli AM, Copik AJ, Soulakova JN, Bracho O, Yan D, Mittal R, Shen R, Telischi FF, Morrison H, Giovannini M, Liu XZ, Chang LS, Fernandez-Valle C. Preclinical assessment of MEK1/2 inhibitors for neurofibromatosis type 2-associated schwannomas reveals differences in efficacy and drug resistance development. Neuro Oncol 2019; 21:486-497. [PMID: 30615146 PMCID: PMC6422635 DOI: 10.1093/neuonc/noz002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Neurofibromatosis type 2 (NF2) is a genetic tumor-predisposition disorder caused by NF2/merlin tumor suppressor gene inactivation. The hallmark of NF2 is formation of bilateral vestibular schwannomas (VS). Because merlin modulates activity of the Ras/Raf/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway, we investigated repurposing drugs targeting MEK1 and/or MEK2 as a treatment for NF2-associated schwannomas. METHODS Mouse and human merlin-deficient Schwann cell lines (MD-MSC/HSC) were screened against 6 MEK1/2 inhibitors. Efficacious drugs were tested in orthotopic allograft and NF2 transgenic mouse models. Pathway and proteome analyses were conducted. Drug efficacy was examined in primary human VS cells with NF2 mutations and correlated with DNA methylation patterns. RESULTS Trametinib, PD0325901, and cobimetinib were most effective in reducing MD-MSC/HSC viability. Each decreased phosphorylated pERK1/2 and cyclin D1, increased p27, and induced caspase-3 cleavage in MD-MSCs. Proteomic analysis confirmed cell cycle arrest and activation of pro-apoptotic pathways in trametinib-treated MD-MSCs. The 3 inhibitors slowed allograft growth; however, decreased pERK1/2, cyclin D1, and Ki-67 levels were observed only in PD0325901 and cobimetinib-treated grafts. Tumor burden and average tumor size were reduced in trametinib-treated NF2 transgenic mice; however, tumors did not exhibit reduced pERK1/2 levels. Trametinib and PD0325901 modestly reduced viability of several primary human VS cell cultures with NF2 mutations. DNA methylation analysis of PD0325901-resistant versus -susceptible VS identified genes that could contribute to drug resistance. CONCLUSION MEK inhibitors exhibited differences in antitumor efficacy resistance in schwannoma models with possible emergence of trametinib resistance. The results support further investigation of MEK inhibitors in combination with other targeted drugs for NF2 schwannomas.
Collapse
Affiliation(s)
- Marisa A Fuse
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida (UCF), Orlando, Florida, USA
| | - Christine T Dinh
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jeremie Vitte
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA and Jonsson Comprehensive Cancer Center, University of California at Los Angeles (UCLA), Los Angeles, California, USA
| | | | - Thomas Mindos
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Stephani Klingeman Plati
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida (UCF), Orlando, Florida, USA
| | - Juan I Young
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jie Huang
- Center for Childhood Cancer and Blood Diseases, Nationwide Children’s Hospital
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | | | - Maria Clara Franco
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida (UCF), Orlando, Florida, USA
| | - Konstantin Brnjos
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida (UCF), Orlando, Florida, USA
| | - Jackson Nagamoto
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida (UCF), Orlando, Florida, USA
| | - Alejandra M Petrilli
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida (UCF), Orlando, Florida, USA
| | - Alicja J Copik
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida (UCF), Orlando, Florida, USA
| | - Julia N Soulakova
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida (UCF), Orlando, Florida, USA
| | - Olena Bracho
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Rulong Shen
- Department of Pathology, The Ohio State University, Columbus, Ohio, USA
| | - Fred F Telischi
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Helen Morrison
- Leibniz Institute on Aging, Fritz Lipmann Institute, Jena, Germany
| | - Marco Giovannini
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA and Jonsson Comprehensive Cancer Center, University of California at Los Angeles (UCLA), Los Angeles, California, USA
| | - Xue-Zhong Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Long-Sheng Chang
- Center for Childhood Cancer and Blood Diseases, Nationwide Children’s Hospital
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Cristina Fernandez-Valle
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida (UCF), Orlando, Florida, USA
| |
Collapse
|
10
|
Oblinger JL, Burns SS, Huang J, Pan L, Ren Y, Shen R, Kinghorn AD, Welling DB, Chang LS. Overexpression of eIF4F components in meningiomas and suppression of meningioma cell growth by inhibiting translation initiation. Exp Neurol 2018; 299:299-307. [PMID: 28610844 PMCID: PMC5723558 DOI: 10.1016/j.expneurol.2017.06.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 06/03/2017] [Accepted: 06/09/2017] [Indexed: 10/19/2022]
Abstract
Meningiomas frequently display activation of the PI3K/AKT/mTOR pathway, leading to elevated levels of phospho-eukaryotic translation initiation factor 4E binding proteins, which enhances protein synthesis; however, it is not known whether inhibition of protein translation is an effective treatment option for meningiomas. We found that human meningiomas expressed high levels of the three components of the eukaryotic initiation factor 4F (eIF4F) translation initiation complex, eIF4A, eIF4E, and eIF4G. The expression of eIF4A and eIF4E was important in sustaining the growth of NF2-deficient benign meningioma Ben-Men-1 cells, as shRNA-mediated knockdown of these proteins strongly reduced cell proliferation. Among a series of 23 natural compounds evaluated, silvestrol, which inhibits eIF4A, was identified as being the most growth inhibitory in both primary meningioma and Ben-Men-1 cells. Silvestrol treatment of meningioma cells prominently induced G2/M arrest. Consistently, silvestrol significantly decreased the amounts of cyclins D1, E1, A, and B, PCNA, and Aurora A. In addition, total and phosphorylated AKT, ERK, and FAK, which have been shown to be important drivers for meningioma cell proliferation, were markedly lower in silvestrol-treated Ben-Men-1 cells. Our findings suggest that inhibiting protein translation could be a potential treatment for meningiomas.
Collapse
Affiliation(s)
- Janet L Oblinger
- Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Otolaryngology-Head and Neck Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Sarah S Burns
- Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Otolaryngology-Head and Neck Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Jie Huang
- Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Otolaryngology-Head and Neck Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Li Pan
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University College of Pharmacy, Columbus, OH, USA
| | - Yulin Ren
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University College of Pharmacy, Columbus, OH, USA
| | - Rulong Shen
- Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, The Ohio State University College of Pharmacy, Columbus, OH, USA
| | - D Bradley Welling
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Long-Sheng Chang
- Center for Childhood Cancer and Blood Diseases, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Otolaryngology-Head and Neck Surgery, The Ohio State University College of Medicine, Columbus, OH, USA; Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
11
|
Fan L, Hong J, Huang H, Fu D, Wu S, Wang Q, Ye Y, Liu Y. High Expression of Phosphorylated Extracellular Signal-Regulated Kinase (ERK1/2) is Associated with Poor Prognosis in Newly Diagnosed Patients with Multiple Myeloma. Med Sci Monit 2017; 23:2636-2643. [PMID: 28557972 PMCID: PMC5461889 DOI: 10.12659/msm.901850] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background Previous research has demonstrated that the extracellular signal-regulated kinase (MEK)/extracellular signal-regulated kinase (ERK) signaling pathway is commonly activated in multiple myeloma (MM) patients. However, the prognostic value of activation of the MEK/ERK signaling pathway in newly diagnosed patients with MM has not been reported. Material/Methods Expression levels of p-ERK1/2 protein in bone marrow biopsy specimens obtained from 60 newly diagnosed patients with MM were analyzed using immunohistochemistry, and classified into 3 groups: high p-ERK1/2 expression, low p-ERK1/2 expression, and negative group. Correlations between clinicopathological characteristics, including expression levels of p-ERK1/2 protein, progression-free survival (PFS), and overall survival (OS), were analyzed using univariate and multivariate analysis. Results Phosphorylated-ERK1/2 protein was positive in 47 bone marrow specimens, including 19 specimens with high p-ERK1/2 expression and 28 specimens with low p-ERK1/2 expression. Univariate Kaplan-Meier analysis showed that in newly diagnosed patients with MM, high p-ERK1/2 expression, high ISS staging, serum creatinine (Scr) ≥177 μmol/l, serum β2-microglobulin (β2-MG) ≥5.5 μmol/l, and serum calcium (Ca) ≥2.75 mmol/l were significantly associated with shorter OS and PFS. Additionally, high ECOG scores (score 2–4) were associated with shorter PFS in newly diagnosed patients with MM. Multivariate Cox regression analysis showed that in newly diagnosed patients with MM, high p-ERK1/2 expression was significantly associated with shorter OS and PFS. Additionally, in newly diagnosed patients with MM, serum Ca ≥2.75 mmol/l was significantly associated with shorter PFS, and serum β2-MG ≥5.5 μmol/l was significantly associated with shorter OS. Conclusions High p-ERK1/2 expression is an independent factor for poor prognosis in newly diagnosed patients with MM.
Collapse
Affiliation(s)
- Liping Fan
- Department of Blood Transfusion, Fujian Medical University Union Hospital, Fuzhou, Fujian, China (mainland)
| | - Jinquan Hong
- Department of Anesthesia, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China (mainland)
| | - Haobo Huang
- Department of Blood Transfusion, Fujian Medical University Union Hospital, Fuzhou, Fujian, China (mainland)
| | - Danhui Fu
- Department of Blood Transfusion, Fujian Medical University Union Hospital, Fuzhou, Fujian, China (mainland).,Department of Hematology, Fujian Medical University Union Hospital and Fujian Institute of Hematology, Fuzhou, Fujian, China (mainland)
| | - Shunquan Wu
- Department of Hematology, Fujian Medical University Union Hospital and Fujian Institute of Hematology, Fuzhou, Fujian, China (mainland)
| | - Qingqing Wang
- Department of Hematology, Fujian Medical University Union Hospital and Fujian Institute of Hematology, Fuzhou, Fujian, China (mainland)
| | - Yamei Ye
- Department of Hematology, Fujian Medical University Union Hospital and Fujian Institute of Hematology, Fuzhou, Fujian, China (mainland)
| | - Yun Liu
- Department of Hematology, Fujian Medical University Union Hospital and Fujian Institute of Hematology, Fuzhou, Fujian, China (mainland)
| |
Collapse
|
12
|
Shi Y, Bollam SR, White SM, Laughlin SZ, Graham GT, Wadhwa M, Chen H, Nguyen C, Vitte J, Giovannini M, Toretsky J, Yi C. Rac1-Mediated DNA Damage and Inflammation Promote Nf2 Tumorigenesis but Also Limit Cell-Cycle Progression. Dev Cell 2016; 39:452-465. [PMID: 27818180 PMCID: PMC5519326 DOI: 10.1016/j.devcel.2016.09.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 06/09/2016] [Accepted: 09/27/2016] [Indexed: 01/04/2023]
Abstract
Merlin encoded by the Nf2 gene is a bona fide tumor suppressor that has been implicated in regulation of both the Hippo-Yap and Rac1-Pak1 pathways. Using genetically engineered murine liver models, we show that co-deletion of Rac1 with Nf2 blocks tumor initiation but paradoxically exacerbates hepatomegaly induced by Nf2 loss, which can be suppressed either by treatment with pro-oxidants or by co-deletion of Yap. Our results suggest that while Yap acts as the central driver of proliferation during Nf2 tumorigenesis, Rac1 primarily functions as an inflammation switch by inducing reactive oxygen species that, on one hand, induce nuclear factor κB signaling and expression of inflammatory cytokines, and on the other activate p53 checkpoint and senescence programs dampening the cyclin D1-pRb-E2F1 pathway. Interestingly, senescence markers are associated with benign NF2 tumors but not with malignant NF2 mutant mesotheliomas, suggesting that senescence may underlie the benign nature of most NF2 tumors.
Collapse
Affiliation(s)
- Yuhao Shi
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Saumya R Bollam
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Shannon M White
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Sean Z Laughlin
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Garrett T Graham
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Mandheer Wadhwa
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Hengye Chen
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Chan Nguyen
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Jeremie Vitte
- Department of Head and Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Marco Giovannini
- Department of Head and Neck Surgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Jeffery Toretsky
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Chunling Yi
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA.
| |
Collapse
|
13
|
Petrilli AM, Fernández-Valle C. Role of Merlin/NF2 inactivation in tumor biology. Oncogene 2016; 35:537-48. [PMID: 25893302 PMCID: PMC4615258 DOI: 10.1038/onc.2015.125] [Citation(s) in RCA: 278] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Revised: 02/20/2015] [Accepted: 03/16/2015] [Indexed: 01/13/2023]
Abstract
Merlin (Moesin-ezrin-radixin-like protein, also known as schwannomin) is a tumor suppressor protein encoded by the neurofibromatosis type 2 gene NF2. Loss of function mutations or deletions in NF2 cause neurofibromatosis type 2 (NF2), a multiple tumor forming disease of the nervous system. NF2 is characterized by the development of bilateral vestibular schwannomas. Patients with NF2 can also develop schwannomas on other cranial and peripheral nerves, as well as meningiomas and ependymomas. The only potential treatment is surgery/radiosurgery, which often results in loss of function of the involved nerve. There is an urgent need for chemotherapies that slow or eliminate tumors and prevent their formation in NF2 patients. Interestingly NF2 mutations and merlin inactivation also occur in spontaneous schwannomas and meningiomas, as well as other types of cancer including mesothelioma, glioma multiforme, breast, colorectal, skin, clear cell renal cell carcinoma, hepatic and prostate cancer. Except for malignant mesotheliomas, the role of NF2 mutation or inactivation has not received much attention in cancer, and NF2 might be relevant for prognosis and future chemotherapeutic approaches. This review discusses the influence of merlin loss of function in NF2-related tumors and common human cancers. We also discuss the NF2 gene status and merlin signaling pathways affected in the different tumor types and the molecular mechanisms that lead to tumorigenesis, progression and pharmacological resistance.
Collapse
Affiliation(s)
- Alejandra M. Petrilli
- Department of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Cristina Fernández-Valle
- Department of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| |
Collapse
|
14
|
Hilton DA, Shivane A, Kirk L, Bassiri K, Enki DG, Hanemann CO. Activation of multiple growth factor signalling pathways is frequent in meningiomas. Neuropathology 2015; 36:250-61. [DOI: 10.1111/neup.12266] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/25/2015] [Accepted: 09/26/2015] [Indexed: 01/14/2023]
Affiliation(s)
- David A Hilton
- Department of Cellular and Anatomical Pathology; Derriford Hospital; Plymouth UK
| | - Aditya Shivane
- Department of Cellular and Anatomical Pathology; Derriford Hospital; Plymouth UK
| | - Leanne Kirk
- Department of Cellular and Anatomical Pathology; Derriford Hospital; Plymouth UK
| | - Kayleigh Bassiri
- Institute of Translational and Stratified Medicine; Plymouth University Peninsula Schools of Medicine & Dentistry; Plymouth UK
| | - Doyo G Enki
- Plymouth University Peninsula Schools of Medicine & Dentistry; Plymouth UK
| | - C Oliver Hanemann
- Institute of Translational and Stratified Medicine; Plymouth University Peninsula Schools of Medicine & Dentistry; Plymouth UK
| |
Collapse
|
15
|
The scaffold protein KSR1, a novel therapeutic target for the treatment of Merlin-deficient tumors. Oncogene 2015; 35:3443-53. [PMID: 26549023 DOI: 10.1038/onc.2015.404] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 09/02/2015] [Accepted: 09/18/2015] [Indexed: 12/27/2022]
Abstract
Merlin has broad tumor-suppressor functions as its mutations have been identified in multiple benign tumors and malignant cancers. In all schwannomas, the majority of meningiomas and 1/3 of ependymomas Merlin loss is causative. In neurofibromatosis type 2, a dominantly inherited tumor disease because of the loss of Merlin, patients suffer from multiple nervous system tumors and die on average around age 40. Chemotherapy is not effective and tumor localization and multiplicity make surgery and radiosurgery challenging and morbidity is often considerable. Thus, a new therapeutic approach is needed for these tumors. Using a primary human in vitro model for Merlin-deficient tumors, we report that the Ras/Raf/mitogen-activated protein, extracellular signal-regulated kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) scaffold, kinase suppressor of Ras 1 (KSR1), has a vital role in promoting schwannomas development. We show that KSR1 overexpression is involved in many pathological phenotypes caused by Merlin loss, namely multipolar morphology, enhanced cell-matrix adhesion, focal adhesion and, most importantly, increased proliferation and survival. Our data demonstrate that KSR1 has a wider role than MEK1/2 in the development of schwannomas because adhesion is more dependent on KSR1 than MEK1/2. Immunoprecipitation analysis reveals that KSR1 is a novel binding partner of Merlin, which suppresses KSR1's function by inhibiting the binding between KSR1 and c-Raf. Our proteomic analysis also demonstrates that KSR1 interacts with several Merlin downstream effectors, including E3 ubiquitin ligase CRL4(DCAF1). Further functional studies suggests that KSR1 and DCAF1 may co-operate to regulate schwannomas formation. Taken together, these findings suggest that KSR1 serves as a potential therapeutic target for Merlin-deficient tumors.
Collapse
|
16
|
Ahmad I, Yue WY, Fernando A, Clark JJ, Woodson EA, Hansen MR. p75NTR is highly expressed in vestibular schwannomas and promotes cell survival by activating nuclear transcription factor κB. Glia 2014; 62:1699-712. [PMID: 24976126 PMCID: PMC4150679 DOI: 10.1002/glia.22709] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 06/03/2014] [Accepted: 06/06/2014] [Indexed: 01/08/2023]
Abstract
Vestibular schwannomas (VSs) arise from Schwann cells (SCs) and result from the loss of function of merlin, the protein product of the NF2 tumor suppressor gene. In contrast to non-neoplastic SCs, VS cells survive long-term in the absence of axons. We find that p75(NTR) is overexpressed in VSs compared with normal nerves, both at the transcript and protein level, similar to the response of non-neoplastic SCs following axotomy. Despite elevated p75(NTR) expression, VS cells are resistant to apoptosis due to treatment with proNGF, a high affinity ligand for p75(NTR) . Furthermore, treatment with proNGF protects VS cells from apoptosis due to c-Jun N-terminal kinase (JNK) inhibition indicating that p75(NTR) promotes VS cell survival. Treatment of VS cells with proNGF activated NF-κB while inhibition of JNK with SP600125 or siRNA-mediated knockdown reduced NF-κB activity. Significantly, proNGF also activated NF-κB in cultures treated with JNK inhibitors. Thus, JNK activity appears to be required for basal levels of NF-κB activity but not for proNGF-induced NF-κB activity. To confirm that the increase in NF-κB activity contributes to the prosurvival effect of proNGF, we infected VS cultures with Ad.IκB.SerS32/36A virus, which inhibits NF-κB activation. Compared with control virus, Ad.IκB.SerS32/36A significantly increased apoptosis including in VS cells treated with proNGF. Thus, in contrast to non-neoplastic SCs, p75(NTR) signaling provides a prosurvival response in VS cells by activating NF-κB independent of JNK. Such differences may contribute to the ability of VS cells to survive long-term in the absence of axons.
Collapse
Affiliation(s)
- Iram Ahmad
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa 52242
| | - Wei Ying Yue
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa 52242
- Department of Otolaryngology-HNS, Mayo Clinic, Rochester, MN
| | - Augusta Fernando
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa 52242
- Department of Otolaryngology-HNS, Northwestern University, Chicago, IL
| | - J. Jason Clark
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa 52242
| | - Erika A. Woodson
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa 52242
- Department of Otolaryngology-HNS, Cleveland Clinic, Cleveland, OH
| | - Marlan R. Hansen
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa 52242
| |
Collapse
|
17
|
Agnihotri S, Gugel I, Remke M, Bornemann A, Pantazis G, Mack SC, Shih D, Singh SK, Sabha N, Taylor MD, Tatagiba M, Zadeh G, Krischek B. Gene-expression profiling elucidates molecular signaling networks that can be therapeutically targeted in vestibular schwannoma. J Neurosurg 2014; 121:1434-45. [PMID: 25245477 DOI: 10.3171/2014.6.jns131433] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Vestibular schwannomas (VS) are common benign tumors of the vestibular nerve that cause significant morbidity. The current treatment strategies for VS include surgery or radiation, with each treatment option having associated complications and side effects. The transcriptional landscape of schwannoma remains largely unknown. METHODS In this study the authors performed gene-expression profiling of 49 schwannomas and 7 normal control vestibular nerves to identify tumor-specific gene-expression patterns. They also interrogated whether schwannomas comprise several molecular subtypes using several transcription-based clustering strategies. The authors also performed in vitro experiments testing therapeutic inhibitors of over-activated pathways in a schwannoma cell line, namely the PI3K/AKT/mTOR pathway. RESULTS The authors identified over 4000 differentially expressed genes between controls and schwannomas with network analysis, uncovering proliferation and anti-apoptotic pathways previously not implicated in VS. Furthermore, using several distinct clustering technologies, they could not reproducibly identify distinct VS subtypes or significant differences between sporadic and germline NF2-associated schwannomas, suggesting that they are highly similar entities. The authors identified overexpression of PI3K/AKT/mTOR signaling networks in their gene-expression study and evaluated this pathway for therapeutic targeting. Testing the compounds BEZ235 and PKI-587, both novel dual inhibitors of PI3K and mTOR, attenuated tumor growth in a preclinical cell line model of schwannoma (HEI-293). In vitro findings demonstrated that pharmacological inhibition of the PI3K/AKT/mTOR pathway with next-generation compounds led to decreased cell viability and increased cell death. CONCLUSIONS These findings implicate aberrant activation of the PI3K/AKT/mTOR pathway as a molecular mechanism of pathogenesis in VS and suggest inhibition of this pathway as a potential treatment strategy.
Collapse
|
18
|
|
19
|
Yue WY, Clark JJ, Telisak M, Hansen MR. Inhibition of c-Jun N-terminal kinase activity enhances vestibular schwannoma cell sensitivity to gamma irradiation. Neurosurgery 2014; 73:506-16. [PMID: 23728448 DOI: 10.1227/01.neu.0000431483.10031.89] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Radiosurgery is increasingly used to treat vestibular schwannomas (VSs). Increasing the sensitivity of VS cells to irradiation (IR) could allow for lower and/or more effective doses of IR, improving safety and efficacy. Persistent c-Jun N-terminal kinase (JNK) activity in VS cells reduces cell death by suppressing the accumulation of reactive oxygen species (ROS), raising the possibility that JNK activity protects against IR-induced VS cell death, which is mediated by ROS. OBJECTIVE To determine the extent to which JNK signaling contributes to VS cell radiosensitivity. METHODS Primary human VS cultures, derived from acutely resected tumors, received single doses (5-40 Gy) of gamma irradiation. Histone 2AX phosphorylation, a marker of IR-induced DNA damage, was assayed by Western blot and immunostaining. ROS levels were quantified by measuring 2',7'-dichlorodihydrofluorescein diacetate (H2DCFDA) fluorescence. Cell apoptosis was determined by terminal deoxynucleotidyl transferase 2'-deoxyuridine, 5'-triphosphate nick end labeling. RESULTS The JNK inhibitors SP6000125 and I-JIP reduced histone 2AX phosphorylation after IR. They also increased H2DCFDA fluorescence in nonirradiated cultures and significantly increased IR-induced (5-10 Gy) H2DCFDA fluorescence 72 hours, but not 2 hours, after IR. Finally, I-JIP (50 μmol/L) significantly increased VS cell apoptosis in cultures treated with 20 to 40 Gy. I-JIP (20 μmol/L), SP600125 (20 μmol/L), and JNK1/2 short interfering RNA knockdown each increased VS cell apoptosis in cultures treated with 30 to 40 Gy, but not lower doses, of IR. CONCLUSION Inhibition of JNK signaling decreases histone 2AX phosphorylation and increases ROS and apoptosis in VS cells after gamma irradiation. These results raise the possibility of using JNK inhibitors to increase the effectiveness of radiosurgery for treatment of VSs.
Collapse
Affiliation(s)
- Wei Ying Yue
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | |
Collapse
|
20
|
Hilton DA, Hanemann CO. Schwannomas and their pathogenesis. Brain Pathol 2014; 24:205-20. [PMID: 24450866 DOI: 10.1111/bpa.12125] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 01/16/2014] [Indexed: 12/18/2022] Open
Abstract
Schwannomas may occur spontaneously, or in the context of a familial tumor syndrome such as neurofibromatosis type 2 (NF2), schwannomatosis and Carney's complex. Schwannomas have a variety of morphological appearances, but they behave as World Health Organization (WHO) grade I tumors, and only very rarely undergo malignant transformation. Central to the pathogenesis of these tumors is loss of function of merlin, either by direct genetic change involving the NF2 gene on chromosome 22 or secondarily to merlin inactivation. The genetic pathways and morphological features of schwannomas associated with different genetic syndromes will be discussed. Merlin has multiple functions, including within the nucleus and at the cell membrane, and this review summarizes our current understanding of the mechanisms by which merlin loss is involved in schwannoma pathogenesis, highlighting potential areas for therapeutic intervention.
Collapse
Affiliation(s)
- David A Hilton
- Department of Cellular and Anatomical Pathology, Derriford Hospital, Plymouth, UK
| | | |
Collapse
|
21
|
Boin A, Couvelard A, Couderc C, Brito I, Filipescu D, Kalamarides M, Bedossa P, De Koning L, Danelsky C, Dubois T, Hupé P, Louvard D, Lallemand D. Proteomic screening identifies a YAP-driven signaling network linked to tumor cell proliferation in human schwannomas. Neuro Oncol 2014; 16:1196-209. [PMID: 24558021 DOI: 10.1093/neuonc/nou020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Inactivation of the NF2 gene predisposes to neurofibromatosis type II and the development of schwannomas. In vitro studies have shown that loss of NF2 leads to the induction of mitogenic signaling mediated by receptor tyrosine kinases (RTKs), MAP kinase, AKT, or Hippo pathways. The goal of our study was to evaluate the expression and activity of these signaling pathways in human schwannomas in order to identify new potential therapeutic targets. METHODS Large sets of human schwannomas, totaling 68 tumors, were analyzed using complementary proteomic approaches. RTK arrays identified the most frequently activated RTKs. The correlation between the expression and activity of signaling pathways and proliferation of tumor cells using Ki67 marker was investigated by reverse-phase protein array (RRPA). Finally, immunohistochemistry was used to evaluate the expression pattern of signaling effectors in the tumors. RESULTS We showed that Her2, Her3, PDGFRß, Axl, and Tie2 are frequently activated in the tumors. Furthermore, RRPA demonstrated that Ki67 levels are linked to YAP, p-Her3, and PDGFRß expression levels. In addition, Her2, Her3, and PDGFRß are transcriptional targets of Yes-associated protein (YAP) in schwannoma cells in culture. Finally, we observed that the expression of these signaling effectors is very variable between tumors. CONCLUSIONS Tumor cell proliferation in human schwannomas is linked to a signaling network controlled by the Hippo effector YAP. Her2, Her3, PDGFRß, Axl, and Tie2, as well as YAP, represent potentially valuable therapeutic targets. However, the variability of their expression between tumors may result in strong differences in the response to targeted therapy.
Collapse
Affiliation(s)
- Alizée Boin
- Centre National de la Recherche Scientifique, Institut Curie, Paris, France (A.B., C.C., D.Lo., D.La.); Institut National de la Santé et de la Recherche Médicale, Paris, France (I.B., P.H.); Mines ParisTech, Fontainebleau, France (P.H.); Breast Cancer Biology Group, Institut Curie, Paris, France (T.D.); Reverse Phase Protein Array Platform, Institut Curie, Paris, France (C.D., L.D.K.); Centre National de la Recherche Scientifique, Institut Curie, Paris, France (D.F.); Department of Neurosurgery, Assistance Publique-Hôpitaux de Paris, Hopital Beaujon, Clichy, France (M.K.); Unité Institut National de la Santé et de la Recherche Médicale, Fondation Jean Dausset, Paris, France (M.K.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Bichat, Paris, France (A.C.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (A.C.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Beaujon, Clichy, France (P.B.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (M.K.)
| | - Anne Couvelard
- Centre National de la Recherche Scientifique, Institut Curie, Paris, France (A.B., C.C., D.Lo., D.La.); Institut National de la Santé et de la Recherche Médicale, Paris, France (I.B., P.H.); Mines ParisTech, Fontainebleau, France (P.H.); Breast Cancer Biology Group, Institut Curie, Paris, France (T.D.); Reverse Phase Protein Array Platform, Institut Curie, Paris, France (C.D., L.D.K.); Centre National de la Recherche Scientifique, Institut Curie, Paris, France (D.F.); Department of Neurosurgery, Assistance Publique-Hôpitaux de Paris, Hopital Beaujon, Clichy, France (M.K.); Unité Institut National de la Santé et de la Recherche Médicale, Fondation Jean Dausset, Paris, France (M.K.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Bichat, Paris, France (A.C.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (A.C.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Beaujon, Clichy, France (P.B.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (M.K.)
| | - Christophe Couderc
- Centre National de la Recherche Scientifique, Institut Curie, Paris, France (A.B., C.C., D.Lo., D.La.); Institut National de la Santé et de la Recherche Médicale, Paris, France (I.B., P.H.); Mines ParisTech, Fontainebleau, France (P.H.); Breast Cancer Biology Group, Institut Curie, Paris, France (T.D.); Reverse Phase Protein Array Platform, Institut Curie, Paris, France (C.D., L.D.K.); Centre National de la Recherche Scientifique, Institut Curie, Paris, France (D.F.); Department of Neurosurgery, Assistance Publique-Hôpitaux de Paris, Hopital Beaujon, Clichy, France (M.K.); Unité Institut National de la Santé et de la Recherche Médicale, Fondation Jean Dausset, Paris, France (M.K.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Bichat, Paris, France (A.C.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (A.C.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Beaujon, Clichy, France (P.B.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (M.K.)
| | - Isabel Brito
- Centre National de la Recherche Scientifique, Institut Curie, Paris, France (A.B., C.C., D.Lo., D.La.); Institut National de la Santé et de la Recherche Médicale, Paris, France (I.B., P.H.); Mines ParisTech, Fontainebleau, France (P.H.); Breast Cancer Biology Group, Institut Curie, Paris, France (T.D.); Reverse Phase Protein Array Platform, Institut Curie, Paris, France (C.D., L.D.K.); Centre National de la Recherche Scientifique, Institut Curie, Paris, France (D.F.); Department of Neurosurgery, Assistance Publique-Hôpitaux de Paris, Hopital Beaujon, Clichy, France (M.K.); Unité Institut National de la Santé et de la Recherche Médicale, Fondation Jean Dausset, Paris, France (M.K.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Bichat, Paris, France (A.C.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (A.C.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Beaujon, Clichy, France (P.B.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (M.K.)
| | - Dan Filipescu
- Centre National de la Recherche Scientifique, Institut Curie, Paris, France (A.B., C.C., D.Lo., D.La.); Institut National de la Santé et de la Recherche Médicale, Paris, France (I.B., P.H.); Mines ParisTech, Fontainebleau, France (P.H.); Breast Cancer Biology Group, Institut Curie, Paris, France (T.D.); Reverse Phase Protein Array Platform, Institut Curie, Paris, France (C.D., L.D.K.); Centre National de la Recherche Scientifique, Institut Curie, Paris, France (D.F.); Department of Neurosurgery, Assistance Publique-Hôpitaux de Paris, Hopital Beaujon, Clichy, France (M.K.); Unité Institut National de la Santé et de la Recherche Médicale, Fondation Jean Dausset, Paris, France (M.K.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Bichat, Paris, France (A.C.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (A.C.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Beaujon, Clichy, France (P.B.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (M.K.)
| | - Michel Kalamarides
- Centre National de la Recherche Scientifique, Institut Curie, Paris, France (A.B., C.C., D.Lo., D.La.); Institut National de la Santé et de la Recherche Médicale, Paris, France (I.B., P.H.); Mines ParisTech, Fontainebleau, France (P.H.); Breast Cancer Biology Group, Institut Curie, Paris, France (T.D.); Reverse Phase Protein Array Platform, Institut Curie, Paris, France (C.D., L.D.K.); Centre National de la Recherche Scientifique, Institut Curie, Paris, France (D.F.); Department of Neurosurgery, Assistance Publique-Hôpitaux de Paris, Hopital Beaujon, Clichy, France (M.K.); Unité Institut National de la Santé et de la Recherche Médicale, Fondation Jean Dausset, Paris, France (M.K.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Bichat, Paris, France (A.C.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (A.C.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Beaujon, Clichy, France (P.B.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (M.K.)
| | - Pierre Bedossa
- Centre National de la Recherche Scientifique, Institut Curie, Paris, France (A.B., C.C., D.Lo., D.La.); Institut National de la Santé et de la Recherche Médicale, Paris, France (I.B., P.H.); Mines ParisTech, Fontainebleau, France (P.H.); Breast Cancer Biology Group, Institut Curie, Paris, France (T.D.); Reverse Phase Protein Array Platform, Institut Curie, Paris, France (C.D., L.D.K.); Centre National de la Recherche Scientifique, Institut Curie, Paris, France (D.F.); Department of Neurosurgery, Assistance Publique-Hôpitaux de Paris, Hopital Beaujon, Clichy, France (M.K.); Unité Institut National de la Santé et de la Recherche Médicale, Fondation Jean Dausset, Paris, France (M.K.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Bichat, Paris, France (A.C.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (A.C.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Beaujon, Clichy, France (P.B.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (M.K.)
| | - Leanne De Koning
- Centre National de la Recherche Scientifique, Institut Curie, Paris, France (A.B., C.C., D.Lo., D.La.); Institut National de la Santé et de la Recherche Médicale, Paris, France (I.B., P.H.); Mines ParisTech, Fontainebleau, France (P.H.); Breast Cancer Biology Group, Institut Curie, Paris, France (T.D.); Reverse Phase Protein Array Platform, Institut Curie, Paris, France (C.D., L.D.K.); Centre National de la Recherche Scientifique, Institut Curie, Paris, France (D.F.); Department of Neurosurgery, Assistance Publique-Hôpitaux de Paris, Hopital Beaujon, Clichy, France (M.K.); Unité Institut National de la Santé et de la Recherche Médicale, Fondation Jean Dausset, Paris, France (M.K.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Bichat, Paris, France (A.C.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (A.C.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Beaujon, Clichy, France (P.B.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (M.K.)
| | - Carine Danelsky
- Centre National de la Recherche Scientifique, Institut Curie, Paris, France (A.B., C.C., D.Lo., D.La.); Institut National de la Santé et de la Recherche Médicale, Paris, France (I.B., P.H.); Mines ParisTech, Fontainebleau, France (P.H.); Breast Cancer Biology Group, Institut Curie, Paris, France (T.D.); Reverse Phase Protein Array Platform, Institut Curie, Paris, France (C.D., L.D.K.); Centre National de la Recherche Scientifique, Institut Curie, Paris, France (D.F.); Department of Neurosurgery, Assistance Publique-Hôpitaux de Paris, Hopital Beaujon, Clichy, France (M.K.); Unité Institut National de la Santé et de la Recherche Médicale, Fondation Jean Dausset, Paris, France (M.K.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Bichat, Paris, France (A.C.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (A.C.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Beaujon, Clichy, France (P.B.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (M.K.)
| | - Thierry Dubois
- Centre National de la Recherche Scientifique, Institut Curie, Paris, France (A.B., C.C., D.Lo., D.La.); Institut National de la Santé et de la Recherche Médicale, Paris, France (I.B., P.H.); Mines ParisTech, Fontainebleau, France (P.H.); Breast Cancer Biology Group, Institut Curie, Paris, France (T.D.); Reverse Phase Protein Array Platform, Institut Curie, Paris, France (C.D., L.D.K.); Centre National de la Recherche Scientifique, Institut Curie, Paris, France (D.F.); Department of Neurosurgery, Assistance Publique-Hôpitaux de Paris, Hopital Beaujon, Clichy, France (M.K.); Unité Institut National de la Santé et de la Recherche Médicale, Fondation Jean Dausset, Paris, France (M.K.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Bichat, Paris, France (A.C.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (A.C.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Beaujon, Clichy, France (P.B.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (M.K.)
| | - Philippe Hupé
- Centre National de la Recherche Scientifique, Institut Curie, Paris, France (A.B., C.C., D.Lo., D.La.); Institut National de la Santé et de la Recherche Médicale, Paris, France (I.B., P.H.); Mines ParisTech, Fontainebleau, France (P.H.); Breast Cancer Biology Group, Institut Curie, Paris, France (T.D.); Reverse Phase Protein Array Platform, Institut Curie, Paris, France (C.D., L.D.K.); Centre National de la Recherche Scientifique, Institut Curie, Paris, France (D.F.); Department of Neurosurgery, Assistance Publique-Hôpitaux de Paris, Hopital Beaujon, Clichy, France (M.K.); Unité Institut National de la Santé et de la Recherche Médicale, Fondation Jean Dausset, Paris, France (M.K.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Bichat, Paris, France (A.C.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (A.C.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Beaujon, Clichy, France (P.B.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (M.K.)
| | - Daniel Louvard
- Centre National de la Recherche Scientifique, Institut Curie, Paris, France (A.B., C.C., D.Lo., D.La.); Institut National de la Santé et de la Recherche Médicale, Paris, France (I.B., P.H.); Mines ParisTech, Fontainebleau, France (P.H.); Breast Cancer Biology Group, Institut Curie, Paris, France (T.D.); Reverse Phase Protein Array Platform, Institut Curie, Paris, France (C.D., L.D.K.); Centre National de la Recherche Scientifique, Institut Curie, Paris, France (D.F.); Department of Neurosurgery, Assistance Publique-Hôpitaux de Paris, Hopital Beaujon, Clichy, France (M.K.); Unité Institut National de la Santé et de la Recherche Médicale, Fondation Jean Dausset, Paris, France (M.K.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Bichat, Paris, France (A.C.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (A.C.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Beaujon, Clichy, France (P.B.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (M.K.)
| | - Dominique Lallemand
- Centre National de la Recherche Scientifique, Institut Curie, Paris, France (A.B., C.C., D.Lo., D.La.); Institut National de la Santé et de la Recherche Médicale, Paris, France (I.B., P.H.); Mines ParisTech, Fontainebleau, France (P.H.); Breast Cancer Biology Group, Institut Curie, Paris, France (T.D.); Reverse Phase Protein Array Platform, Institut Curie, Paris, France (C.D., L.D.K.); Centre National de la Recherche Scientifique, Institut Curie, Paris, France (D.F.); Department of Neurosurgery, Assistance Publique-Hôpitaux de Paris, Hopital Beaujon, Clichy, France (M.K.); Unité Institut National de la Santé et de la Recherche Médicale, Fondation Jean Dausset, Paris, France (M.K.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Bichat, Paris, France (A.C.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (A.C.); Pathology Department Beaujon-Bichat, AP-HP, Hôpital Beaujon, Clichy, France (P.B.); Université Paris Diderot, Sorbonne Paris Cité, Paris, France (M.K.)
| |
Collapse
|
22
|
Abstract
Sarcomas are cancers arising from the mesenchymal layer that affect children, adolescents, young adults, and adults. Although most sarcomas are localized, many display a remarkable predilection for metastasis to the lungs, liver, bones, subcutaneous tissue, and lymph nodes. Additionally, many sarcoma patients presenting initially with localized disease may relapse at metastatic sites. While localized sarcomas can often be cured through surgery and often radiation, controversies exist over optimal management of patients with metastatic sarcoma. Combinations of chemotherapy are the most effective in many settings, and many promising new agents are under active investigation or are being explored in preclinical models. Metastatic sarcomas are excellent candidates for novel approaches with additional agents as they have demonstrated chemosensitivity and affect a portion of the population that is motivated toward curative therapy. In this paper, we provide an overview on the common sarcomas of childhood (rhabdomyosarcoma), adolescence, and young adults (osteosarcoma, Ewing sarcoma, synovial sarcoma, and malignant peripheral nerve sheath tumor) and older adults (leiomyosarcoma, liposarcoma, and undifferentiated high grade sarcoma) in terms of the epidemiology, current therapy, promising therapeutic directions and outcome with a focus on metastatic disease. Potential advances in terms of promising therapy and biologic insights may lead to more effective and safer therapies; however, more clinical trials and research are needed for patients with metastatic sarcoma.
Collapse
Affiliation(s)
- Ernest K Amankwah
- Department of Cancer Epidemiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Anthony P Conley
- Sarcoma Department, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Damon R Reed
- Sarcoma Department, H Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
23
|
Shivane A, Parkinson DB, Ammoun S, Hanemann CO. Expression of c-Jun and Sox-2 in human schwannomas and traumatic neuromas. Histopathology 2013; 62:651-6. [PMID: 23362975 DOI: 10.1111/his.12062] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 11/04/2012] [Indexed: 12/24/2022]
Abstract
AIMS Schwann cells myelinate axons of the peripheral nervous system. This process of myelination is regulated by various transcription factors. c-Jun and Sox-2 are negative regulators of myelination and control Schwann cell differentiation and plasticity. Schwannoma cells within tumours no longer express myelin markers, and show increased proliferation and decreased apoptosis. We have shown previously that several signalling pathways are activated in schwannoma cells in situ, in particular the c-Jun N-terminal kinase (JNK) pathway. Both in vitro and in vivo we have demonstrated that c-Jun and Sox-2 are co-regulated in Schwann cells and evidence shows that both these proteins regulate myelination negatively. In this study, we aimed to characterize the expression of c-Jun and Sox-2 in schwannoma and traumatic neuroma. METHODS AND RESULTS Immunohistochemistry using antibodies to c-Jun and Sox-2 was applied to six schwannomas, and the results were compared with those seen in traumatic neuroma and normal nerve. Increased expression of c-Jun and Sox-2 was seen in schwannoma. CONCLUSIONS We have demonstrated increased expression of c-Jun and Sox-2 in schwannoma compared to traumatic neuroma. There was no expression of c-Jun and Sox-2 in a histologically normal peripheral nerve.
Collapse
Affiliation(s)
- Aditya Shivane
- Department of Cellular and Anatomical Pathology, Derriford Hospital, Plymouth, UK.
| | | | | | | |
Collapse
|
24
|
Axl/Gas6/NFκB signalling in schwannoma pathological proliferation, adhesion and survival. Oncogene 2013; 33:336-46. [PMID: 23318455 DOI: 10.1038/onc.2012.587] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 10/02/2012] [Accepted: 10/29/2012] [Indexed: 01/01/2023]
Abstract
TAM family receptor tyrosine kinases comprising Tyro3 (Sky), Axl, and Mer are overexpressed in some cancers, correlate with multidrug resistance and contribute to tumourigenesis by regulating invasion, angiogenesis, cell survival and tumour growth. Mutations in the gene coding for a tumour suppressor merlin cause development of multiple tumours of the nervous system such as schwannomas, meningiomas and ependymomas occurring spontaneously or as part of a hereditary disease neurofibromatosis type 2. The benign character of merlin-deficient tumours makes them less responsive to chemotherapy. We previously showed that, amongst other growth factor receptors, TAM family receptors (Tyro3, Axl and Mer) are significantly overexpressed in schwannoma tissues. As Axl is negatively regulated by merlin and positively regulated by E3 ubiquitin ligase CRL4DCAF1, previously shown to be a key regulator in schwannoma growth we hypothesized that Axl is a good target to study in merlin-deficient tumours. Moreover, Axl positively regulates the oncogene Yes-associated protein, which is known to be under merlin regulation in schwannoma and is involved in increased proliferation of merlin-deficient meningioma and mesothelioma. Here, we demonstrated strong overexpression and activation of Axl receptor as well as its ligand Gas6 in human schwannoma primary cells compared to normal Schwann cells. We show that Gas6 is mitogenic and increases schwannoma cell-matrix adhesion and survival acting via Axl in schwannoma cells. Stimulation of the Gas6/Axl signalling pathway recruits Src, focal adhesion kinase (FAK) and NFκB. We showed that NFκB mediates Gas6/Axl-mediated overexpression of survivin, cyclin D1 and FAK, leading to enhanced survival, cell-matrix adhesion and proliferation of schwannoma. We conclude that Axl/FAK/Src/NFκB pathway is relevant in merlin-deficient tumours and is a potential therapeutic target for schwannoma and other merlin-deficient tumours.
Collapse
|
25
|
Morrow KA, Shevde LA. Merlin: the wizard requires protein stability to function as a tumor suppressor. Biochim Biophys Acta Rev Cancer 2012; 1826:400-6. [PMID: 22750751 DOI: 10.1016/j.bbcan.2012.06.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 06/18/2012] [Accepted: 06/20/2012] [Indexed: 11/27/2022]
Abstract
Neurofibromatosis type 2 (NF2), characterized by tumors of the nervous system, is a result of functional loss of the NF2 gene. The NF2 gene encodes Merlin (moesin-ezrin-radixin-like protein), an ERM (Ezrin, Radixin, Moesin) protein family member. Merlin functions as a tumor suppressor through impacting mechanisms related to proliferation, apoptosis, survival, motility, adhesion, and invasion. Several studies have summarized the tumor intrinsic mutations in Merlin. Given the fact that tumor cells are not in isolation, but rather in an intricate, mutually sustaining synergy with their surrounding stroma, the dialog between the tumor cells and the stroma can potentially impact the molecular homeostasis and promote evolution of the malignant phenotype. This review summarizes the epigenetic modifications, transcript stability, and post-translational modifications that impact Merlin. We have reviewed the role of extrinsic factors originating from the tumor milieu that influence the availability of Merlin inside the cell. Information regarding Merlin regulation could lead to novel therapeutics by stabilizing Merlin protein in tumors that have reduced Merlin protein expression without displaying any NF2 genetic alterations.
Collapse
Affiliation(s)
- K Adam Morrow
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | | |
Collapse
|
26
|
James MF, Stivison E, Beauchamp R, Han S, Li H, Wallace MR, Gusella JF, Stemmer-Rachamimov AO, Ramesh V. Regulation of mTOR complex 2 signaling in neurofibromatosis 2-deficient target cell types. Mol Cancer Res 2012; 10:649-59. [PMID: 22426462 DOI: 10.1158/1541-7786.mcr-11-0425-t] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Inactivating mutations in the neurofibromatosis 2 (NF2) tumor suppressor gene results in the development of schwannomas and meningiomas. Using NF2-deficient meningioma cells and tumors, together with the normal cellular counterparts that meningiomas derive, arachnoid cells, we identified merlin as a novel negative regulator of mTOR complex 1 (mTORC1). We now show that merlin positively regulates the kinase activity of mTORC2, a second functionally distinct mTOR complex, and that downstream phosphorylation of mTORC2 substrates, including Akt, is reduced upon acute merlin deficiency in cells. In response to general growth factor stimulation, Akt signaling is attenuated in merlin RNA interference-suppressed human arachnoid and Schwann cells by mechanisms mediated by hyperactive mTORC1 and impaired mTORC2. Moreover, Akt signaling is impaired differentially in a cell type-dependent manner in response to distinct growth factor stimuli. However, contrary to activation of mTORC1, the attenuated mTORC2 signaling profiles exhibited by normal arachnoid and Schwann cells in response to acute merlin loss were not consistently reflected in NF2-deficient meningiomas and schwannomas, suggesting additional genetic events may have been acquired in tumors after initial merlin loss. This finding contrasts with another benign tumor disorder, tuberous sclerosis complex, which exhibits attenuated mTORC2 signaling profiles in both cells and tumors. Finally, we examined rapamycin, as well as the mTOR kinase inhibitor, Torin1, targeting both mTOR complexes to identify the most efficacious class of compounds for blocking mTOR-mediated signaling and proliferation in merlin-deficient meningioma cells. These studies may ultimately aid in the development of suitable therapeutics for NF2-associated tumors.
Collapse
Affiliation(s)
- Marianne F James
- Center for Human Genetic Research, Massachusetts General Hospital, Richard B. Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Molecular Pathogenesis of Vestibular Schwannomas: Insights for the Development of Novel Medical Therapies. Otolaryngol Pol 2012; 66:84-95. [DOI: 10.1016/s0030-6657(12)70754-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 02/13/2012] [Indexed: 11/21/2022]
|
28
|
Multistep phosphorylation by oncogenic kinases enhances the degradation of the NF2 tumor suppressor merlin. Neoplasia 2011; 13:643-52. [PMID: 21750658 DOI: 10.1593/neo.11356] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 05/13/2011] [Accepted: 05/16/2011] [Indexed: 11/18/2022] Open
Abstract
Mutations in the Neurofibromatosis 2 gene (NF2) predispose to tumors of the nervous system, mainly schwannomas and meningiomas. The NF2 gene encodes for the tumor suppressor protein merlin (moesin-ezrin-radixin-like protein), which functions as a linker between the plasma membrane and the cytoskeleton. Carboxyterminal phosphorylation affects merlin activity, but many open questions on the regulation of merlin function still remain. The phosphoinositide 3-kinase/Akt pathway is activated in human vestibular schwannoma, suggesting a role for Akt-dependent merlin regulation in the formation of these tumors. In this study, we identify merlin serine 10 as a novel substrate for Akt phosphorylation. We demonstrate that this N-terminal phosphorylation directs merlin for proteasome-mediated degradation and affects merlin binding to the E3 ligase component DCAF1. Our data indicate that sequential phosphorylation of merlin C- and N-terminus by different oncogenic kinases targets merlin for degradation and thus downregulates its activity. On the basis of these findings, we propose a model for a posttranslational mechanism of merlin inactivation.
Collapse
|
29
|
Jacob A, Oblinger J, Bush ML, Brendel V, Santarelli G, Chaudhury AR, Kulp S, La Perle KMD, Chen CS, Chang LS, Welling DB. Preclinical validation of AR42, a novel histone deacetylase inhibitor, as treatment for vestibular schwannomas. Laryngoscope 2011; 122:174-89. [PMID: 22109824 DOI: 10.1002/lary.22392] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 09/07/2011] [Accepted: 09/14/2011] [Indexed: 01/17/2023]
Abstract
OBJECTIVES/HYPOTHESIS Recent studies indicate that vestibular schwannomas (VSs) rely on phosphatidylinositol 3-kinase/AKT activation to promote cell proliferation and survival; therefore, targeting AKT may provide new therapeutic options. We have previously shown that AR42, a novel histone deacetylase inhibitor, potently suppresses VS growth in vitro at doses correlating with AKT inactivation. The objectives of the current study were translational: 1) to examine the end biologic effects of AR42 on tumor growth in vivo, 2) to validate AKT as its in vivo molecular target, 3) to determine whether AR42 penetrates the blood-brain barrier (BBB), and 4) to study the pharmacotoxicity profile of AR42. STUDY DESIGN In vivo mouse studies. METHODS AR42 was dosed orally in murine schwannoma allografts and human VS xenografts. Magnetic resonance imaging was used to quantify changes in tumor volume, and intracellular molecular targets were analyzed using immunohistochemistry. BBB penetration was assayed, and both blood-chemistry measurements and histology studies were used to evaluate toxicity. RESULTS Growth of schwannoma implants was dramatically decreased by AR42 at doses correlating with AKT dephosphorylation, cell cycle arrest, and apoptosis. AR42 penetrated the BBB, and wild-type mice fed AR42 for 6 months behaved normally and gained weight appropriately. Blood-chemistry studies and organ histology performed after 3 and 6 months of AR42 treatment demonstrated no clinically significant abnormalities. CONCLUSIONS AR42 suppresses schwannoma growth at doses correlating with AKT pathway inhibition. This orally bioavailable drug penetrates the BBB, is well tolerated, and represents a novel candidate for translation to human VS clinical trials.
Collapse
Affiliation(s)
- Abraham Jacob
- Department of Surgery, Division of Otolaryngology, University of Arizona, Tucson, Arizona 85724, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Ammoun S, Schmid MC, Zhou L, Ristic N, Ercolano E, Hilton DA, Perks CM, Hanemann CO. Insulin-like growth factor-binding protein-1 (IGFBP-1) regulates human schwannoma proliferation, adhesion and survival. Oncogene 2011; 31:1710-22. [PMID: 21892205 DOI: 10.1038/onc.2011.357] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Merlin is a tumour suppressor involved in the development of a variety of tumours including mesotheliomas. Neurofibromatosis type 2 (NF2), a dominantly inherited tumour disease, is also caused by loss of merlin. NF2 patients suffer from multiple genetically well-defined tumours, schwannomas are most frequent among those. Using our in vitro model for human schwannoma, we found that schwannoma cells display enhanced proliferation because of the overexpression/activation of platelet-derived growth factor receptor and ErbB2/3, increased cell-matrix adhesion because of the overexpression of integrins, and decreased apoptosis. Mechanisms underlying schwannomas basal proliferation and cell-matrix adhesion are not understood. Here, we investigated insulin-like growth factor-binding protein-1 (IGFBP-1), which is expressed and released from central nervous system tumours and strongly overexpressed in schwannoma at the mRNA level. IGFBP-1 acts via β1-integrin and focal-adhesion-kinase (FAK), which are strongly overexpressed and basally activated in schwannoma. Using short hairpin RNA knockdown, small inhibitors and recombinant IGFBP-1, we demonstrate that schwannoma cells, in contrast to Schwann cells, release IGFBP-1 that activates the Src/FAK pathway, via integrin β1, potentiating schwannoma's proliferation and cell-matrix adhesion. We show that FAK localizes to the nucleus and Src triggers IGFBP-1 production. Further, we observed downregulation of the tumour-suppressor phosphatase and tensin homolog in schwannoma cells leading to increased activity of anti-apoptotic AKT. Thus, IGFBP-1/integrin β1/Src/FAK pathway has a crucial role in merlin-related tumourigenesis and therefore represents an important therapeutic target in the treatment of merlin-deficient tumours.
Collapse
Affiliation(s)
- S Ammoun
- Clinical Neurobiology, Peninsula College for Medicine and Dentistry, Plymouth, UK
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Bush ML, Oblinger J, Brendel V, Santarelli G, Huang J, Akhmametyeva EM, Burns SS, Wheeler J, Davis J, Yates CW, Chaudhury AR, Kulp S, Chen CS, Chang LS, Welling DB, Jacob A. AR42, a novel histone deacetylase inhibitor, as a potential therapy for vestibular schwannomas and meningiomas. Neuro Oncol 2011; 13:983-99. [PMID: 21778190 DOI: 10.1093/neuonc/nor072] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Neurofibromatosis type 2 (NF2) is an autosomal-dominant disease that results in the formation of bilateral vestibular schwannomas (VSs) and multiple meningiomas. Treatment options for NF2-associated tumors are limited, and to date, no medical therapies are FDA approved. The ideal chemotherapeutic agent would inhibit both VS and meningiomas simultaneously. The objectives of this study are (1) to test the efficacy of AR42, a novel histone deacetylase inhibitor, to inhibit VS and meningioma growth and (2) to investigate this drug's mechanisms of action. Primary cultures of human VS and meningioma cells were established. Nf2-deficient mouse schwannoma and benign human meningioma Ben-Men-1 cells were also cultured. Cells were treated with AR42, and the drug's effects on proliferation and the cell cycle were analyzed using a methanethiosulfonate assay and flow cytometry, respectively. Human phospho-kinase arrays and Western blots were used to evaluate the effects of AR42 on intracellular signaling. The in vivo efficacy of AR42 was investigated using schwannoma xenografts. Tumor volumes were quantified using high-field, volumetric MRI, and molecular target analysis was performed using immunohistochemistry. AR42 inhibited the growth of primary human VS and Nf2-deficient mouse schwannoma cells with a half maximal inhibitory concentration (IC(50)) of 500 nM and 250-350 nM, respectively. AR42 also inhibited primary meningioma cells and the benign meningioma cell line, Ben-Men-1, with IC(50) values of 1.5 µM and 1.0 µM, respectively. AR42 treatment induced cell-cycle arrest at G(2) and apoptosis in both VS and meningioma cells. Also, AR42 exposure decreased phosphorylated Akt in schwannoma and meningioma cells. In vivo treatment with AR42 inhibited the growth of schwannoma xenografts, induced apoptosis, and decreased Akt activation. The potent growth inhibitory activity of AR42 in schwannoma and meningioma cells suggests that AR42 should be further evaluated as a potential treatment for NF2-associated tumors.
Collapse
Affiliation(s)
- Matthew L Bush
- Department of Otolaryngology–Head and Neck Surgery, The Ohio State University, Columbus, OH 43212, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Yue WY, Clark JJ, Fernando A, Domann F, Hansen MR. Contribution of persistent C-Jun N-terminal kinase activity to the survival of human vestibular schwannoma cells by suppression of accumulation of mitochondrial superoxides. Neuro Oncol 2011; 13:961-73. [PMID: 21697181 DOI: 10.1093/neuonc/nor068] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Vestibular schwannomas (VSs) result from inactivating mutations in the merlin tumor suppressor gene. The merlin protein suppresses a variety of progrowth kinase-signaling cascades, including extracellular regulated kinase/mitogen-activated protein kinase (ERK/MAPK), c-Jun N-terminal kinase (JNK), and phosphatidyl-inositol 3-kinase (PI3-K)/Akt. Recent studies indicate that ERKs and Akt are active in human VSs, and here we show that JNKs are also persistently active in human VS cells. With use of cultures of human VSs, we investigated the contribution of each of these signals to the proliferative and survival response of VS cells. Inhibition of ERK or Akt signaling reduced VS cell proliferation but did not increase apoptosis, whereas inhibition of JNK with SP600125, I-JIP, or siRNA knock-down reduced VS cell proliferation and survival by inducing apoptosis. By contrast, JNK activity promotes apoptosis in normal Schwann cells. Inhibition of JNK increased the fluorescence intensity of VS cells loaded with 5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate (H(2)DCFDA), a fluorescent probe for reactive oxygen species (ROS). Furthermore, ebselen, a ROS scavenger, rescued VS cells with suppressed JNK from apoptosis, suggesting that JNK activity protects VS cells from apoptosis by limiting accumulation of ROS. VS cultures treated with JNK inhibitors demonstrated significantly higher levels of MitoSOX Red fluorescence, implying that persistent JNK activity specifically suppresses superoxide production in the mitochondria. Overexpression of superoxide dismutase 2 (MnSOD; mitochondrial SOD) prevented apoptosis in VS cells with suppressed JNK signaling. Taken together, these results indicate that persistent JNK activity enhances VS cell survival, at least in part, by suppressing accumulation of mitochondrial superoxides.
Collapse
Affiliation(s)
- Wei Ying Yue
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa 52242, USA
| | | | | | | | | |
Collapse
|
33
|
Ammoun S, Hanemann CO. Emerging therapeutic targets in schwannomas and other merlin-deficient tumors. Nat Rev Neurol 2011; 7:392-9. [DOI: 10.1038/nrneurol.2011.82] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
34
|
Ammoun S, Cunliffe CH, Allen JC, Chiriboga L, Giancotti FG, Zagzag D, Hanemann CO, Karajannis MA. ErbB/HER receptor activation and preclinical efficacy of lapatinib in vestibular schwannoma. Neuro Oncol 2010; 12:834-43. [PMID: 20511180 DOI: 10.1093/neuonc/noq012] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Vestibular schwannomas (VS) arising sporadically or in patients with neurofibromatosis type 2 (NF2) consistently lack expression of Merlin, a tumor suppressor. Conventional treatment options include surgery and radiotherapy but there is no validated medical option. Recent evidence suggests that Merlin deficiency may result in abnormal activation of receptor tyrosine kinases (RTKs) and downstream signaling, promoting tumor growth. Although small-molecule RTK inhibitors are widely available for clinical use, no such therapy has been validated in patients with VS. To screen for RTK activation, surgical VS specimens from patients with and without NF2 were analyzed by phospho-RTK profiling arrays. Downstream signaling pathway activation was analyzed by phospho-MAPK arrays. Activated RTKs and downstream kinases were validated immunohistochemically in corresponding formalin-fixed, paraffin-embedded tissues. Phospho-RTK arrays and immunohistochemistry showed consistent overexpression and activation of EGFR family receptors and evidence of ERK1/2 downstream signaling was observed in all samples analyzed (n = 11). Based on the findings, the small-molecule EGFR/ErbB2 kinase inhibitor lapatinib was selected for evaluation of target inhibition and treatment efficacy in our in vitro human schwannoma model. EGFR/ErbB2 targeted therapy with lapatinib inhibited ErbB2 phosphorylation and survivin upregulation, as well as downstream ERK1/2 and AKT activation, resulting in decreased proliferation. We conclude that EGFR family receptor activation is a consistent feature of both sporadic and NF2-related VS. Molecular targeted therapy with lapatinib downregulates survivin and has antiproliferative activity in a preclinical VS model. Based on these findings, a clinical trial with lapatinib for the treatment of VS is currently underway.
Collapse
Affiliation(s)
- Sylwia Ammoun
- Division of Pediatric Hematology/Oncology and NYU Cancer Institute, Hassenfeld Children's Center for Cancer and Blood Disorders, 160 East 32nd Street, 2nd Floor, New York, NY 10016, USA
| | | | | | | | | | | | | | | |
Collapse
|