Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Oncogene ; 43(13): 921-930, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38336988

ABSTRACT

Neurofibromatosis Type 2 (NF2)-related schwannomatosis is a genetic disorder that causes development of multiple types of nervous system tumors. The primary and diagnostic tumor type is bilateral vestibular schwannoma. There is no cure or drug therapy for NF2. Recommended treatments include surgical resection and radiation, both of which can leave patients with severe neurological deficits or increase the risk of future malignant tumors. Results of our previous pilot high-throughput drug screen identified phosphoinositide 3-kinase (PI3K) inhibitors as strong candidates based on loss of viability of mouse merlin-deficient Schwann cells (MD-SCs). Here we used novel human schwannoma model cells to conduct combination drug screens. We identified a class I PI3K inhibitor, pictilisib and p21 activated kinase (PAK) inhibitor, PF-3758309 as the top combination due to high synergy in cell viability assays. Both single and combination therapies significantly reduced growth of mouse MD-SCs in an orthotopic allograft mouse model. The inhibitor combination promoted cell cycle arrest and apoptosis in mouse merlin-deficient Schwann (MD-SCs) cells and cell cycle arrest in human MD-SCs. This study identifies the PI3K and PAK pathways as potential targets for combination drug treatment of NF2-related schwannomatosis.


Subject(s)
Indazoles , Neurilemmoma , Neurofibromatoses , Neurofibromatosis 2 , Skin Neoplasms , Sulfonamides , Humans , Animals , Mice , Neurofibromatosis 2/drug therapy , Neurofibromatosis 2/genetics , Neurofibromatosis 2/metabolism , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Phosphatidylinositol 3-Kinases , p21-Activated Kinases/genetics , Phosphatidylinositol 3-Kinase/therapeutic use , Neurilemmoma/drug therapy , Neurilemmoma/genetics
2.
Oncotarget ; 13: 890-904, 2022.
Article in English | MEDLINE | ID: mdl-35875610

ABSTRACT

Neurofibromatosis Type 2 (NF2) is a rare tumor disorder caused by pathogenic variants of the merlin tumor suppressor encoded by NF2. Patients develop vestibular schwannomas (VS), peripheral schwannomas, meningiomas, and ependymomas. There are no approved drug therapies for NF2. Previous work identified phosphoinositide-3 kinase (PI3K) as a druggable target. Here we screened PI3K pathway inhibitors for efficacy in reducing viability of human schwannoma cells. The lead compound, CUDC907, a dual histone deacetylase (HDAC)/PI3K inhibitor, was further evaluated for its effects on isolated and nerve-grafted schwannoma model cells, and primary VS cells. CUDC907 (3 nM IG50) reduced human merlin deficient Schwann cell (MD-SC) viability and was 5-100 fold selective for MD over WT-SCs. CUDC907 (10 nM) promoted cell cycle arrest and caspase-3/7 activation within 24 h in human MD-SCs. Western blots confirmed a dose-dependent increase in acetylated lysine and decreases in pAKT and YAP. CUDC907 decreased tumor growth rate by 44% in a 14-day treatment regimen, modulated phospho-target levels, and decreased YAP levels. In five primary VS, CUDC907 decreased viability, induced caspase-3/7 cleavage, and reduced YAP levels. Its efficacy correlated with basal phospho-HDAC2 levels. CUDC907 has cytotoxic activity in NF2 schwannoma models and primary VS cells and is a candidate for clinical trials.


Subject(s)
Neurilemmoma , Neurofibromatosis 2 , Humans , Apoptosis , Caspase 3 , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases , Lysine , Neurilemmoma/pathology , Neurofibromatosis 2/drug therapy , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/pathology , Neurofibromin 2 , Phosphatidylinositol 3-Kinases , Phosphatidylinositols/pharmacology , Phosphatidylinositols/therapeutic use , Phosphoinositide-3 Kinase Inhibitors
3.
World Neurosurg ; 157: e66-e76, 2022 01.
Article in English | MEDLINE | ID: mdl-34587518

ABSTRACT

BACKGROUND: Vestibular schwannoma is the third most common benign intracranial tumor that can occur sporadically or be associated with neurofibromatosis type 2 (neurofibromatosis type 2 vestibular schwannoma [NF2-VS]). The aim of this study is to provide a comprehensive bioinformatic analysis of methylated-differentially expressed genes (MDEGs) in NF2-VS. METHODS: Transcriptional sequencing datasets (GSE141801 and GSE108524) and gene methylation microarrays (GSE56598) from the Gene Expression Omnibus database were used to identify and analyze MDEGs in NF2-VS. A protein-protein interaction (PPI) network was built, and the hub genes and modules were identified. Finally, potential pharmacotherapy targeting MDEGs were extracted for NF2-VS. RESULTS: A total of 57 hypermethylation-low expression genes and 88 hypomethylation-high expression genes were identified. Pathways associated with aberrantly MDEGs included P13K-AKT, MAPK, and Ras, which were also involved in NF2-VS. Six hub genes (EGFR, CCND1, CD53, CSF1R, PLAU, and FGFR1) were identified from the PPI network. Modification of the aforementioned genes altered cell-to-cell communication, response to stimulus, cellular regulation, and membrane and protein bindings. Thirty drugs targeting these pathways were selected based on the hub genes. CONCLUSIONS: Analysis of MDEGs may enrich the understanding of the molecular mechanisms of NF2-VS pathogenesis and lay the groundwork for potential biomarkers and therapeutic targets for NF2-VS.


Subject(s)
Computational Biology/methods , DNA Methylation/physiology , Neurofibromatosis 2/genetics , Neuroma, Acoustic/genetics , Protein Interaction Maps/genetics , Transcriptome/genetics , Databases, Genetic , Humans , Neurofibromatosis 2/metabolism , Neuroma, Acoustic/metabolism
4.
Hum Mol Genet ; 30(17): 1607-1617, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34075397

ABSTRACT

Neurofibromatosis Type II (NF2) is an autosomal dominant cancer predisposition syndrome in which germline haploinsufficiency at the NF2 gene confers a greatly increased propensity for tumor development arising from tissues of neural crest derived origin. NF2 encodes the tumor suppressor, Merlin, and its biochemical function is incompletely understood. One well-established function of Merlin is as a negative regulator of group A serine/threonine p21-activated kinases (PAKs). In these studies we explore the role of PAK1 and its closely related paralog, PAK2, both pharmacologically and genetically, in Merlin-deficient Schwann cells and in a genetically engineered mouse model (GEMM) that develops spontaneous vestibular and spinal schwannomas. We demonstrate that PAK1 and PAK2 are both hyper activated in Merlin-deficient murine schwannomas. In preclinical trials, a pan Group A PAK inhibitor, FRAX-1036, transiently reduced PAK1 and PAK2 phosphorylation in vitro, but had insignificant efficacy in vivo. NVS-PAK1-1, a PAK1 selective inhibitor, had a greater but still minimal effect on our GEMM phenotype. However, genetic ablation of Pak1 but not Pak2 reduced tumor formation in our NF2 GEMM. Moreover, germline genetic deletion of Pak1 was well tolerated, while conditional deletion of Pak2 in Schwann cells resulted in significant morbidity and mortality. These data support the further development of PAK1-specific small molecule inhibitors and the therapeutic targeting of PAK1 in vestibular schwannomas and argue against PAK1 and PAK2 existing as functionally redundant protein isoforms in Schwann cells.


Subject(s)
Neurofibromatosis 2/genetics , p21-Activated Kinases/metabolism , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Genes, Tumor Suppressor/drug effects , Indoles , Longevity , Mice , Neurilemmoma/genetics , Neurofibromatosis 2/metabolism , Neurofibromin 2/genetics , Phosphorylation , Piperidines , Pyrimidines , Schwann Cells/metabolism , p21-Activated Kinases/genetics
5.
Ann Clin Transl Neurol ; 8(7): 1508-1514, 2021 07.
Article in English | MEDLINE | ID: mdl-34053190

ABSTRACT

Schwannomas are benign neoplasms that can cause gain- and loss-of-function neurological phenotypes, including severe, intractable pain. To investigate the molecular mechanisms underlying schwannoma-associated pain we compared the RNA sequencing profile of painful and non-painful schwannomas from NF2 patients. Distinct segregation of painful and non-painful tumors by gene expression patterns was observed. Differential expression analysis showed the upregulation of fibroblast growth factor 7 (FGF7) in painful schwannomas. Behavioral support for this finding was observed using a xenograft human NF2-schwannoma model in nude mice. In this model, over-expression of FGF7 in intra-sciatically implanted NF2 tumor cells generated pain behavior compared with controls.


Subject(s)
Fibroblast Growth Factor 7/genetics , Neurilemmoma/genetics , Neurofibromatosis 2/genetics , Pain/genetics , Sequence Analysis, RNA/methods , Transcriptome/genetics , Animals , Cell Line, Tumor , Female , Fibroblast Growth Factor 7/biosynthesis , Humans , Male , Mice , Mice, Nude , Neurilemmoma/metabolism , Neurilemmoma/pathology , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/pathology , Pain/metabolism , Pain/pathology , Sciatic Neuropathy/genetics , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/pathology , Xenograft Model Antitumor Assays/methods
6.
Clin Nucl Med ; 46(6): e312-e316, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33826573

ABSTRACT

PURPOSE: Neurofibromatosis type 2 (NF2) is a genetic disorder that is associated with multiple tumors of the nervous system, and approximately one half of patients present with meningiomas. For patients with multifocal disease, somatostatin receptor-targeted peptide receptor radionuclide therapy (PRRT) might be a suitable systemic treatment option. PATIENTS AND METHODS: Between March 2015 and August 2017, 11 NF2 patients (7 females and 4 males; mean age, 39 ± 12 years) with multifocal, progressive meningiomas underwent a median of 4 cycles of PRRT (range, 2-6 cycles). Acute and chronic adverse events were recorded according to National Institutes of Health's Common Toxicity Criteria (CTC) version 5.0. Follow-up MRIs (every 3 to 6 months), using the Response Assessment in Neuro-Oncology response criteria for meningiomas, were used to assess treatment responses. RESULTS: Peptide receptor radionuclide therapy was well tolerated in all patients without any relevant acute adverse effects. Transient hematologic toxicity (CTC grade 3) was observed in 2 subjects. Somatostatin receptor-directed radiopeptide therapy resulted in radiological disease stabilization in 6 of 11 patients. Median progression-free survival was 12 months (range, 1-55 months), and overall survival was 37 months (range, 5-61 months). CONCLUSIONS: Based on our retrospective pilot data, PRRT is feasible and well-tolerated in NF2 patients. It might offer a suitable treatment option in subjects with multiple, recurrent, or treatment-refractory meningiomas.


Subject(s)
Neurofibromatosis 2/radiotherapy , Receptors, Somatostatin/metabolism , Adult , Aged , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neurofibromatosis 2/diagnostic imaging , Neurofibromatosis 2/metabolism , Octreotide/therapeutic use , Retrospective Studies
7.
J Neurooncol ; 146(2): 265-273, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31897926

ABSTRACT

BACKGROUND: Neurofibromatosis type 2 (NF2) patients uniformly develop multiple schwannomas. The tumor-microenvironment (TME) is associated with hypoxia and consists of immunosuppressive cells, including regulatory T cells (Tregs) and tumor-associated macrophages (TAMs). The hypoxic TME of NF2 schwannomas remains unclear. In addition, no comparative study has investigated immunosuppressive cells in NF2 and sporadic schwannomas. METHODS: In 22 NF2 and 21 sporadic schwannomas, we analyzed the immunohistochemistry for Ki-67, hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor receptor 1 (VEGFR1) and VEGFR2, platelet derived growth factor receptor-beta (PDGFR-ß), programmed cell death-1 (PD-1)/ programmed cell death ligand-1 (PD-L1), Foxp3, CD163, CD3, and CD8 to assess the immunosuppressive TME. RESULTS: Most vessels in sporadic schwannomas exhibited slight or negative VEGFR1 and 2 expressions with pericytes coverage. In contrast, large vessels in NF2 schwannomas exhibited strong VEGFR1 and 2 expressions without pericytes. The number of CD3+, CD8+, and CD163+ cells was significantly higher in NF2 schwannomas than in sporadic ones. The expression of PD-L1 and nestin positive cell ratio was higher in NF2 schwannomas than that in sporadic ones. The number of CD163+ cells, nestin positive cell ratio, and HIF-1α expression were significantly associated with shorter progression-free survival in NF2 schwannomas. CONCLUSIONS: This study presents the clinicopathological features of the differences in immunosuppressive cells and the expression of immune checkpoint molecules between NF2 and sporadic schwannomas. Hypoxic TME was first detected in NF2-schwannomas, which was associated with the tumor progression.


Subject(s)
Biomarkers, Tumor/metabolism , Hypoxia , Immunosuppression Therapy , Neurilemmoma/immunology , Neurofibromatosis 2/immunology , Tumor Microenvironment/immunology , Adolescent , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/metabolism , Female , Humans , Male , Middle Aged , Neurilemmoma/metabolism , Neurilemmoma/pathology , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/pathology , Prognosis , Survival Rate , Tumor Cells, Cultured , Young Adult
8.
Oncogene ; 38(36): 6370-6381, 2019 09.
Article in English | MEDLINE | ID: mdl-31312020

ABSTRACT

Inactivation of the tumor suppressor NF2/merlin underlies neurofibromatosis type 2 (NF2) and some sporadic tumors. Previous studies have established that merlin mediates contact inhibition of proliferation; however, the exact mechanisms remain obscure and multiple pathways have been implicated. We have previously reported that merlin inhibits Ras and Rac activity during contact inhibition, but how merlin regulates Ras activity has remained elusive. Here we demonstrate that merlin can directly interact with both Ras and p120RasGAP (also named RasGAP). While merlin does not increase the catalytic activity of RasGAP, the interactions with Ras and RasGAP may fine-tune Ras signaling. In vivo, loss of RasGAP in Schwann cells, unlike the loss of merlin, failed to promote tumorigenic growth in an orthotopic model. Therefore, modulation of Ras signaling through RasGAP likely contributes to, but is not sufficient to account for, merlin's tumor suppressor activity. Our study provides new insight into the mechanisms of merlin-dependent Ras regulation and may have additional implications for merlin-dependent regulation of other small GTPases.


Subject(s)
Neurofibromin 2/physiology , Proto-Oncogene Proteins p21(ras)/metabolism , ras GTPase-Activating Proteins/metabolism , Animals , Cells, Cultured , GTPase-Activating Proteins/metabolism , Genes, Tumor Suppressor , HEK293 Cells , Humans , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Neurofibromatosis 2/genetics , Neurofibromatosis 2/metabolism , Neurofibromin 2/metabolism , Protein Binding , Signal Transduction/genetics
9.
Endocrinol Metab (Seoul) ; 34(2): 203-212, 2019 06.
Article in English | MEDLINE | ID: mdl-31257748

ABSTRACT

BACKGROUND: We previously reported the frequent neurofibromatosis 2 (NF2) gene mutations in anaplastic thyroid cancers in association with the BRAFV600E mutation. We aimed to investigate the role of NF2 in thyroid cancer with BRAF mutation. METHODS: To identify the function of NF2 in thyroid cancers, we investigated the changes in cell proliferation, colon formation, migration and invasion of thyroid cancer cells (8505C, BHT101, and KTC-1) with BRAFV600E mutation after overexpression and knock-down of NF2. We also examined how cell proliferation changed when NF2 was mutagenized. Human NF2 expression in papillary thyroid carcinoma (PTC) was analyzed using the The Cancer Genome Atlas (TCGA) data. RESULTS: First, NF2 was overexpressed in 8505C and KTC-1 cells. Compared to control, NF2 overexpressed group of both thyroid cancer cells showed significant inhibition in cell proliferation and colony formation. These results were also confirmed by cell migration and invasion assay. After knock-down of NF2 in 8505C cells, there were no significant changes in cell proliferation and colony formation, compared with the control group. However, after mutagenized S288* and Q470* sites of NF2 gene, the cell proliferation increased compared to NF2 overexpression group. In the analysis of TCGA data, the mRNA expression of NF2 was significantly decreased in PTCs with lateral cervical lymph node (LN) metastasis compared with PTCs without LN metastasis. CONCLUSION: Our study suggests that NF2 might play a role as a tumor suppressor in thyroid cancer with BRAF mutation. More studies are needed to elucidate the mechanism how NF2 acts in thyroid cancer with BRAF mutation.


Subject(s)
Neurofibromatosis 2/metabolism , Neurofibromin 2/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Thyroid Neoplasms/metabolism , Adult , Cell Enlargement , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Female , Gene Knockdown Techniques , Humans , Male , Middle Aged , Neoplasm Invasiveness , Neurofibromatosis 2/genetics , Neurofibromin 2/genetics , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/physiopathology
10.
Dev Cell ; 49(3): 425-443.e9, 2019 05 06.
Article in English | MEDLINE | ID: mdl-31063758

ABSTRACT

Merlin/NF2 is a bona fide tumor suppressor whose mutations underlie inherited tumor syndrome neurofibromatosis type 2 (NF2), as well as various sporadic cancers including kidney cancer. Multiple Merlin/NF2 effector pathways including the Hippo-YAP/TAZ pathway have been identified. However, the molecular mechanisms underpinning the growth and survival of NF2-mutant tumors remain poorly understood. Using an inducible orthotopic kidney tumor model, we demonstrate that YAP/TAZ silencing is sufficient to induce regression of pre-established NF2-deficient tumors. Mechanistically, YAP/TAZ depletion diminishes glycolysis-dependent growth and increases mitochondrial respiration and reactive oxygen species (ROS) buildup, resulting in oxidative-stress-induced cell death when challenged by nutrient stress. Furthermore, we identify lysosome-mediated cAMP-PKA/EPAC-dependent activation of RAF-MEK-ERK signaling as a resistance mechanism to YAP/TAZ inhibition. Finally, unbiased analysis of TCGA primary kidney tumor transcriptomes confirms a positive correlation of a YAP/TAZ signature with glycolysis and inverse correlations with oxidative phosphorylation and lysosomal gene expression, supporting the clinical relevance of our findings.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Neurofibromin 2/deficiency , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Glycolysis , Heterografts , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , MAP Kinase Signaling System , Mice , Mice, SCID , Neurofibromatosis 2/genetics , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/pathology , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Oxidative Phosphorylation , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Trans-Activators , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
11.
Sci Signal ; 12(578)2019 04 23.
Article in English | MEDLINE | ID: mdl-31015291

ABSTRACT

Neurofibromatosis type 2 is an inherited, neoplastic disease associated with schwannomas, meningiomas, and ependymomas and that is caused by inactivation of the tumor suppressor gene NF2 The NF2 gene product, Merlin, has no intrinsic catalytic activity; its tumor suppressor function is mediated through the proteins with which it interacts. We used proximity biotinylation followed by mass spectrometry and direct binding assays to identify proteins that associated with wild-type and various mutant forms of Merlin in immortalized Schwann cells. We defined a set of 52 proteins in close proximity to wild-type Merlin. Most of the Merlin-proximal proteins were components of cell junctional signaling complexes, suggesting that additional potential interaction partners may exist in adherens junctions, tight junctions, and focal adhesions. With mutant forms of Merlin that cannot bind to phosphatidylinositol 4,5-bisphosphate (PIP2) or that constitutively adopt a closed conformation, we confirmed a critical role for PIP2 binding in Merlin function and identified a large cohort of proteins that specifically interacted with Merlin in the closed conformation. Among these proteins, we identified a previously unreported Merlin-binding protein, apoptosis-stimulated p53 protein 2 (ASPP2, also called Tp53bp2), that bound to closed-conformation Merlin predominately through the FERM domain. Our results demonstrate that Merlin is a component of cell junctional mechanosensing complexes and defines a specific set of proteins through which it acts.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Intercellular Junctions/metabolism , Mechanotransduction, Cellular , Schwann Cells/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Biotinylation , Cell Line, Transformed , Humans , Intercellular Junctions/genetics , Mice , Neurofibromatosis 2/genetics , Neurofibromatosis 2/metabolism , Phosphatidylinositol 4,5-Diphosphate/genetics , Phosphatidylinositol 4,5-Diphosphate/metabolism , Protein Domains , Tumor Suppressor Proteins/genetics
12.
Am J Med Genet A ; 179(6): 1098-1106, 2019 06.
Article in English | MEDLINE | ID: mdl-30908866

ABSTRACT

The neurofibromatoses are inherited, tumor suppressor disorders that are characterized by multiple, benign peripheral nerve sheath tumors and other nervous system tumors. Each disease is associated with a distinct genetic mutation and with a different pathogenesis and clinical course. Neurofibromatosis 1 (NF1) is common and epitomized by multiple neurofibromas with widespread complications. NF2 and schwannomatosis are rare diseases that are typified by multiple schwannomas that are particularly painful in people with schwannomatosis. Since 1985, the Children's Tumor Foundation (formerly the National Neurofibromatosis Foundation) has hosted an international Neurofibromatosis Conference, bringing together international participants who are focused on NF research and clinical care. The 2017 Conference, held in Washington, DC, was among the largest gatherings of NF researchers to date and included presentations from clinicians and basic scientists, highlighting new data regarding the molecular and cellular mechanisms underlying each of these diseases as well as results from clinical studies and clinical trials. This article summarizes the findings presented at the meeting and represents the current state-of-the art for NF research.


Subject(s)
Neurilemmoma/etiology , Neurofibromatoses/etiology , Neurofibromatosis 1/etiology , Neurofibromatosis 2/etiology , Skin Neoplasms/etiology , Animals , Disease Susceptibility , Humans , Neurilemmoma/diagnosis , Neurilemmoma/metabolism , Neurilemmoma/therapy , Neurofibromatoses/diagnosis , Neurofibromatoses/metabolism , Neurofibromatoses/therapy , Neurofibromatosis 1/diagnosis , Neurofibromatosis 1/metabolism , Neurofibromatosis 1/therapy , Neurofibromatosis 2/diagnosis , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/therapy , Skin Neoplasms/diagnosis , Skin Neoplasms/metabolism , Skin Neoplasms/therapy
14.
J Cancer Res Clin Oncol ; 145(2): 523-533, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30603902

ABSTRACT

PURPOSE: There is no targeted drug therapy for NF2 patients, and surgery or radiosurgery is not always effective. Therefore, the exploration of new therapeutic pathways is urgently needed. METHODS: We analyzed the expression of cytokines in the serum of NF2 patients and determined the percentage of HLA-DR-CD33+CD11b+ cells in blood and NF2-associated schwannomas. Furthermore, we analyzed the role of HLA-DR-CD33+CD11b+ cells in inhibiting T-cell proliferation, cytokine production, and transforming growth factor expression. RESULTS: NF2 patients are in an immunosuppressed state with elevated IL-10 and TGF-ß expression in plasma and the lymphocytes from NF2 patients secrete less IFN-γ and CD3+ T cells proliferate slower than normal healthy donors. HLA-DR-CD33+CD11b+ cells frequency significantly increased in the PBMCs and infiltrated in the tumor, these cells express higher iNOS, NOX2 and TGF-ß, and induce TGF-ß secretion to inhibit CD8+ T-cell proliferation, and induce T-cell transformation to a CD4+CD25+Foxp3+ regulatory T cells phenotype. NF2-associated schwannoma cells induced monocytes transformation into an HLA-DR-CD33+CD11b+ phenotype, and surgical removal of the tumor reduced the percentage of these cells. CONCLUSIONS: HLA-DR-CD33+CD11b+ cells may represent a population of MDSCs in NF2 patients. Dissecting the mechanisms behind these suppressive mechanisms will be helpful for the design of effective immunotherapeutic protocols and likely provide a new effective treatment for NF2 patients.


Subject(s)
Biomarkers, Tumor/metabolism , Immunosuppressive Agents , Myeloid-Derived Suppressor Cells/immunology , Neurofibromatosis 2/immunology , T-Lymphocytes/immunology , Adult , Case-Control Studies , Cell Proliferation , Cytokines/metabolism , Female , Follow-Up Studies , Humans , Lymphocyte Activation , Male , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/pathology , Prognosis , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
15.
Arch Pathol Lab Med ; 142(12): 1549-1553, 2018 12.
Article in English | MEDLINE | ID: mdl-30059257

ABSTRACT

CONTEXT.­: The separation of reactive from malignant mesothelial proliferations is often a difficult morphologic problem. There is contradictory information in the literature on whether methylthioadenosine phosphorylase (MTAP) immunohistochemistry can be used for this purpose. OBJECTIVE.­: To determine the utility of MTAP immunohistochemistry in distinguishing reactive from malignant mesothelial proliferations. DESIGN.­: We stained a tissue microarray containing 20 epithelioid malignant mesotheliomas and 17 reactive mesothelial proliferations. For the mesotheliomas, comparisons were made between MTAP staining and BRCA-associated nuclear protein 1 (BAP1) immunohistochemistry, cyclin-dependent kinase inhibitor 2A ( CDKN2A) fluorescence in situ hybridization, and neurofibromin 2 ( NF2) fluorescence in situ hybridization, which are established techniques for making this separation. RESULTS.­: Loss of MTAP was seen in 0 of 17 reactive mesothelial proliferations and 13/20 (65%) malignant mesotheliomas. Almost all cases with loss showed loss in 100% of mesothelial cells. Background inflammatory and stromal cells served as a positive internal control. CDKN2A fluorescence in situ hybridization on the mesotheliomas showed concordance with MTAP staining in 14 of 17 evaluable cases. BAP1 immunohistochemistry showed loss of nuclear staining in 11 of 20 mesotheliomas (55%). No cases showed loss of NF2. A total of 18 of 20 mesotheliomas (90%) showed loss of either MTAP or BAP1. CONCLUSIONS.­: In the context of a mesothelial proliferation, loss of MTAP staining is 100% specific for malignant mesothelioma. In this study the combination of MTAP and BAP1 immunohistochemical staining allowed separation of reactive from epithelial malignant mesothelial proliferations in 90% of cases.


Subject(s)
Biomarkers, Tumor/metabolism , Lung Neoplasms/diagnosis , Mesothelioma/diagnosis , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Epithelioid Cells/metabolism , Epithelium/metabolism , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Mesothelioma, Malignant , Neurofibromatosis 2/metabolism , Purine-Nucleoside Phosphorylase/metabolism , Tissue Array Analysis , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism
16.
PLoS One ; 13(5): e0196726, 2018.
Article in English | MEDLINE | ID: mdl-29715273

ABSTRACT

The neurofibromatosis type 2 (NF2) tumor suppressor protein Merlin functions as a negative regulator of cell growth and actin dynamics in different cell types amongst which Schwann cells have been extensively studied. In contrast, the presence and the role of Merlin in oligodendrocytes, the myelin forming cells within the CNS, have not been elucidated. In this work, we demonstrate that Merlin immunoreactivity was broadly distributed in the white matter throughout the central nervous system. Following Merlin expression during development in the cerebellum, Merlin could be detected in the cerebellar white matter tract at early postnatal stages as shown by its co-localization with Olig2-positive cells as well as in adult brain sections where it was aligned with myelin basic protein containing fibers. This suggests that Merlin is expressed in immature and mature oligodendrocytes. Expression levels of Merlin were low in oligodendrocytes as compared to astrocytes and neurons throughout development. Expression of Merlin in oligodendroglia was further supported by its identification in either immortalized cell lines of oligodendroglial origin or in primary oligodendrocyte cultures. In these cultures, the two main splice variants of Nf2 could be detected. Merlin was localized in clusters within the nuclei and in the cytoplasm. Overexpressing Merlin in oligodendrocyte cell lines strengthened reduced impedance in XCELLigence measurements and Ki67 stainings in cultures over time. In addition, the initiation and elongation of cellular projections were reduced by Merlin overexpression. Consistently, cell migration was retarded in scratch assays done on Nf2-transfected oligodendrocyte cell lines. These data suggest that Merlin actively modulates process outgrowth and migration in oligodendrocytes.


Subject(s)
Cell Proliferation/physiology , Neurofibromatosis 2/metabolism , Neurofibromin 2/metabolism , Oligodendroglia/metabolism , Animals , Astrocytes/metabolism , Cell Line , Cell Movement/physiology , Cell Nucleus/metabolism , Central Nervous System/metabolism , Cytoplasm/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Neurons/metabolism , Schwann Cells/metabolism , Transfection/methods
17.
Methods Mol Biol ; 1739: 161-173, 2018.
Article in English | MEDLINE | ID: mdl-29546707

ABSTRACT

Schwannomas are benign nerve tumors that occur sporadically in the general population and in those with neurofibromatosis type 2 (NF2), a tumor predisposition genetic disorder. NF2-associated schwannomas and most sporadic schwannomas are caused by inactivating mutations in Schwann cells in the neurofibromatosis type 2 gene (NF2) that encodes the merlin tumor suppressor. Despite their benign nature, schwannomas and especially vestibular schwannomas cause considerable morbidity. The primary available therapies are surgery or radiosurgery which usually lead to loss of function of the compromised nerve. Thus, there is a need for effective chemotherapies. We established an untransformed merlin-deficient human Schwann cell line for use in drug discovery studies for NF2-associated schwannomas. We describe the generation of human Schwann cells (HSCs) with depletion of merlin and their application in high-throughput screening of chemical libraries to identify compounds that decrease their viability. This NF2-HSC model is amenable for use in independent labs and high-throughput screening (HTS) facilities.


Subject(s)
Neurofibromin 2/metabolism , Schwann Cells/cytology , Cell Culture Techniques/methods , Cell Survival/genetics , Cells, Cultured , Drug Discovery , Humans , Neurilemmoma/metabolism , Neurofibromatosis 2/metabolism , Neurofibromin 2/deficiency , Neurofibromin 2/genetics , Schwann Cells/metabolism , Small Molecule Libraries
18.
Neurochirurgie ; 64(5): 335-341, 2018 Nov.
Article in English | MEDLINE | ID: mdl-26073919

ABSTRACT

OBJECTIVE: Neurofibromatosis type 2 (NF2) affects about one in 25,000 to 40,000 people. Most NF2 patients have private loss-of-function mutations scattered along the NF2 gene. Here, we present our NF2 investigation strategy. MATERIAL AND METHODS: We report a comprehensive NF2 mutation analysis of 221 NF2 French patients: 134 unrelated typical NF2 patients fulfilling the Manchester criteria and 87 unrelated patients presenting symptoms that partially fulfilled the Manchester criteria. RESULTS: A NF2 mutation was identified in 56 of the 221 patients, giving a global mutation detection rate of 25%. This rate reached 37% (49/134) for typical NF2 patients fulfilling the Manchester criteria and only 8% (7/87) for patients presenting symptoms suggestive of NF2. Six of these seven patients were under 25 of age. Our approach showed that 77% of NF2 identified variants were detected by coding exons sequencing. Multiplex ligation-dependent probe amplification allowed the identification of restricted rearrangements (23% of NF2 identified variants corresponding to complete deletion or partial deletion/duplication of NF2). CONCLUSION: High mutation detection rate can be achieved if well phenotyped NF2 patients are studied with multiple complementary and optimized techniques. NF2 somatic mosaicism detection was improved by frozen tumor samples molecular analysis.


Subject(s)
Genes, Neurofibromatosis 2/physiology , Mutation/genetics , Neoplasms/diagnosis , Neurofibromatosis 2/genetics , Neurofibromatosis 2/metabolism , Adult , Cohort Studies , DNA Mutational Analysis/methods , Female , Humans , Male , Neoplasms/genetics , Neoplasms/metabolism , Neurofibromatosis 2/diagnosis , Pathology, Molecular
19.
Neurol Res ; 39(12): 1086-1093, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28934903

ABSTRACT

Objective To study the function and potential mechanism of RAC1 inhibitors in NF2-associated schwannoma. Methods In this study, we the downregulation of RAC1 activity and tumor cell phenotypes by RAC1 inhibitor NSC23766 in vitro. And we further validated the anti-proliferation effect by this RAC1 inhibitor in subcutaneous xenograft tumor model and sciatic nerve model. Results Pharmacological inhibition of RAC1 could significantly inhibit the proliferation of both RT4 cells and human NF2-associated primary schwannoma cells by inducing apoptosis. Pharmacological inhibition of RAC1 effectively reduced Rac1 activity and down-regulated the pathway downstream of Rac. Moreover, pharmacological inhibition of RAC1 showed a potential antitumor effect, with low toxicity in vivo. Conclusion RAC1 inhibitors may play a therapeutic role in patients with schwannoma.


Subject(s)
Apoptosis/physiology , Cell Proliferation/physiology , Neurilemmoma/metabolism , Neurofibromatosis 2/metabolism , rac1 GTP-Binding Protein/metabolism , Aminoquinolines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Enzyme Inhibitors/pharmacology , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neurilemmoma/drug therapy , Neurilemmoma/pathology , Neurofibromatosis 2/drug therapy , Neurofibromatosis 2/pathology , Peripheral Nervous System Neoplasms/drug therapy , Peripheral Nervous System Neoplasms/metabolism , Peripheral Nervous System Neoplasms/pathology , Pyrimidines/pharmacology , Random Allocation , Rats , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , rac1 GTP-Binding Protein/antagonists & inhibitors
20.
J Exp Clin Cancer Res ; 36(1): 100, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28764788

ABSTRACT

BACKGROUND: MiRNAs are involved in aberrant DNA methylation through regulation of DNA methyltransferases (DNMTs) in the pathogenesis and progression of glioblastomas (GBM). MiR-152-3p was down-expressed in human malignancies, and served as a tumor suppressor. Neurofibromatosis type 2 (NF2) was significantly decreased in GBM tissues with a high level of methylation. However, the link between miR-152-3p, DNMT1 and methylation of NF2 in GBM is not clearly established. This study was conducted to detect the mechanism between miR-152-3p, DNMT1 and NF2 in GBM. METHODS: The levels of DNMT1 and NF2 expression were studied by qRT-PCR, Western blot, immunofluorescence, and immumohistochemical staining. Methylation in the promoter region of NF2 was detected by methylation-specific PCR and bisulfate genomic sequencing PCR. Cell proliferation was examined by Cell-Counting Kit-8 and 5-ethynyl-2'-deoxyuridine assay, and cell invasion was evaluated by transwell assay. Flow cytomery and Hoechst staining were used to analyze cell apoptosis. A dual luciferase system was used to confirm the relationship between miR-152-3p and DNMT1. RESULTS: Methylation of NF2 and DNMT1 was markedly increased, and miR-152-3p was downregulated in GBM tissues and glioma cells. Both knockdown of DNMT1 and overexpression miR-152-3p showed that demethylation activated the expression of NF2. Furthermore, miR-152-3p directly targeted DNMT1. Both miR-152-3p overexpression and DNMT1 knockdown significantly induced cell apoptosis and inhibited invasive activity. This was also observed after NF2 overexpression. CONCLUSIONS: These results indicated that miR-152-3p can inhibit glioma cell proliferation and invasion activities by decreasing DNMT1. The restoration of miR-152-3p may have therapeutic application in the treatment of GBM.


Subject(s)
Brain Neoplasms/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Glioblastoma/genetics , MicroRNAs/genetics , Neurofibromatosis 2/genetics , Apoptosis/physiology , Brain Neoplasms/enzymology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Glioblastoma/enzymology , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , MicroRNAs/metabolism , Neoplasm Invasiveness , Neurofibromatosis 2/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...