Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
Add more filters

Country/Region as subject
Publication year range
1.
FASEB J ; 38(13): e23809, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38967126

ABSTRACT

The neurofibromatosis type 2 (NF2) gene, known for encoding the tumor suppressor protein Merlin, is central to the study of tumorigenesis and associated cellular processes. This review comprehensively examines the multifaceted role of NF2/Merlin, detailing its structural characteristics, functional diversity, and involvement in various signaling pathways such as Wnt/ß-catenin, Hippo, TGF-ß, RTKs, mTOR, Notch, and Hedgehog. These pathways are crucial for cellular growth, proliferation, and differentiation. NF2 mutations are specifically linked to the development of schwannomas, meningiomas, and ependymomas, although the precise mechanisms of tumor formation in these specific cell types remain unclear. Additionally, the review explores Merlin's role in embryogenesis, highlighting the severe developmental defects and embryonic lethality caused by NF2 deficiency. The potential therapeutic strategies targeting these genetic aberrations are also discussed, emphasizing inhibitors of mTOR, HDAC, and VEGF as promising avenues for treatment. This synthesis of current knowledge underscores the necessity for ongoing research to elucidate the detailed mechanisms of NF2/Merlin and develop effective therapeutic strategies, ultimately aiming to improve the prognosis and quality of life for individuals with NF2 mutations.


Subject(s)
Carcinogenesis , Neurofibromin 2 , Humans , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Neurofibromatosis 2/genetics , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/pathology , Signal Transduction , Mutation
2.
Int J Mol Sci ; 25(12)2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38928264

ABSTRACT

NF2-related schwannomatosis (NF2) is a genetic syndrome characterized by the growth of benign tumors in the nervous system, particularly bilateral vestibular schwannomas, meningiomas, and ependymomas. This review consolidates the current knowledge on NF2 syndrome, emphasizing the molecular pathology associated with the mutations in the gene of the same name, the NF2 gene, and the subsequent dysfunction of its product, the Merlin protein. Merlin, a tumor suppressor, integrates multiple signaling pathways that regulate cell contact, proliferation, and motility, thereby influencing tumor growth. The loss of Merlin disrupts these pathways, leading to tumorigenesis. We discuss the roles of another two proteins potentially associated with NF2 deficiency as well as Merlin: Yes-associated protein 1 (YAP), which may promote tumor growth, and Raf kinase inhibitory protein (RKIP), which appears to suppress tumor development. Additionally, this review discusses the efficacy of various treatments, such as molecular therapies that target specific pathways or inhibit neomorphic protein-protein interaction caused by NF2 deficiency. This overview not only expands on the fundamental understanding of NF2 pathophysiology but also explores the potential of novel therapeutic targets that affect the clinical approach to NF2 syndrome.


Subject(s)
Neurilemmoma , Neurofibromatoses , Neurofibromin 2 , Skin Neoplasms , Humans , Neurofibromatoses/therapy , Neurofibromatoses/genetics , Neurofibromatoses/metabolism , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Neurilemmoma/genetics , Neurilemmoma/therapy , Neurilemmoma/metabolism , Neurilemmoma/pathology , Skin Neoplasms/genetics , Skin Neoplasms/therapy , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Animals , Neurofibromatosis 2/genetics , Neurofibromatosis 2/therapy , Neurofibromatosis 2/metabolism , Mutation , Signal Transduction , Molecular Targeted Therapy
3.
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
4.
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
5.
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
6.
Lab Invest ; 97(7): 843-853, 2017 07.
Article in English | MEDLINE | ID: mdl-28581486

ABSTRACT

The Hippo signaling pathway and the Notch signaling pathway are evolutionary conserved signaling cascades that have important roles in embryonic development of many organs. In murine liver, disruption of either pathway impairs intrahepatic bile duct development. Recent studies suggested that the Notch signaling receptor Notch2 is a direct transcriptional target of the Hippo signaling pathway effector YAP, and the Notch signaling is a major mediator of the Hippo signaling in maintaining biliary cell characteristics in adult mice. However, it remains to be determined whether the Hippo signaling pathway functions through the Notch signaling in intrahepatic bile duct development. We found that loss of the Hippo signaling pathway tumor suppressor Nf2 resulted in increased expression levels of the Notch signaling pathway receptor Notch2 in cholangiocytes but not in hepatocytes. When knocking down Notch2 on the background of Nf2 deficiency in mouse livers, the excessive bile duct development induced by Nf2 deficiency was suppressed by heterozygous and homozygous deletion of Notch2 in a dose-dependent manner. These results implicated that Notch signaling is one of the downstream effectors of the Hippo signaling pathway in regulating intrahepatic bile duct development.


Subject(s)
Bile Ducts, Intrahepatic/growth & development , Neurofibromatosis 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, Notch2/metabolism , Animals , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Female , Hippo Signaling Pathway , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurofibromatosis 2/genetics , Protein Serine-Threonine Kinases/genetics , Receptor, Notch2/genetics
7.
Development ; 141(21): 4182-93, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25336744

ABSTRACT

The corpus callosum connects cerebral hemispheres and is the largest axon tract in the mammalian brain. Callosal malformations are among the most common congenital brain anomalies and are associated with a wide range of neuropsychological deficits. Crossing of the midline by callosal axons relies on a proper midline environment that harbors guidepost cells emitting guidance cues to instruct callosal axon navigation. Little is known about what controls the formation of the midline environment. We find that two components of the Hippo pathway, the tumor suppressor Nf2 (Merlin) and the transcriptional coactivator Yap (Yap1), regulate guidepost development and expression of the guidance cue Slit2 in mouse. During normal brain development, Nf2 suppresses Yap activity in neural progenitor cells to promote guidepost cell differentiation and prevent ectopic Slit2 expression. Loss of Nf2 causes malformation of midline guideposts and Slit2 upregulation, resulting in callosal agenesis. Slit2 heterozygosity and Yap deletion both restore callosal formation in Nf2 mutants. Furthermore, selectively elevating Yap activity in midline neural progenitors is sufficient to disrupt guidepost formation, upregulate Slit2 and prevent midline crossing. The Hippo pathway is known for its role in controlling organ growth and tumorigenesis. Our study identifies a novel role of this pathway in axon guidance. Moreover, by linking axon pathfinding and neural progenitor behaviors, our results provide an example of the intricate coordination between growth and wiring during brain development.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Corpus Callosum/embryology , Neurofibromatosis 2/metabolism , Phosphoproteins/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Corpus Callosum/metabolism , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Female , Limbic Lobe/embryology , Limbic Lobe/metabolism , Mice , Nervous System , Neurofibromatosis 2/genetics , Phosphoproteins/genetics , Pregnancy , Transcription Factors/genetics , YAP-Signaling Proteins
8.
Neurochem Res ; 42(8): 2363-2371, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28397069

ABSTRACT

Lithium is considered a first-line therapy for the treatment of bipolar disorder and was recently shown to be associated with a reduced overall cancer risk. A growing body of evidence has indicated the potential antitumor benefits of this drug. Lithium likely functions as an antitumor agent. In this study, we found that lithium chloride (LiCl) significantly inhibits the proliferation of both RT4 cells and human NF2-associated primary schwannoma cells by inhibiting the expression of apoptosis-related proteins. LiCl-induced cell death exhibits ultrastructural features of necrosis and is reversed by the RIPK1-specific inhibitor necrostatin-1 in a dose-dependent manner, indicating that LiCl induces the necroptosis type of cell death. Moreover, LiCl treatment induces ROS generation and activates the AKT/mTOR pathway, which is reversed by necrostatin-1 treatment. Based on our results, LiCl treatment may induce the programmed cell death of schwannoma cells through AKT- and mTOR-mediated necroptosis, potentially representing a new mechanism by which LiCl induces tumor cell death. Moreover, LiCl may prove to be a new drug for treating schwannoma.


Subject(s)
Apoptosis/physiology , Lithium Chloride/pharmacology , Neurilemmoma/metabolism , Neurofibromatosis 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Humans , Lithium Chloride/therapeutic use , Necrosis/metabolism , Neurilemmoma/drug therapy , Neurilemmoma/pathology , Neurofibromatosis 2/drug therapy , Neurofibromatosis 2/pathology , Rats
9.
HNO ; 65(5): 362-372, 2017 May.
Article in German | MEDLINE | ID: mdl-27421984

ABSTRACT

Schwannomas are benign Schwann cell-derived tumors of the peripheral nerve sheath often involving the vestibular cranial nerve (vestibular schwannoma). Histologically, they consist of bipolar spindle cells and show a moderate cellularity. Typically, Antoni A regions with a storiform pattern and loose Antoni B regions are intermingled. Verocay bodies are the pathognomonic palisading structures. Malignant transformation is rare. Merlin (schwannomin), the protein product of NF2, is inactivated by mutations, loss of heterozygosity or methylation. Within neurofibromatosis type 2, a germline mutation is present in about half of cases, whereas tumors demonstrate an additional second hit of the NF2 gene. A loss of chromosome 22 or 22q is common. Merlin links the cell membrane with the cytoskeleton and regulates intracellular signaling pathways leading to dysorganization when merlin is inactivated. Loss of merlin activates Rac1 and Ras, and the PAK1, mTORC1, EGFR-Ras-ERK, PI3K-Akt, WNT and Hippo pathways as well as receptor tyrosine kinases. Furthermore, merlin locates to the nucleus and inhibits E3 ubiquitin ligase CRL4DCAF1. Besides biallelic inactivation of NF2 in schwannomas, other genes are involved in the pathogenesis of schwannomatosis-associated schwannomas such as LZTR1, SMARCB1, COQ6 indicating an important role of SWI/SNF chromatin-remodeling complex for schwannoma development. Our own investigations point to deregulation of BAF170, another essential SWI/SNF complex component. Knowledge of mechanisms allows targeted molecular therapy, especially in vestibular schwannomas, using antagonists against mTOR (rapamycin/sirolmus/everolimus), EGFR (lapatinib) or VEGF (bevacizumab), although clinical studies have been in part disappointing so far.


Subject(s)
Biomarkers, Tumor/metabolism , Neurofibromatosis 2/metabolism , Neurofibromin 2/metabolism , Neuroma, Acoustic/metabolism , Neuroma, Acoustic/pathology , Gene Expression Regulation, Neoplastic , Humans
10.
Mod Pathol ; 29(1): 14-24, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26493618

ABSTRACT

Cytoreductive surgery and hyperthermic intraperitoneal chemoperfusion for patients with malignant peritoneal mesothelioma has resulted in improved disease control and increased survival. Despite these results, there are significant perioperative risks associated with this aggressive procedure that necessitate consideration of prognostic markers during patient selection. The molecular pathogenesis of peritoneal mesothelioma remains relatively unknown, but extrapolation of findings from their pleural counterpart would suggest frequent alterations in CDKN2A, NF2, and BAP1. Homozygous deletions in CDKN2A portend a worse overall survival in peritoneal mesothelioma. However, the prevalence and prognostic significance of NF2 and BAP1 abnormalities has not been studied. Dual-color fluorescence in situ hybridization using CDKN2A and NF2 locus-specific probes and BAP1 immunohistochemistry identified homozygous CDKN2A deletions (n=25, 29%), hemizygous NF2 loss (n=30, 35%), and/or loss of BAP1 protein expression (n=49, 57%) in 68 of 86 (79%) peritoneal mesotheliomas. Homozygous CDKN2A deletions or hemizygous NF2 loss correlated with shorter progression-free survival (P<0.02) and poor overall survival (P<0.03). Moreover, the significance of these findings was cumulative. Patients harboring both homozygous CDKN2A deletions and hemizygous NF2 loss had a 2-year progression-free survival rate of 9% with a median of 6 months (P<0.01) and overall survival rate of 18% with a median of 8 months (P<0.01). By multivariate analysis, combined homozygous CDKN2A deletions and hemizygous NF2 loss was a negative prognostic factor for both progression-free survival and overall survival, independent of patient age, peritoneal cancer index, completeness of cytoreduction, and extent of invasion. In contrast, loss of BAP1 was not associated with clinical outcome. In summary, homozygous deletions in CDKN2A and hemizygous loss of NF2 as detected by fluorescence in situ hybridization would confer a poor clinical outcome and may guide future treatment decisions for patients with peritoneal mesothelioma.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Lung Neoplasms/metabolism , Mesothelioma/metabolism , Neurofibromatosis 2/metabolism , Peritoneal Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Mesothelioma/mortality , Mesothelioma/pathology , Mesothelioma, Malignant , Middle Aged , Peritoneal Neoplasms/mortality , Peritoneal Neoplasms/pathology , Prognosis , Survival Rate , Young Adult
11.
Hum Genomics ; 8: 10, 2014 Jun 30.
Article in English | MEDLINE | ID: mdl-24980480

ABSTRACT

BACKGROUND: Schwannomas are the most common neurofibromatosis type 2 (NF2)-associated tumors with significant phenotypic heterogeneity in patients. The most severe subtype has an early and rapid progression and the mild type has a later onset and a less aggressive course. The aim of this study was to elucidate the underlying molecular differences between these groups. We compared the gene expression pattern between patients with early to late age of onset. RESULTS: A gene signature of 21 genes was constructed to differentiate between early-onset and late-onset patients. We confirmed these results by real-time PCR for SNF1LK2, NGFRAP1L1 (BEX 5), GMNN, and EPHA2. CONCLUSION: Genes identified here may be additional aberrations in merlin-depleted cells that govern the disease onset. A significant number of these genes have been suggested as having a role in carcinogenesis and are used as biomarkers for prognosis in several other cancers. The role of these genes in NF2 carcinogenesis and their potential as biomarkers or drug target are worthwhile exploring.


Subject(s)
Biomarkers, Tumor/metabolism , Neurofibromatosis 2/metabolism , Transcriptome , Adolescent , Adult , Age Factors , Aged , Biomarkers, Tumor/genetics , Child , Gene Expression Profiling , Humans , Middle Aged , Molecular Targeted Therapy , Neurofibromatosis 2/diagnosis , Neurofibromatosis 2/genetics
12.
J Neurooncol ; 122(3): 451-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25672645

ABSTRACT

Brain-invasive growth of a subset of meningiomas is associated with less favorable prognosis. The molecular mechanisms causing invasiveness are only partially understood, however, the expression of matrix metalloproteinases (MMPs) has been identified as a contributing factor. We have previously found that loss of density enhanced phosphatase-1 (DEP-1, also designated PTPRJ), a transmembrane protein-tyrosine phosphatase, promotes meningioma cell motility and invasive growth in an orthotopic xenotransplantation model. We have now analyzed potential alterations of the expression of genes involved in motility control, caused by DEP-1 loss in meningioma cell lines. DEP-1 depleted cells exhibited increased expression of mRNA encoding MMP-9, and the growth factors EGF and FGF-2. The increase of MMP-9 expression in DEP-1 depleted cells was also readily detectable at the protein level by zymography. MMP-9 upregulation was sensitive to chemical inhibitors of growth factor signal transduction. Conversely, MMP-9 mRNA levels could be stimulated with growth factors (e.g. EGF) and inflammatory cytokines (e.g. TNFα). Increase of MMP-9 expression by DEP-1 depletion, or growth factor/cytokine stimulation qualitatively correlated with increased invasiveness in vitro scored as transmigration through matrigel-coated membranes. The studies suggest induction of MMP-9 expression promoted by DEP-1 deficiency, or potentially by growth factors and inflammatory cytokines, as a mechanism contributing to meningioma brain invasiveness.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Glioma/metabolism , Matrix Metalloproteinase 9/metabolism , Analysis of Variance , Cell Line, Tumor , Cytokines/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Glioma/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Neurofibromatosis 2/genetics , Neurofibromatosis 2/metabolism , RNA, Messenger/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/deficiency , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics , Transfection
13.
Brain ; 137(Pt 2): 420-32, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24309211

ABSTRACT

Axonal surface proteins encompass a group of heterogeneous molecules, which exert a variety of different functions in the highly interdependent relationship between axons and Schwann cells. We recently revealed that the tumour suppressor protein merlin, mutated in the hereditary tumour syndrome neurofibromatosis type 2, impacts significantly on axon structure maintenance in the peripheral nervous system. We now report on a role of neuronal merlin in the regulation of the axonal surface protein neuregulin 1 important for modulating Schwann cell differentiation and myelination. Specifically, neuregulin 1 type III expression is reduced in sciatic nerve tissue of neuron-specific knockout animals as well as in biopsies from seven patients with neurofibromatosis type 2. In vitro experiments performed on both the P19 neuronal cell line and primary dorsal root ganglion cells demonstrate the influence of merlin on neuregulin 1 type III expression. Moreover, expression of ERBB2, a Schwann cell receptor for neuregulin 1 ligands is increased in nerve tissue of both neuron-specific merlin knockout animals and patients with neurofibromatosis type 2, demonstrating for the first time that axonal merlin indirectly regulates Schwann cell behaviour. Collectively, we have identified that neuronally expressed merlin can influence Schwann cell activity in a cell-extrinsic manner.


Subject(s)
Neuregulin-1/physiology , Neurofibromin 2/physiology , Neurons/physiology , Receptor, ErbB-2/biosynthesis , Schwann Cells/metabolism , Signal Transduction/physiology , Adult , Aged , Animals , Cell Line , Cells, Cultured , Female , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/pathology , Neurons/pathology , Schwann Cells/pathology
14.
Future Oncol ; 11(12): 1751-8, 2015.
Article in English | MEDLINE | ID: mdl-26075443

ABSTRACT

AIM: Neurofibromatosis type 2 (NF2)-associated vestibular schwannomas have variable size at presentation which presents a unique challenge in NF2 patient management. Therefore, we investigated the molecular signature characteristic of the differences in size for improved individualized precise therapy. MATERIALS & METHODS: RNA expression analysis was performed on 15 small and 27 large NF2-associated vestibular schwannoma tumors using a microarray analyzing over 47,000 transcripts. RESULTS: A signature of 11 genes was found to be correlated with NF2 tumor size. CONCLUSION: We have identified the genetic hallmark that differentiates large NF2-associated tumors from smaller tumors. This is the first time that these genes have been shown to be the hallmark for NF2 tumor size.


Subject(s)
Neurofibromatosis 2/metabolism , Neuroma, Acoustic/metabolism , Transcriptome , Adolescent , Adult , Female , Humans , Male , Neurofibromatosis 2/genetics , Neurofibromatosis 2/pathology , Neuroma, Acoustic/genetics , Neuroma, Acoustic/pathology , Tumor Burden , Young Adult
15.
Eur Arch Otorhinolaryngol ; 272(12): 3627-33, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25421643

ABSTRACT

The hallmark of neurofibromatosis type 2 (NF2) is bilateral vestibular schwannomas (VS) and severe hearing loss is common in NF2 patients. Vascular endothelial growth factor (VEGF) expression level in NF2 correlates with tumour growth rate and bevacizumab, a VEGF-binding antibody, has previously been shown to induce tumour shrinkage and improve hearing. We retrospectively reviewed the effect of bevacizumab on hearing and VS tumour size in 12 consecutive NF2 patients. Bevacizumab 10 mg/kg was administered intravenously every second week for 6 months; hereafter, bevacizumab 15 mg/kg was administered every third week. Patients were evaluated with repeated audiometries, MR scans and clinical evaluations. Radiological response was defined as a 20 % or greater reduction in VS volume. A total of 398 treatments (median 36) were administered and the median duration on therapy was 22 months (range 7-34). We observed a radiological response (≥20 % tumour shrinkage) in seven out of 18 tumours (39 %) in six out of 12 patients (50 %). Sustained radiological responses were maintained in six tumours (33 %) for more than 2 months. Three patients had objectively improved hearing and five patients reported subjective benefit in neurological symptoms, including improved hearing. Toxicity was in general manageable; however, one patient died from cerebral haemorrhage which was possibly related to therapy. In conclusion, bevacizumab improved hearing and reduced the size of VS in some patients with progressive NF2 which corroborates previous findings; however, the risk of severe side effects should be carefully considered and discussed with the patients prior to treatment.


Subject(s)
Bevacizumab , Cerebral Hemorrhage/etiology , Hearing/drug effects , Neurofibromatosis 2 , Neuroma, Acoustic , Tumor Burden/drug effects , Adult , Aged , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/adverse effects , Audiometry/methods , Bevacizumab/administration & dosage , Bevacizumab/adverse effects , Female , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Neoplasm Staging , Neurofibromatosis 2/drug therapy , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/pathology , Neuroma, Acoustic/drug therapy , Neuroma, Acoustic/metabolism , Neuroma, Acoustic/pathology , Retrospective Studies , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
17.
Brain ; 136(Pt 2): 549-63, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23413263

ABSTRACT

Loss of the Merlin tumour suppressor causes abnormal de-differentiation and proliferation of Schwann cells and formation of schwannoma tumours in patients with neurofibromatosis type 2. Within the mature peripheral nerve the normal development, differentiation and maintenance of myelinating and non-myelinating Schwann cells is regulated by a network of transcription factors that include SOX10, OCT6 (now known as POU3F1), NFATC4 and KROX20 (also known as Egr2). We have examined for the first time how their regulation of Schwann cell development is disrupted in primary human schwannoma cells. We find that induction of both KROX20 and OCT6 is impaired, whereas enforced expression of KROX20 drives both myelin gene expression and cell cycle arrest in Merlin-null cells. Importantly, we show that human schwannoma cells have reduced expression of SOX10 protein and messenger RNA. Analysis of mouse SOX10-null Schwann cells shows they display many of the characteristics of human schwannoma cells, including increased expression of platelet derived growth factor receptor beta (PDGFRB) messenger RNA and protein, enhanced proliferation, increased focal adhesions and schwannoma-like morphology. Correspondingly, reintroduction of SOX10 into human Merlin-null cells restores the ability of these cells to induce KROX20 and myelin protein zero (MPZ), localizes NFATC4 to the nucleus, reduces cell proliferation and suppresses PDGFRB expression. Thus, we propose that loss of the SOX10 protein, which is vital for normal Schwann cell development, is also key to the pathology of Merlin-null schwannoma tumours.


Subject(s)
Gene Knockdown Techniques , Neurilemmoma/genetics , Neurofibromatosis 2/genetics , Neurofibromin 2/deficiency , Phenotype , SOXE Transcription Factors/deficiency , Animals , Cells, Cultured , Humans , Mice , Mice, Transgenic , Neurilemmoma/metabolism , Neurilemmoma/pathology , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/pathology , Neurofibromin 2/genetics , SOXE Transcription Factors/physiology
18.
Proc Natl Acad Sci U S A ; 108(12): 4980-5, 2011 Mar 22.
Article in English | MEDLINE | ID: mdl-21383154

ABSTRACT

Neurofibromatosis type 2 (NF2) is a multiple neoplasia syndrome and is caused by a mutation of the NF2 tumor suppressor gene that encodes for the tumor suppressor protein merlin. Biallelic NF2 gene inactivation results in the development of central nervous system tumors, including schwannomas, meningiomas, ependymomas, and astrocytomas. Although a wide variety of missense germline mutations in the coding sequences of the NF2 gene can cause loss of merlin function, the mechanism of this functional loss is unknown. To gain insight into the mechanisms underlying loss of merlin function in NF2, we investigated mutated merlin homeostasis and function in NF2-associated tumors and cell lines. Quantitative protein and RT-PCR analysis revealed that whereas merlin protein expression was significantly reduced in NF2-associated tumors, mRNA expression levels were unchanged. Transfection of genetic constructs of common NF2 missense mutations into NF2 gene-deficient meningioma cell lines revealed that merlin loss of function is due to a reduction in mutant protein half-life and increased protein degradation. Transfection analysis also demonstrated that recovery of tumor suppressor protein function is possible, indicating that these mutants maintain intrinsic functional capacity. Further, increased expression of mutant protein is possible after treatment with specific proteostasis regulators, implicating protein quality control systems in the degradative fate of mutant tumor suppressor proteins. These findings provide direct insight into protein function and tumorigenesis in NF2 and indicate a unique treatment paradigm for this disorder.


Subject(s)
Central Nervous System Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Genes, Neurofibromatosis 2 , Mutation, Missense , Neurofibromatosis 2/metabolism , Neurofibromin 2/biosynthesis , Cell Line, Tumor , Central Nervous System Neoplasms/genetics , Gene Silencing , Humans , Neurofibromatosis 2/genetics , Neurofibromin 2/genetics , Reverse Transcriptase Polymerase Chain Reaction
19.
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
20.
Nat Commun ; 15(1): 5115, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879607

ABSTRACT

Neurofibromatosis Type II (NFII) is a genetic condition caused by loss of the NF2 gene, resulting in activation of the YAP/TAZ pathway and recurrent Schwann cell tumors, as well as meningiomas and ependymomas. Unfortunately, few pharmacological options are available for NFII. Here, we undertake a genome-wide CRISPR/Cas9 screen to search for synthetic-lethal genes that, when inhibited, cause death of NF2 mutant Schwann cells but not NF2 wildtype cells. We identify ACSL3 and G6PD as two synthetic-lethal partners for NF2, both involved in lipid biogenesis and cellular redox. We find that NF2 mutant Schwann cells are more oxidized than control cells, in part due to reduced expression of genes involved in NADPH generation such as ME1. Since G6PD and ME1 redundantly generate cytosolic NADPH, lack of either one is compatible with cell viability, but not down-regulation of both. Since genetic deficiency for G6PD is tolerated in the human population, G6PD could be a good pharmacological target for NFII.


Subject(s)
CRISPR-Cas Systems , Coenzyme A Ligases , Glucosephosphate Dehydrogenase , Neurofibromin 2 , Schwann Cells , Synthetic Lethal Mutations , Schwann Cells/metabolism , Humans , Glucosephosphate Dehydrogenase/metabolism , Glucosephosphate Dehydrogenase/genetics , Neurofibromin 2/metabolism , Neurofibromin 2/genetics , Coenzyme A Ligases/metabolism , Coenzyme A Ligases/genetics , Animals , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/genetics , NADP/metabolism , Mice , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL