Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 393
Filter
1.
Nat Commun ; 15(1): 7873, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251601

ABSTRACT

Meningiomas are associated with inactivation of NF2/Merlin, but approximately one-third of meningiomas with favorable clinical outcomes retain Merlin expression. Biochemical mechanisms underlying Merlin-intact meningioma growth are incompletely understood, and non-invasive biomarkers that may be used to guide treatment de-escalation or imaging surveillance are lacking. Here, we use single-cell RNA sequencing, proximity-labeling proteomic mass spectrometry, mechanistic and functional approaches, and magnetic resonance imaging (MRI) across meningioma xenografts and patients to define biochemical mechanisms and an imaging biomarker that underlie Merlin-intact meningiomas. We find Merlin serine 13 (S13) dephosphorylation drives meningioma Wnt signaling and tumor growth by attenuating inhibitory interactions with ß-catenin and activating the Wnt pathway. MRI analyses show Merlin-intact meningiomas with S13 phosphorylation and favorable clinical outcomes are associated with high apparent diffusion coefficient (ADC). These results define mechanisms underlying a potential imaging biomarker that could be used to guide treatment de-escalation or imaging surveillance for patients with Merlin-intact meningiomas.


Subject(s)
Magnetic Resonance Imaging , Meningeal Neoplasms , Meningioma , Neurofibromin 2 , Wnt Signaling Pathway , Meningioma/diagnostic imaging , Meningioma/metabolism , Meningioma/pathology , Meningioma/genetics , Humans , Phosphorylation , Neurofibromin 2/metabolism , Neurofibromin 2/genetics , Animals , Magnetic Resonance Imaging/methods , Meningeal Neoplasms/diagnostic imaging , Meningeal Neoplasms/metabolism , Meningeal Neoplasms/pathology , Meningeal Neoplasms/genetics , Mice , Cell Line, Tumor , beta Catenin/metabolism , beta Catenin/genetics , Female , Serine/metabolism , Male , Proteomics/methods , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics
2.
EMBO Rep ; 25(9): 4033-4061, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39160347

ABSTRACT

Cells sense and respond to various mechanical forces from the extracellular matrix primarily by modulating the actin cytoskeleton. Mechanical forces can be translated into biochemical signals in a process called mechanotransduction. Yes-associated protein (YAP) is an effector of Hippo signaling and a mediator of mechanotransduction, but how mechanical forces regulate Hippo signaling is still an open question. We propose that retinoic acid-induced protein 14 (RAI14) responds to mechanical forces and regulates Hippo signaling. RAI14 positively regulates the activity of YAP. RAI14 interacts with NF2, a key component of the Hippo pathway, and the interaction occurs on filamentous actin. When mechanical forces are kept low in cells, NF2 dissociates from RAI14 and filamentous actin, resulting in increased interactions with LATS1 and activation of the Hippo pathway. Clinical data show that tissue stiffness and expression of RAI14 and YAP are upregulated in tumor tissues and that RAI14 is strongly associated with adverse outcome in patients with gastric cancer. Our data suggest that RAI14 links mechanotransduction with Hippo signaling and mediates Hippo-related biological functions such as cancer progression.


Subject(s)
Hippo Signaling Pathway , Protein Serine-Threonine Kinases , Transcription Factors , Humans , Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Mechanotransduction, Cellular , Neurofibromin 2/metabolism , Neurofibromin 2/genetics , Phosphoproteins/metabolism , Phosphoproteins/genetics , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Transcription Factors/metabolism , YAP-Signaling Proteins/metabolism
3.
Science ; 385(6709): eadf4478, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39116228

ABSTRACT

Despite recent studies implicating liquid-like biomolecular condensates in diverse cellular processes, many biomolecular condensates exist in a solid-like state, and their function and regulation are less understood. We show that the tumor suppressor Merlin, an upstream regulator of the Hippo pathway, localizes to both cell junctions and medial apical cortex in Drosophila epithelia, with the latter forming solid-like condensates that activate Hippo signaling. Merlin condensation required phosphatidylinositol-4-phosphate (PI4P)-mediated plasma membrane targeting and was antagonistically controlled by Pez and cytoskeletal tension through plasma membrane PI4P regulation. The solid-like material properties of Merlin condensates are essential for physiological function and protect the condensates against external perturbations. Collectively, these findings uncover an essential role for solid-like condensates in normal physiology and reveal regulatory mechanisms for their formation and disassembly.


Subject(s)
Biomolecular Condensates , Drosophila Proteins , Drosophila melanogaster , Hippo Signaling Pathway , Neurofibromin 2 , Animals , Cell Membrane/metabolism , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Intercellular Junctions/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Neurofibromin 2/metabolism , Neurofibromin 2/genetics , Phosphatidylinositol Phosphates/metabolism , Protein Serine-Threonine Kinases/metabolism , Biomolecular Condensates/metabolism
4.
PLoS One ; 19(7): e0305121, 2024.
Article in English | MEDLINE | ID: mdl-39083549

ABSTRACT

Neurofibromatosis type 2 is a genetic disorder that results in the formation and progressive growth of schwannomas, ependymomas, and/or meningiomas. The NF2 gene encodes the Merlin protein, which links cell cortical elements to the actin cytoskeleton and regulates a number of key enzymes including Group I p21-activated kinases (PAKs), the Hippo-pathway kinase LATS, and mTORC. While PAK1 and PAK2 directly bind Merlin and transmit proliferation and survival signals when Merlin is mutated or absent, inhibition of Group 1 PAKs alone has not proven sufficient to completely stop the growth of NF2-deficient meningiomas or schwannomas in vivo, suggesting the need for a second pathway inhibitor. As the Hippo pathway is also activated in NF2-deficient cells, several inhibitors of the Hippo pathway have recently been developed in the form of YAP-TEAD binding inhibitors. These inhibitors prevent activation of pro-proliferation and anti-apoptotic Hippo pathway effectors. In this study, we show that PAK inhibition slows cell proliferation while TEAD inhibition promotes apoptotic cell death. Finally, we demonstrate the efficacy of PAK and TEAD inhibitor combinations in several NF2-deficient Schwannoma cell lines.


Subject(s)
Cell Proliferation , Hippo Signaling Pathway , Neurilemmoma , Neurofibromin 2 , Protein Serine-Threonine Kinases , Signal Transduction , Transcription Factors , p21-Activated Kinases , Humans , Neurilemmoma/metabolism , Neurilemmoma/genetics , Neurilemmoma/pathology , p21-Activated Kinases/metabolism , p21-Activated Kinases/antagonists & inhibitors , p21-Activated Kinases/genetics , Cell Proliferation/drug effects , Neurofibromin 2/genetics , Neurofibromin 2/deficiency , Neurofibromin 2/metabolism , Signal Transduction/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/antagonists & inhibitors , Cell Line, Tumor , Transcription Factors/metabolism , Transcription Factors/genetics , Apoptosis/drug effects , Drug Synergism , Neurofibromatosis 2/metabolism , Neurofibromatosis 2/genetics , YAP-Signaling Proteins/metabolism , YAP-Signaling Proteins/genetics , Protein Kinase Inhibitors/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics
5.
Development ; 151(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39077779

ABSTRACT

The Hippo pathway plays a crucial role in cell proliferation and differentiation during tumorigenesis, tissue homeostasis and early embryogenesis. Scaffold proteins from the ezrin-radixin-moesin (ERM) family, including neurofibromin 2 (NF2; Merlin), regulate the Hippo pathway through cell polarity. However, the mechanisms underlying Hippo pathway regulation via cell polarity in establishing outer cells remain unclear. In this study, we generated artificial Nf2 mutants in the N-terminal FERM domain (L64P) and examined Hippo pathway activity by assessing the subcellular localization of YAP1 in early embryos expressing these mutant mRNAs. The L64P-Nf2 mutant inhibited NF2 localization around the cell membrane, resulting in YAP1 cytoplasmic translocation in the polar cells. L64P-Nf2 expression also disrupted the apical centralization of both large tumor suppressor 2 (LATS2) and ezrin in the polar cells. Furthermore, Lats2 mutants in the FERM binding domain (L83K) inhibited YAP1 nuclear translocation. These findings demonstrate that NF2 subcellular localization mediates cell polarity establishment involving ezrin centralization. This study provides previously unreported insights into how the orchestration of the cell-surface components, including NF2, LATS2 and ezrin, modulates the Hippo pathway during cell polarization.


Subject(s)
Adaptor Proteins, Signal Transducing , Cell Polarity , Cytoskeletal Proteins , Hippo Signaling Pathway , Neurofibromin 2 , Protein Serine-Threonine Kinases , Tumor Suppressor Proteins , YAP-Signaling Proteins , Neurofibromin 2/metabolism , Neurofibromin 2/genetics , Animals , Mice , YAP-Signaling Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Signal Transduction , Embryo, Mammalian/metabolism , Mutation/genetics , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Protein Transport , Cell Membrane/metabolism , Phosphoproteins/metabolism , Phosphoproteins/genetics
6.
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
8.
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
9.
CNS Neurosci Ther ; 30(6): e14784, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38828669

ABSTRACT

INTRODUCTION: Programmed death-ligand 1 (PD-L1) expression is an immune evasion mechanism that has been demonstrated in many tumors and is commonly associated with a poor prognosis. Over the years, anti-PD-L1 agents have gained attention as novel anticancer therapeutics that induce durable tumor regression in numerous malignancies. They may be a new treatment choice for neurofibromatosis type 2 (NF2) patients. AIMS: The aims of this study were to detect the expression of PD-L1 in NF2-associated meningiomas, explore the effect of PD-L1 downregulation on tumor cell characteristics and T-cell functions, and investigate the possible pathways that regulate PD-L1 expression to further dissect the possible mechanism of immune suppression in NF2 tumors and to provide new treatment options for NF2 patients. RESULTS: PD-L1 is heterogeneously expressed in NF2-associated meningiomas. After PD-L1 knockdown in NF2-associated meningioma cells, tumor cell proliferation was significantly inhibited, and the apoptosis rate was elevated. When T cells were cocultured with siPD-L1-transfected NF2-associated meningioma cells, the expression of CD69 on both CD4+ and CD8+ T cells was partly reversed, and the capacity of CD8+ T cells to kill siPD-L1-transfected tumor cells was partly restored. Results also showed that the PI3K-AKT-mTOR pathway regulates PD-L1 expression, and the mTOR inhibitor rapamycin rapidly and persistently suppresses PD-L1 expression. In vivo experimental results suggested that anti-PD-L1 antibody may have a synergetic effect with the mTOR inhibitor in reducing tumor cell proliferation and that reduced PD-L1 expression could contribute to antitumor efficacy. CONCLUSIONS: Targeting PD-L1 could be helpful for restoring the function of tumor-infiltrating lymphocytes and inducing apoptosis to inhibit tumor proliferation in NF2-associated meningiomas. Dissecting the mechanisms of the PD-L1-driven tumorigenesis of NF2-associated meningioma will help to improve our understanding of the mechanisms underlying tumor progression and could facilitate further refinement of current therapies to improve the treatment of NF2 patients.


Subject(s)
B7-H1 Antigen , Cell Proliferation , Meningeal Neoplasms , Meningioma , Neurofibromatosis 2 , T-Lymphocytes , Meningioma/metabolism , Meningioma/immunology , Meningioma/pathology , Humans , B7-H1 Antigen/metabolism , Cell Proliferation/drug effects , Cell Proliferation/physiology , Meningeal Neoplasms/metabolism , Meningeal Neoplasms/pathology , Meningeal Neoplasms/immunology , Animals , T-Lymphocytes/metabolism , T-Lymphocytes/drug effects , Neurofibromatosis 2/metabolism , Mice , Male , Female , Neurofibromin 2/metabolism , Neurofibromin 2/genetics , Cell Line, Tumor , Middle Aged , Mice, Nude , Apoptosis/drug effects , Apoptosis/physiology
10.
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
11.
Commun Biol ; 7(1): 533, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710747

ABSTRACT

Insect wing development is a fascinating and intricate process that involves the regulation of wing size through cell proliferation and apoptosis. In this study, we find that Ter94, an AAA-ATPase, is essential for proper wing size dependently on its ATPase activity. Loss of Ter94 enables the suppression of Hippo target genes. When Ter94 is depleted, it results in reduced wing size and increased apoptosis, which can be rescued by inhibiting the Hippo pathway. Biochemical experiments reveal that Ter94 reciprocally binds to Mer, a critical upstream component of the Hippo pathway, and disrupts its interaction with Ex and Kib. This disruption prevents the formation of the Ex-Mer-Kib complex, ultimately leading to the inactivation of the Hippo pathway and promoting proper wing development. Finally, we show that hVCP, the human homolog of Ter94, is able to substitute for Ter94 in modulating Drosophila wing size, underscoring their functional conservation. In conclusion, Ter94 plays a positive role in regulating wing size by interfering with the Ex-Mer-Kib complex, which results in the suppression of the Hippo pathway.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Membrane Proteins , Protein Serine-Threonine Kinases , Signal Transduction , Tumor Suppressor Proteins , Wings, Animal , Animals , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/genetics , Apoptosis , Drosophila/genetics , Drosophila/growth & development , Drosophila/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Neurofibromin 2/metabolism , Neurofibromin 2/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Wings, Animal/growth & development , Wings, Animal/metabolism
12.
Glia ; 72(8): 1518-1540, 2024 08.
Article in English | MEDLINE | ID: mdl-38794866

ABSTRACT

In the central nervous system, the formation of myelin by oligodendrocytes (OLs) relies on the switch from the polymerization of the actin cytoskeleton to its depolymerization. The molecular mechanisms that trigger this switch have yet to be elucidated. Here, we identified P21-activated kinase 1 (PAK1) as a major regulator of actin depolymerization in OLs. Our results demonstrate that PAK1 accumulates in OLs in a kinase-inhibited form, triggering actin disassembly and, consequently, myelin membrane expansion. Remarkably, proteomic analysis of PAK1 binding partners enabled the identification of NF2/Merlin as its endogenous inhibitor. Our findings indicate that Nf2 knockdown in OLs results in PAK1 activation, actin polymerization, and a reduction in OL myelin membrane expansion. This effect is rescued by treatment with a PAK1 inhibitor. We also provide evidence that the specific Pak1 loss-of-function in oligodendroglia stimulates the thickening of myelin sheaths in vivo. Overall, our data indicate that the antagonistic actions of PAK1 and NF2/Merlin on the actin cytoskeleton of the OLs are critical for proper myelin formation. These findings have broad mechanistic and therapeutic implications in demyelinating diseases and neurodevelopmental disorders.


Subject(s)
Myelin Sheath , Oligodendroglia , p21-Activated Kinases , p21-Activated Kinases/metabolism , Oligodendroglia/metabolism , Animals , Myelin Sheath/metabolism , Neurofibromin 2/metabolism , Neurofibromin 2/genetics , Rats , Actins/metabolism , Cells, Cultured , Mice , Mice, Inbred C57BL , Actin Cytoskeleton/metabolism
13.
Circulation ; 149(25): 1960-1979, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38752370

ABSTRACT

BACKGROUND: Cardiomyocyte differentiation involves a stepwise clearance of repressors and fate-restricting regulators through the modulation of BMP (bone morphogenic protein)/Wnt-signaling pathways. However, the mechanisms and how regulatory roadblocks are removed with specific developmental signaling pathways remain unclear. METHODS: We conducted a genome-wide CRISPR screen to uncover essential regulators of cardiomyocyte specification in human embryonic stem cells using a myosin heavy chain 6 (MYH6)-GFP (green fluorescence protein) reporter system. After an independent secondary single guide ribonucleic acid validation of 25 candidates, we identified NF2 (neurofibromin 2), a moesin-ezrin-radixin like (MERLIN) tumor suppressor, as an upstream driver of early cardiomyocyte lineage specification. Independent monoclonal NF2 knockouts were generated using CRISPR-Cas9, and cell states were inferred through bulk RNA sequencing and protein expression analysis across differentiation time points. Terminal lineage differentiation was assessed by using an in vitro 2-dimensional-micropatterned gastruloid model, trilineage differentiation, and cardiomyocyte differentiation. Protein interaction and post-translation modification of NF2 with its interacting partners were assessed using site-directed mutagenesis, coimmunoprecipitation, and proximity ligation assays. RESULTS: Transcriptional regulation and trajectory inference from NF2-null cells reveal the loss of cardiomyocyte identity and the acquisition of nonmesodermal identity. Sustained elevation of early mesoderm lineage repressor SOX2 and upregulation of late anticardiac regulators CDX2 and MSX1 in NF2 knockout cells reflect a necessary role for NF2 in removing regulatory roadblocks. Furthermore, we found that NF2 and AMOT (angiomotin) cooperatively bind to YAP (yes-associated protein) during mesendoderm formation, thereby preventing YAP activation, independent of canonical MST (mammalian sterile 20-like serine-threonine protein kinase)-LATS (large tumor suppressor serine-threonine protein kinase) signaling. Mechanistically, cardiomyocyte lineage identity was rescued by wild-type and NF2 serine-518 phosphomutants, but not NF2 FERM (ezrin-radixin-meosin homology protein) domain blue-box mutants, demonstrating that the critical FERM domain-dependent formation of the AMOT-NF2-YAP scaffold complex at the adherens junction is required for early cardiomyocyte lineage differentiation. CONCLUSIONS: These results provide mechanistic insight into the essential role of NF2 during early epithelial-mesenchymal transition by sequestering the repressive effect of YAP and relieving regulatory roadblocks en route to cardiomyocytes.


Subject(s)
Cell Differentiation , Cell Lineage , Myocytes, Cardiac , Neurofibromin 2 , Humans , Myocytes, Cardiac/metabolism , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , CRISPR-Cas Systems , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology
14.
J Pathol ; 263(2): 257-269, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38613194

ABSTRACT

Genomic rearrangements of the neurotrophic receptor tyrosine kinase genes (NTRK1, NTRK2, and NTRK3) are the most common mechanism of oncogenic activation for this family of receptors, resulting in sustained cancer cell proliferation. Several targeted therapies have been approved for tumours harbouring NTRK fusions and a new generation of TRK inhibitors has already been developed due to acquired resistance. We established a patient-derived LMNA::NTRK1-rearranged soft-tissue sarcoma cell model ex vivo with an acquired resistance to targeted TRK inhibition. Molecular profiling of the resistant clones revealed an acquired NF2 loss of function mutation that was absent in the parental cell model. Parental cells showed continuous sensitivity to TRK-targeted treatment, whereas the resistant clones were insensitive. Furthermore, resistant clones showed upregulation of the MAPK and mTOR/AKT pathways in the gene expression based on RNA sequencing data and increased sensitivity to MEK and mTOR inhibitor therapy. Drug synergy was seen using trametinib and rapamycin in combination with entrectinib. Medium-throughput drug screening further identified small compounds as potential drug candidates to overcome resistance as monotherapy or in combination with entrectinib. In summary, we developed a comprehensive model of drug resistance in an LMNA::NTRK1-rearranged soft-tissue sarcoma and have broadened the understanding of acquired drug resistance to targeted TRK therapy. Furthermore, we identified drug combinations and small compounds to overcome acquired drug resistance and potentially guide patient care in a functional precision oncology setting. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Drug Resistance, Neoplasm , Gene Rearrangement , Lamin Type A , Mutation , Neurofibromin 2 , Protein Kinase Inhibitors , Receptor, trkA , Sarcoma , Humans , Lamin Type A/genetics , Lamin Type A/metabolism , Drug Resistance, Neoplasm/genetics , Receptor, trkA/genetics , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/metabolism , Sarcoma/genetics , Sarcoma/drug therapy , Sarcoma/pathology , Sarcoma/metabolism , Protein Kinase Inhibitors/pharmacology , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Pyridones/pharmacology , Benzamides/pharmacology , Pyrimidinones/pharmacology , Sirolimus/pharmacology , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/drug therapy , Soft Tissue Neoplasms/pathology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Signal Transduction/drug effects , Drug Synergism , Indazoles
15.
J Biol Chem ; 300(5): 107212, 2024 May.
Article in English | MEDLINE | ID: mdl-38522513

ABSTRACT

As an output effector of the Hippo signaling pathway, the TEAD transcription factor and co-activator YAP play crucial functions in promoting cell proliferation and organ size. The tumor suppressor NF2 has been shown to activate LATS1/2 kinases and interplay with the Hippo pathway to suppress the YAP-TEAD complex. However, whether and how NF2 could directly regulate TEAD remains unknown. We identified a direct link and physical interaction between NF2 and TEAD4. NF2 interacted with TEAD4 through its FERM domain and C-terminal tail and decreased the protein stability of TEAD4 independently of LATS1/2 and YAP. Furthermore, NF2 inhibited TEAD4 palmitoylation and induced the cytoplasmic translocation of TEAD4, resulting in ubiquitination and dysfunction of TEAD4. Moreover, the interaction with TEAD4 is required for NF2 function to suppress cell proliferation. These findings reveal an unanticipated role of NF2 as a binding partner and inhibitor of the transcription factor TEAD, shedding light on an alternative mechanism of how NF2 functions as a tumor suppressor through the Hippo signaling cascade.


Subject(s)
Hippo Signaling Pathway , Neurofibromin 2 , Protein Serine-Threonine Kinases , Signal Transduction , TEA Domain Transcription Factors , Humans , Cell Proliferation , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , HEK293 Cells , Lipoylation , Neurofibromin 2/metabolism , Neurofibromin 2/genetics , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Stability , TEA Domain Transcription Factors/metabolism , Tumor Suppressor Proteins , Ubiquitination
16.
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
17.
Int J Mol Sci ; 25(3)2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38338806

ABSTRACT

Solid tumours can universally evade contact inhibition of proliferation (CIP), a mechanism halting cell proliferation when cell-cell contact occurs. Merlin, an ERM-like protein, crucially regulates CIP and is frequently deactivated in various cancers, indicating its significance as a tumour suppressor in cancer biology. Despite extensive investigations into Merlin's role in cancer, its lack of intrinsic catalytic activity and frequent conformation changes have made it notoriously challenging to study. To address this challenge, we harnessed innovative luciferase technologies to create and validate a NanoBiT split-luciferase biosensor system in which Merlin is cloned between two split components (LgBiT and SmBiT) of NanoLuc luciferase. This system enables precise quantification of Merlin's conformation and activity both in vitro and within living cells. This biosensor significantly enhances the study of Merlin's molecular functions, serving as a potent tool for exploring its contributions to CIP and tumorigenesis.


Subject(s)
Biosensing Techniques , Neoplasms , Neurofibromin 2 , Humans , Cell Transformation, Neoplastic , Genes, Tumor Suppressor , Luciferases , Neurofibromin 2/chemistry , Neurofibromin 2/metabolism , Biosensing Techniques/methods
18.
Int J Mol Sci ; 24(19)2023 Sep 30.
Article in English | MEDLINE | ID: mdl-37834234

ABSTRACT

The type I phosphatidylinositol 4-phosphate 5-kinase (PIP5K) family produces the critical lipid regulator phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) in the plasma membrane (PM). Here, we investigated the potential role of PIP5Kγ, a PIP5K isoform, in the Hippo pathway. The ectopic expression of PIP5Kγ87 or PIP5Kγ90, two major PIP5Kγ splice variants, activated large tumor suppressor kinase 1 (LATS1) and inhibited Yes-associated protein (YAP), whereas PIP5Kγ knockdown yielded opposite effects. The regulatory effects of PIP5Kγ were dependent on its catalytic activity and the presence of Merlin and LATS1. PIP5Kγ knockdown weakened the restoration of YAP phosphorylation upon stimulation with epidermal growth factor or lysophosphatidic acid. We further found that PIP5Kγ90 bound to the Merlin's band 4.1/ezrin/radixin/moesin (FERM) domain, forming a complex with PI(4,5)P2 and LATS1 at the PM. Notably, PIP5Kγ90, but not its kinase-deficient mutant, potentiated Merlin-LATS1 interaction and recruited LATS1 to the PM. Consistently, PIP5Kγ knockdown or inhibitor (UNC3230) enhanced colony formation in carcinoma cell lines YAP-dependently. In addition, PIP5Kγ90 interacted with heat shock cognate 71-kDa protein (Hsc70), which also contributed to Hippo pathway activation. Collectively, our results suggest that PIP5Kγ regulates the Hippo-YAP pathway by forming a functional complex with Merlin and LATS1 at the PI(4,5)P2-rich PM and via interplay with Hsc70.


Subject(s)
Hippo Signaling Pathway , Neurofibromin 2 , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Cell Proliferation/physiology , Signal Transduction
19.
Mol Cancer Ther ; 22(11): 1280-1289, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37527526

ABSTRACT

Neurofibromatosis Type 2 (NF2) is a tumor predisposition syndrome caused by germline inactivating mutations in the NF2 gene encoding the merlin tumor suppressor. Patients develop multiple benign tumor types in the nervous system including bilateral vestibular schwannomas (VS). Standard treatments include surgery and radiation therapy, which may lead to loss of hearing, impaired facial nerve function, and other complications. Kinase inhibitor monotherapies have been evaluated clinically for NF2 patients with limited success, and more effective nonsurgical therapies are urgently needed. Schwannoma model cells treated with PI3K inhibitors upregulate activity of the focal adhesion kinase (FAK) family as a compensatory survival pathway. We screened combinations of 13 clinically relevant PI3K and FAK inhibitors using human isogenic normal and merlin-deficient Schwann cell lines. The most efficacious combination was PI3K/mTOR inhibitor omipalisib with SRC/FAK inhibitor dasatinib. Sub-GI50 doses of the single drugs blocked phosphorylation of their major target proteins. The combination was superior to either single agent in promoting a G1 cell-cycle arrest and produced a 44% decrease in tumor growth over a 2-week period in a pilot orthotopic allograft model. Evaluation of single and combination drugs in six human primary VS cell models revealed the combination was superior to the monotherapies in 3 of 6 VS samples, highlighting inter-tumor variability between patients consistent with observations from clinical trials with other molecular targeted agents. Dasatinib alone performed as well as the combination in the remaining three samples. Preclinically validated combination therapies hold promise for NF2 patients and warrants further study in clinical trials.


Subject(s)
Antineoplastic Agents , Neurilemmoma , Neurofibromatosis 2 , Humans , Neurofibromatosis 2/drug therapy , Neurofibromatosis 2/genetics , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Phosphatidylinositol 3-Kinases/pharmacology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Dasatinib/pharmacology , Phosphatidylinositol 3-Kinase/pharmacology , Phosphatidylinositol 3-Kinase/therapeutic use , Neurilemmoma/drug therapy , Neurilemmoma/genetics , Antineoplastic Agents/pharmacology , Cell Proliferation
20.
Apoptosis ; 28(9-10): 1484-1495, 2023 10.
Article in English | MEDLINE | ID: mdl-37368176

ABSTRACT

Neutrophil and neutrophil extracellular traps (NETs) were reported to be associated with tumor development, but the exact role and concrete mechanisms are still poorly understood, especially in triple negative breast cancer (TNBC). In this study, our results exhibited that NETs formation in TNBC tissues was higher than that in non-TNBC tissues, and NETs formation was distinctly correlated with tumor size, ki67 level and lymph node metastasis in TNBC patients. Subsequent in vivo experiments demonstrated that NETs inhibition could suppress TNBC tumor growth and lung metastasis. Further in vitro experiments uncovered that oncogenic function of NETs on TNBC cells were possibly dependent on TLR9 expression. We also found that neutrophils from peripheral blood of TNBC patients with postoperative fever were prone to form NETs and could enhance the proliferation and invasion of TNBC cells. Mechanistically, we revealed that NETs could interact with TLR9 to decrease Merlin phosphorylation which contributed to TNBC cell ferroptosis resistance. Our work provides a novel insight into the mechanism of NETs promoting TNBC progression and blocking the key modulator of NETs might be a promising therapeutic strategy in TNBC.


Subject(s)
Extracellular Traps , Ferroptosis , Triple Negative Breast Neoplasms , Humans , Extracellular Traps/metabolism , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism , Neurofibromin 2/metabolism , Ferroptosis/genetics , Cell Line, Tumor , Triple Negative Breast Neoplasms/pathology , Apoptosis , Neutrophils/pathology , Cell Proliferation
SELECTION OF CITATIONS
SEARCH DETAIL