Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 896
Filtrar
1.
Nat Commun ; 15(1): 7873, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39251601

RESUMEN

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.


Asunto(s)
Imagen por Resonancia Magnética , Neoplasias Meníngeas , Meningioma , Neurofibromina 2 , Vía de Señalización Wnt , Meningioma/diagnóstico por imagen , Meningioma/metabolismo , Meningioma/patología , Meningioma/genética , Humanos , Fosforilación , Neurofibromina 2/metabolismo , Neurofibromina 2/genética , Animales , Imagen por Resonancia Magnética/métodos , Neoplasias Meníngeas/diagnóstico por imagen , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/genética , Ratones , Línea Celular Tumoral , beta Catenina/metabolismo , beta Catenina/genética , Femenino , Serina/metabolismo , Masculino , Proteómica/métodos , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/genética
2.
Chin Clin Oncol ; 13(Suppl 1): AB063, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39295381

RESUMEN

BACKGROUND: Meningioma is the most common type of primary central nervous system tumor. The development of meningioma is often influenced by the neurofibromin-2 (NF2) gene. Studies have demonstrated that the genetic profile of meningioma is linked to their location. However, it is currently unclear whether there is an association between NF2 expression and meningioma location in the Indonesian population. METHODS: This retrospective cross-sectional analysis included 153 histologically confirmed meningioma patients admitted to Dr. Sardjito General Hospital in Yogyakarta, Indonesia. The study utilized immunohistochemistry (IHC) staining to determine the NF2/Merlin expression. Meningioma locations were established using brain magnetic resonance imaging (MRI) or computerized tomography (CT) scans before surgery. The data on other variables were collected from medical records. RESULTS: Of all the patients, 118 (77.1%) tested positive for NF2/Merlin expression. The subjects in the study comprised 126 (82.4%) World Health Organization (WHO) grade I tumors and 27 (17.6%) WHO grade II and III tumors. The research showed that 85 (55.6%) of the tumors were located in the spheno-orbital region. The study found a significant association between NF2 positive expression and meningioma location in the spheno-orbital region [odds ratio (OR) 2.51, P=0.02]. Therefore, the patients who tested positive for NF2 had higher chances of visual impairment (P=0.04). CONCLUSIONS: This study showed positive NF2 expression is associated with meningioma location in spheno-orbital and patients' visual symptoms.


Asunto(s)
Neoplasias Meníngeas , Meningioma , Humanos , Meningioma/genética , Femenino , Masculino , Estudios Retrospectivos , Estudios Transversales , Persona de Mediana Edad , Indonesia , Adulto , Neoplasias Meníngeas/genética , Neurofibromina 2/genética , Anciano , Adulto Joven
4.
EMBO Rep ; 25(9): 4033-4061, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39160347

RESUMEN

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.


Asunto(s)
Vía de Señalización Hippo , Proteínas Serina-Treonina Quinasas , Factores de Transcripción , Humanos , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Mecanotransducción Celular , Neurofibromina 2/metabolismo , Neurofibromina 2/genética , Fosfoproteínas/metabolismo , Fosfoproteínas/genética , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Neoplasias Gástricas/genética , Factores de Transcripción/metabolismo , Proteínas Señalizadoras YAP/metabolismo
5.
Science ; 385(6709): eadf4478, 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39116228

RESUMEN

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.


Asunto(s)
Condensados Biomoleculares , Proteínas de Drosophila , Drosophila melanogaster , Vía de Señalización Hippo , Neurofibromina 2 , Animales , Membrana Celular/metabolismo , Drosophila melanogaster/metabolismo , Drosophila melanogaster/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Uniones Intercelulares/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Neurofibromina 2/metabolismo , Neurofibromina 2/genética , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Condensados Biomoleculares/metabolismo
6.
Oncogene ; 43(40): 2995-3002, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39209965

RESUMEN

Neurofibromatosis type 2 (NF2) is a rare disorder that causes vestibular schwannomas (VS), meningiomas and ependymomas. To date, there is no FDA approved drug-based treatment for NF2. We have previously identified that BET inhibition can selectively reduce growth of the NF2-null schwannoma and Schwann cells in vitro and tumorigenesis in vivo and, separately, reported that inhibition of Focal Adhesion Kinase 1 (FAK1) via crizotinib has antiproliferative effects in NF2-null Schwann cells. The current study was aimed at determining whether combined BET and FAK inhibition can synergize and to identify the mechanisms of action. A panel of normal and NF2-null Schwann and schwannoma cell lines were used to characterize the effects of combined BET and FAK inhibition in vitro and in vivo using pharmacological and genetic approaches. The mechanism of action was explored by chromatin immunoprecipitation, ChIP-PCR, western blotting, and functional approaches. We find that combined BET and FAK inhibition are synergistic and inhibit the proliferation of NF2-null schwannoma and Schwann cell lines in vitro and in vivo, by arresting cells in the G1/S and G2/M phases of the cell cycle. Further, we identify the mechanism of action through the downregulation of FAK1 transcription by BET inhibition, which potentiates inhibition of FAK by 100-fold. Our findings suggest that combined targeting of BET and FAK1 may offer a potential therapeutic option for the treatment of NF2-related schwannomas.


Asunto(s)
Proliferación Celular , Quinasa 1 de Adhesión Focal , Neurilemoma , Neurofibromina 2 , Neuroma Acústico , Animales , Humanos , Ratones , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Quinasa 1 de Adhesión Focal/genética , Neuroma Acústico/patología , Neuroma Acústico/genética , Neuroma Acústico/tratamiento farmacológico , Neuroma Acústico/metabolismo , Neurilemoma/patología , Neurilemoma/genética , Neurilemoma/tratamiento farmacológico , Neurilemoma/metabolismo , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Proliferación Celular/efectos de los fármacos , Línea Celular Tumoral , Sinergismo Farmacológico , Neurofibromatosis/tratamiento farmacológico , Neurofibromatosis/genética , Neurofibromatosis/patología , Neurofibromatosis 2/genética , Neurofibromatosis 2/tratamiento farmacológico , Neurofibromatosis 2/patología , Neurofibromatosis 2/metabolismo , Células de Schwann/efectos de los fármacos , Células de Schwann/metabolismo , Células de Schwann/patología , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Pathol Res Pract ; 261: 155459, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39083879

RESUMEN

Succinate dehydrogenase (SDH)-deficient renal cell carcinoma (RCC) is a rare subtype of RCC classified as a molecularly defined RCC in the fifth edition of the WHO. Most gene alterations in patients with SDH-deficient RCC involve the SDHB subunit, with less involvement of the SDHC, SDHA, and SDHD subunits. Four cases of SDHA-deficient RCC have been reported in the literature, of which one case was associated with an NF2 gene mutation. Herein, we report six novel SDHA-deficient RCC cases, including two cases with NF2 gene mutations. In contrast to the typical morphology of SDH-deficient RCC, the six tumors mainly displayed glandular, sheet-like, or papillary growth patterns with prominent nucleoli (Grades 2-3), among which two cases with NF2 mutations had prominent nucleoli (Grade 3), large transparent vacuoles in the cytoplasm, and a large number of lymphocytes in the stroma. Six tumors showed negative immunohistochemical staining for SDHA and SDHB, and three cases presented with high expression of PD-L1. Second-generation sequencing revealed novel pathogenic somatic SDHA gene mutation and NF2 gene mutations in six and two tumors, respectively. Follow-up data were collected for the six patients with a follow-up time ranging from 7 to 268 months, and all six patients have survived to date. One patient received targeted therapy for tumor metastasis to the lungs after seven months, and another patient with an NF2 gene mutation received immunotherapy for lymph node metastasis revealed during surgery. SDHA-deficient RCCs with NF2 gene mutations have the ability to metastasize but might respond well to immunotherapy. For the first time, we report the largest number of SDHA-deficient RCC cases and comprehensively investigate their clinicopathological and molecular features to provide important guidance for diagnosis and clinical immunotherapy.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Succinato Deshidrogenasa , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Masculino , Femenino , Persona de Mediana Edad , Succinato Deshidrogenasa/deficiencia , Succinato Deshidrogenasa/genética , Mutación , Adulto , Anciano , Biomarcadores de Tumor/genética , Neurofibromina 2 , Complejo II de Transporte de Electrones
8.
Development ; 151(14)2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39077779

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Polaridad Celular , Proteínas del Citoesqueleto , Vía de Señalización Hippo , Neurofibromina 2 , Proteínas Serina-Treonina Quinasas , Proteínas Supresoras de Tumor , Proteínas Señalizadoras YAP , Neurofibromina 2/metabolismo , Neurofibromina 2/genética , Animales , Ratones , Proteínas Señalizadoras YAP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Transducción de Señal , Embrión de Mamíferos/metabolismo , Mutación/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Transporte de Proteínas , Membrana Celular/metabolismo , Fosfoproteínas/metabolismo , Fosfoproteínas/genética
9.
Cancer Res Commun ; 4(8): 2133-2146, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38994676

RESUMEN

Immunotherapies have demonstrated limited clinical efficacy in malignant mesothelioma treatment. We conducted multiplex immunofluorescence analyses on tissue microarrays (n = 3) from patients with malignant pleural mesothelioma (MPM, n = 88) and malignant peritoneal mesothelioma (MPeM, n = 25). Our study aimed to elucidate spatial distributions of key immune cell populations and their association with lymphocyte activation gene 3 (LAG3), BRCA1-associated protein 1 (BAP1), neurofibromatosis type 2 (NF2), and methylthioadenosine phosphorylase (MTAP), with MTAP serving as a cyclin-dependent kinase inhibitor 2A/2B (CDKN2A/B) surrogate marker. Additionally, we examined the relationship between the spatial distribution of major immune cell types and prognosis and clinical characteristics of patients with malignant mesothelioma. We observed a higher degree of interaction between immune cells and tumor cells in MPM compared with MPeM. Notably, within MPM tumors, we detected a significantly increased interaction between tumor cells and CD8+ T cells in tumors with low BAP1 expression compared with those with high BAP1 expression. To support the broader research community, we have developed The Human Spatial Atlas of Malignant Mesothelioma, containing hematoxylin and eosin and multiplex immunofluorescence images with corresponding metadata. SIGNIFICANCE: Considering the limited therapeutic options available to patients with malignant mesothelioma, there is substantial translational potential in understanding the correlation between the spatial architecture of the malignant mesothelioma tumor immune microenvironment and tumor biology. Our investigation reveals critical cell-cell interactions that may influence the immune response against malignant mesothelioma tumors, potentially contributing to the differential behaviors observed in MPM and MPeM. These findings represent a valuable resource for the malignant mesothelioma cancer research community.


Asunto(s)
Mesotelioma Maligno , Mesotelioma , Neoplasias Peritoneales , Neoplasias Pleurales , Purina-Nucleósido Fosforilasa , Microambiente Tumoral , Ubiquitina Tiolesterasa , Humanos , Microambiente Tumoral/inmunología , Mesotelioma Maligno/inmunología , Mesotelioma Maligno/patología , Neoplasias Pleurales/inmunología , Neoplasias Pleurales/patología , Masculino , Femenino , Neoplasias Peritoneales/inmunología , Neoplasias Peritoneales/patología , Purina-Nucleósido Fosforilasa/metabolismo , Purina-Nucleósido Fosforilasa/genética , Persona de Mediana Edad , Ubiquitina Tiolesterasa/metabolismo , Mesotelioma/inmunología , Mesotelioma/patología , Anciano , Proteínas Supresoras de Tumor/metabolismo , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neurofibromina 2/genética , Biomarcadores de Tumor , Pronóstico , Antígenos CD , Linfocitos T CD8-positivos/inmunología
11.
FASEB J ; 38(13): e23809, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38967126

RESUMEN

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.


Asunto(s)
Carcinogénesis , Neurofibromina 2 , Humanos , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Neurofibromatosis 2/genética , Neurofibromatosis 2/metabolismo , Neurofibromatosis 2/patología , Transducción de Señal , Mutación
12.
FASEB J ; 38(13): e23737, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38953724

RESUMEN

Meningiomas are the most common primary intracranial tumors and account for nearly 30% of all nervous system tumors. Approximately half of meningioma patients exhibit neurofibromin 2 (NF2) gene inactivation. Here, NF2 was shown to interact with the endoplasmic reticulum (ER) calcium (Ca2+) channel inositol 1,4,5-trisphosphate receptor 1 (IP3R1) in IOMM-Lee, a high-grade malignant meningioma cell line, and the F1 subdomain of NF2 plays a critical role in this interaction. Functional assays indicated that NF2 promotes the phosphorylation of IP3R (Ser 1756) and IP3R-mediated endoplasmic reticulum (ER) Ca2+ release by binding to IP3R1, which results in Ca2+-dependent apoptosis. Knockout of NF2 decreased Ca2+ release and promoted resistance to apoptosis, which was rescued by wild-type NF2 overexpression but not by F1 subdomain deletion truncation overexpression. The effects of NF2 defects on the development of tumors were further studied in mouse models. The decreased expression level of NF2 caused by NF2 gene knockout or mutation affects the activity of the IP3R channel, which reduces Ca2+-dependent apoptosis, thereby promoting the development of tumors. We elucidated the interaction patterns of NF2 and IP3R1, revealed the molecular mechanism through which NF2 regulates IP3R1-mediated Ca2+ release, and elucidated the new pathogenic mechanism of meningioma-related NF2 variants. Our study broadens the current understanding of the biological function of NF2 and provides ideas for drug screening of NF2-associated meningioma.


Asunto(s)
Apoptosis , Señalización del Calcio , Calcio , Receptores de Inositol 1,4,5-Trifosfato , Neoplasias Meníngeas , Meningioma , Animales , Humanos , Ratones , Calcio/metabolismo , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/genética , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/genética , Meningioma/metabolismo , Meningioma/patología , Meningioma/genética , Neurofibromina 2
13.
PLoS One ; 19(7): e0305121, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39083549

RESUMEN

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.


Asunto(s)
Proliferación Celular , Vía de Señalización Hippo , Neurilemoma , Neurofibromina 2 , Proteínas Serina-Treonina Quinasas , Transducción de Señal , Factores de Transcripción , Quinasas p21 Activadas , Humanos , Neurilemoma/metabolismo , Neurilemoma/genética , Neurilemoma/patología , Quinasas p21 Activadas/metabolismo , Quinasas p21 Activadas/antagonistas & inhibidores , Quinasas p21 Activadas/genética , Proliferación Celular/efectos de los fármacos , Neurofibromina 2/genética , Neurofibromina 2/deficiencia , Neurofibromina 2/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Línea Celular Tumoral , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Apoptosis/efectos de los fármacos , Sinergismo Farmacológico , Neurofibromatosis 2/metabolismo , Neurofibromatosis 2/genética , Proteínas Señalizadoras YAP/metabolismo , Proteínas Señalizadoras YAP/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética
14.
J Pathol ; 264(2): 129-131, 2024 10.
Artículo en Inglés | MEDLINE | ID: mdl-39072755

RESUMEN

In a recent issue of The Journal of Pathology, Chen and colleagues established novel patient-derived ex vivo models of NTRK fusion-positive soft tissue sarcoma to characterize resistance mechanisms against targeted therapy with tyrosine kinase inhibitors. Prolonged exposure to escalating concentrations of the tyrosine kinase inhibitor, entrectinib, ultimately led to the occurrence of resistant clones that harbored an inactivating mutation in the NF2 gene, not previously described in this context, accompanied by increased PI3K/AKT/mTOR and Ras/Raf/MEK/ERK signaling. Finally, an inhibitor screen identified, among others, MEK and mTOR inhibitors as potential combination agents. © 2024 The Pathological Society of Great Britain and Ireland.


Asunto(s)
Resistencia a Antineoplásicos , Inhibidores de Proteínas Quinasas , Humanos , Resistencia a Antineoplásicos/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Neurofibromina 2/genética , Proteínas de Fusión Oncogénica/genética , Benzamidas/uso terapéutico , Benzamidas/farmacología , Receptor trkA/genética , Receptor trkA/metabolismo , Transducción de Señal/genética , Indazoles/uso terapéutico , Indazoles/farmacología , Mutación , Sarcoma/genética , Sarcoma/tratamiento farmacológico , Sarcoma/patología , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
15.
J Med Genet ; 61(9): 856-860, 2024 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-38925914

RESUMEN

OBJECTIVES: New diagnostic criteria for NF2-related schwannomatosis (NF2) were published in 2022. An updated UK prevalence was generated in accordance with these, with an emphasis on the rate of de novo NF2 (a 50% frequency is widely quoted in genetic counselling). The distribution of variant types among de novo and familial NF2 cases was also assessed. METHODS: The UK National NF2 database identifies patients meeting updated NF2 criteria from a highly ascertained population cared for by England's specialised service. Diagnostic prevalence was assessed on 1 February 2023. Molecular analysis of blood and, where possible, tumour specimens for NF2, LZTR1 and SMARCB1 was performed. RESULTS: 1084 living NF2 patients were identified on prevalence day (equivalent to 1 in 61 332). The proportion with NF2 inherited from an affected parent was only 23% in England. If people without a confirmed molecular diagnosis or bilateral vestibular schwannoma are excluded, the frequency of de novo NF2 remains high (72%). Of the identified de novo cases, almost half were mosaic. The most common variant type was nonsense variants, accounting for 173/697 (24.8%) of people with an established variant, but only 18/235 (7.7%) with an inherited NF2 pathogenic variant (p<0.0001). Missense variants had the highest proportion of familial association (56%). The prevalence of LZTR1-related schwannomatosis and SMARCB1-related schwannomatosis was 1 in 527 000 and 1 in 1.1M, respectively, 8.4-18.4 times lower than NF2. CONCLUSIONS: This work confirms a much higher rate of de novo NF2 than previously reported and highlights the benefits of maintaining patient databases for accurate counselling.


Asunto(s)
Neurilemoma , Neurofibromatosis , Neurofibromatosis 2 , Neurofibromina 2 , Proteína SMARCB1 , Neoplasias Cutáneas , Humanos , Neurilemoma/genética , Neurilemoma/epidemiología , Neurilemoma/patología , Neurofibromatosis/genética , Neurofibromatosis/epidemiología , Neurofibromatosis/patología , Neurofibromatosis 2/genética , Neurofibromatosis 2/epidemiología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/epidemiología , Neoplasias Cutáneas/patología , Masculino , Femenino , Proteína SMARCB1/genética , Neurofibromina 2/genética , Factores de Transcripción/genética , Prevalencia , Adulto , Mutación/genética , Persona de Mediana Edad , Predisposición Genética a la Enfermedad , Adolescente
16.
Nat Commun ; 15(1): 5115, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38879607

RESUMEN

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.


Asunto(s)
Sistemas CRISPR-Cas , Coenzima A Ligasas , Glucosafosfato Deshidrogenasa , Neurofibromina 2 , Células de Schwann , Mutaciones Letales Sintéticas , Células de Schwann/metabolismo , Humanos , Glucosafosfato Deshidrogenasa/metabolismo , Glucosafosfato Deshidrogenasa/genética , Neurofibromina 2/metabolismo , Neurofibromina 2/genética , Coenzima A Ligasas/metabolismo , Coenzima A Ligasas/genética , Animales , Neurofibromatosis 2/metabolismo , Neurofibromatosis 2/genética , NADP/metabolismo , Ratones , Oxidación-Reducción
17.
Int J Mol Sci ; 25(12)2024 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-38928264

RESUMEN

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.


Asunto(s)
Neurilemoma , Neurofibromatosis , Neurofibromina 2 , Neoplasias Cutáneas , Humanos , Neurofibromatosis/terapia , Neurofibromatosis/genética , Neurofibromatosis/metabolismo , Neurofibromina 2/genética , Neurofibromina 2/metabolismo , Neurilemoma/genética , Neurilemoma/terapia , Neurilemoma/metabolismo , Neurilemoma/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/terapia , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Animales , Neurofibromatosis 2/genética , Neurofibromatosis 2/terapia , Neurofibromatosis 2/metabolismo , Mutación , Transducción de Señal , Terapia Molecular Dirigida
18.
CNS Neurosci Ther ; 30(6): e14784, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38828669

RESUMEN

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.


Asunto(s)
Antígeno B7-H1 , Proliferación Celular , Neoplasias Meníngeas , Meningioma , Neurofibromatosis 2 , Linfocitos T , Meningioma/metabolismo , Meningioma/inmunología , Meningioma/patología , Humanos , Antígeno B7-H1/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/inmunología , Animales , Linfocitos T/metabolismo , Linfocitos T/efectos de los fármacos , Neurofibromatosis 2/metabolismo , Ratones , Masculino , Femenino , Neurofibromina 2/metabolismo , Neurofibromina 2/genética , Línea Celular Tumoral , Persona de Mediana Edad , Ratones Desnudos , Apoptosis/efectos de los fármacos , Apoptosis/fisiología
19.
Artículo en Ruso | MEDLINE | ID: mdl-38884429

RESUMEN

OBJECTIVE: To identify the characteristics of pain syndrome in patients with schwannomas depending on genetic predisposition. MATERIAL AND METHODS: The study included 46 patients with peripheral, spinal and intracranial schwannomas, corresponding to the schwannomatosis phenotype according to the 2022 clinical criteria. All patients underwent sequencing of the LZRT1, Nf2 and SMARCB1 and a copy number study in the NF2. RESULTS: The most severe widespread pain was observed in patients with pathogenic LZRT1 variants, while patients with mosaic variants may not even have local tumor-related pain. Patients with SMARCB1variants may have no pain or have localized pain that responds well to surgical treatment. CONCLUSION: Further studies of the molecular features of schwannomatosis and driver mutations in the pathogenesis of pain are necessary to improve the effectiveness of pain therapy in this group of patients. Schwannomatosis is a disease from the group of neurofibromatosis, manifested by the development of multiple schwannomas. Neuropathic pain is one of the main symptoms characteristic of peripheral schwannomas, however, the severity and prevalence of the pain syndrome does not always correlate with the location of the tumors. According to modern concepts, the key factors influencing the characteristics of the pain syndrome are the target gene and the type of pathogenic variant. The most severe widespread pain is observed in patients with pathogenic variants in the LZRT1 gene, while patients with mosaic variants may not even have local pain associated with tumors. Patients with variants in SMARCB1 may have no pain or localized pain that responds well to surgical treatment.


Asunto(s)
Neurilemoma , Neurofibromatosis , Proteína SMARCB1 , Humanos , Neurilemoma/genética , Neurilemoma/complicaciones , Neurilemoma/diagnóstico , Neurofibromatosis/complicaciones , Neurofibromatosis/genética , Masculino , Femenino , Adulto , Proteína SMARCB1/genética , Persona de Mediana Edad , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/complicaciones , Neurofibromina 2/genética , Factores de Transcripción/genética , Mutación , Neuralgia/genética , Neuralgia/etiología , Neuralgia/diagnóstico , Predisposición Genética a la Enfermedad , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA