Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 150(4): 816-30, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22901811

RESUMEN

Germline mutations in the RAS/ERK signaling pathway underlie several related developmental disorders collectively termed neuro-cardio-facial-cutaneous (NCFC) syndromes. NCFC patients manifest varying degrees of cognitive impairment, but the developmental basis of their brain abnormalities remains largely unknown. Neurofibromatosis type 1 (NF1), an NCFC syndrome, is caused by loss-of-function heterozygous mutations in the NF1 gene, which encodes neurofibromin, a RAS GTPase-activating protein. Here, we show that biallelic Nf1 inactivation promotes Erk-dependent, ectopic Olig2 expression specifically in transit-amplifying progenitors, leading to increased gliogenesis at the expense of neurogenesis in neonatal and adult subventricular zone (SVZ). Nf1-deficient brains exhibit enlarged corpus callosum, a structural defect linked to severe learning deficits in NF1 patients. Strikingly, these NF1-associated developmental defects are rescued by transient treatment with an MEK/ERK inhibitor during neonatal stages. This study reveals a critical role for Nf1 in maintaining postnatal SVZ-derived neurogenesis and identifies a potential therapeutic window for treating NF1-associated brain abnormalities.


Asunto(s)
Encéfalo/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células-Madre Neurales/patología , Neurofibromatosis 1/patología , Neurofibromina 1/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Cuerpo Calloso/patología , Humanos , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Neurofibromatosis 1/embriología , Neurofibromatosis 1/metabolismo , Neurofibromina 1/genética , Neuroglía/patología , Factor de Transcripción 2 de los Oligodendrocitos
2.
Planta Med ; 87(12-13): 937-948, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33784769

RESUMEN

With about 120 species, Aglaia is one of the largest genera of the plant family Meliaceae (the mahogany plants). It is native to the tropical rainforests of the Indo-Australian region, ranging from India and Sri Lanka eastward to Polynesia and Micronesia. Various Aglaia species have been investigated since the 1960s for their phytochemical constituents and biological properties, with the cyclopenta[b]benzofurans (rocaglates or flavaglines) being of particular interest. Phytochemists, medicinal chemists, and biologists have conducted extensive research in establishing these secondary metabolites as potential lead compounds with antineoplastic and antiviral effects, among others. The varied biological properties of rocaglates can be attributed to their unusual structures and their ability to act as inhibitors of the eukaryotic translation initiation factor 4A (eIF4A), affecting protein translation. The present review provides an update on the recently reported phytochemical constituents of Aglaia species, focusing on rocaglate derivatives. Furthermore, laboratory work performed on investigating the biological activities of these chemical constituents is also covered.


Asunto(s)
Aglaia , Benzofuranos , Australia , Factor 4A Eucariótico de Iniciación , Fitoquímicos/farmacología
3.
Neurooncol Adv ; 6(1): vdae024, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38476930

RESUMEN

Background: NF2-associated meningiomas are progressive, highly morbid, and nonresponsive to chemotherapies, highlighting the need for improved treatments. We have established aberrant activation of the mechanistic target of rapamycin (mTOR) signaling in NF2-deficient tumors, leading to clinical trials with first- and second-generation mTOR inhibitors. However, results have been mixed, showing stabilized tumor growth without shrinkage offset by adverse side effects. To address these limitations, here we explored the potential of third-generation, bi-steric mTOR complex 1 (mTORC1) inhibitors using the preclinical tool compound RMC-6272. Methods: Employing human NF2-deficient meningioma lines, we compared mTOR inhibitors rapamycin (first-generation), INK128 (second-generation), and RMC-6272 (third-generation) using in vitro dose-response testing, cell-cycle analysis, and immunoblotting. Furthermore, the efficacy of RMC-6272 was assessed in NF2-null 3D-spheroid meningioma models, and its in vivo potential was evaluated in 2 orthotopic meningioma mouse models. Results: Treatment of meningioma cells revealed that, unlike rapamycin, RMC-6272 demonstrated superior growth inhibitory effects, cell-cycle arrest, and complete inhibition of phosphorylated 4E-BP1 (mTORC1 readout). Moreover, RMC-6272 had a longer retention time than INK128 and inhibited the expression of several eIF4E-sensitive targets on the protein level. RMC-6272 treatment of NF2 spheroids showed significant shrinkage in size as well as reduced proliferation. Furthermore, in vivo studies in mice revealed effective blockage of meningioma growth by RMC-6272, compared with vehicle controls. Conclusions: Our study in preclinical models of NF2 supports possible future clinical evaluation of third-generation, investigational mTORC1 inhibitors, such as RMC-5552, as a potential treatment strategy for NF2.

4.
Sci Rep ; 14(1): 19349, 2024 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-39164287

RESUMEN

Inhibition of translation initiation using eIF4A inhibitors like (-)-didesmethylrocaglamide [(-)-DDR] and (-)-rocaglamide [(-)-Roc] is a potential cancer treatment strategy as they simultaneously diminish multiple oncogenic drivers. We showed that human and dog osteosarcoma cells expressed higher levels of eIF4A1/2 compared with mesenchymal stem cells. Genetic depletion of eIF4A1 and/or 2 slowed osteosarcoma cell growth. To advance preclinical development of eIF4A inhibitors, we demonstrated the importance of (-)-chirality in DDR for growth-inhibitory activity. Bromination of DDR at carbon-5 abolished growth-inhibitory activity, while acetylating DDR at carbon-1 was tolerated. Like (-)-DDR, (±)-DDR, and (-)-Roc, (±)-DDR-acetate increased γH2A.X levels and induced G2/M arrest and apoptosis. Consistent with translation inhibition, these rocaglates decreased the levels of several mitogenic kinases, the STAT3 transcription factor, and the stress-activated protein kinase p38. However, phosphorylated p38 was greatly enhanced in treated cells, suggesting activation of stress response pathways. RNA sequencing identified RHOB as a top upregulated gene in both (-)-DDR- and (-)-Roc-treated osteosarcoma cells, but the Rho inhibitor Rhosin did not enhance the growth-inhibitory activity of (-)-DDR or (-)-Roc. Nonetheless, these rocaglates potently suppressed tumor growth in a canine osteosarcoma patient-derived xenograft model. These results suggest that these eIF4A inhibitors can be leveraged to treat both human and dog osteosarcomas.


Asunto(s)
Factor 4A Eucariótico de Iniciación , Osteosarcoma , Perros , Animales , Humanos , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Osteosarcoma/metabolismo , Línea Celular Tumoral , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Factor 4A Eucariótico de Iniciación/metabolismo , Ratones , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Neoplasias Óseas/metabolismo , Neoplasias Óseas/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Proliferación Celular/efectos de los fármacos , Benzofuranos/farmacología
5.
Neuro Oncol ; 25(9): 1617-1630, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-36806881

RESUMEN

BACKGROUND: Neurofibromatosis 2 (NF2) is an inherited disorder caused by bi-allelic inactivation of the NF2 tumor suppressor gene. NF2-associated tumors, including schwannoma and meningioma, are resistant to chemotherapy, often recurring despite surgery and/or radiation, and have generally shown cytostatic response to signal transduction pathway inhibitors, highlighting the need for improved cytotoxic therapies. METHODS: Leveraging data from our previous high-throughput drug screening in NF2 preclinical models, we identified a class of compounds targeting the ubiquitin-proteasome pathway (UPP), and undertook studies using candidate UPP inhibitors, ixazomib/MLN9708, pevonedistat/MLN4924, and TAK-243/MLN7243. Employing human primary and immortalized meningioma (MN) cell lines, CRISPR-modified Schwann cells (SCs), and mouse Nf2-/- SCs, we performed dose response testing, flow cytometry-based Annexin V and cell cycle analyses, and RNA-sequencing to identify potential underlying mechanisms of apoptosis. In vivo efficacy was also assessed in orthotopic NF2-deficient meningioma and schwannoma tumor models. RESULTS: Testing of three UPP inhibitors demonstrated potent reduction in cell viability and induction of apoptosis for ixazomib or TAK-243, but not pevonedistat. In vitro analyses revealed that ixazomib or TAK-243 downregulates expression of c-KIT and PDGFRα, as well as the E3 ubiquitin ligase SKP2 while upregulating genes associated with endoplasmic reticulum stress-mediated activation of the unfolded protein response (UPR). In vivo treatment of mouse models revealed delayed tumor growth, suggesting a therapeutic potential. CONCLUSIONS: This study demonstrates the efficacy of proteasomal pathway inhibitors in meningioma and schwannoma preclinical models and lays the groundwork for use of these drugs as a promising novel treatment strategy for NF2 patients.


Asunto(s)
Neoplasias Meníngeas , Meningioma , Neurilemoma , Neurofibromatosis 2 , Animales , Humanos , Ratones , Neoplasias Meníngeas/genética , Meningioma/genética , Neurilemoma/tratamiento farmacológico , Neurilemoma/genética , Neurofibromatosis 2/tratamiento farmacológico , Neurofibromina 2/genética
6.
Sci Adv ; 8(44): eabo5442, 2022 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-36322658

RESUMEN

Malignant peripheral nerve sheath tumor (MPNST), a highly aggressive Schwann cell (SC)-derived soft tissue sarcoma, arises from benign neurofibroma (NF); however, the identity, heterogeneity and origins of tumor populations remain elusive. Nestin+ cells have been implicated as tumor stem cells in MPNST; unexpectedly, single-cell profiling of human NF and MPNST and their animal models reveal a broad range of nestin-expressing SC lineage cells and dynamic acquisition of discrete cancer states during malignant transformation. We uncover a nestin-negative mesenchymal neural crest-like subpopulation as a previously unknown malignant stem-like state common to murine and human MPNSTs, which correlates with clinical severity. Integrative multiomics profiling further identifies unique regulatory networks and druggable targets against the malignant subpopulations in MPNST. Targeting key epithelial-mesenchymal transition and stemness regulators including ZEB1 and ALDH1A1 impedes MPNST growth. Together, our studies reveal the underlying principles of tumor cell-state evolution and their regulatory circuitries during NF-to-MPNST transformation, highlighting a hitherto unrecognized mesenchymal stem-like subpopulation in MPNST disease progression.


Asunto(s)
Neoplasias de la Vaina del Nervio , Neurofibroma , Neurofibrosarcoma , Humanos , Animales , Ratones , Neoplasias de la Vaina del Nervio/patología , Nestina , Transformación Celular Neoplásica/genética
7.
Front Oncol ; 11: 698192, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34604034

RESUMEN

The neurofibromatosis syndromes, including NF1, NF2, and schwannomatosis, are tumor suppressor syndromes characterized by multiple nervous system tumors, particularly Schwann cell neoplasms. NF-related tumors are mainly treated by surgery, and some of them have been treated by but are refractory to conventional chemotherapy. Recent advances in molecular genetics and genomics alongside the development of multiple animal models have provided a better understanding of NF tumor biology and facilitated target identification and therapeutic evaluation. Many targeted therapies have been evaluated in preclinical models and patients with limited success. One major advance is the FDA approval of the MEK inhibitor selumetinib for the treatment of NF1-associated plexiform neurofibroma. Due to their anti-neoplastic, antioxidant, and anti-inflammatory properties, selected natural compounds could be useful as a primary therapy or as an adjuvant therapy prior to or following surgery and/or radiation for patients with tumor predisposition syndromes, as patients often take them as dietary supplements and for health enhancement purposes. Here we review the natural compounds that have been evaluated in NF models. Some have demonstrated potent anti-tumor effects and may become viable treatments in the future.

8.
Laryngoscope Investig Otolaryngol ; 6(5): 1008-1019, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34667843

RESUMEN

OBJECTIVES: Two pilot studies of AR-42, a pan-histone deacetylase inhibitor, in human neurofibromatosis type 2 (NF2), vestibular schwannomas (VS), and meningiomas are presented. Primary endpoints included safety, and intra-tumoral pharmacokinetics (PK) and pharmacodynamics (PD). METHODS: Pilot 1 is a subset analysis of a phase 1 study of AR-42 in solid tumors, which included NF2 or sporadic meningiomas. Tumor volumes and treatment-related adverse events (TRAEs) are reported (NCT01129193).Pilot 2 is a phase 0 surgical study of AR-42 assessing intra-tumoral PK and PD. AR-42 was administered for 3 weeks pre-operatively. Plasma and tumor drug concentrations and p-AKT expression were measured (NCT02282917). RESULTS: Pilot 1: Five patients with NF2 and two with sporadic meningiomas experienced a similar incidence of TRAEs to the overall phase I trial. The six evaluable patients had 15 tumors (8 VS, 7 meningiomas). On AR-42, tumor volume increased in six, remained stable in eight, and decreased in one tumor. The annual percent growth rate decreased in eight, remained stable in three, and increased in four tumors. Pilot 2: Four patients with sporadic VS and one patient with meningioma experienced no grade 3/4 toxicities. Expression of p-AKT decreased in three of four VS. All tumors had higher AR-42 concentrations than plasma. CONCLUSIONS: AR-42 is safe. Tumor volumes showed a mixed response, but most slowed growth. On a 40-mg regimen, drug concentrated in tumors and growth pathways were suppressed in most tumors, suggesting this may be a well-tolerated and effective dose. A phase 2 study of AR-42 for NF2-associated tumors appears warranted. LEVEL OF EVIDENCE: 1b, 4.

9.
PLoS One ; 16(7): e0252048, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34264955

RESUMEN

Neurofibromatosis Type 2 (NF2) is an autosomal dominant genetic syndrome caused by mutations in the NF2 tumor suppressor gene resulting in multiple schwannomas and meningiomas. There are no FDA approved therapies for these tumors and their relentless progression results in high rates of morbidity and mortality. Through a combination of high throughput screens, preclinical in vivo modeling, and evaluation of the kinome en masse, we identified actionable drug targets and efficacious experimental therapeutics for the treatment of NF2 related schwannomas and meningiomas. These efforts identified brigatinib (ALUNBRIG®), an FDA-approved inhibitor of multiple tyrosine kinases including ALK, to be a potent inhibitor of tumor growth in established NF2 deficient xenograft meningiomas and a genetically engineered murine model of spontaneous NF2 schwannomas. Surprisingly, neither meningioma nor schwannoma cells express ALK. Instead, we demonstrate that brigatinib inhibited multiple tyrosine kinases, including EphA2, Fer and focal adhesion kinase 1 (FAK1). These data demonstrate the power of the de novo unbiased approach for drug discovery and represents a major step forward in the advancement of therapeutics for the treatment of NF2 related malignancies.


Asunto(s)
Neoplasias Meníngeas/genética , Meningioma/genética , Neurilemoma/genética , Neurofibromina 2/deficiencia , Neurofibromina 2/genética , Compuestos Organofosforados/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Proliferación Celular , Humanos , Mutación , Neurilemoma/patología
10.
Cancer Sci ; 101(9): 1997-2004, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20550523

RESUMEN

MicroRNAs (miRNAs) are frequently deregulated in human tumors, and play important roles in tumor development and progression. The pathological roles of miRNAs in neurofibromatosis type 1 (NF1) tumorigenesis are largely unknown. We demonstrated that miR-10b was up-regulated in primary Schwann cells isolated from NF1 neurofibromas and in cell lines and tumor tissues from malignant peripheral nerve sheath tumors (MPNSTs). Intriguingly, a significantly high level of miR-10b correlated with low neurofibromin expression was found in a neuroectodermal cell line: Ewing's sarcoma SK-ES-1 cells. Antisense inhibiting miR-10b in NF1 MPNST cells reduced cell proliferation, migration and invasion. Furthermore, we showed that NF1 mRNA was the target for miR-10b. Overexpression of miR-10b in 293T cells suppressed neurofibromin expression and activated RAS signaling. Antisense inhibition of miR-10b restored neurofibromin expression in SK-ES-1 cells, and decreased RAS signaling independent of neurofibromin in NF1 MPNST cells. These results suggest that miR-10b may play an important role in NF1 tumorigenesis through targeting neurofibromin and RAS signaling.


Asunto(s)
MicroARNs/genética , Neurofibromatosis 1/genética , Transducción de Señal , Proteínas ras/genética , Regiones no Traducidas 3'/genética , Western Blotting , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Neurofibromatosis 1/metabolismo , Neurofibromatosis 1/patología , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , ARN sin Sentido/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Schwann/metabolismo , Células de Schwann/patología , Células Tumorales Cultivadas , Proteínas ras/metabolismo
11.
Mol Cancer Ther ; 19(3): 731-741, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31848295

RESUMEN

Malignant peripheral nerve sheath tumors (MPNST) frequently overexpress eukaryotic initiation factor 4F components, and the eIF4A inhibitor silvestrol potently suppresses MPNST growth. However, silvestrol has suboptimal drug-like properties, including a bulky structure, poor oral bioavailability (<2%), sensitivity to MDR1 efflux, and pulmonary toxicity in dogs. We compared ten silvestrol-related rocaglates lacking the dioxanyl ring and found that didesmethylrocaglamide (DDR) and rocaglamide (Roc) had growth-inhibitory activity comparable with silvestrol. Structure-activity relationship analysis revealed that the dioxanyl ring present in silvestrol was dispensable for, but may enhance, cytotoxicity. Both DDR and Roc arrested MPNST cells at G2-M, increased the sub-G1 population, induced cleavage of caspases and PARP, and elevated the levels of the DNA-damage response marker γH2A.X, while decreasing the expression of AKT and ERK1/2, consistent with translation inhibition. Unlike silvestrol, DDR and Roc were not sensitive to MDR1 inhibition. Pharmacokinetic analysis confirmed that Roc had 50% oral bioavailability. Importantly, Roc, when administered intraperitoneally or orally, showed potent antitumor effects in an orthotopic MPNST mouse model and did not induce pulmonary toxicity in dogs as found with silvestrol. Treated tumors displayed degenerative changes and had more cleaved caspase-3-positive cells, indicative of increased apoptosis. Furthermore, Roc effectively suppressed the growth of osteosarcoma, Ewing sarcoma, and rhabdomyosarcoma cells and patient-derived xenografts. Both Roc- and DDR-treated sarcoma cells showed decreased levels of multiple oncogenic kinases, including insulin-like growth factor-1 receptor. The more favorable drug-like properties of DDR and Roc and the potent antitumor activity of Roc suggest that these rocaglamides could become viable treatments for MPNST and other sarcomas.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Benzofuranos/química , Benzofuranos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neurofibrosarcoma/tratamiento farmacológico , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Aglaia/química , Animales , Apoptosis , Caspasa 3/metabolismo , Ciclo Celular , Proliferación Celular , Humanos , Ratones , Neurofibrosarcoma/metabolismo , Neurofibrosarcoma/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Methods Mol Biol ; 493: 163-77, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18839347

RESUMEN

Recent advances in molecular biology have led to a better understanding of the etiology of vestibular schwannomas. The underlying purpose of vestibular schwannoma research is the development of new treatment options; however, such options have not yet been established. A fundamental understanding of the underlying molecular events leading to tumor formation began when mutations in the neurofibromatosis type 2 (NF2) tumor suppressor gene were identified in vestibular schwannomas. The clinical characteristics of vestibular schwannomas and neurofibromatosis type 2 (NF2) syndromes have both been related to alterations in the NF2 gene. Genetic screening for NF2 is now available. When utilized with clinical screening, such as magnetic resonance imaging (MRI), conventional audiometry, and auditory brainstem response (ABR), the early detection of NF2 can be made, which consequently makes a significant difference in the ability to successfully treat vestibular schwannomas. Additionally, the signaling pathways affected by merlin, the product of the NF2 gene, are becoming better understood. Nf2-transgenic and knockout mice as well as vestibular schwannoma xenograft models are now ready for novel therapeutic testing. Hopefully, better treatment options will be forthcoming soon.


Asunto(s)
Mutación , Neurofibromina 2/genética , Neuroma Acústico/genética , Animales , Análisis Mutacional de ADN , Predisposición Genética a la Enfermedad , Humanos , Imagen por Resonancia Magnética , Ratones , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Neurofibromatosis 2/genética , Neurofibromatosis 2/patología , Neuroma Acústico/patología
13.
Neuro Oncol ; 21(4): 486-497, 2019 03 18.
Artículo en Inglés | MEDLINE | ID: mdl-30615146

RESUMEN

BACKGROUND: Neurofibromatosis type 2 (NF2) is a genetic tumor-predisposition disorder caused by NF2/merlin tumor suppressor gene inactivation. The hallmark of NF2 is formation of bilateral vestibular schwannomas (VS). Because merlin modulates activity of the Ras/Raf/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway, we investigated repurposing drugs targeting MEK1 and/or MEK2 as a treatment for NF2-associated schwannomas. METHODS: Mouse and human merlin-deficient Schwann cell lines (MD-MSC/HSC) were screened against 6 MEK1/2 inhibitors. Efficacious drugs were tested in orthotopic allograft and NF2 transgenic mouse models. Pathway and proteome analyses were conducted. Drug efficacy was examined in primary human VS cells with NF2 mutations and correlated with DNA methylation patterns. RESULTS: Trametinib, PD0325901, and cobimetinib were most effective in reducing MD-MSC/HSC viability. Each decreased phosphorylated pERK1/2 and cyclin D1, increased p27, and induced caspase-3 cleavage in MD-MSCs. Proteomic analysis confirmed cell cycle arrest and activation of pro-apoptotic pathways in trametinib-treated MD-MSCs. The 3 inhibitors slowed allograft growth; however, decreased pERK1/2, cyclin D1, and Ki-67 levels were observed only in PD0325901 and cobimetinib-treated grafts. Tumor burden and average tumor size were reduced in trametinib-treated NF2 transgenic mice; however, tumors did not exhibit reduced pERK1/2 levels. Trametinib and PD0325901 modestly reduced viability of several primary human VS cell cultures with NF2 mutations. DNA methylation analysis of PD0325901-resistant versus -susceptible VS identified genes that could contribute to drug resistance. CONCLUSION: MEK inhibitors exhibited differences in antitumor efficacy resistance in schwannoma models with possible emergence of trametinib resistance. The results support further investigation of MEK inhibitors in combination with other targeted drugs for NF2 schwannomas.


Asunto(s)
Azetidinas/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Neuroma Acústico , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridonas/farmacología , Pirimidinonas/farmacología , Animales , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Ratones , Neurofibromatosis 2/complicaciones , Neuroma Acústico/etiología
14.
BMC Cell Biol ; 9: 1, 2008 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-18186933

RESUMEN

BACKGROUND: Drosophila Merlin, the homolog of the human Neurofibromatosis 2 (NF2) gene, is important for the regulation of cell proliferation and receptor endocytosis. Male flies carrying a Mer3 allele, a missense mutation (Met177-->Ile) in the Merlin gene, are viable but sterile; however, the cause of sterility is unknown. RESULTS: Testis examination reveals that hemizygous Mer3 mutant males have small seminal vesicles that contain only a few immotile sperm. By cytological and electron microscopy analyses of the Mer3, Mer4 (Gln170-->stop), and control testes at various stages of spermatogenesis, we show that Merlin mutations affect meiotic cytokinesis of spermatocytes, cyst polarization and nuclear shaping during spermatid elongation, and spermatid individualization. We also demonstrate that the lethality and sterility phenotype of the Mer4 mutant is rescued by the introduction of a wild-type Merlin gene. Immunostaining demonstrates that the Merlin protein is redistributed to the area associated with the microtubules of the central spindle in telophase and its staining is less in the region of the contractile ring during meiotic cytokinesis. At the onion stage, Merlin is concentrated in the Nebenkern of spermatids, and this mitochondrial localization is maintained throughout sperm formation. Also, Merlin exhibits punctate staining in the acrosomal region of mature sperm. CONCLUSION: Merlin mutations affect spermatogenesis at multiple stages. The Merlin protein is dynamically redistributed during meiosis of spermatocytes and is concentrated in the Nebenkern of spermatids. Our results demonstrated for the first time the mitochondrial localization of Merlin and suggest that Merlin may play a role in mitochondria formation and function during spermatogenesis.


Asunto(s)
Drosophila/genética , Infertilidad Masculina/genética , Proteínas de la Membrana/genética , Mitocondrias/metabolismo , Neurofibromina 2/genética , Espermatogénesis/genética , Animales , Masculino , Mitocondrias/genética , Mutación Missense
15.
Otol Neurotol ; 29(1): 58-68, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18199958

RESUMEN

HYPOTHESIS: The neurofibromatosis 2 gene, which encodes the tumor suppressor protein merlin, is frequently mutated in vestibular schwannomas (VS). Merlin can inhibit phosphatidylinositol 3 kinase (PI3 kinase) by binding to PI3 kinase enhancer long isoform. Therefore, we hypothesized that the PI3 kinase/AKT pathway is activated in VS. BACKGROUND: Despite advances in diagnosis and treatment, VS continue to cause patient morbidity. A more thorough understanding of the signaling pathways deregulated in VS will aid in the development of novel medical therapeutics. Activation of the PI3 kinase/AKT pathway increases cell survival and cell proliferation and has been observed in a variety of human cancers. However, whether the PI3 kinase/AKT pathway is activated in human VS has not been reported. METHODS: Complementary deoxyribonucleic acid microarrays were performed using cultured Schwann cells, 4 VS specimens, and 2 paired normal vestibular nerves. Immunohistochemical analysis using antibodies to activated phosphorylated-AKT was performed on 14 VS tissue sections. Western blots using various antibodies to components of the PI3 kinase/AKT pathways were conducted. RESULTS: Microarray analysis demonstrated that total AKT gene expression was upregulated in VS, compared with normal vestibular nerves. Immunohistochemical analysis of 14 VS tissue sections detected positive staining for activated AKT phosphorylated at both serine-473 and threonine-308 in all tumors. Western blots comparing VS specimens with normal vestibular nerves showed that the AKT pathway was activated in VS but not in normal nerve. Total AKT, phosphorylated-AKT, PI3-kinase, phosphorylated-phosphatase and tensin homologue deleted on chromosome 10, phosphorylated-phosphoinositide-dependent protein kinase 1, phosphorylated-forkhead box O, phosphorylated-glycogen synthase kinase 3beta, and phosphorylated-mammalian target of rapamycin were upregulated in VS. CONCLUSION: The PI3 kinase/AKT pathway is activated in VS. Using our recently reported quantifiable VS xenograft model, novel inhibitors of the PI3 kinase/AKT pathway may be tested for VS growth inhibition in vivo.


Asunto(s)
Neuroma Acústico/genética , Proteína Oncogénica v-akt/genética , Neoplasias del Sistema Nervioso Periférico/genética , Fosfatidilinositol 3-Quinasas/genética , Transducción de Señal/fisiología , Western Blotting , Colforsina/farmacología , ADN de Neoplasias/genética , Humanos , Inmunohistoquímica , Neuroma Acústico/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Adhesión en Parafina , Neoplasias del Sistema Nervioso Periférico/patología , Tubulina (Proteína)/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Exp Neurol ; 299(Pt B): 299-307, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28610844

RESUMEN

Meningiomas frequently display activation of the PI3K/AKT/mTOR pathway, leading to elevated levels of phospho-eukaryotic translation initiation factor 4E binding proteins, which enhances protein synthesis; however, it is not known whether inhibition of protein translation is an effective treatment option for meningiomas. We found that human meningiomas expressed high levels of the three components of the eukaryotic initiation factor 4F (eIF4F) translation initiation complex, eIF4A, eIF4E, and eIF4G. The expression of eIF4A and eIF4E was important in sustaining the growth of NF2-deficient benign meningioma Ben-Men-1 cells, as shRNA-mediated knockdown of these proteins strongly reduced cell proliferation. Among a series of 23 natural compounds evaluated, silvestrol, which inhibits eIF4A, was identified as being the most growth inhibitory in both primary meningioma and Ben-Men-1 cells. Silvestrol treatment of meningioma cells prominently induced G2/M arrest. Consistently, silvestrol significantly decreased the amounts of cyclins D1, E1, A, and B, PCNA, and Aurora A. In addition, total and phosphorylated AKT, ERK, and FAK, which have been shown to be important drivers for meningioma cell proliferation, were markedly lower in silvestrol-treated Ben-Men-1 cells. Our findings suggest that inhibiting protein translation could be a potential treatment for meningiomas.


Asunto(s)
Antineoplásicos/farmacología , Factor 4A Eucariótico de Iniciación/biosíntesis , Factor 4E Eucariótico de Iniciación/biosíntesis , Factor 4G Eucariótico de Iniciación/biosíntesis , Neoplasias Meníngeas/tratamiento farmacológico , Meningioma/tratamiento farmacológico , Proteínas de Neoplasias/biosíntesis , Iniciación de la Cadena Peptídica Traduccional/efectos de los fármacos , Triterpenos/farmacología , Antineoplásicos/uso terapéutico , Aurora Quinasa A/biosíntesis , Aurora Quinasa A/genética , Ciclinas/biosíntesis , Ciclinas/genética , Ensayos de Selección de Medicamentos Antitumorales , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Factor 4A Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/genética , Factor 4G Eucariótico de Iniciación/genética , Femenino , Fase G2/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Meningioma/genética , Meningioma/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Antígeno Nuclear de Célula en Proliferación/biosíntesis , Antígeno Nuclear de Célula en Proliferación/genética , ARN Interferente Pequeño/farmacología , Triterpenos/uso terapéutico , Células Tumorales Cultivadas
17.
Neuro Oncol ; 20(9): 1185-1196, 2018 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-29982664

RESUMEN

Background: Meningiomas are the most common primary brain tumor in adults, and somatic loss of the neurofibromatosis 2 (NF2) tumor suppressor gene is a frequent genetic event. There is no effective treatment for tumors that recur or continue to grow despite surgery and/or radiation. Therefore, targeted therapies that either delay tumor progression or cause tumor shrinkage are much needed. Our earlier work established mammalian target of rapamycin complex mTORC1/mTORC2 activation in NF2-deficient meningiomas. Methods: High-throughput kinome analyses were performed in NF2-null human arachnoidal and meningioma cell lines to identify functional kinome changes upon NF2 loss. Immunoblotting confirmed the activation of kinases and demonstrated effectiveness of drugs to block the activation. Drugs, singly and in combination, were screened in cells for their growth inhibitory activity. Antitumor drug efficacy was tested in an orthotopic meningioma model. Results: Erythropoietin-producing hepatocellular receptor tyrosine kinases (EPH RTKs), c-KIT, and Src family kinase (SFK) members, which are biological targets of dasatinib, were among the top candidates activated in NF2-null cells. Dasatinib significantly inhibited phospho-EPH receptor A2 (pEPHA2), pEPHB1, c-KIT, and Src/SFK in NF2-null cells, showing no cross-talk with mTORC1/2 signaling. Posttreatment kinome analyses showed minimal adaptive changes. While dasatinib treatment showed some activity, dual mTORC1/2 inhibitor and its combination with dasatinib elicited stronger growth inhibition in meningiomas. Conclusion: Co-targeting mTORC1/2 and EPH RTK/SFK pathways could be a novel effective treatment strategy for NF2-deficient meningiomas.


Asunto(s)
Antineoplásicos/farmacología , Biomarcadores de Tumor/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Meníngeas/patología , Meningioma/patología , Neurofibromina 2/deficiencia , Receptores de la Familia Eph/antagonistas & inhibidores , Animales , Apoptosis , Proliferación Celular , Humanos , Neoplasias Meníngeas/tratamiento farmacológico , Neoplasias Meníngeas/metabolismo , Meningioma/tratamiento farmacológico , Meningioma/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptores de la Familia Eph/genética , Receptores de la Familia Eph/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
PLoS One ; 13(6): e0197350, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29897904

RESUMEN

Neurofibromatosis 2 (NF2) is a rare tumor suppressor syndrome that manifests with multiple schwannomas and meningiomas. There are no effective drug therapies for these benign tumors and conventional therapies have limited efficacy. Various model systems have been created and several drug targets have been implicated in NF2-driven tumorigenesis based on known effects of the absence of merlin, the product of the NF2 gene. We tested priority compounds based on known biology with traditional dose-concentration studies in meningioma and schwann cell systems. Concurrently, we studied functional kinome and gene expression in these cells pre- and post-treatment to determine merlin deficient molecular phenotypes. Cell viability results showed that three agents (GSK2126458, Panobinostat, CUDC-907) had the greatest activity across schwannoma and meningioma cell systems, but merlin status did not significantly influence response. In vivo, drug effect was tumor specific with meningioma, but not schwannoma, showing response to GSK2126458 and Panobinostat. In culture, changes in both the transcriptome and kinome in response to treatment clustered predominantly based on tumor type. However, there were differences in both gene expression and functional kinome at baseline between meningioma and schwannoma cell systems that may form the basis for future selective therapies. This work has created an openly accessible resource (www.synapse.org/SynodosNF2) of fully characterized isogenic schwannoma and meningioma cell systems as well as a rich data source of kinome and transcriptome data from these assay systems before and after treatment that enables single and combination drug discovery based on molecular phenotype.


Asunto(s)
Neoplasias Meníngeas/genética , Neurilemoma/genética , Neurofibromatosis 2/genética , Neurofibromina 2/genética , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Meníngeas/tratamiento farmacológico , Neoplasias Meníngeas/patología , Ratones , Morfolinas/farmacología , Neurilemoma/tratamiento farmacológico , Neurilemoma/patología , Neurofibromatosis 2/tratamiento farmacológico , Neurofibromatosis 2/patología , Panobinostat/farmacología , Piridazinas , Pirimidinas/farmacología , Quinolinas/farmacología , Sulfonamidas/farmacología , Biología de Sistemas , Transcriptoma/genética
19.
Mol Cell Biol ; 23(21): 7585-99, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14560005

RESUMEN

The TAL1 (or SCL) gene, originally identified from its involvement by a recurrent chromosomal translocation, encodes a basic helix-loop-helix transcription factor essential for erythropoiesis. Although presumed to regulate transcription, its target genes are largely unknown. We show here that a nuclear complex containing TAL1, its DNA-binding partner E47, zinc finger transcription factor GATA-1, LIM domain protein LMO2, and LIM domain-binding protein Ldb1 transactivates the protein 4.2 (P4.2) gene through two E box GATA elements in its proximal promoter. Binding of this complex to DNA was dependent on the integrity of both E box and GATA sites and was demonstrated to occur on the P4.2 promoter in cells. Maximal transcription in transiently transfected cells required both E box GATA elements and expression of all five components of the complex. This complex was shown, in addition, to be capable of linking in solution double-stranded oligonucleotides corresponding to the two P4.2 E box GATA elements. This DNA-linking activity required Ldb1 and increased with dimethyl sulfoxide-induced differentiation of murine erythroleukemia (MEL) cells. In contrast, enforced expression in MEL cells of dimerization-defective mutant Ldb1, as well as wild-type Ldb1, significantly decreased E box GATA DNA-binding activities, P4.2 promoter activity, and accumulation of P4.2 and beta-globin mRNAs. These studies define a physiologic target for a TAL1- and GATA-1-containing ternary complex and reveal a positive role for Ldb1 in erythroid gene expression and differentiation.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Diferenciación Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Eritrocitos/metabolismo , Factores de Unión al ADN Específico de las Células Eritroides , Factor de Transcripción GATA1 , Genes Reguladores , Secuencias Hélice-Asa-Hélice , Proteínas con Dominio LIM , Leucemia Eritroblástica Aguda , Proteínas de la Membrana , Ratones , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Proteína 1 de la Leucemia Linfocítica T Aguda , Factores de Transcripción/genética , Células Tumorales Cultivadas
20.
Curr Opin Otolaryngol Head Neck Surg ; 15(5): 341-6, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17823551

RESUMEN

PURPOSE OF REVIEW: To summarize advances in understanding the molecular biology of vestibular schwannomas over the past year. RECENT FINDINGS: The role of the neurofibromatosis type 2 protein, denoted as merlin or schwannomin, in embryonic development, cellular adherence, and in cell proliferation has become better elucidated in the past year. Likewise, the role of merlin in Schwann cell-axon interaction has been studied. Additionally, two comprehensive analyses of the spectrum of human neurofibromatosis type 2 mutations have been compiled which make up a valuable resource in understanding critical regions of the neurofibromatosis type 2 gene. Neurofibromatosis type 2 screening guidelines for young patients with solitary vestibular schwannomas have been published. The role of electromagnetic radiation via cellular and portable telephones as a predisposing factor to vestibular schwannoma formation has also been the topic of several studies. Based on increased knowledge of the pathways in which merlin functions and the available transgenic and xenograft mouse models, preliminary data regarding directed pharmacotherapy are also summarized. SUMMARY: With increased knowledge of the pathologic mechanisms and interacting proteins associated with merlin, the research community is poised to begin trials of targeted interventions in vitro and in the current mouse models.


Asunto(s)
Neuroma Acústico/genética , Animales , Teléfono Celular , Desarrollo Embrionario , Pruebas Genéticas , Humanos , Mutación , Neurofibromatosis 2/genética , Neurofibromina 2/metabolismo , Neuroma Acústico/tratamiento farmacológico , Análisis de Secuencia por Matrices de Oligonucleótidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA