Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
J Biol Chem ; 295(29): 9948-9958, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32471868

RESUMEN

Neurofibromatosis type 1 (NF1) is a common cancer predisposition syndrome caused by mutations in the NF1 tumor suppressor gene. NF1 encodes neurofibromin, a GTPase-activating protein for RAS proto-oncogene GTPase (RAS). Plexiform neurofibromas are a hallmark of NF1 and result from loss of heterozygosity of NF1 in Schwann cells, leading to constitutively activated p21RAS. Given the inability to target p21RAS directly, here we performed an shRNA library screen of all human kinases and Rho-GTPases in a patient-derived NF1-/- Schwann cell line to identify novel therapeutic targets to disrupt PN formation and progression. Rho family members, including Rac family small GTPase 1 (RAC1), were identified as candidates. Corroborating these findings, we observed that shRNA-mediated knockdown of RAC1 reduces cell proliferation and phosphorylation of extracellular signal-regulated kinase (ERK) in NF1-/- Schwann cells. Genetically engineered Nf1flox/flox;PostnCre+ mice, which develop multiple PNs, also exhibited increased RAC1-GTP and phospho-ERK levels compared with Nf1flox/flox;PostnCre- littermates. Notably, mice in which both Nf1 and Rac1 loci were disrupted (Nf1flox/floxRac1flox/flox;PostnCre+) were completely free of tumors and had normal phospho-ERK activity compared with Nf1flox/flox ;PostnCre+ mice. We conclude that the RAC1-GTPase is a key downstream node of RAS and that genetic disruption of the Rac1 allele completely prevents PN tumor formation in vivo in mice.


Asunto(s)
Técnicas de Silenciamiento del Gen , Neoplasias Primarias Secundarias , Neurofibroma Plexiforme , Neurofibromatosis 1 , Neuropéptidos/deficiencia , Proteína de Unión al GTP rac1/deficiencia , Animales , Ratones , Ratones Noqueados , Neoplasias Primarias Secundarias/enzimología , Neoplasias Primarias Secundarias/genética , Neoplasias Primarias Secundarias/patología , Neoplasias Primarias Secundarias/prevención & control , Neurofibroma Plexiforme/enzimología , Neurofibroma Plexiforme/genética , Neurofibroma Plexiforme/prevención & control , Neurofibromatosis 1/enzimología , Neurofibromatosis 1/genética , Neurofibromatosis 1/patología , Neurofibromina 1/deficiencia , Neurofibromina 1/metabolismo , Neuropéptidos/metabolismo , Proto-Oncogenes Mas , Proteína de Unión al GTP rac1/metabolismo
2.
Curr Opin Oncol ; 33(2): 110-119, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33395032

RESUMEN

PURPOSE OF REVIEW: An early understanding of the role of the Ras/Raf/MEK/ERK signalling pathway in regulating cell proliferation has set the stage for the development of several potent and selective MEK inhibitors (MEKi). MEKi represent promising therapies for RAS-driven neoplasias and RASopathies associated with increased Ras/MAPK activity. RECENT FINDINGS: Neurofibromatosis 1 (NF1) is a prototypic RASopathy in which early-phase clinical trials with MEKi have been successful in the treatment of plexiform neurofibromas (pNF) and low-grade gliomas (LGGs). The phase 2 trial (SPRINT) of selumetinib in pNF resulted in at least 20% reduction in the size of pNF from baseline in 71% of patients and was associated with clinically meaningful improvements. On the basis of this trial, selumetinib (Koselugo) received FDA approval for children 2 years of age and older with inoperable, symptomatic pNF. The phase 2 trial of selumetinib in LGG resulted in 40% partial response and 96% of patients had 2 years of progression-free survival. SUMMARY: Given the potential of MEK inhibition as an effective and overall well tolerated medical treatment, the use of targeted agents in the NF1 population is likely to increase considerably. Future work on non-NF1 RASopathies should focus on developing preclinical models and defining endpoints for measurement of efficacy in order to conduct clinical trials.


Asunto(s)
Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Neurofibroma Plexiforme/tratamiento farmacológico , Neurofibromatosis 1/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Animales , Ensayos Clínicos Fase II como Asunto , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Neurofibroma Plexiforme/enzimología , Neurofibroma Plexiforme/metabolismo , Neurofibromatosis 1/enzimología , Neurofibromatosis 1/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Ensayos Clínicos Controlados Aleatorios como Asunto , Proteínas ras/metabolismo
3.
Hum Mol Genet ; 23(25): 6712-21, 2014 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-25070947

RESUMEN

Neurofibromatosis type 1 (NF1) is a common neurodevelopmental disorder in which affected individuals are prone to learning, attention and behavioral problems. Previous studies in mice and flies have yielded conflicting results regarding the specific effector pathways responsible for NF1 protein (neurofibromin) regulation of neuronal function, with both cyclic AMP (cAMP)- and RAS-dependent mechanisms described. Herein, we leverage a combination of induced pluripotent stem cell-derived NF1 patient neural progenitor cells and Nf1 genetically engineered mice to establish, for the first time, that neurofibromin regulation of cAMP requires RAS activation in human and mouse neurons. However, instead of involving RAS-mediated MEK/AKT signaling, RAS regulation of cAMP homeostasis operates through the activation of atypical protein kinase C zeta, leading to GRK2-driven Gαs inactivation. These findings reveal a novel mechanism by which RAS can regulate cAMP levels in the mammalian brain.


Asunto(s)
AMP Cíclico/metabolismo , Células-Madre Neurales/enzimología , Neurofibromatosis 1/genética , Neurofibromina 1/genética , Neuronas/enzimología , Proteína Quinasa C/genética , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Animales , Diferenciación Celular , Cromograninas , Activación Enzimática , Quinasa 2 del Receptor Acoplado a Proteína-G/genética , Quinasa 2 del Receptor Acoplado a Proteína-G/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Regulación de la Expresión Génica , Hipocampo/enzimología , Hipocampo/patología , Humanos , Células Madre Pluripotentes Inducidas/enzimología , Células Madre Pluripotentes Inducidas/patología , Ratones , Ratones Transgénicos , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Células-Madre Neurales/patología , Neurofibromatosis 1/enzimología , Neurofibromatosis 1/patología , Neurofibromina 1/metabolismo , Neuronas/patología , Cultivo Primario de Células , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , Transducción de Señal , Proteínas ras/metabolismo
4.
Proc Natl Acad Sci U S A ; 110(12): 4574-9, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23487764

RESUMEN

Defects in the RAS small G protein or its associated network of regulatory proteins that disrupt GTPase cycling are a major cause of cancer and developmental RASopathy disorders. Lack of robust functional assays has been a major hurdle in RAS pathway-targeted drug development. We used NMR to obtain detailed mechanistic data on RAS cycling defects conferred by oncogenic mutations, or full-length RASopathy-derived regulatory proteins. By monitoring the conformation of wild-type and oncogenic RAS in real-time, we show that opposing properties integrate with regulators to hyperactivate oncogenic RAS mutants. Q61L and G13D exhibited rapid nucleotide exchange and an unexpected susceptibility to GAP-mediated hydrolysis, in direct contrast with G12V, indicating different approaches must be taken to inhibit these oncoproteins. An NMR methodology was established to directly monitor RAS cycling by intact, multidomain proteins encoded by RASopathy genes in mammalian cell extracts. By measuring GAP activity from tumor cells, we demonstrate how loss of neurofibromatosis type 1 (NF1) increases RAS-GTP levels in NF1-derived cells. We further applied this methodology to profile Noonan Syndrome (NS)-derived SOS1 mutants. Combining NMR with cell-based assays allowed us to differentiate defects in catalysis, allosteric regulation, and membrane targeting of individual mutants, while revealing a membrane-dependent compensatory effect that attenuates dramatic increases in RAS activation shown by Y337C, L550P, and I252T. Our NMR method presents a precise and robust measure of RAS activity, providing mechanistic insights that facilitate discovery of therapeutics targeted against the RAS signaling network.


Asunto(s)
Mutación Missense , Neoplasias/metabolismo , Neurofibromatosis 1/enzimología , Proteína Oncogénica p21(ras)/metabolismo , Transducción de Señal , Sustitución de Aminoácidos , Línea Celular Tumoral , Activación Enzimática/genética , Células HEK293 , Humanos , Neoplasias/química , Neoplasias/genética , Neurofibromatosis 1/genética , Neurofibromatosis 1/patología , Resonancia Magnética Nuclear Biomolecular , Proteína Oncogénica p21(ras)/química , Proteína Oncogénica p21(ras)/genética
5.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 32(3): 318-22, 2015 Jun.
Artículo en Zh | MEDLINE | ID: mdl-26037340

RESUMEN

OBJECTIVE: To identify the genetic etiology in a Chinese patient with neurofibromatosis type 1 (NF-1). METHODS: All coding exons and the flanking sequences of neurofibromin 1 (NF1) gene from the patient were captured, individually barcoded and subjected to HiSeq2000 high-throughput sequencing. Suspected mutation was validated in the nuclear family members with Sanger sequencing. RESULTS: A novel indel mutation, c.789_790delAGinsT, was identified in the exon 8 of the NF1 gene in the patient but not in her asymptomatic parents. The mutation was predicted to have caused shifting of the reading frame and a premature downstream stop codon (p.K263Nfs*18). Two known polymorphisms, c.888+108 C>T (rs2953000) and c.888+118 G>T (rs2952999), was detected in the flanking of the indel mutation in the patient and her father. Sequencing chromatogram for the family indicates that above changes are located on the same chromosome. CONCLUSION: The c.789_790delAGinsT, as a de novo mutation occurring on the paternally derived chromosome, is most likely to be causative for the disease. Compared with Sanger sequencing, targeted next-generation sequencing is more efficient and can dramatically reduce the cost for the genetic testing of NF-1.


Asunto(s)
Neurofibromatosis 1/enzimología , Neurofibromina 1/genética , Mutación Puntual , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Femenino , Humanos , Datos de Secuencia Molecular , Neurofibromatosis 1/genética , Neurofibromina 1/metabolismo
6.
Hum Mol Genet ; 19(22): 4515-28, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20826448

RESUMEN

Learning and behavioral abnormalities are among the most common clinical problems in children with the neurofibromatosis-1 (NF1) inherited cancer syndrome. Recent studies using Nf1 genetically engineered mice (GEM) have been instructive for partly elucidating the cellular and molecular defects underlying these cognitive deficits; however, no current model has shed light on the more frequently encountered attention system abnormalities seen in children with NF1. Using an Nf1 optic glioma (OPG) GEM model, we report novel defects in non-selective and selective attention without an accompanying hyperactivity phenotype. Specifically, Nf1 OPG mice exhibit reduced rearing in response to novel objects and environmental stimuli. Similar to children with NF1, the attention system dysfunction in these mice is reversed by treatment with methylphenidate (MPH), suggesting a defect in brain catecholamine homeostasis. We further demonstrate that this attention system abnormality is the consequence of reduced dopamine (DA) levels in the striatum, which is normalized following either MPH or l-dopa administration. The reduction in striatal DA levels in Nf1 OPG mice is associated with reduced striatal expression of tyrosine hydroxylase, the rate-limited enzyme in DA synthesis, without any associated dopaminergic cell loss in the substantia nigra. Moreover, we demonstrate a cell-autonomous defect in Nf1+/- dopaminergic neuron growth cone areas and neurite extension in vitro, which results in decreased dopaminergic cell projections to the striatum in Nf1 OPG mice in vivo. Collectively, these data establish abnormal DA homeostasis as the primary biochemical defect underlying the attention system dysfunction in Nf1 GEM relevant to children with NF1.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/genética , Atención , Dopamina/metabolismo , Neurofibromatosis 1/genética , Neurofibromatosis 1/metabolismo , Animales , Encéfalo/metabolismo , Niño , Cuerpo Estriado/metabolismo , Dopamina/genética , Genes de Neurofibromatosis 1 , Humanos , Levodopa/genética , Levodopa/metabolismo , Metilfenidato/metabolismo , Metilfenidato/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Actividad Motora/genética , Neurofibromatosis 1/enzimología , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Neuronas/metabolismo , Glioma del Nervio Óptico/genética , Glioma del Nervio Óptico/metabolismo , Sustancia Negra/metabolismo
8.
Cell Death Dis ; 12(6): 513, 2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-34011935

RESUMEN

Neurofibromatosis type 1 is an autosomal dominant genetic disorder caused by mutation in the neurofibromin 1 (NF1) gene. Its hallmarks are cutaneous findings including neurofibromas, benign peripheral nerve sheath tumors. We analyzed the collagen and matrix metalloproteinase 1 (MMP1) expression in Neurofibromatosis 1 cutaneous neurofibroma and found excessive expression of collagen and reduced expression of MMP1. To identify new therapeutic drugs for neurofibroma, we analyzed phosphorylation of components of the Ras pathway, which underlies NF1 regulation, and applied treatments to block this pathway (PD184352, U0126, and rapamycin) and lysosomal processes (chloroquine (CQ), hydroxychloroquine (HCQ), and bafilomycin A (BafA)) in cultured Neurofibromatosis 1 fibroblasts. We found that downregulation of the MMP1 protein was a key abnormal feature in the neurofibromatosis 1 fibroblasts and that the decreased MMP1 was restored by the lysosomal blockers CQ and HCQ, but not by the blockers of the Ras pathway. Moreover, the MMP1-upregulating activity of those lysosomal blockers was dependent on aryl hydrocarbon receptor (AHR) activation and ERK phosphorylation. Our findings suggest that lysosomal blockers are potential candidates for the treatment of Neurofibromatosis 1 neurofibroma.


Asunto(s)
Cloroquina/farmacología , Hidroxicloroquina/farmacología , Metaloproteinasa 1 de la Matriz/metabolismo , Neurofibromatosis 1/tratamiento farmacológico , Antimaláricos/farmacología , Células Cultivadas , Regulación hacia Abajo , Fibroblastos , Humanos , Neurofibromatosis 1/enzimología , Neurofibromatosis 1/patología , Transfección
9.
J Clin Oncol ; 39(7): 797-806, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33507822

RESUMEN

PURPOSE: Patients with neurofibromatosis type 1 (NF1) frequently develop plexiform neurofibromas (PNs), which can cause significant morbidity. We performed a phase II trial of the MAPK/ERK kinase inhibitor, mirdametinib (PD-0325901), in patients with NF1 and inoperable PNs. The primary objective was response rate based on volumetric magnetic resonance imaging analysis. METHODS: Inclusion criteria included age ≥ 16 years and a PN that was either progressive or causing significant morbidity. First-dose pharmacokinetics were performed. Patients completed patient-reported outcome measures. Patients received mirdametinib by mouth twice a day at 2 mg/m2/dose (maximum dose = 4 mg twice a day) in a 3-week on/1-week off sequence. Each course was 4 weeks in duration. Evaluations were performed after four courses for the first year and then after every six courses. Patients could receive a maximum of 24 total courses. RESULTS: Nineteen patients were enrolled, and all 19 received mirdametinib. The median age was 24 years (range, 16-39 years); the median baseline tumor volume was 363.8 mL (range, 3.9-5,161 mL). Eight of the 19 patients (42%) achieved a partial response of the target PN by course 12, and 10 (53%) had stable disease. One patient (5%) developed progressive disease at course 8. Significant and durable decreases were observed in pain ratings. CONCLUSION: To our knowledge, this analysis represents the first characterization of the activity and pharmacokinetics of mirdametinib in patients with NF1 and PNs and is the first published response study for MAPK/ERK kinase inhibitors in adults with NF1 and PNs. Mirdametinib given at 2 mg/m2/dose (maximum dose, 4 mg) twice daily in a 3-week on/1-week off sequence resulted in a 42% partial response rate with preliminary evidence of reduction in pain.


Asunto(s)
Benzamidas/uso terapéutico , Difenilamina/análogos & derivados , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Neurofibroma Plexiforme/tratamiento farmacológico , Neurofibromatosis 1/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Adolescente , Adulto , Benzamidas/efectos adversos , Benzamidas/farmacocinética , Difenilamina/efectos adversos , Difenilamina/farmacocinética , Difenilamina/uso terapéutico , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Neurofibroma Plexiforme/diagnóstico por imagen , Neurofibroma Plexiforme/enzimología , Neurofibromatosis 1/diagnóstico por imagen , Neurofibromatosis 1/enzimología , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/farmacocinética , Factores de Tiempo , Resultado del Tratamiento , Estados Unidos , Adulto Joven
10.
Protein Expr Purif ; 65(1): 30-7, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19111619

RESUMEN

The protein giant neurofibromin (320kDa) is the protein product of the NF1 tumor suppressor gene, alterations of which are responsible for the pathogenesis of neurofibromatosis type 1 (NF1). Neurofibromin is a Ras-specific GTPase activating protein (RasGAP) that, 15 years after the cloning of the gene, remains the only clearly defined function of the protein. In a structural proteomics approach, we aimed at defining functions beyond RasGAP activity based on the discovery of structural modules. Given the poor outcome of domain prediction tools, we have undertaken a fragment solubility survey covering the full protein sequence, with the aim of defining new domain boundaries or fragments that could be investigated by biochemical methods including structural analysis. More than 200 constructs have been expressed and tested for solubility in small scale assays. Boundaries were chosen based upon secondary structure predictions, sequence conservation among neurofibromin orthologues and chemical properties of amino acids. Using this strategy we recently discovered a novel bipartite module in neurofibromin. We have expanded our study to include ESPRIT, a library-based construct screen, to perform fragment testing at a finer level with respect to the choice of terminal residues. Our study confirms earlier notions about the challenges neurofibromin presents to the biochemist and points to strategies whereby the success rate may be enhanced in the future.


Asunto(s)
Neurofibromatosis 1/enzimología , Neurofibromina 1/química , Humanos , Neurofibromina 1/biosíntesis , Neurofibromina 1/genética , Neurofibromina 1/aislamiento & purificación , Estabilidad Proteica , Estructura Terciaria de Proteína/fisiología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Homología de Secuencia de Aminoácido , Solubilidad
11.
J Pharmacol Exp Ther ; 326(1): 1-11, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18367665

RESUMEN

Neurofibromatosis type 1 (NF1) is a genetic disorder that is driven by the loss of neurofibromin (Nf) protein function. Nf contains a Ras-GTPase-activating protein domain, which directly regulates Ras signaling. Numerous clinical manifestations are associated with the loss of Nf and increased Ras activity. Ras proteins must be prenylated to traffic and functionally localize with target membranes. Hence, Ras is a potential therapeutic target for treating NF1. We have tested the efficacy of two novel farnesyl transferase inhibitors (FTIs), 1 and 2, alone or in combination with lovastatin, on two NF1 malignant peripheral nerve sheath tumor (MPNST) cell lines, NF90-8 and ST88-14. Single treatments of 1, 2, or lovastatin had no effect on Ras prenylation or MPNST cell proliferation. However, low micromolar combinations of 1 or 2 with lovastatin (FTI/lovastatin) reduced Ras prenylation in both MPNST cell lines. Furthermore, this FTI/lovastatin combination treatment reduced cell proliferation and induced an apoptotic response as shown by morphological analysis, procaspase-3/-7 activation, loss of mitochondrial membrane potential, and accumulation of cells with sub-G(1) DNA content. Little to no detectable toxicity was observed in normal rat Schwann cells following FTI/lovastatin combination treatment. These data support the hypothesis that combination FTI plus lovastatin therapy may be a potential treatment for NF1 MPNSTs.


Asunto(s)
Apoptosis/efectos de los fármacos , Farnesiltransferasa/antagonistas & inhibidores , Lovastatina/administración & dosificación , Neoplasias de la Vaina del Nervio/tratamiento farmacológico , Neoplasias de la Vaina del Nervio/patología , Neurofibromatosis 1/tratamiento farmacológico , Neurofibromatosis 1/patología , Animales , Animales Recién Nacidos , Apoptosis/fisiología , Línea Celular , Línea Celular Transformada , Línea Celular Tumoral , Quimioterapia Combinada , Inhibidores Enzimáticos/administración & dosificación , Farnesiltransferasa/metabolismo , Lovastatina/química , Neurofibromatosis 1/enzimología , Ratas , Ratas Sprague-Dawley
12.
Cancer Res ; 65(21): 9843-50, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16267007

RESUMEN

Individuals affected with the neurofibromatosis 1 (NF1) tumor predisposition syndrome are prone to the development of multiple nervous system tumors, including optic pathway gliomas (OPG). The NF1 tumor suppressor gene product, neurofibromin, functions as a Ras GTPase-activating protein, and has been proposed to regulate cell growth by inhibiting Ras activity. Recent studies from our laboratory have shown that neurofibromin also regulates the mammalian target of rapamycin activity in a Ras-dependent fashion, and that the rapamycin-mediated mammalian target of rapamycin inhibition ameliorates the Nf1-/- astrocyte growth advantage. Moreover, Nf1-deficient astrocytes exhibit increased protein translation. As part of a larger effort to identify protein markers for NF1-associated astrocytomas that could be exploited for therapeutic drug design, we did an objective proteomic analysis of the cerebrospinal fluid from genetically engineered Nf1 mice with optic glioma. One of the proteins found to be increased in the cerebrospinal fluid of OPG-bearing mice was the eukaryotic initiation factor-2alpha binding protein, methionine aminopeptidase 2 (MetAP2). In this study, we show that Nf1 mouse OPGs and NF1-associated human astrocytic tumors, but not sporadic pilocytic or other low-grade astrocytomas, specifically expressed high levels of MetAP2. In addition, we show that Nf1-deficient astrocytes overexpress MetAP2 in vitro and in vivo, and that treatment with the MetAP2 inhibitor fumagillin significantly reduces Nf1-/- astrocyte proliferation in vitro. These observations suggest that MetAP2 is regulated by neurofibromin, and that MetAP2 inhibitors could be potentially employed to treat NF1-associated tumor proliferation.


Asunto(s)
Aminopeptidasas/líquido cefalorraquídeo , Glioma/líquido cefalorraquídeo , Metaloendopeptidasas/líquido cefalorraquídeo , Neurofibromatosis 1/líquido cefalorraquídeo , Neoplasias del Nervio Óptico/líquido cefalorraquídeo , Secuencia de Aminoácidos , Aminopeptidasas/antagonistas & inhibidores , Aminopeptidasas/biosíntesis , Aminopeptidasas/genética , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/patología , Astrocitoma/líquido cefalorraquídeo , Astrocitoma/complicaciones , Astrocitoma/enzimología , Astrocitoma/genética , Procesos de Crecimiento Celular/efectos de los fármacos , Ciclohexanos , Ácidos Grasos Insaturados/farmacología , Silenciador del Gen , Glioma/complicaciones , Glioma/enzimología , Glioma/genética , Glicoproteínas/antagonistas & inhibidores , Glicoproteínas/biosíntesis , Glicoproteínas/líquido cefalorraquídeo , Glicoproteínas/genética , Humanos , Metaloendopeptidasas/antagonistas & inhibidores , Metaloendopeptidasas/biosíntesis , Metaloendopeptidasas/genética , Metionil Aminopeptidasas , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Neurofibromatosis 1/complicaciones , Neurofibromatosis 1/enzimología , Neurofibromatosis 1/genética , Neurofibromina 1/deficiencia , Neurofibromina 1/genética , Neoplasias del Nervio Óptico/complicaciones , Neoplasias del Nervio Óptico/enzimología , Neoplasias del Nervio Óptico/genética , Proteómica , Sesquiterpenos , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteínas Supresoras de Tumor/genética
13.
Behav Brain Res ; 321: 148-156, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28057529

RESUMEN

Heterozygous Neurofibromatosis 1 (NF1) loss of function mutations occur in approximately 90% of patients with neurofibromatosis. A major, disabling phenotypic consequence of reduced NF1 function is cognitive impairment; a possibly related behavioral phenotype is impaired sleep. Recent results in Drosophila have demonstrated a genetic interaction between Anaplastic Lymphoma Kinase (Alk) and NF1 for both associative learning and sleep. Inhibition of Alk improves associative learning and sleep in heterozygous NF1 mutant flies. The results in Drosophila provide a strong motivation to investigate NF1/Alk genetic interactions in mice. In Drosophila, activation of Alk by its ligand, Jelly belly (Jeb), is the physiologically relevant target of negative regulation by NF1. Therefore, we tested whether genetic inhibition of Alk in heterozygous NF1 mutant mice attenuates or rescues cognitive impairments in mice. Our results are consistent with the hypothesis that NF1 functions in mice biochemically to inhibit signaling from Alk through Ras. The cognitive phenotypes observed in heterozygous NF1 mutant mice are rescued or ameliorated by genetic inhibition of Alk activity. In two tests of hippocampus-dependent learning, the Morris water maze and extinction of contextual fear, mutation of one or both alleles of Alk was sufficient to improve performance to wild type or near wild type levels in NF1-/+ mice. In addition, in NF1 mice genetic inhibition of Alk improves circadian activity levels. These data are intriguing in light of the circadian alterations seen in NF1 patients and indicate that inhibition of Alk activity may cognitively benefit patients with Neurofibromatosis 1.


Asunto(s)
Disfunción Cognitiva/enzimología , Disfunción Cognitiva/terapia , Neurofibromatosis 1/enzimología , Neurofibromatosis 1/psicología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Quinasa de Linfoma Anaplásico , Animales , Ritmo Circadiano/fisiología , Disfunción Cognitiva/etiología , Condicionamiento Psicológico/fisiología , Extinción Psicológica/fisiología , Miedo/fisiología , Femenino , Masculino , Aprendizaje por Laberinto/fisiología , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Neurofibromatosis 1/complicaciones , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Memoria Espacial/fisiología
14.
Behav Brain Res ; 332: 337-342, 2017 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-28629962

RESUMEN

Heterozygous Neurofibromatosis 1 (NF1) loss of function mutations are found in 90% of patients with neurofibromatosis, a syndrome associated with disabling cognitive impairment. Drosophila studies have demonstrated a genetic interaction between Anaplastic Lymphoma Kinase (Alk) and NF1 in cognitive performance. In addition, pharmacologic inhibition of Alk improves cognitive performance in heterozygous NF1 mutant flies. In this study, we tested whether pharmacological inhibition of Alk in heterozygous NF1 mutant mice attenuates or rescues cognitive impairments. Cognitive impairment of spatial memory retention observed in heterozygous NF1 mutant mice was rescued by the Alk inhibitor. These data support the hypothesis that inhibition of Alk may cognitively benefit patients with Neurofibromatosis 1.


Asunto(s)
Carbazoles/farmacología , Trastornos de la Memoria/tratamiento farmacológico , Neurofibromatosis 1/tratamiento farmacológico , Nootrópicos/farmacología , Piperidinas/farmacología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Memoria Espacial/efectos de los fármacos , Quinasa de Linfoma Anaplásico , Animales , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/enzimología , Modelos Animales de Enfermedad , Femenino , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/enzimología , Ratones Endogámicos C57BL , Ratones Noqueados , Neurofibromatosis 1/enzimología , Neurofibromatosis 1/psicología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas Receptoras/deficiencia , Proteínas Tirosina Quinasas Receptoras/genética , Memoria Espacial/fisiología , Natación/fisiología , Natación/psicología
15.
Cancer Res ; 55(16): 3569-75, 1995 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-7627966

RESUMEN

Neurofibromatosis type I (NF1) is a hereditary tumor and developmental disorder whose defective gene was cloned previously. The protein product of the NF1 gene, neurofibromin, contains a domain that shows significant sequence homology to the known catalytic domains of mammalian Ras GTPase-activating proteins (GAP) and the yeast IRA1 and IRA2 proteins. This homologous region of neurofibromin has been shown to exhibit GAP activity toward Ras proteins. Malignant schwannoma cell lines from NF1 patients contain normal levels of GAP and nonmutated Ras proteins but barely detectable levels of neurofibromin, based on genetic mutations in the NF1 gene. Because these cells contain constitutively activated Ras.GTP, it has been proposed that neurofibromin may be the sole negative regulator of Ras in these cells. Overall, these results have implied an important role of the Ras signaling pathway in NF1 malignant schwannomas. Recently, several laboratories have developed small molecule inhibitors of Ras function that inhibit the enzyme farnesyltransferase (FT). FT-mediated post-translational farnesylation of Ras proteins is absolutely necessary for Ras function since this modification is required for the anchoring of Ras proteins to the plasma cell membrane. Although previous studies have shown that FT inhibitors can block the growth of tumor cells carrying mutant Ras proteins, it remained unclear how this class of inhibitors would affect tumor cells such as in NF1, whose malignant growth appears to be mediated by up-regulation of wild-type Ras activity. Thus, in the current study, we investigated whether BMS-186511, a bisubstrate analogue inhibitor of FT, would inhibit the malignant growth properties of a cell line established from malignant schwannoma of an NF1 patient. Our results indicate that the malignant growth properties of ST88-14 cells, the most malignant cell line among several well-characterized NF1 cells, are inhibited by BMS-186511 in a concentration-dependent manner. Following treatment with BMS-186511, ST88-14 cells became flat, nonrefractile, were contact-inhibited, and lost their ability to grow in soft agar. In the drug-exposed cells, Ras proteins were prevented from FT-mediated membrane association. BMS-186511 was found to specifically inhibit FT, but not geranylgeranyltransferase I, a closely related enzyme. Thus, it is conceivable that FT inhibitors may ultimately become the first generation of drugs against the malignant phenotype in NF1 based on rational insights into the mechanism of action of neurofibromin.


Asunto(s)
Transferasas Alquil y Aril , Neurofibromatosis 1/enzimología , Oligopéptidos/farmacología , Ácidos Fosfínicos/farmacología , Prenilación de Proteína/efectos de los fármacos , Transferasas/antagonistas & inhibidores , Secuencia de Aminoácidos , Adhesión Celular/efectos de los fármacos , Compartimento Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Farnesiltransferasa , Humanos , Técnicas In Vitro , Datos de Secuencia Molecular , Neurofibromatosis 1/patología , Oligopéptidos/química , Fenotipo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Especificidad por Sustrato , Células Tumorales Cultivadas
16.
Neurology ; 31(9): 1184-8, 1981 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-6115338

RESUMEN

We examined 15 dermal neurofibromas from five adults with disseminated neurofibromatosis. All tumors contained axons that reacted for catecholamines and tyrosine hydroxylase on histochemical stains. Assay of tissue homogenates identified norepinephrine as the catecholamine. Assays for dopamine and choline acetyltransferase were negative. Some axonal components of dermal neurofibromas may originate in sympathetic adrenergic neurons. Most dermal neurofibromas do not contain neuronal cell bodies, and some of the axons may maintain functional connections with proximal sympathetic neuronal cell bodies. Sympathetic denervation may therefore affect the growth of these dermal neurofibromas.


Asunto(s)
Neurofibromatosis 1/metabolismo , Neurotransmisores/metabolismo , Neoplasias Cutáneas/metabolismo , Adulto , Colina O-Acetiltransferasa/metabolismo , Dopamina/metabolismo , Femenino , Histocitoquímica , Humanos , Masculino , Persona de Mediana Edad , Neurofibromatosis 1/enzimología , Norepinefrina/metabolismo , Neoplasias Cutáneas/enzimología , Tirosina 3-Monooxigenasa/metabolismo
17.
Am J Med Genet ; 39(4): 502-8, 1991 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-1908637

RESUMEN

To explore a possible relationship between neurofibromatosis and abnormalities of galactose metabolism, we examined the activity and characteristics of red cell galactokinase and galactose-1-phosphate uridyl transferase in 14 white women with type 1 neurofibromatosis and in 28 control women. Women with NF1 had a significantly lower activity of galactokinase and were more likely to have the 3-band pattern on electrophoresis of their transferase enzyme known as the Duarte variant. In addition, we found that women with NF1 were more likely to have reproductive histories suggestive of hypogonadism, including irregular menses, infertility, ovarian cysts, and early menopause. We conclude that abnormalities of galactose metabolism may be linked to abnormalities of both germ cell and neural crest development.


Asunto(s)
Galactosa/metabolismo , Neurofibromatosis 1/fisiopatología , Reproducción/fisiología , Adulto , Eritrocitos/enzimología , Femenino , Humanos , Menopausia Prematura/fisiología , Persona de Mediana Edad , Neurofibromatosis 1/enzimología , Neurofibromatosis 1/etiología
18.
Anticancer Res ; 17(6D): 4515-7, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9494560

RESUMEN

Cytochrome P450 CYP2D6 polymorphism is an autosomal recessive trait leading to impaired sparteine/ debrisoquine metabolism in 5-10% of the Caucasian population. Previous studies have associated affected individuals (poor metabolizers = PM) with susceptibility to bladder cancer and various forms of leukemia. In many other cancer forms, the data remain contradictory. A PCR assay allows the convenient screening of about 90% of known mutations resulting in the PM phenotype. Since in patients with neurofibromatosis type 2, we had observed a significantly increased rate of CYP2D6 mutations leading to PM and apparently predisposing for NF2, we extended our investigation to tumor and peripheral blood samples obtained from NF1 patients. Although the number of cases investigated remains low, the study indicated that during tumor formation no changes occurred at the mutational hot spot within the CYP2D6 sequence. Moreover, no loss of heterozygosity was notable. However, the frequency of the mutated allele in the NF1 individuals is comparable to that of neurofibromatosis type 2 and above that observed in breast and colon cancer, or meningiomas.


Asunto(s)
Citocromo P-450 CYP2D6/genética , Neurofibromatosis 1/enzimología , Neurofibromatosis 1/genética , Polimorfismo Genético , Adolescente , Adulto , Alelos , Exones , Femenino , Genes Recesivos , Humanos , Intrones , Leucemia/enzimología , Leucemia/genética , Masculino , Neoplasias Meníngeas/enzimología , Neoplasias Meníngeas/genética , Meningioma/enzimología , Meningioma/genética , Persona de Mediana Edad , Neurofibromatosis 2/enzimología , Neurofibromatosis 2/genética , Enfermedad de Parkinson/enzimología , Enfermedad de Parkinson/genética , Mutación Puntual , Reacción en Cadena de la Polimerasa , Eliminación de Secuencia
19.
J Dermatol ; 29(11): 713-7, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12484433

RESUMEN

Neurofibromas of neurofibromatosis 1 (NF1) are highly vascular. Because the number of PDGF beta receptors in neurofibroma-derived cultured cells (NF-derived cells) has been reported to be increased, we tested whether platelet-derived growth factor BB (PDGF-BB) could induce expression of vascular endothelial growth factor (VEGF) in NF-derived cells. When analysed by reverse transcription-polymerase chain reaction, VEGF mRNA expression was found to be stimulated by PDGF-BB and TGF-beta1. Those growth factors stimulated the secretion of VEGF from NF-derived cells. PDGF-BB furthermore induced the mitogen-activated protein kinase phosphorylation in NF-derived cells from patients with NF1. In conclusion, PDGF-BB stimulated VEGF secretion in NF-derived cells, and this stimulation is probably important in neurofibroma hypervascularization.


Asunto(s)
Factores de Crecimiento Endotelial/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Linfocinas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neurofibromatosis 1/enzimología , Neurofibromatosis 1/fisiopatología , Factor de Crecimiento Derivado de Plaquetas/farmacología , ARN Mensajero/análisis , Anciano , Secuencia de Bases , Becaplermina , Células Cultivadas , Humanos , Masculino , Persona de Mediana Edad , Proteínas Quinasas Activadas por Mitógenos/efectos de los fármacos , Datos de Secuencia Molecular , Fosforilación , Proteínas Proto-Oncogénicas c-sis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Sensibilidad y Especificidad , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
20.
J Clin Invest ; 123(1): 340-7, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23221341

RESUMEN

Neurofibromatosis type 1 (NF1) patients develop benign neurofibromas and malignant peripheral nerve sheath tumors (MPNST). These incurable peripheral nerve tumors result from loss of NF1 tumor suppressor gene function, causing hyperactive Ras signaling. Activated Ras controls numerous downstream effectors, but specific pathways mediating the effects of hyperactive Ras in NF1 tumors are unknown. We performed cross-species transcriptome analyses of mouse and human neurofibromas and MPNSTs and identified global negative feedback of genes that regulate Ras/Raf/MEK/ERK signaling in both species. Nonetheless, ERK activation was sustained in mouse and human neurofibromas and MPNST. We used a highly selective pharmacological inhibitor of MEK, PD0325901, to test whether sustained Ras/Raf/MEK/ERK signaling contributes to neurofibroma growth in a neurofibromatosis mouse model (Nf1(fl/fl);Dhh-Cre) or in NF1 patient MPNST cell xenografts. PD0325901 treatment reduced aberrantly proliferating cells in neurofibroma and MPNST, prolonged survival of mice implanted with human MPNST cells, and shrank neurofibromas in more than 80% of mice tested. Our data demonstrate that deregulated Ras/ERK signaling is critical for the growth of NF1 peripheral nerve tumors and provide a strong rationale for testing MEK inhibitors in NF1 clinical trials.


Asunto(s)
Benzamidas/farmacología , Difenilamina/análogos & derivados , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Neurofibromatosis 1/tratamiento farmacológico , Neurofibromatosis 1/enzimología , Neoplasias del Sistema Nervioso Periférico/tratamiento farmacológico , Neoplasias del Sistema Nervioso Periférico/enzimología , Animales , Niño , Preescolar , Difenilamina/farmacología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Mutantes , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Trasplante de Neoplasias , Neurofibromatosis 1/genética , Neurofibromatosis 1/patología , Proteína Oncogénica p21(ras)/genética , Proteína Oncogénica p21(ras)/metabolismo , Neoplasias del Sistema Nervioso Periférico/genética , Neoplasias del Sistema Nervioso Periférico/patología , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasas raf/genética , Quinasas raf/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA