Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Trends Pharmacol Sci ; 44(12): 963-977, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37940430

RESUMEN

Autism spectrum disorder (ASD) is a complex neurodevelopmental condition with uncertain origins. Understanding of the mechanisms underlying ASD remains limited, and treatments are lacking. Genetic diversity complicates drug development. Given the complexity and severity of ASD symptoms and the rising number of diagnoses, exploring novel therapeutic strategies is essential. Here, we focus on shared molecular pathways between ASD and cancer and highlight recent progress on the repurposing of cancer drugs for ASD treatment, such as mTOR inhibitors, histone deacetylase inhibitors, and anti-inflammatory agents. We discuss how to improve trial design considering drug dose and patient age. Lastly, the discussion explores the critical aspects of side effects, commercial factors, and the efficiency of drug-screening pipelines; all of which are essential considerations in the pursuit of repurposing cancer drugs for addressing core features of ASD.


Asunto(s)
Antineoplásicos , Trastorno del Espectro Autista , Neoplasias , Humanos , Trastorno del Espectro Autista/tratamiento farmacológico , Trastorno del Espectro Autista/diagnóstico , Trastorno del Espectro Autista/genética , Reposicionamiento de Medicamentos , Desarrollo de Medicamentos , Evaluación Preclínica de Medicamentos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico
2.
Neuron ; 111(11): 1760-1775.e8, 2023 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-36996810

RESUMEN

The proteome of glutamatergic synapses is diverse across the mammalian brain and involved in neurodevelopmental disorders (NDDs). Among those is fragile X syndrome (FXS), an NDD caused by the absence of the functional RNA-binding protein FMRP. Here, we demonstrate how the brain region-specific composition of postsynaptic density (PSD) contributes to FXS. In the striatum, the FXS mouse model shows an altered association of the PSD with the actin cytoskeleton, reflecting immature dendritic spine morphology and reduced synaptic actin dynamics. Enhancing actin turnover with constitutively active RAC1 ameliorates these deficits. At the behavioral level, the FXS model displays striatal-driven inflexibility, a typical feature of FXS individuals, which is rescued by exogenous RAC1. Striatal ablation of Fmr1 is sufficient to recapitulate behavioral impairments observed in the FXS model. These results indicate that dysregulation of synaptic actin dynamics in the striatum, a region largely unexplored in FXS, contributes to the manifestation of FXS behavioral phenotypes.


Asunto(s)
Síndrome del Cromosoma X Frágil , Animales , Ratones , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Actinas/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Ratones Noqueados , Espinas Dendríticas/metabolismo , Mamíferos/metabolismo
3.
Cell Death Dis ; 13(8): 719, 2022 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-35982038

RESUMEN

Converging evidence indicates that the Fragile X Messenger Ribonucleoprotein (FMRP), which absent or mutated in Fragile X Syndrome (FXS), plays a role in many types of cancers. However, while FMRP roles in brain development and function have been extensively studied, its involvement in the biology of brain tumors remains largely unexplored. Here we show, in human glioblastoma (GBM) biopsies, that increased expression of FMRP directly correlates with a worse patient outcome. In contrast, reductions in FMRP correlate with a diminished tumor growth and proliferation of human GBM stem-like cells (GSCs) in vitro in a cell culture model and in vivo in mouse brain GSC xenografts. Consistently, increased FMRP levels promote GSC proliferation. To characterize the mechanism(s) by which FMRP regulates GSC proliferation, we performed GSC transcriptome analyses in GSCs expressing high levels of FMRP, and in these GSCs after knockdown of FMRP. We show that the WNT signalling is the most significantly enriched among the published FMRP target genes and genes involved in ASD. Consistently, we find that reductions in FMRP downregulate both the canonical WNT/ß-Catenin and the non-canonical WNT-ERK1/2 signalling pathways, reducing the stability of several key transcription factors (i.e. ß-Catenin, CREB and ETS1) previously implicated in the modulation of malignant features of glioma cells. Our findings support a key role for FMRP in GBM cancer progression, acting via regulation of WNT signalling.


Asunto(s)
Neoplasias Encefálicas , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Glioblastoma , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioblastoma/patología , Humanos , Ratones , Células Madre Neoplásicas/metabolismo , Ribonucleoproteínas , Vía de Señalización Wnt/genética , beta Catenina/metabolismo
4.
Science ; 376(6599): eabf9088, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35709258

RESUMEN

The centrosome provides an intracellular anchor for the cytoskeleton, regulating cell division, cell migration, and cilia formation. We used spatial proteomics to elucidate protein interaction networks at the centrosome of human induced pluripotent stem cell-derived neural stem cells (NSCs) and neurons. Centrosome-associated proteins were largely cell type-specific, with protein hubs involved in RNA dynamics. Analysis of neurodevelopmental disease cohorts identified a significant overrepresentation of NSC centrosome proteins with variants in patients with periventricular heterotopia (PH). Expressing the PH-associated mutant pre-mRNA-processing factor 6 (PRPF6) reproduced the periventricular misplacement in the developing mouse brain, highlighting missplicing of transcripts of a microtubule-associated kinase with centrosomal location as essential for the phenotype. Collectively, cell type-specific centrosome interactomes explain how genetic variants in ubiquitous proteins may convey brain-specific phenotypes.


Asunto(s)
Centrosoma , Células-Madre Neurales , Neurogénesis , Neuronas , Heterotopia Nodular Periventricular , Mapas de Interacción de Proteínas , Empalme Alternativo , Animales , Encéfalo/anomalías , Centrosoma/metabolismo , Humanos , Células Madre Pluripotentes Inducidas , Ratones , Microtúbulos/metabolismo , Neuronas/metabolismo , Heterotopia Nodular Periventricular/metabolismo , Proteoma/metabolismo , Factores de Empalme de ARN/metabolismo , Factores de Transcripción/metabolismo
5.
Neuron ; 110(7): 1085-1087, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-35390284

RESUMEN

Mutations in epigenetic factors are associated with autism spectrum disorder (ASD). In this issue of Neuron, Yan et al. (2022) show that the antagonism of ASH1L and PRC2 switches the equilibrium of histone methylation at the ephrin receptor A7 locus, causing decreased EphA7 expression, deficits in synaptic pruning, and ASD-like behaviors.


Asunto(s)
Trastorno del Espectro Autista , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Epigénesis Genética , Epigenómica , Humanos , Mutación/genética
6.
Oncogene ; 40(23): 4033-4049, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34017076

RESUMEN

Intrahepatic cholangiocarcinoma (iCCA) is a rare malignancy of the intrahepatic biliary tract with a very poor prognosis. Although some clinicopathological parameters can be prognostic factors for iCCA, the molecular prognostic markers and potential mechanisms of iCCA have not been well investigated. Here, we report that the Fragile X mental retardation protein (FMRP), a RNA binding protein functionally absent in patients with the Fragile X syndrome (FXS) and also involved in several types of cancers, is overexpressed in human iCCA and its expression is significantly increased in iCCA metastatic tissues. The silencing of FMRP in metastatic iCCA cell lines affects cell migration and invasion, suggesting a role of FMRP in iCCA progression. Moreover, we show evidence that FMRP is localized at the invasive front of human iCCA neoplastic nests and in pseudopodia and invadopodia protrusions of migrating and invading iCCA cancer cells. Here FMRP binds several mRNAs encoding key proteins involved in the formation and/or function of these protrusions. In particular, we find that FMRP binds to and regulates the expression of Cortactin, a critical regulator of invadopodia formation. Altogether, our findings suggest that FMRP could promote cell invasiveness modulating membrane plasticity and invadopodia formation at the leading edges of invading iCCA cells.


Asunto(s)
Neoplasias de los Conductos Biliares/metabolismo , Colangiocarcinoma/metabolismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Animales , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/patología , Línea Celular Tumoral , Plasticidad de la Célula/fisiología , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Cortactina/metabolismo , Humanos , Masculino , Ratones Desnudos , Metástasis de la Neoplasia , Podosomas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Transl Psychiatry ; 11(1): 112, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33547274

RESUMEN

In fragile X syndrome (FXS) the lack of the fragile X mental retardation protein (FMRP) leads to exacerbated signaling through the metabotropic glutamate receptors 5 (mGlu5Rs). The adenosine A2A receptors (A2ARs), modulators of neuronal damage, could play a role in FXS. A synaptic colocalization and a strong permissive interaction between A2A and mGlu5 receptors in the hippocampus have been previously reported, suggesting that blocking A2ARs might normalize the mGlu5R-mediated effects of FXS. To study the cross-talk between A2A and mGlu5 receptors in the absence of FMRP, we performed extracellular electrophysiology experiments in hippocampal slices of Fmr1 KO mouse. The depression of field excitatory postsynaptic potential (fEPSPs) slope induced by the mGlu5R agonist CHPG was completely blocked by the A2AR antagonist ZM241385 and strongly potentiated by the A2AR agonist CGS21680, suggesting that the functional synergistic coupling between the two receptors could be increased in FXS. To verify if chronic A2AR blockade could reverse the FXS phenotypes, we treated the Fmr1 KO mice with istradefylline, an A2AR antagonist. We found that hippocampal DHPG-induced long-term depression (LTD), which is abnormally increased in FXS mice, was restored to the WT level. Furthermore, istradefylline corrected aberrant dendritic spine density, specific behavioral alterations, and overactive mTOR, TrkB, and STEP signaling in Fmr1 KO mice. Finally, we identified A2AR mRNA as a target of FMRP. Our results show that the pharmacological blockade of A2ARs partially restores some of the phenotypes of Fmr1 KO mice, both by reducing mGlu5R functioning and by acting on other A2AR-related downstream targets.


Asunto(s)
Síndrome del Cromosoma X Frágil , Receptor de Adenosina A2A , Adenosina , Animales , Cognición , Modelos Animales de Enfermedad , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/tratamiento farmacológico , Síndrome del Cromosoma X Frágil/genética , Hipocampo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Adenosina A2A/genética
8.
Cell Mol Gastroenterol Hepatol ; 11(2): 639-658, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33091622

RESUMEN

BACKGROUND & AIMS: The fragile X mental retardation protein (FMRP) affects multiple steps of the mRNA metabolism during brain development and in different neoplastic processes. However, the contribution of FMRP in colon carcinogenesis has not been investigated. METHODS: FMR1 mRNA transcript and FMRP protein expression were analyzed in human colon samples derived from patients with sporadic colorectal cancer (CRC) and healthy subjects. We used a well-established mouse model of sporadic CRC induced by azoxymethane to determine the possible role of FMRP in CRC. To address whether FMRP controls cancer cell survival, we analyzed cell death pathway in CRC human epithelial cell lines and in patient-derived colon cancer organoids in presence or absence of a specific FMR1 antisense oligonucleotide or siRNA. RESULTS: We document a significant increase of FMRP in human CRC relative to non-tumor tissues. Next, using an inducible mouse model of CRC, we observed a reduction of colonic tumor incidence and size in the Fmr1 knockout mice. The abrogation of FMRP induced spontaneous cell death in human CRC cell lines activating the necroptotic pathway. Indeed, specific immunoprecipitation experiments on human cell lines and CRC samples indicated that FMRP binds receptor-interacting protein kinase 1 (RIPK1) mRNA, suggesting that FMRP acts as a regulator of necroptosis pathway through the surveillance of RIPK1 mRNA metabolism. Treatment of human CRC cell lines and patient-derived colon cancer organoids with the FMR1 antisense resulted in up-regulation of RIPK1. CONCLUSIONS: Altogether, these data support a role for FMRP  in controlling RIPK1 expression and necroptotic activation in CRC.


Asunto(s)
Neoplasias Colorrectales/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Recurrencia Local de Neoplasia/epidemiología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Animales , Azoximetano/administración & dosificación , Azoximetano/toxicidad , Carcinogénesis/genética , Estudios de Casos y Controles , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Colon/patología , Colon/cirugía , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/cirugía , Conjuntos de Datos como Asunto , Supervivencia sin Enfermedad , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/antagonistas & inhibidores , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Voluntarios Sanos , Humanos , Masculino , Ratones , Ratones Noqueados , Necroptosis/genética , Recurrencia Local de Neoplasia/genética , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Organoides , Pronóstico
9.
Mov Disord ; 34(3): 323-334, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30624801

RESUMEN

BACKGROUND: Friedreich's ataxia is an autosomal-recessive cerebellar ataxia caused by mutation of the frataxin gene, resulting in decreased frataxin expression, mitochondrial dysfunction, and oxidative stress. Currently, no treatment is available for Friedreich's ataxia patients. Given that levels of residual frataxin critically affect disease severity, the main goal of a specific therapy for Friedreich's ataxia is to increase frataxin levels. OBJECTIVES: With the aim to accelerate the development of a new therapy for Friedreich's ataxia, we took a drug repositioning approach to identify market-available drugs able to increase frataxin levels. METHODS: Using a cell-based reporter assay to monitor variation in frataxin amount, we performed a high-throughput screening of a library containing 853 U.S. Food and Drug Administration-approved drugs. RESULTS: Among the potentially interesting candidates isolated from the screening, we focused our attention on etravirine, an antiviral drug currently in use as an anti-human immunodeficiency virus therapy. Here, we show that etravirine can promote a significant increase in frataxin levels in cells derived from Friedreich's ataxia patients, by enhancing frataxin messenger RNA translation. Importantly, frataxin accumulation in treated patient cell lines is comparable to frataxin levels in unaffected carrier cells, suggesting that etravirine could be therapeutically relevant. Indeed, etravirine treatment restores the activity of the iron-sulphur cluster containing enzyme aconitase and confers resistance to oxidative stress in cells derived from Friedreich's ataxia patients. CONCLUSIONS: Considering its excellent safety profile along with its ability to increase frataxin levels and correct some of the disease-related defects, etravirine represents a promising candidate as a therapeutic for Friedreich's ataxia. © 2019 International Parkinson and Movement Disorder Society.


Asunto(s)
Ataxia de Friedreich/tratamiento farmacológico , Proteínas de Unión a Hierro/metabolismo , Piridazinas/uso terapéutico , Línea Celular , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Ataxia de Friedreich/genética , Ataxia de Friedreich/metabolismo , Humanos , Proteínas de Unión a Hierro/genética , Nitrilos , Pirimidinas , Frataxina
11.
Hum Mol Genet ; 27(12): 2039-2051, 2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29590342

RESUMEN

Fragile X syndrome (FXS) is a monogenic form of intellectual disability and autism spectrum disorder caused by the absence of the fragile X mental retardation protein (FMRP). In biological models for the disease, this leads to upregulated mRNA translation and as a consequence, deficits in synaptic architecture and plasticity. Preclinical studies revealed that pharmacological interventions restore those deficits, which are thought to mediate the FXS cognitive and behavioral symptoms. Here, we characterized the de novo rate of protein synthesis in patients with FXS and their relationship with clinical severity. We measured the rate of protein synthesis in fibroblasts derived from 32 individuals with FXS and from 17 controls as well as in fibroblasts and primary neurons of 27 Fmr1 KO mice and 20 controls. Here, we show that levels of protein synthesis are increased in fibroblasts of individuals with FXS and Fmr1 KO mice. However, this cellular phenotype displays a broad distribution and a proportion of fragile X individuals and Fmr1 KO mice do not show increased levels of protein synthesis, having measures in the normal range. Because the same Fmr1 KO animal measures in fibroblasts predict those in neurons we suggest the validity of this peripheral biomarker. Our study offers a potential explanation for the comprehensive drug development program undertaken thus far yielding negative results and suggests that a significant proportion, but not all individuals with FXS, may benefit from the reduction of excessive levels of protein synthesis.


Asunto(s)
Trastorno del Espectro Autista/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Adolescente , Adulto , Anciano , Animales , Trastorno del Espectro Autista/fisiopatología , Niño , Modelos Animales de Enfermedad , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/biosíntesis , Síndrome del Cromosoma X Frágil/fisiopatología , Hipocampo/metabolismo , Hipocampo/fisiopatología , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Neuronas/metabolismo , Neuronas/patología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...