Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Neurobiol Dis ; 191: 106393, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38154608

RESUMO

Phosphodiesterase 2 A (PDE2A) is an enzyme involved in the homeostasis of cAMP and cGMP and is the most highly expressed PDE in human brain regions critical for socio-cognitive behavior. In cerebral cortex and hippocampus, PDE2A expression level is upregulated in Fmr1-KO mice, a model of the Fragile X Syndrome (FXS), the most common form of inherited intellectual disability (ID) and autism spectrum disorder (ASD). Indeed, PDE2A translation is negatively modulated by FMRP, whose functional absence causes FXS. While the pharmacological inhibition of PDE2A has been associated to its pro-cognitive role in normal animals and in models of ID and ASD, homozygous PDE2A mutations have been identified in patients affected by ID, ASD and epilepsy. To clarify this apparent paradox about the role of PDE2A in brain development, we characterized here Pde2a+/- mice (homozygote animals being not viable) at the behavioral, cellular, molecular and electrophysiological levels. Pde2a+/- females display a milder form of the disorder with reduced cognitive performance in adulthood, conversely males show severe socio-cognitive deficits throughout their life. In males, these phenotypes are associated with microglia activation, elevated glutathione levels and increased externalization of Glutamate receptor (GluR1) in CA1, producing reduced mGluR-dependent Long-term Depression. Overall, our results reveal molecular targets of the PDE2A-dependent pathway underlying socio-cognitive performance. These results clarify the mechanism of action of pro-cognitive drugs based on PDE2A inactivation, which have been shown to be promising therapeutic approaches for Alzheimer's disease, schizophrenia, FXS as well as other forms of ASD.


Assuntos
Transtorno do Espectro Autista , Síndrome do Cromossomo X Frágil , Animais , Feminino , Humanos , Masculino , Camundongos , Cognição , Proteína do X Frágil da Deficiência Intelectual/genética , Camundongos Knockout , Microglia/metabolismo , Diester Fosfórico Hidrolases/metabolismo
2.
Genome Res ; 30(11): 1633-1642, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32973039

RESUMO

To gain better insight into the dynamic interaction between cells and their environment, we developed the agonist-induced functional analysis and cell sorting (aiFACS) technique, which allows the simultaneous recording and sorting of cells in real-time according to their immediate and individual response to a stimulus. By modulating the aiFACS selection parameters, testing different developmental times, using various stimuli, and multiplying the analysis of readouts, it is possible to analyze cell populations of any normal or pathological tissue. The association of aiFACS with single-cell transcriptomics allows the construction of functional tissue cartography based on specific pharmacological responses of cells. As a proof of concept, we used aiFACS on the dissociated mouse brain, a highly heterogeneous tissue, enriching it in interneurons by stimulation with KCl or with AMPA, an agonist of the glutamate receptors, followed by sorting based on calcium levels. After AMPA stimulus, single-cell transcriptomics of these aiFACS-selected interneurons resulted in a nine-cluster classification. Furthermore, we used aiFACS on interneurons derived from the brain of the Fmr1-KO mouse, a rodent model of fragile X syndrome. We showed that these interneurons manifest a generalized defective response to AMPA compared with wild-type cells, affecting all the analyzed cell clusters at one specific postnatal developmental time.


Assuntos
Encéfalo/metabolismo , Separação Celular/métodos , Citometria de Fluxo/métodos , Interneurônios/metabolismo , RNA-Seq , Análise de Célula Única , Encéfalo/citologia , Agonistas de Aminoácidos Excitatórios/farmacologia , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/metabolismo , Técnicas de Inativação de Genes , Interneurônios/efeitos dos fármacos , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia
3.
Cereb Cortex ; 29(8): 3241-3252, 2019 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-30137253

RESUMO

The fragile X mental retardation protein (FMRP) is an RNA-binding protein involved in translational regulation of mRNAs that play key roles in synaptic morphology and plasticity. The functional absence of FMRP causes the fragile X syndrome (FXS), the most common form of inherited intellectual disability and the most common monogenic cause of autism. No effective treatment is available for FXS. We recently identified the Phosphodiesterase 2A (Pde2a) mRNA as a prominent target of FMRP. PDE2A enzymatic activity is increased in the brain of Fmr1-KO mice, a recognized model of FXS, leading to decreased levels of cAMP and cGMP. Here, we pharmacologically inhibited PDE2A in Fmr1-KO mice and observed a rescue both of the maturity of dendritic spines and of the exaggerated hippocampal mGluR-dependent long-term depression. Remarkably, PDE2A blockade rescued the social and communicative deficits of both mouse and rat Fmr1-KO animals. Importantly, chronic inhibition of PDE2A in newborn Fmr1-KO mice followed by a washout interval, resulted in the rescue of the altered social behavior observed in adolescent mice. Altogether, these results reveal the key role of PDE2A in the physiopathology of FXS and suggest that its pharmacological inhibition represents a novel therapeutic approach for FXS.


Assuntos
Comunicação Animal , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/metabolismo , Espinhas Dendríticas/efeitos dos fármacos , Síndrome do Cromossomo X Frágil/enzimologia , Hipocampo/efeitos dos fármacos , Imidazóis/farmacologia , Depressão Sináptica de Longo Prazo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Inibidores de Fosfodiesterase/farmacologia , Comportamento Social , Triazinas/farmacologia , Animais , Animais Recém-Nascidos , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/antagonistas & inibidores , Espinhas Dendríticas/patologia , Embrião de Mamíferos , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/patologia , Síndrome do Cromossomo X Frágil/fisiopatologia , Técnicas de Inativação de Genes , Hipocampo/metabolismo , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Neurônios/patologia , Cultura Primária de Células , Ratos , Receptores de Glutamato Metabotrópico/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/metabolismo
4.
Nucleic Acids Res ; 46(12): 6344-6355, 2018 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-29668986

RESUMO

Fragile X syndrome (FXS), the most common form of inherited intellectual disability, is due to the functional deficiency of the fragile X mental retardation protein (FMRP), an RNA-binding protein involved in translational regulation of many messenger RNAs, playing key roles in synaptic morphology and plasticity. To date, no effective treatment for FXS is available. We searched for FMRP targets by HITS-CLIP during early development of multiple mouse brain regions (hippocampus, cortex and cerebellum) at a time of brain development when FMRP is most highly expressed and synaptogenesis reaches a peak. We identified the largest dataset of mRNA targets of FMRP available in brain and we defined their cellular origin. We confirmed the G-quadruplex containing structure as an enriched motif in FMRP RNA targets. In addition to four less represented motifs, our study points out that, in the brain, CTGKA is the prominent motif bound by FMRP, which recognizes it when not engaged in Watson-Crick pairing. All of these motifs negatively modulated the expression level of a reporter protein. While the repertoire of FMRP RNA targets in cerebellum is quite divergent, the ones of cortex and hippocampus are vastly overlapping. In these two brain regions, the Phosphodiesterase 2a (Pde2a) mRNA is a prominent target of FMRP, which modulates its translation and intracellular transport. This enzyme regulates the homeostasis of cAMP and cGMP and represents a novel and attractive therapeutic target to treat FXS.


Assuntos
Encéfalo/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , RNA Mensageiro/metabolismo , Animais , Encéfalo/crescimento & desenvolvimento , Cerebelo/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Hipocampo/metabolismo , Imunoprecipitação , Masculino , Camundongos , Motivos de Nucleotídeos , Ligação Proteica , RNA Mensageiro/química , Análise de Sequência de RNA
5.
RNA ; 23(6): 899-909, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28258160

RESUMO

Spinal muscular atrophy (SMA) is caused by mutations and/or deletions of the survival motor neuron gene (SMN1). Besides its function in the biogenesis of spliceosomal snRNPs, SMN might possess a motor neuron specific role and could function in the transport of axonal mRNAs and in the modulation of local protein translation. Accordingly, SMN colocalizes with axonal mRNAs of differentiated NSC-34 motor neuron-like cells. We recently showed that SMN depletion gives rise to a decrease in the axonal transport of the mRNAs encoding Annexin A2 (Anxa2). In this work, we have characterized the structural features of the Anxa2 mRNA required for its axonal targeting by SMN. We found that a G-rich motif located near the 3'UTR is essential for axonal localization of the Anxa2 transcript. We also show that mutations in the motif sequence abolish targeting of Anxa2 reporter mRNAs in axon-like structures of differentiated NSC-34 cells. Finally, localization of both wild-type and mutated Anxa2 reporters is restricted to the cell body in SMN-depleted cells. Altogether, our studies show that this G-motif represents a novel and essential determinant for axonal localization of the Anxa2 mRNA mediated by the SMN complex.


Assuntos
Anexina A2/genética , Anexina A2/metabolismo , Axônios/metabolismo , Motivos de Nucleotídeos , RNA Mensageiro , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Regiões 3' não Traduzidas , Animais , Anexina A2/química , Sequência de Bases , Linhagem Celular , Quadruplex G , Expressão Gênica , Genes Reporter , Humanos , Camundongos , Neurônios Motores/metabolismo , Ligação Proteica , Transporte Proteico , Transporte de RNA
6.
Nature ; 481(7380): 195-8, 2011 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-22198669

RESUMO

Fundamental questions remain unanswered about the transcriptional networks that control the identity and self-renewal of neural stem cells (NSCs), a specialized subset of astroglial cells that are endowed with stem properties and neurogenic capacity. Here we report that the zinc finger protein Ars2 (arsenite-resistance protein 2; also known as Srrt) is expressed by adult NSCs from the subventricular zone (SVZ) of mice, and that selective knockdown of Ars2 in cells expressing glial fibrillary acidic protein within the adult SVZ depletes the number of NSCs and their neurogenic capacity. These phenotypes are recapitulated in the postnatal SVZ of hGFAP-cre::Ars2(fl/fl) conditional knockout mice, but are more severe. Ex vivo assays show that Ars2 is necessary and sufficient to promote NSC self-renewal, and that it does so by positively regulating the expression of Sox2. Although plant and animal orthologues of Ars2 are known for their conserved roles in microRNA biogenesis, we unexpectedly observed that Ars2 retains its capacity to promote self-renewal in Drosha and Dicer1 knockout NSCs. Instead, chromatin immunoprecipitation revealed that Ars2 binds a specific region within the 6-kilobase NSC enhancer of Sox2. This association is RNA-independent, and the region that is bound is required for Ars2-mediated activation of Sox2. We used gel-shift analysis to refine the Sox2 region bound by Ars2 to a specific conserved DNA sequence. The importance of Sox2 as a critical downstream effector is shown by its ability to restore the self-renewal and multipotency defects of Ars2 knockout NSCs. Our findings reveal Ars2 as a new transcription factor that controls the multipotent progenitor state of NSCs through direct activation of the pluripotency factor Sox2.


Assuntos
Encéfalo/citologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , Animais , Proliferação de Células , Células Cultivadas , Imunoprecipitação da Cromatina , Sequência Conservada/genética , RNA Helicases DEAD-box/deficiência , Proteínas de Ligação a DNA , Ensaio de Desvio de Mobilidade Eletroforética , Elementos Facilitadores Genéticos/genética , Proteína Glial Fibrilar Ácida/metabolismo , Camundongos , Camundongos Knockout , Neurogênese/genética , Proteínas Nucleares/química , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Bulbo Olfatório/citologia , Ribonuclease III/deficiência , Fatores de Transcrição/química , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Dedos de Zinco
7.
Nucleic Acids Res ; 43(17): 8540-50, 2015 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-26250109

RESUMO

Fragile X syndrome (FXS), the most common form of inherited intellectual disability, is caused by the silencing of the FMR1 gene encoding an RNA-binding protein (FMRP) mainly involved in translational control. We characterized the interaction between FMRP and the mRNA of GRK4, a member of the guanine nucleotide-binding protein (G protein)-coupled receptor kinase super-family, both in vitro and in vivo. While the mRNA level of GRK4 is unchanged in the absence or in the presence of FMRP in different regions of the brain, GRK4 protein level is increased in Fmr1-null cerebellum, suggesting that FMRP negatively modulates the expression of GRK4 at the translational level in this brain region. The C-terminal region of FMRP interacts with a domain of GRK4 mRNA, that we called G4RIF, that is folded in four stem loops. The SL1 stem loop of G4RIF is protected by FMRP and is part of the S1/S2 sub-domain that directs translation repression of a reporter mRNA by FMRP. These data confirm the role of the G4RIF/FMRP complex in translational regulation. Considering the role of GRK4 in GABAB receptors desensitization, our results suggest that an increased GRK4 levels in FXS might contribute to cerebellum-dependent phenotypes through a deregulated desensitization of GABAB receptors.


Assuntos
Cerebelo/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Quinase 4 de Receptor Acoplado a Proteína G/genética , RNA Mensageiro/metabolismo , Animais , Sítios de Ligação , Proteína do X Frágil da Deficiência Intelectual/química , Proteína do X Frágil da Deficiência Intelectual/genética , Quinase 4 de Receptor Acoplado a Proteína G/metabolismo , Humanos , Camundongos , Camundongos Knockout , Ligação Proteica , Biossíntese de Proteínas , Domínios e Motivos de Interação entre Proteínas , RNA Mensageiro/química
8.
Nucleic Acids Res ; 42(3): 1987-2002, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24220090

RESUMO

Although Dicer is essential for general microRNA (miRNA) biogenesis, vertebrate mir-451 is Dicer independent. Instead, its short pre-miRNA hairpin is 'sliced' by Ago2, then 3'-resected into mature miRNAs. Here, we show that Drosophila cells and animals generate functional small RNAs from mir-451-type precursors. However, their bulk maturation arrests as Ago-cleaved pre-miRNAs, which mostly associate with the RNAi effector AGO2. Routing of pre-mir-451 hairpins to the miRNA effector AGO1 was inhibited by Dicer-1 and its partner Loqs. Loss of these miRNA factors promoted association of pre-mir-451 with AGO1, which sliced them and permitted maturation into ∼ 23-26 nt products. The difference was due to the 3' modification of single-stranded species in AGO2 by Hen1 methyltransferase, whose depletion permitted 3' trimming of Ago-cleaved pre-miRNAs in AGO2. Surprisingly, Nibbler, a 3'-5' exoribonuclease that trims 'long' mature miRNAs in AGO1, antagonized miR-451 processing. We used an in vitro reconstitution assay to identify a soluble, EDTA-sensitive activity that resects sliced pre-miRNAs in AGO1 complexes. Finally, we use deep sequencing to show that depletion of dicer-1 increases the diversity of small RNAs in AGO1, including some candidate mir-451-like loci. Altogether, we document unexpected aspects of miRNA biogenesis and Ago sorting, and provide insights into maturation of Argonaute-cleaved miRNA substrates.


Assuntos
Proteínas Argonautas/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila/genética , MicroRNAs/metabolismo , Precursores de RNA/metabolismo , Processamento Pós-Transcricional do RNA , Animais , Células Cultivadas , Drosophila/metabolismo , Exorribonucleases/metabolismo , Metiltransferases/metabolismo , RNA Helicases/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribonuclease III/metabolismo
9.
RNA ; 18(5): 945-57, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22461413

RESUMO

Until recently, a Dicer-class RNase III enzyme was believed to be essential for microRNA (miRNA) biogenesis in all animals. The conserved vertebrate locus mir-451 defies this expectation and instead matures by direct cleavage of its pre-miRNA hairpin via the Slicer activity of Argonaute2 (Ago2). In this study, we used structure-function analysis to define the functional parameters of Ago2-mediated miRNA biogenesis. These include (1) the requirement for base-pairing at most, but not all, positions within the pre-mir-451 stem; (2) surprisingly little influence of the 5'-nucleotide on Ago sorting; (3) substantial influence of Ago protein stoichiometry on mir-451 maturation; (4) strong influence of G:C content in the distal stem on 3' resection of cleaved mir-451 substrates; and (5) the influence of hairpin length on substrate utilization by Ago2 and Dicer. Unexpectedly, we find that certain hairpin lengths confer competence to mature via both Dicer-mediated and Ago2-mediated pathways, and we show, in fact, that a conventional shRNA can traverse the Dicer-independent pathway. Altogether, these data inform the design of effective Dicer-independent substrates for gene silencing and reveal novel aspects of substrate handling by Ago proteins.


Assuntos
MicroRNAs/metabolismo , Ribonuclease III/metabolismo , Proteínas Argonautas/química , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Composição de Bases , Pareamento de Bases , Sequência de Bases , Linhagem Celular , Humanos , Sequências Repetidas Invertidas , MicroRNAs/química , MicroRNAs/genética , Dados de Sequência Molecular , Mutação , Ribonuclease III/genética , Alinhamento de Sequência
10.
RNA ; 18(12): 2166-73, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23097423

RESUMO

RNase III enzymes are fundamental to the biogenesis of microRNAs (miRNAs) and small interfering RNAs (siRNAs) in all species studied. Although alternative miRNA pathways independent of Drosha or Dicer exist, each still requires one RNase III-type enzyme. Here, we describe two strategies that marry either RNase Z or the Integrator complex with the slicing activity of Argonaute2 to generate highly functional mature miRNAs. We provide stringent validation of their RNase III independence by demonstrating efficient miRNA biogenesis and activity in Drosha and Dicer knockout cells. These data provide proof-of-principle evidence for additional mechanistic possibilities for efficient generation of small regulatory RNAs, and represent novel silencing triggers that may be exploited for technical purposes.


Assuntos
MicroRNAs/biossíntese , Ribonuclease III/metabolismo , Animais , Proteínas Argonautas/deficiência , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Sequência de Bases , Linhagem Celular , RNA Helicases DEAD-box/deficiência , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Endorribonucleases/deficiência , Endorribonucleases/genética , Endorribonucleases/metabolismo , Técnicas de Inativação de Genes , Células HEK293 , Células HeLa , Herpesvirus Saimiriíneo 2/genética , Herpesvirus Saimiriíneo 2/metabolismo , Humanos , Camundongos , MicroRNAs/genética , RNA Nuclear Pequeno/genética , RNA Nuclear Pequeno/metabolismo , RNA Viral/genética , RNA Viral/metabolismo , Ribonuclease III/deficiência , Ribonuclease III/genética
11.
Proc Natl Acad Sci U S A ; 107(34): 15163-8, 2010 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-20699384

RESUMO

Canonical animal microRNAs (miRNAs) are generated by sequential cleavage of precursor substrates by the Drosha and Dicer RNase III enzymes. Several variant pathways exploit other RNA metabolic activities to generate functional miRNAs. However, all of these pathways culminate in Dicer cleavage, suggesting that this is a unifying feature of miRNA biogenesis. Here, we show that maturation of miR-451, a functional miRNA that is perfectly conserved among vertebrates, is independent of Dicer. Instead, structure-function and knockdown studies indicate that Drosha generates a short pre-mir-451 hairpin that is directly cleaved by Ago2 and followed by resection of its 3' terminus. We provide stringent evidence for this model by showing that Dicer knockout cells can generate mature miR-451 but not other miRNAs, whereas Ago2 knockout cells reconstituted with wild-type Ago2, but not Slicer-deficient Ago2, can process miR-451. Finally, we show that the mir-451 backbone is amenable to reprogramming, permitting vector-driven expression of diverse functional miRNAs in the absence of Dicer. Beyond the demonstration of an alternative strategy to direct gene silencing, these observations open the way for transgenic rescue of Dicer conditional knockouts.


Assuntos
Fator de Iniciação 2 em Eucariotos/metabolismo , MicroRNAs/biossíntese , Animais , Proteínas Argonautas , Sequência de Bases , Sequência Conservada , RNA Helicases DEAD-box/metabolismo , Primers do DNA/genética , Endorribonucleases/metabolismo , Fator de Iniciação 2 em Eucariotos/deficiência , Fator de Iniciação 2 em Eucariotos/genética , Células HeLa , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/antagonistas & inibidores , MicroRNAs/química , MicroRNAs/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Conformação de Ácido Nucleico , RNA Interferente Pequeno/genética , Ribonuclease III/metabolismo , Homologia de Sequência do Ácido Nucleico
12.
Clin Microbiol Infect ; 28(9): 1286.e1-1286.e8, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35398511

RESUMO

OBJECTIVE: Antibiotic susceptibility testing (AST) is necessary in order to adjust empirical antibiotic treatment, but the interpretation of results requires experience and knowledge. We have developed a machine learning software that is capable of reading AST images without any human intervention and that automatically interprets the AST, based on a database of antibiograms that have been clinically validated with European Committee on Antimicrobial Susceptibility Testing rules. METHODS: We built a database of antibiograms that were labelled by senior microbiologists for three species: Escherichia coli, Klebsiella pneumoniae, and Staphylococcus aureus. We then developed Antilogic, a Python software based on an original image segmentation module and supervised learning models that we trained against the database. Finally, we blind tested Antilogic against a validation set of 5100 photos of antibiograms. RESULTS: We trained Antilogic against a database of 18072 pictures of antibiograms. Overall agreement against the validation set reached 97% (16 855/17 281) regarding phenotypes. The severity rate of errors was also evaluated: 1.66% (287/17 281) were major errors and 0.80% (136/17 281) were very major errors. After implementation of uncertainty quantifications, the rate of errors decreased to 0.80% (114/13 451) and 0.42% (51/13 451) for major and very major errors respectively. DISCUSSION: Antilogic is the first machine learning software that has been developed for AST interpretation. It is based on a novel approach that differs from the typical diameter measurement and expert system approach. Antilogic is a proof of concept that artificial intelligence can contribute to faster and easier diagnostic methods in the field of clinical microbiology.


Assuntos
Antibacterianos , Inteligência Artificial , Antibacterianos/farmacologia , Bactérias , Escherichia coli , Humanos , Testes de Sensibilidade Microbiana , Software , Aprendizado de Máquina Supervisionado
13.
Blood ; 113(2): 412-21, 2009 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-18941112

RESUMO

Micro(mi)RNAs are small noncoding RNAs that orchestrate many key aspects of cell physiology and their deregulation is often linked to distinct diseases including cancer. Here, we studied the contribution of miRNAs in a well-characterized human myeloid leukemia, acute promyelocytic leukemia (APL), targeted by retinoic acid and trioxide arsenic therapy. We identified several miRNAs transcriptionally repressed by the APL-associated PML-RAR oncogene which are released after treatment with all-trans retinoic acid. These coregulated miRNAs were found to control, in a coordinated manner, crucial pathways linked to leukemogenesis, such as HOX proteins and cell adhesion molecules whose expressions are thereby repressed by the chemotherapy. Thus, APL appears linked to transcriptional perturbation of miRNA genes, and clinical protocols able to successfully eradicate cancer cells may do so by restoring miRNA expression. The identification of abnormal miRNA biogenesis in cancer may therefore provide novel biomarkers and therapeutic targets in myeloid leukemias.


Assuntos
Biomarcadores Tumorais/biossíntese , Regulação Leucêmica da Expressão Gênica , Leucemia Promielocítica Aguda/metabolismo , MicroRNAs/biossíntese , Proteínas de Fusão Oncogênica/metabolismo , RNA Neoplásico/biossíntese , Transcrição Gênica , Antineoplásicos/uso terapêutico , Arsênio/uso terapêutico , Biomarcadores Tumorais/genética , Moléculas de Adesão Celular/biossíntese , Moléculas de Adesão Celular/genética , Linhagem Celular Tumoral , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Leucemia Promielocítica Aguda/tratamento farmacológico , Leucemia Promielocítica Aguda/genética , MicroRNAs/genética , Proteínas de Fusão Oncogênica/genética , RNA Neoplásico/genética , Transcrição Gênica/efeitos dos fármacos , Tretinoína/uso terapêutico
14.
Mol Ther Nucleic Acids ; 18: 546-553, 2019 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-31671347

RESUMO

Fragile X-associated tremor ataxia syndrome (FXTAS) is a rare disorder associated to the presence of the fragile X premutation, a 55-200 CGG repeat expansion in the 5' UTR of the FMR1 gene. Two main neurological phenotypes have been described in carriers of the CGG premutation: (1) neurodevelopmental disorders characterized by anxiety, attention deficit hyperactivity disorder (ADHD), social deficits, or autism spectrum disorder (ASD); and (2) after 50 years old, the FXTAS phenotype. This neurodegenerative disorder is characterized by ataxia and a form of parkinsonism. The molecular pathology of this disorder is characterized by the presence of elevated levels of Fragile X Mental Retardation 1 (FMR1) mRNA, presence of a repeat-associated non-AUG (RAN) translated peptide, and FMR1 mRNA-containing nuclear inclusions. Whereas in the past FXTAS was mainly considered as a late-onset disorder, some phenotypes of patients and altered learning and memory behavior of a mouse model of FXTAS suggested that this disorder involves neurodevelopment. To better understand the physiopathological role of the increased levels of Fmr1 mRNA during neuronal differentiation, we used a small interfering RNA (siRNA) approach to reduce the abundance of this mRNA in cultured cortical neurons from the FXTAS mouse model. Morphological alterations of neurons were rescued by this approach. This cellular phenotype is associated to differentially expressed proteins that we identified by mass spectrometry analysis. Interestingly, phenotype rescue is also associated to the rescue of the abundance of 29 proteins that are involved in various pathways, which represent putative targets for early therapeutic approaches.

15.
J Mol Biol ; 367(3): 702-14, 2007 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-17292399

RESUMO

Secreted phospholipases A(2) (sPLA(2)s) represent a new class of human immunodeficiency virus (HIV) inhibitors that block the early steps of virus entry into cells. Here, we applied an in vitro evolution/selection procedure to select, from primary HIV isolates, an emerging variant (HIV(RBV-3)) able to actively infect cells in the presence of sPLA(2)s. HIV(RBV-3) represents a very atypical HIV-1 isolate because, in contrast to others, this virus requires a functional endocytic machinery to infect cells. Indeed, endocytosis inhibitors that affect endosome acidification (bafilomycin A(1), monensin) and/or endosomal trafficking (nocodazole, latrunculin A) drastically reduced HIV(RBV-3) replication. Using a standardized PCR-assay, we showed that endocytosis inhibitors block HIV(RBV-3) entry just before the reverse transcription step. Concurrently, to identify the viral proteins responsible for the HIV(RBV-3) atypical behaviour, we constructed a HIV-1 molecular chimera bearing different HIV(RBV-3) proteins. We demonstrated that the sole presence of the HIV(RBV-3) envelope glycoprotein is enough, not only to confer the resistance to sPLA(2)s, but also to direct HIV(RBV-3) to the endosomal-dependent entry pathway. Interestingly, HIV(RBV-3) envelope glycoprotein sequencing revealed an unusual structural pattern with the presence of rare mutations in the N-terminal region and V1-V2 envelope loop sequence extensions. Taken together, we conclude that HIV-1 may escape from entry inhibitors, such as sPLA2s, through the selection of a particular HIV-1 envelope glycoprotein that allows HIV to infect cells via an alternative entry route that relies on endosome trafficking.


Assuntos
HIV-1/fisiologia , HIV-1/patogenicidade , Fosfolipases A/fisiologia , Sequência de Aminoácidos , Linhagem Celular , Endocitose/efeitos dos fármacos , Variação Genética , HIV-1/genética , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Macrolídeos/farmacologia , Fusão de Membrana , Dados de Sequência Molecular , Monensin/farmacologia , Fosfolipases A2 , Homologia de Sequência de Aminoácidos , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/fisiologia , Replicação Viral/efeitos dos fármacos
16.
BMJ Open ; 8(7): e023330, 2018 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-29980548

RESUMO

INTRODUCTION: Early-onset schizophrenia (EOS) is a rare and severe condition. A higher rate of neurodevelopmental abnormalities, such as intellectual or communication impairments as well as attention deficit hyperactivity disorder, is observed in EOS compared with adult-onset schizophrenia. Early signs of autism spectrum disorders (ASD) are present in about 30% of patients. Genetic abnormalities, including copy number variations, are frequent in neurodevelopmental disorders and have been associated to ASD physiopathology. Implicated genes encode proteins involved in brain development, synapses morphology and plasticity and neurogenesis. In addition, an increasing number of genetic abnormalities are shared by EOS and ASD, underlying the neurodevelopmental hypothesis of EOS.The main objective of our study is to identify disease-causing genetic mutations in a cohort of patients affected by both EOS and ASD. Special attention will be paid to genes involved in neurodevelopmental pathways. METHODS AND ANALYSIS: We describe a multicentric study in a paediatric population. The study started in April 2014. Inclusion criteria are: age 7-22 years, diagnosis of EOS with comorbid ASD and IQ >50; Parents and siblings are also enrolled. We perform psychiatric assessments (Mini International Neuropsychiatric Interview, Kiddie Schedule for Affective Disorders and Schizophrenia -Present and Lifetime Version, Positive and Negative Syndrome Scale and Scale for the Assessment of Negative Symptoms) together with neurocognitive evaluations (IQ, Trail Making Test A/B and verbal fluency). Then, we study variants of the coding part of DNA (exome), using next-generation sequencing process on trio (mother, father and child). Bioinformatics tools (RVIS and PolyPhen-2) are used to prioritise disease-causing mutations in candidate genes. The inclusion period will end in November 2019. ETHICS AND DISSEMINATION: The study protocol was approved by the Local Ethic Committee and by the French National Agency for Medicines and Health Products Safety. All patients signed informed consent on enrolment in the study. Results of the present study should help to unravel the molecular pathology of EOS, paving the way for an early therapeutic intervention. TRIAL REGISTRATION NUMBER: NCT0256552; Pre-results.


Assuntos
Transtorno do Espectro Autista/genética , Esquizofrenia/genética , Psicologia do Esquizofrênico , Adolescente , Idade de Início , Transtorno do Espectro Autista/fisiopatologia , Transtorno do Espectro Autista/psicologia , Criança , França , Genótipo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Pais , Fenótipo , Esquizofrenia/fisiopatologia , Análise de Sequência de DNA , Adulto Jovem
17.
Front Mol Neurosci ; 11: 342, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30319351

RESUMO

Fragile X syndrome (FXS), the most common form of inherited intellectual disability (ID) and a leading cause of autism, results from the loss of expression of the Fmr1 gene which encodes the RNA-binding protein Fragile X Mental Retardation Protein (FMRP). Among the thousands mRNA targets of FMRP, numerous encode regulators of ion homeostasis. It has also been described that FMRP directly interacts with Ca2+ channels modulating their activity. Collectively these findings suggest that FMRP plays critical roles in Ca2+ homeostasis during nervous system development. We carried out a functional analysis of Ca2+ regulation using a calcium imaging approach in Fmr1-KO cultured neurons and we show that these cells display impaired steady state Ca2+ concentration and an altered entry of Ca2+ after KCl-triggered depolarization. Consistent with these data, we show that the protein product of the Cacna1a gene, the pore-forming subunit of the Cav2.1 channel, is less expressed at the plasma membrane of Fmr1-KO neurons compared to wild-type (WT). Thus, our findings point out the critical role that Cav2.1 plays in the altered Ca2+ flux in Fmr1-KO neurons, impacting Ca2+ homeostasis of these cells. Remarkably, we highlight a new phenotype of cultured Fmr1-KO neurons that can be considered a novel cellular biomarker and is amenable to small molecule screening and identification of new drugs to treat FXS.

18.
Nat Commun ; 9(1): 757, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29472612

RESUMO

Fragile X syndrome (FXS) is the most frequent inherited cause of intellectual disability and the best-studied monogenic cause of autism. FXS results from the functional absence of the fragile X mental retardation protein (FMRP) leading to abnormal pruning and consequently to synaptic communication defects. Here we show that FMRP is a substrate of the small ubiquitin-like modifier (SUMO) pathway in the brain and identify its active SUMO sites. We unravel the functional consequences of FMRP sumoylation in neurons by combining molecular replacement strategy, biochemical reconstitution assays with advanced live-cell imaging. We first demonstrate that FMRP sumoylation is promoted by activation of metabotropic glutamate receptors. We then show that this increase in sumoylation controls the homomerization of FMRP within dendritic mRNA granules which, in turn, regulates spine elimination and maturation. Altogether, our findings reveal the sumoylation of FMRP as a critical activity-dependent regulatory mechanism of FMRP-mediated neuronal function.


Assuntos
Espinhas Dendríticas/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Sumoilação , Sequência de Aminoácidos , Animais , Células Cultivadas , Espinhas Dendríticas/genética , Espinhas Dendríticas/patologia , Feminino , Proteína do X Frágil da Deficiência Intelectual/química , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/metabolismo , Síndrome do Cromossomo X Frágil/patologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Modelos Neurológicos , Fenótipo , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Vesículas Secretórias/metabolismo , Homologia de Sequência de Aminoácidos
20.
Artigo em Inglês | MEDLINE | ID: mdl-29163124

RESUMO

Fragile X Syndrome (FXS) is the most common form of intellectual disability and a primary cause of autism. It originates from the lack of the Fragile X Mental Retardation Protein (FMRP), which is an RNA-binding protein encoded by the Fragile X Mental Retardation Gene 1 (FMR1) gene. Multiple roles have been attributed to this protein, ranging from RNA transport (from the nucleus to the cytoplasm, but also along neurites) to translational control of mRNAs. Over the last 20 years many studies have found a large number of FMRP mRNA targets, but it is still not clear which are those playing a critical role in the etiology of FXS. So far, no therapy for FXS has been found, making the quest for novel targets of considerable importance. Several pharmacological approaches have been attempted, but, despite some promising preclinical results, no strategy gave successful outcomes, due either to the induction of major side effects or to the lack of improvement of the phenotypes. However, these studies suggested that, in order to measure the effectiveness of a specific treatment, trials should be redesigned and new endpoints defined in FXS patients. Nevertheless, the search for new therapeutic targets for FXS is very active. In this context, the advances in animal modeling, coupled with better understanding of neurobiology and physiopathology of FXS, are of crucial importance in developing new selected treatments. Here, we discuss the pathways that were recently linked to the physiopathology of FXS (mGluR, GABAR, insulin, Insulin-like Growth Factor 1 (IGF-1), MPP-9, serotonin, oxytocin and endocannabinoid signaling) and that suggest new approaches to find an effective therapy for this disorder. Our goal with this review article is to summarize some recent relevant findings on FXS treatment strategies in order to have a clearer view of the different pathways analyzed to date emphasizing those shared with other synaptic disorders.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa