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










Intervalo de año de publicación
1.
J Neuroinflammation ; 18(1): 270, 2021 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-34789272

RESUMEN

Predisposition to autoimmunity and inflammatory disorders is observed in patients with fragile X-associated syndromes. These patients have increased numbers of CGG triplets in the 5' UTR region of FMR1 (Fragile X Mental Retardation 1) gene, that affects its expression. FMR1 is decreased in the thymus of myasthenia gravis (MG) patients, a prototypical autoimmune disease. We thus analyzed the number of CGG triplets in FMR1 in MG, and explored the regulatory mechanisms affecting thymic FMR1 expression. We measured the number of CGGs using thymic DNA from MG and controls, but no abnormalities in CGGs were found in MG that could explain thymic decrease of FMR1. We next analyzed by RT-PCR the expression of FMR1 and its transcription factors in thymic samples, and in thymic epithelial cell cultures in response to inflammatory stimuli. In control thymuses, FMR1 expression was higher in males than females, and correlated with CTCF (CCCTC-binding factor) expression. In MG thymuses, decreased expression of FMR1 was correlated with both CTCF and MAX (Myc-associated factor X) expression. Changes in FMR1 expression were supported by western blot analyses for FMRP. In addition, we demonstrated that FMR1, CTCF and MAX expression in thymic epithelial cells was also sensitive to inflammatory signals. Our results suggest that FMR1 could play a central role in the thymus and autoimmunity. First, in relation with the higher susceptibility of females to autoimmune diseases. Second, due to the modulation of its expression by inflammatory signals that are known to be altered in MG thymuses.


Asunto(s)
Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Miastenia Gravis/metabolismo , Timo/metabolismo , Adolescente , Adulto , Autoinmunidad/genética , Factor de Unión a CCCTC/biosíntesis , Factor de Unión a CCCTC/genética , Células Cultivadas , ADN/química , ADN/genética , Células Epiteliales/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Caracteres Sexuales , Adulto Joven
2.
J Neuroinflammation ; 18(1): 17, 2021 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-33407653

RESUMEN

BACKGROUND: FMRP is a selective mRNA-binding protein that regulates protein synthesis at synapses, and its loss may lead to the impairment of trace fear memory. Previously, we found that FMRP levels in the hippocampus of rats with post-traumatic stress disorder (PTSD) were decreased. However, the mechanism underlying these changes remains unclear. METHODS: Forty-eight male Sprague-Dawley rats were randomly divided into four groups. The experimental groups were treated with the single-prolonged stress (SPS) procedure and injected with a lentivirus-mediated inhibitor of miR-142-5p. Behavior test as well as morphology and molecular biology experiments were performed to detect the effect of miR-142 downregulation on PTSD, which was further verified by in vitro experiments. RESULTS: We found that silence of miRNA-142 (miR-142), an upstream regulator of FMRP, could alleviate PTSD-like behaviors of rats exposed to the SPS paradigm. MiR-142 silence not only decreased the levels of proinflammatory mediators, such as interleukin-1ß, interleukin-6, and tumor necrosis factor-α, but also increased the expressive levels of synaptic proteins including PSD95 and synapsin I in the hippocampus, which was one of the key brain regions associated with PTSD. We further detected that miR-142 silence also downregulated the transportation of nuclear factor kappa-B (NF-κB) into the nuclei of neurons and might further affect the morphology of neurons. CONCLUSIONS: The results revealed miR-142 downregulation could alleviate PTSD-like behaviors through attenuating neuroinflammation in the hippocampus of SPS rats by binding to FMRP.


Asunto(s)
Apoptosis/fisiología , Citocinas/biosíntesis , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Hipocampo/metabolismo , MicroARNs/biosíntesis , Trastornos por Estrés Postraumático/metabolismo , Animales , Células Cultivadas , Citocinas/antagonistas & inhibidores , Citocinas/genética , Regulación hacia Abajo/fisiología , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Expresión Génica , Inflamación/genética , Inflamación/metabolismo , Inflamación/prevención & control , Masculino , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Células PC12 , Ratas , Ratas Sprague-Dawley , Trastornos por Estrés Postraumático/genética , Trastornos por Estrés Postraumático/prevención & control , Regulación hacia Arriba/fisiología
3.
J Comp Neurol ; 529(3): 481-500, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32449186

RESUMEN

Afferent activity dynamically regulates neuronal properties and connectivity in the central nervous system. The Fragile X mental retardation protein (FMRP) is an RNA-binding protein that regulates cellular and synaptic properties in an activity-dependent manner. Whether and how FMRP level and localization are regulated by afferent input remains sparsely examined and how such regulation is associated with neuronal response to changes in sensory input is unknown. We characterized changes in FMRP level and localization in the chicken nucleus magnocellularis (NM), a primary cochlear nucleus, following afferent deprivation by unilateral cochlea removal. We observed rapid (within 2 hr) aggregation of FMRP immunoreactivity into large granular structures in a subset of deafferented NM neurons. Neurons that exhibited persistent FMRP aggregation at 12-24 hr eventually lost cytoplasmic Nissl substance, indicating cell death. A week later, FMRP expression in surviving neurons regained its homeostasis, with a slightly reduced immunostaining intensity and enhanced heterogeneity. Correlation analyses under the homeostatic status (7-14 days) revealed that neurons expressing relatively more FMRP had a higher capability of maintaining cell body size and ribosomal activity, as well as a better ability to detach inactive presynaptic terminals. Additionally, the intensity of an inhibitory postsynaptic protein, gephyrin, was reduced following deafferentation and was positively correlated with FMRP intensity, implicating an involvement of FMRP in synaptic dynamics in response to reduced afferent inputs. Collectively, this study demonstrates that afferent input regulates FMRP expression and localization in ways associated with multiple types of neuronal responses and synaptic rearrangements.


Asunto(s)
Cóclea/metabolismo , Nervio Coclear/metabolismo , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Sinapsis/metabolismo , Vías Aferentes/química , Vías Aferentes/metabolismo , Animales , Pollos , Cóclea/química , Nervio Coclear/química , Electroporación/métodos , Femenino , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/análisis , Masculino , Sinapsis/química
4.
Nat Neurosci ; 23(3): 386-397, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32066985

RESUMEN

Repeat-associated non-AUG-initiated translation of expanded CGG repeats (CGG RAN) from the FMR1 5'-leader produces toxic proteins that contribute to neurodegeneration in fragile X-associated tremor/ataxia syndrome. Here we describe how unexpanded CGG repeats and their translation play conserved roles in regulating fragile X protein (FMRP) synthesis. In neurons, CGG RAN acts as an inhibitory upstream open reading frame to suppress basal FMRP production. Activation of mGluR5 receptors enhances FMRP synthesis. This enhancement requires both the CGG repeat and CGG RAN initiation sites. Using non-cleaving antisense oligonucleotides (ASOs), we selectively blocked CGG RAN. This ASO blockade enhanced endogenous FMRP expression in human neurons. In human and rodent neurons, CGG RAN-blocking ASOs suppressed repeat toxicity and prolonged survival. These findings delineate a native function for CGG repeats and RAN translation in regulating basal and activity-dependent FMRP synthesis, and they demonstrate the therapeutic potential of modulating CGG RAN translation in fragile X-associated disorders.


Asunto(s)
Expansión de las Repeticiones de ADN/genética , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Repeticiones de Trinucleótidos/genética , Animales , Línea Celular , Supervivencia Celular/genética , Femenino , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Células Madre Pluripotentes Inducidas , Masculino , Ratones , Neuronas/metabolismo , Oligonucleótidos Antisentido/farmacología , Biosíntesis de Proteínas , Ratas , Ratas Long-Evans , Ratas Sprague-Dawley , Receptor del Glutamato Metabotropico 5/biosíntesis , Receptor del Glutamato Metabotropico 5/genética
5.
Acta Neuropathol Commun ; 7(1): 27, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30808398

RESUMEN

The fragile X premutation is a CGG trinucleotide repeat expansion between 55 and 200 repeats in the 5'-untranslated region of the fragile X mental retardation 1 (FMR1) gene. Human carriers of the premutation allele are at risk of developing the late-onset neurodegenerative disorder, fragile X-associated tremor/ataxia syndrome (FXTAS). Characteristic neuropathology associated with FXTAS includes intranuclear inclusions in neurons and astroglia. Previous studies recapitulated these histopathological features in neurons in a knock-in mouse model, but without significant astroglial pathology. To determine the role of astroglia in FXTAS, we generated a transgenic mouse line (Gfa2-CGG99-eGFP) that selectively expresses a 99-CGG repeat expansion linked to an enhanced green fluorescent protein (eGFP) reporter in astroglia throughout the brain, including cerebellar Bergmann glia. Behaviorally these mice displayed impaired motor performance on the ladder-rung test, but paradoxically better performance on the rotarod. Immunocytochemical analysis revealed that CGG99-eGFP co-localized with GFAP and S-100ß, but not with NeuN, Iba1, or MBP, indicating that CGG99-eGFP expression is specific to astroglia. Ubiquitin-positive intranuclear inclusions were found in eGFP-expressing glia throughout the brain. In addition, intracytoplasmic ubiquitin-positive inclusions were found outside the nucleus in distal astrocyte processes. Intriguingly, intranuclear inclusions, in the absence of eGFP mRNA and eGFP fluorescence, were present in neurons of the hypothalamus and neocortex. Furthermore, intranuclear inclusions in both neurons and astrocytes displayed immunofluorescent labeling for the polyglycine peptide FMRpolyG, implicating FMRpolyG in the pathology found in Gfa2-CGG99 mice. Considered together, these results show that Gfa2-CGG99 expression in mice is sufficient to induce key features of FXTAS pathology, including formation of intranuclear inclusions, translation of FMRpolyG, and deficits in motor function.


Asunto(s)
Astrocitos/fisiología , Ataxia/genética , Comunicación Celular/fisiología , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Trastornos de la Destreza Motora/genética , Temblor/genética , Expansión de Repetición de Trinucleótido/genética , Animales , Astrocitos/metabolismo , Astrocitos/patología , Ataxia/metabolismo , Ataxia/patología , Secuencia de Bases , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Síndrome del Cromosoma X Frágil/metabolismo , Síndrome del Cromosoma X Frágil/patología , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trastornos de la Destreza Motora/metabolismo , Trastornos de la Destreza Motora/patología , Temblor/metabolismo , Temblor/patología
6.
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 del Retraso Mental 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 del Retraso Mental 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
7.
Hum Mol Genet ; 26(5): 955-968, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28073926

RESUMEN

Renpenning syndrome is a group of X-linked intellectual disability syndromes caused by mutations in human polyglutamine-binding protein 1 (PQBP1) gene. Little is known about the molecular pathogenesis of the various mutations that cause the notable variability in patients. In this study, we examine the cellular and synaptic functions of the most common mutations found in the patients: c.461_462delAG, c.459_462delAGAG and c.463_464dupAG in an AG hexamer in PQBP1 exon 4. We discovered that PQBP1 c.459_462delAGAG and c.463_464dupAG mutations encode a new C-terminal epitope that preferentially binds non-phosphorylated fragile X mental retardation protein (FMRP) and promotes its ubiquitin-mediated degradation. Impairment of FMRP function up-regulates its targets such as MAP1B, and disrupts FMRP-dependent synaptic scaling in primary cultured neurons. In Drosophila neuromuscular junction model, PQBP1 c.463_464dupAG transgenic flies showed remarkable defects of synaptic over-growth, which can be rescued by exogenously expressing dFMRP. Our data strongly support a gain-of-function pathogenic mechanism of PQBP1 c.459_462delAGAG and c.463_464dupAG mutations, and suggest that therapeutic strategies to restore FMRP function may be beneficial for those patients.


Asunto(s)
Proteínas Portadoras/genética , Parálisis Cerebral/genética , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Discapacidad Intelectual/genética , Retraso Mental Ligado al Cromosoma X/genética , Proteínas Nucleares/genética , Animales , Animales Modificados Genéticamente , Proteínas Portadoras/biosíntesis , Parálisis Cerebral/metabolismo , Parálisis Cerebral/patología , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Drosophila/genética , Epítopos/genética , Epítopos/inmunología , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Humanos , Discapacidad Intelectual/inmunología , Discapacidad Intelectual/patología , Retraso Mental Ligado al Cromosoma X/metabolismo , Retraso Mental Ligado al Cromosoma X/patología , Proteínas Asociadas a Microtúbulos/genética , Mutación , Unión Neuromuscular , Proteínas Nucleares/biosíntesis , Péptidos/genética , Proteolisis , Ubiquitina/genética
8.
J Comp Neurol ; 525(4): 818-849, 2017 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-27539535

RESUMEN

The fragile X mental retardation protein (FMRP) plays an important role in normal brain development. Absence of FMRP results in abnormal neuronal morphologies in a selected manner throughout the brain, leading to intellectual deficits and sensory dysfunction in the fragile X syndrome (FXS). Despite FMRP importance for proper brain function, its overall expression pattern in the mammalian brain at the resolution of individual neuronal cell groups is not known. In this study we used FMR1 knockout and isogenic wildtype mice to systematically map the distribution of FMRP expression in the entire mouse brain. Using immunocytochemistry and cellular quantification analyses, we identified a large number of prominent cell groups expressing high levels of FMRP at the subcortical levels, in particular sensory and motor neurons in the brainstem and thalamus. In contrast, many cell groups in the midbrain and hypothalamus exhibit low FMRP levels. More important, we describe differential patterns of FMRP distribution in both cortical and subcortical brain regions. Almost all major brain areas contain high and low levels of FMRP cell groups adjacent to each other or between layers of the same cortical areas. These differential patterns indicate that FMRP expression appears to be specific to individual neuronal cell groups instead of being associated with all neurons in distinct brain regions, as previously considered. Taken together, these findings support the notion of FMRP differential neuronal regulation and strongly implicate the contribution of fundamental sensory and motor processing at subcortical levels to FXS pathology. J. Comp. Neurol. 525:818-849, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Encéfalo/metabolismo , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Animales , Western Blotting , Síndrome del Cromosoma X Frágil/metabolismo , Perfilación de la Expresión Génica , Inmunohistoquímica , Ratones , Ratones Noqueados , Transcriptoma
9.
J Neurochem ; 139(6): 1081-1092, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27365114

RESUMEN

Dynamic changes in synaptic strength rely on de novo protein synthesis and protein degradation by the ubiquitin proteasome system (UPS). Disruption of either of these cellular processes will result in significant impairments in synaptic plasticity and memory formation. Mutations in several genes encoding regulators of mRNA translation and members of the UPS have been associated with an increased risk for the development of autism spectrum disorders. It is possible that these mutations result in a similar imbalance in protein homeostasis (proteostasis) at the synapse. This review will summarize recent work investigating the role of the UPS in synaptic plasticity at glutamatergic synapses, and propose that dysfunctional proteostasis is a common consequence of several genetic mutations linked to autism spectrum disorders. Dynamic changes in synaptic strength rely on de novo protein synthesis and protein degradation by the ubiquitin proteasome system (UPS). Disruption of either of these cellular processes will result in significant impairments in synaptic plasticity and memory formation. Mutations in several genes encoding regulators of mRNA translation (i.e. FMR1) and protein degradation (i.e. UBE3A) have been associated with an increased risk for autism spectrum disorders and intellectual disability (ASD/ID). These mutations similarly disrupt protein homeostasis (proteostasis). Compensatory changes that reset the rate of proteostasis may contribute to the neurological symptoms of ASD/ID. This review summarizes recent work investigating the role of the UPS in synaptic plasticity at glutamatergic synapses, and proposes that dysfunctional proteostasis is a common consequence of several genetic mutations linked to ASD. This article is part of a mini review series: "Synaptic Function and Dysfunction in Brain Diseases".


Asunto(s)
Trastorno del Espectro Autista/metabolismo , Homeostasis/fisiología , Complejo de la Endopetidasa Proteasomal/biosíntesis , Biosíntesis de Proteínas/fisiología , Ubiquitina/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Trastorno del Espectro Autista/genética , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Humanos , Aprendizaje/fisiología , Complejo de la Endopetidasa Proteasomal/genética , Ubiquitina/genética
10.
Mol Cell ; 62(2): 314-322, 2016 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-27041225

RESUMEN

Repeat-associated non-AUG (RAN) translation produces toxic polypeptides from nucleotide repeat expansions in the absence of an AUG start codon and contributes to neurodegenerative disorders such as ALS and fragile X-associated tremor/ataxia syndrome. How RAN translation occurs is unknown. Here we define the critical sequence and initiation factors that mediate CGG repeat RAN translation in the 5' leader of fragile X mRNA, FMR1. Our results reveal that CGG RAN translation is 30%-40% as efficient as AUG-initiated translation, is m(7)G cap and eIF4E dependent, requires the eIF4A helicase, and is strongly influenced by repeat length. However, it displays a dichotomous requirement for initiation site selection between reading frames, with initiation in the +1 frame, but not the +2 frame, occurring at near-cognate start codons upstream of the repeat. These data support a model in which RAN translation at CGG repeats uses cap-dependent ribosomal scanning, yet bypasses normal requirements for start codon selection.


Asunto(s)
Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Degeneración Nerviosa , Biosíntesis de Proteínas , ARN Mensajero/genética , Repeticiones de Trinucleótidos , Factor 4E Eucariótico de Iniciación/genética , Factor 4E Eucariótico de Iniciación/metabolismo , Síndrome del Cromosoma X Frágil/diagnóstico , Síndrome del Cromosoma X Frágil/patología , Sistema de Lectura Ribosómico , Genes Reporteros , Predisposición Genética a la Enfermedad , Células HeLa , Humanos , Neuronas/metabolismo , Neuronas/patología , Sistemas de Lectura Abierta , Fenotipo , ARN Mensajero/metabolismo , Ribosomas/metabolismo , Sitio de Iniciación de la Transcripción , Transfección , Expansión de Repetición de Trinucleótido
11.
Exp Eye Res ; 146: 72-82, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26719241

RESUMEN

Fragile X Mental Retardation Protein (FMRP) is a RNA-binding protein that modulates protein synthesis at the synapse and its function is regulated by glutamate. The retina is the first structure that participates in vision, and uses glutamate to transduce electromagnetic signals from light to electrochemical signals to neurons. FMRP has been previously detected in the retina, but its localization has not been studied yet. In this work, our objectives were to describe the localization of FMRP in the retina, to determine whether different exposure to dark or light stimulus alters FMRP expression in the retina, and to compare the pattern in two different species, the mouse and chick. We found that both FMRP mRNA and protein are expressed in the retina. By immunohistochemistry analysis we found that both mouse and chick present similar FMRP expression localized mainly in both plexiform layers and the inner retina. It was also observed that FMRP is down-regulated by 24 h dark adaptation compared to its expression in the retina of animals that were exposed to light for 1 h after 24 h in the dark. We conclude that FMRP is likely to participate in retinal physiology, since its expression changes with light exposure. In addition, the expression pattern and regulation by light of FMRP seems well conserved since it was similar in both mouse and chick.


Asunto(s)
Adaptación a la Oscuridad/fisiología , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Regulación de la Expresión Génica , Luz , ARN/genética , Retina/metabolismo , Animales , Pollos , Femenino , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/efectos de la radiación , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL
12.
PLoS Genet ; 11(12): e1005702, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26637167

RESUMEN

Fragile X syndrome (FXS) is the most frequent inherited form of mental retardation. The cause for this X-linked disorder is the silencing of the fragile X mental retardation 1 (fmr1) gene and the absence of the fragile X mental retardation protein (Fmrp). The RNA-binding protein Fmrp represses protein translation, particularly in synapses. In Drosophila, Fmrp interacts with the adenosine deaminase acting on RNA (Adar) enzymes. Adar enzymes convert adenosine to inosine (A-to-I) and modify the sequence of RNA transcripts. Utilizing the fmr1 zebrafish mutant (fmr1-/-), we studied Fmrp-dependent neuronal circuit formation, behavior, and Adar-mediated RNA editing. By combining behavior analyses and live imaging of single axons and synapses, we showed hyperlocomotor activity, as well as increased axonal branching and synaptic density, in fmr1-/- larvae. We identified thousands of clustered RNA editing sites in the zebrafish transcriptome and showed that Fmrp biochemically interacts with the Adar2a protein. The expression levels of the adar genes and Adar2 protein increased in fmr1-/- zebrafish. Microfluidic-based multiplex PCR coupled with deep sequencing showed a mild increase in A-to-I RNA editing levels in evolutionarily conserved neuronal and synaptic Adar-targets in fmr1-/- larvae. These findings suggest that loss of Fmrp results in increased Adar-mediated RNA editing activity on target-specific RNAs, which, in turn, might alter neuronal circuit formation and behavior in FXS.


Asunto(s)
Adenosina Desaminasa/genética , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Proteínas de Unión al ARN/genética , Proteínas de Pez Cebra/genética , Adenosina Desaminasa/biosíntesis , Animales , Axones/metabolismo , Axones/patología , Modelos Animales de Enfermedad , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Síndrome del Cromosoma X Frágil/patología , Regulación del Desarrollo de la Expresión Génica , Humanos , Actividad Motora/genética , Neuronas/metabolismo , Neuronas/patología , Edición de ARN/genética , Proteínas de Unión al ARN/biosíntesis , Sinapsis/metabolismo , Sinapsis/patología , Transcriptoma/genética , Pez Cebra , Proteínas de Pez Cebra/biosíntesis
13.
J Neurosci ; 35(45): 15073-81, 2015 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-26558778

RESUMEN

Previous studies have hypothesized that diverse genetic causes of intellectual disability (ID) and autism spectrum disorders (ASDs) converge on common cellular pathways. Testing this hypothesis requires detailed phenotypic analyses of animal models with genetic mutations that accurately reflect those seen in the human condition (i.e., have structural validity) and which produce phenotypes that mirror ID/ASDs (i.e., have face validity). We show that SynGAP haploinsufficiency, which causes ID with co-occurring ASD in humans, mimics and occludes the synaptic pathophysiology associated with deletion of the Fmr1 gene. Syngap(+/-) and Fmr1(-/y) mice show increases in basal protein synthesis and metabotropic glutamate receptor (mGluR)-dependent long-term depression that, unlike in their wild-type controls, is independent of new protein synthesis. Basal levels of phosphorylated ERK1/2 are also elevated in Syngap(+/-) hippocampal slices. Super-resolution microscopy reveals that Syngap(+/-) and Fmr1(-/y) mice show nanoscale alterations in dendritic spine morphology that predict an increase in biochemical compartmentalization. Finally, increased basal protein synthesis is rescued by negative regulators of the mGlu subtype 5 receptor and the Ras-ERK1/2 pathway, indicating that therapeutic interventions for fragile X syndrome may benefit patients with SYNGAP1 haploinsufficiency. SIGNIFICANCE STATEMENT: As the genetics of intellectual disability (ID) and autism spectrum disorders (ASDs) are unraveled, a key issue is whether genetically divergent forms of these disorders converge on common biochemical/cellular pathways and hence may be amenable to common therapeutic interventions. This study compares the pathophysiology associated with the loss of fragile X mental retardation protein (FMRP) and haploinsufficiency of synaptic GTPase-activating protein (SynGAP), two prevalent monogenic forms of ID. We show that Syngap(+/-) mice phenocopy Fmr1(-/y) mice in the alterations in mGluR-dependent long-term depression, basal protein synthesis, and dendritic spine morphology. Deficits in basal protein synthesis can be rescued by pharmacological interventions that reduce the mGlu5 receptor-ERK1/2 signaling pathway, which also rescues the same deficit in Fmr1(-/y) mice. Our findings support the hypothesis that phenotypes associated with genetically diverse forms of ID/ASDs result from alterations in common cellular/biochemical pathways.


Asunto(s)
Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Hipocampo/metabolismo , Hipocampo/fisiopatología , Proteínas Activadoras de ras GTPasa/biosíntesis , Animales , Espinas Dendríticas/metabolismo , Espinas Dendríticas/patología , Potenciales Postsinápticos Excitadores/fisiología , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Proteínas Activadoras de ras GTPasa/genética
14.
Rev. panam. salud pública ; 38(5): 347-354, Nov. 2015. ilus, tab
Artículo en Inglés | LILACS | ID: lil-772129

RESUMEN

OBJECTIVE: To explore distributional inequality of key health outcomes as determined by access coverage to water and sanitation (WS) between countries in the Region of the Americas. METHODS: An ecological study was designed to explore the magnitude and change-over-time of standard gap and gradient metrics of environmental inequalities in health at the country level in 1990 and 2010 among the 35 countries of the Americas. Access to drinking water and access to improved sanitation facilities were selected as equity stratifiers. Five dependent variables were: total and healthy life expectancies at birth, and infant, under-5, and maternal mortality. RESULTS: Access to WS correlated with survival and mortality, and strong gradients were seen in both 1990 and 2010. Higher WS access corresponded to higher life expectancy and healthy life expectancy and lower infant, under-5, and maternal mortality risks. Burden of life lost was unequally distributed, steadily concentrated among the most environmentally disadvantaged, who carried up to twice the burden than they would if WS were fairly distributed. Population averages in life expectancy and specific mortality improved, but whereas absolute inequalities decreased, relative inequalities remained mostly invariant. CONCLUSIONS: Even with the Region on track to meet MDG 7 on water and sanitation, large environmental gradients and health inequities among countries remain hidden by Regional averages. As the post-2015 development agenda unfolds, policies and actions focused on health equity-mainly on the most socially and environmentally deprived-will be needed in order to secure the right for universal access to water and sanitation.


OBJETIVO:Explorar la desigualdad distributiva de resultados clave en salud determinada por la cobertura de acceso a agua y saneamiento (AS) entre países en la Región de las Américas. MÉTODOS: Se diseñó un estudio ecológico para explorar la magnitud y el cambio en el tiempo de métricas estándar de brecha y gradiente de desigualdades ambientales en salud a nivel país en 1990 y 2010 entre los 35 países de las Américas. El acceso a agua potable y el acceso a instalaciones sanitarias mejoradas fueron seleccionados como estratificadores de equidad. Las cinco variables dependientes fueron: expectativa de vida al nacer total y saludable, mortalidad infantil, en menores de cinco años y materna. RESULTADOS: El acceso a AS se correlacionó con la supervivencia y mortalidad y se observaron intensos gradientes tanto en 1990 como en 2010. Un acceso a AS más alto se correspondió con más alta expectativa de vida al nacer total y saludable y con más bajos riesgos de muerte infantil, en menores de 5 años y materna. La carga de vida perdida se distribuyó inequitativamente, concentrándose de manera sostenida entre los más desaventajados ambientalmente, quienes acarrearon hasta dos veces la carga que hubieran acarreado si el acceso a AS hubiese estado equitativamente distribuido. Los promedios poblacionales en la expectativa de vida y la mortalidad específica mejoraron pero, mientras que las desigualdades absolutas se redujeron, las desigualdades relativas se mantuvieron esencialmente invariantes. CONCLUSIONES: Aún cuando la Región está en curso para alcanzar el ODM 7 sobre agua y saneamiento, los promedios regionales siguen ocultando grandes gradientes ambientales y desigualdades en salud entre países. A medida que se despliega la agenda de desarrollo post-2015, serán necesarias políticas y acciones orientadas a la equidad en salud -principalmente hacia aquellos con mayor privación social y ambiental- a fin de asegurar el derecho por el acceso universal al agua y saneamiento.


Asunto(s)
Humanos , Animales , Ratones , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Homeostasis/genética , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Síndrome del Cromosoma X Frágil/fisiopatología , Expresión Génica , ARN Mensajero/biosíntesis , ARN Mensajero/genética
15.
Nat Rev Neurosci ; 16(10): 595-605, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26350240

RESUMEN

Fragile X syndrome (FXS), the most-frequently inherited form of intellectual disability and the most-prevalent single-gene cause of autism, results from a lack of fragile X mental retardation protein (FMRP), an RNA-binding protein that acts, in most cases, to repress translation. Multiple pharmacological and genetic manipulations that target receptors, scaffolding proteins, kinases and translational control proteins can rescue neuronal morphology, synaptic function and behavioural phenotypes in FXS model mice, presumably by reducing excessive neuronal translation to normal levels. Such rescue strategies might also be explored in the future to identify the mRNAs that are critical for FXS pathophysiology.


Asunto(s)
Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Homeostasis/genética , Animales , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Síndrome del Cromosoma X Frágil/fisiopatología , Expresión Génica , Humanos , Ratones , ARN Mensajero/biosíntesis , ARN Mensajero/genética
16.
Sci Rep ; 5: 10468, 2015 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-26020477

RESUMEN

Fragile X mental retardation protein (FMRP) is an RNA-binding protein important for the control of translation and synaptic function. The mutation or silencing of FMRP causes Fragile X syndrome (FXS), which leads to intellectual disability and social impairment. γ-Aminobutyric acid (GABA) is the major inhibitory neurotransmitter of the mammalian central nervous system, and its metabotropic GABAB receptor has been implicated in various mental disorders. The GABAB receptor agonist baclofen has been shown to improve FXS symptoms in a mouse model and in human patients, but the signaling events linking the GABAB receptor and FMRP are unknown. In this study, we found that GABAB receptor activation upregulated cAMP response element binding protein-dependent Fmrp expression in cultured mouse cerebellar granule neurons via two distinct mechanisms: the transactivation of insulin-like growth factor-1 receptor and activation of protein kinase C. In addition, a positive allosteric modulator of the GABAB receptor, CGP7930, stimulated Fmrp expression in neurons. These results suggest a role for GABAB receptor in Fmrp regulation and a potential interest of GABAB receptor signaling in FXS improvement.


Asunto(s)
Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Síndrome del Cromosoma X Frágil/genética , Receptores de GABA-B/genética , Ácido gamma-Aminobutírico/genética , Animales , Baclofeno/administración & dosificación , Modelos Animales de Enfermedad , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/tratamiento farmacológico , Síndrome del Cromosoma X Frágil/fisiopatología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Receptores de GABA-B/biosíntesis , Receptores de Somatomedina/biosíntesis , Receptores de Somatomedina/genética , Transducción de Señal/efectos de los fármacos , Ácido gamma-Aminobutírico/metabolismo
17.
Biochimie ; 114: 147-54, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25701550

RESUMEN

Translational control is a common mechanism used to regulate gene expression and occur in bacteria to mammals. Typically in translational control, an RNA-binding protein binds to a unique sequence in the mRNA to regulate protein synthesis by the ribosomes. Alternatively, a protein may bind to or modify a translation factor to globally regulate protein synthesis by the cell. Here, we review translational control by the fragile X mental retardation protein (FMRP), the absence of which causes the neurological disease, fragile X syndrome (FXS).


Asunto(s)
Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Biosíntesis de Proteínas , Animales , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/tratamiento farmacológico , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Regulación de la Expresión Génica , Humanos , Terapia Molecular Dirigida , Proteínas de Unión al ARN/fisiología
18.
PLoS Genet ; 9(10): e1003890, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24204304

RESUMEN

Fragile X syndrome is caused by loss of function of a single gene encoding the Fragile X Mental Retardation Protein (FMRP). This RNA-binding protein, widely expressed in mammalian tissues, is particularly abundant in neurons and is a component of messenger ribonucleoprotein (mRNP) complexes present within the translational apparatus. The absence of FMRP in neurons is believed to cause translation dysregulation and defects in mRNA transport essential for local protein synthesis and for synaptic development and maturation. A prevalent model posits that FMRP is a nucleocytoplasmic shuttling protein that transports its mRNA targets from the nucleus to the translation machinery. However, it is not known which of the multiple FMRP isoforms, resulting from the numerous alternatively spliced FMR1 transcripts variants, would be involved in such a process. Using a new generation of anti-FMRP antibodies and recombinant expression, we show here that the most commonly expressed human FMRP isoforms (ISO1 and 7) do not localize to the nucleus. Instead, specific FMRP isoforms 6 and 12 (ISO6 and 12), containing a novel C-terminal domain, were the only isoforms that localized to the nuclei in cultured human cells. These isoforms localized to specific p80-coilin and SMN positive structures that were identified as Cajal bodies. The Cajal body localization signal was confined to a 17 amino acid stretch in the C-terminus of human ISO6 and is lacking in a mouse Iso6 variant. As FMRP is an RNA-binding protein, its presence in Cajal bodies suggests additional functions in nuclear post-transcriptional RNA metabolism. Supporting this hypothesis, a missense mutation (I304N), known to alter the KH2-mediated RNA binding properties of FMRP, abolishes the localization of human FMRP ISO6 to Cajal bodies. These findings open unexplored avenues in search for new insights into the pathophysiology of Fragile X Syndrome.


Asunto(s)
Cuerpos Enrollados/genética , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Isoformas de Proteínas/biosíntesis , Animales , Núcleo Celular/genética , Núcleo Celular/ultraestructura , Cuerpos Enrollados/ultraestructura , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Síndrome del Cromosoma X Frágil/patología , Regulación de la Expresión Génica , Humanos , Ratones , Neuronas/metabolismo , Isoformas de Proteínas/ultraestructura , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Ribonucleoproteínas/genética
19.
JAMA Neurol ; 70(8): 1022-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23753897

RESUMEN

IMPORTANCE: Individuals with the fragile X premutation express expanded CGG repeats (repeats 55-200) in the FMR1 gene and elevated FMR1 messenger RNA (mRNA) levels, both of which may underlie the occurrence of the late-onset neurodegenerative disorder fragile X-associated tremor/ataxia syndrome (FXTAS). Because the core feature of FXTAS is motor impairment, determining the influence of FMR1 mRNA levels on structural connectivity of motor fiber tracts is critical for a better understanding of the pathologic features of FXTAS. OBJECTIVE: To examine the associations of CGG repeat and FMR1 mRNA with motor-related fiber tracts in males with premutation alleles. DESIGN AND SETTING: A case-control study conducted at the University of California, Davis, from April 1, 2008, through August 31, 2009. All data were collected masked to the carrier status of the FMR1 gene. PARTICIPANTS: Thirty-six male premutation carriers with FXTAS and 26 male premutation carriers without FXTAS were recruited through their family relationships with children affected by fragile X syndrome. The controls were 34 unaffected family members and healthy volunteers from the local community. MAIN OUTCOMES AND MEASURES: The CGG repeat lengths and FMR1 mRNA expression levels in peripheral blood lymphocytes, motor functioning, and white matter structural integrity that were estimated using diffusion tensor imaging. After data collection, we selected 4 motor tracts to reconstruct using diffusion tensor tractography, namely, the middle and superior cerebellar peduncles, descending motor tracts (containing the corticospinal, corticobulbar, and corticopontine tracts), and the anterior body of the corpus callosum. RESULTS: All fiber tracts exhibited weaker structural connectivity in the FXTAS group (decreased 5%-53% from controls, P ≤ .02). Genetic imaging correlation analysis revealed negative associations of CGG repeat length and FMR1 mRNA with connectivity strength of the superior cerebellar peduncles in both premutation groups (partial r² = 0.23-0.33, P ≤ .004). In addition, the measurements from the corpus callosum and superior cerebellar peduncles revealed a high correlation with motor functioning in all 3 groups (r between partial least square predicted and actual test scores = 0.41-0.56, P ≤ .04). CONCLUSIONS AND RELEVANCE: Distinct pathophysiologic processes may underlie the structural impairment of the motor tracts in FXTAS. Although both the corpus callosum and superior cerebellar peduncles were of great importance to motor functioning, only the superior cerebellar peduncles exhibited an association with the elevated RNA levels in the blood of fragile X premutation carriers.


Asunto(s)
Alelos , Ataxia/genética , Química Encefálica/genética , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Mutación/genética , Temblor/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Ataxia/patología , Ataxia/fisiopatología , Estudios de Casos y Controles , Imagen de Difusión Tensora/instrumentación , Imagen de Difusión Tensora/métodos , Vías Eferentes/metabolismo , Vías Eferentes/patología , Vías Eferentes/fisiopatología , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Síndrome del Cromosoma X Frágil/patología , Síndrome del Cromosoma X Frágil/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , ARN Mensajero/biosíntesis , Temblor/patología , Temblor/fisiopatología , Expansión de Repetición de Trinucleótido/genética , Adulto Joven
20.
Hum Mol Genet ; 22(10): 1971-82, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23390134

RESUMEN

While FMR1 is silenced in Fragile X syndrome (FXS) patients carrying the full mutation, its expression is elevated (2-8 fold) in premutated individuals. These people may develop the Fragile X-associated Tremor/Ataxia syndrome (FXTAS), a late onset neurodegenerative disorder characterized by ataxia and parkinsonism. In addition, people carrying the premutation can be affected by a set of neurological and behavioral disorders during young age. Problems of memory have been detected in these patients as well as in the mouse models for FXTAS. To date little is known concerning the metabolism of FMR1 mRNA, notwithstanding the importance of the finely tuned regulation of the expression of this gene. In the present study, we identified three microRNAs that specifically target the 3' UTR of FMR1 and can modulate its expression throughout the brain particularly at the synapse where their expression is very high. The expression level of miR-221 is reduced in synaptosomal preparations of young FXTAS mice suggesting a general deregulation of transcripts located at the synapse of these mice. By transcriptome analysis, we show here a robust deregulation of the expression levels of genes involved in learning, memory and autistic behavior, Parkinson disease and neurodegeneration. These findings suggest the presence of a synaptopathy in these animals. Interestingly, many of those deregulated mRNAs are target of the same microRNAs that modulate the expression of FMR1 at the synapse.


Asunto(s)
Regiones no Traducidas 3' , Ataxia/metabolismo , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/biosíntesis , Síndrome del Cromosoma X Frágil/metabolismo , MicroARNs/metabolismo , Sinapsis/metabolismo , Temblor/metabolismo , Animales , Ataxia/genética , Ataxia/patología , Células COS , Chlorocebus aethiops , Modelos Animales de Enfermedad , Proteína del Retraso Mental del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/patología , Células HeLa , Humanos , Ratones , MicroARNs/genética , Células 3T3 NIH , Sinapsis/genética , Sinapsis/patología , Temblor/genética , Temblor/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...