Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Cell ; 148(1-2): 296-308, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22265417

RESUMEN

Although uridine-rich small nuclear RNAs (U-snRNAs) are essential for pre-mRNA splicing, little is known regarding their function in the regulation of alternative splicing or of the biological consequences of their dysfunction in mammals. Here, we demonstrate that mutation of Rnu2-8, one of the mouse multicopy U2 snRNA genes, causes ataxia and neurodegeneration. Coincident with the observed pathology, the level of mutant U2 RNAs was highest in the cerebellum and increased after granule neuron maturation. Furthermore, neuron loss was strongly dependent on the dosage of mutant and wild-type snRNA genes. Comprehensive transcriptome analysis identified a group of alternative splicing events, including the splicing of small introns, which were disrupted in the mutant cerebellum. Our results suggest that the expression of mammalian U2 snRNA genes, previously presumed to be ubiquitous, is spatially and temporally regulated, and dysfunction of a single U2 snRNA causes neuron degeneration through distortion of pre-mRNA splicing.


Asunto(s)
Empalme Alternativo , ARN Nuclear Pequeño/genética , Animales , Ataxia/genética , Secuencia de Bases , Cerebelo/citología , Cerebelo/metabolismo , Perfilación de la Expresión Génica , Ratones , Datos de Secuencia Molecular , Mutagénesis , Mutación , Enfermedades Neurodegenerativas/genética , Alineación de Secuencia
2.
Nucleic Acids Res ; 51(19): 10768-10781, 2023 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-37739431

RESUMEN

Translational readthrough of UGA stop codons by selenocysteine-specific tRNA (tRNASec) enables the synthesis of selenoproteins. Seryl-tRNA synthetase (SerRS) charges tRNASec with serine, which is modified into selenocysteine and delivered to the ribosome by a designated elongation factor (eEFSec in eukaryotes). Here we found that components of the human selenocysteine incorporation machinery (SerRS, tRNASec, and eEFSec) also increased translational readthrough of non-selenocysteine genes, including VEGFA, to create C-terminally extended isoforms. SerRS recognizes target mRNAs through a stem-loop structure that resembles the variable loop of its cognate tRNAs. This function of SerRS depends on both its enzymatic activity and a vertebrate-specific domain. Through eCLIP-seq, we identified additional SerRS-interacting mRNAs as potential readthrough genes. Moreover, SerRS overexpression was sufficient to reverse premature termination caused by a pathogenic nonsense mutation. Our findings expand the repertoire of selenoprotein biosynthesis machinery and suggest an avenue for therapeutic targeting of nonsense mutations using endogenous factors.


Asunto(s)
Biosíntesis de Proteínas , Serina-ARNt Ligasa , Humanos , Codón sin Sentido , Codón de Terminación , ARN Mensajero/metabolismo , Selenocisteína/genética , Selenocisteína/metabolismo , Selenoproteínas/genética , Serina-ARNt Ligasa/genética
3.
Proc Natl Acad Sci U S A ; 119(10): e2119529119, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35238631

RESUMEN

SignificanceUnderstanding and treating neurological disorders are global priorities. Some of these diseases are engendered by mutations that cause defects in the cellular synthesis of transfer RNAs (tRNAs), which function as adapter molecules that translate messenger RNAs into proteins. During tRNA biogenesis, ribonuclease P catalyzes removal of the transcribed sequence upstream of the mature tRNA. Here, we focus on a cytoplasmic tRNAArgUCU that is expressed specifically in neurons and, when harboring a particular point mutation, contributes to neurodegeneration in mice. Our results suggest that this mutation favors stable alternative structures that are not cleaved by mouse ribonuclease P and motivate a paradigm that may help to understand the molecular basis for disease-associated mutations in other tRNAs.


Asunto(s)
Homeostasis , Neuronas/metabolismo , Conformación de Ácido Nucleico , ARN de Transferencia/metabolismo , Animales , Emparejamiento Base , Corteza Cerebral/enzimología , Magnesio/metabolismo , Ratones , Modelos Moleculares , Mutación Puntual , Procesamiento Proteico-Postraduccional , ARN de Transferencia/química , ARN de Transferencia/genética , Ribonucleasa P/aislamiento & purificación , Ribonucleasa P/metabolismo , Especificidad por Sustrato
4.
Nature ; 560(7720): E35, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29925958

RESUMEN

In the Fig. 3b western blot of this Article, 'Myc-AlaRS' in row one should have been 'Myc-AAD Aars', 'AlaRS' in row two should have been 'Aars' and 'ANKRD16' in row four should have been 'Ankrd16'. In Fig. 4f, 'ANKRD16' and 'ANKRD16(3xR)' should have been 'Ankrd16' and 'Ankrd163xR; and in Fig. 3c the position of the molecular mass markers had shifted. These figures have been corrected online, and see Supplementary Information to the accompanying Amendment for the original figure.

5.
Nature ; 557(7706): 510-515, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29769718

RESUMEN

Editing domains of aminoacyl tRNA synthetases correct tRNA charging errors to maintain translational fidelity. A mutation in the editing domain of alanyl tRNA synthetase (AlaRS) in Aars sti mutant mice results in an increase in the production of serine-mischarged tRNAAla and the degeneration of cerebellar Purkinje cells. Here, using positional cloning, we identified Ankrd16, a gene that acts epistatically with the Aars sti mutation to attenuate neurodegeneration. ANKRD16, a vertebrate-specific protein that contains ankyrin repeats, binds directly to the catalytic domain of AlaRS. Serine that is misactivated by AlaRS is captured by the lysine side chains of ANKRD16, which prevents the charging of serine adenylates to tRNAAla and precludes serine misincorporation in nascent peptides. The deletion of Ankrd16 in the brains of Aarssti/sti mice causes widespread protein aggregation and neuron loss. These results identify an amino-acid-accepting co-regulator of tRNA synthetase editing as a new layer of the machinery that is essential to the prevention of severe pathologies that arise from defects in editing.


Asunto(s)
Alanina-ARNt Ligasa/genética , Alanina-ARNt Ligasa/metabolismo , Mutación , Biosíntesis de Proteínas , Células de Purkinje/enzimología , Células de Purkinje/patología , Alanina/metabolismo , Alanina-ARNt Ligasa/química , Animales , Dominio Catalítico , Muerte Celular , Femenino , Lisina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Células de Purkinje/metabolismo , Serina/metabolismo
6.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34548404

RESUMEN

Homozygous mutation of the RNA kinase CLP1 (cleavage factor polyribonucleotide kinase subunit 1) causes pontocerebellar hypoplasia type 10 (PCH10), a pediatric neurodegenerative disease. CLP1 is associated with the transfer RNA (tRNA) splicing endonuclease complex and the cleavage and polyadenylation machinery, but its function remains unclear. We generated two mouse models of PCH10: one homozygous for the disease-associated Clp1 mutation, R140H, and one heterozygous for this mutation and a null allele. Both models exhibit loss of lower motor neurons and neurons of the deep cerebellar nuclei. To explore whether Clp1 mutation impacts tRNA splicing, we profiled the products of intron-containing tRNA genes. While mature tRNAs were expressed at normal levels in mutant mice, numerous other products of intron-containing tRNA genes were dysregulated, with pre-tRNAs, introns, and certain tRNA fragments up-regulated, and other fragments down-regulated. However, the spatiotemporal patterns of dysregulation do not correlate with pathogenicity for most altered tRNA products. To elucidate the effect of Clp1 mutation on precursor messenger RNA (pre-mRNA) cleavage, we analyzed poly(A) site (PAS) usage and gene expression in Clp1R140H/- spinal cord. PAS usage was shifted from proximal to distal sites in the mutant mouse, particularly in short and closely spaced genes. Many such genes were also expressed at lower levels in the Clp1R140H/- mouse, possibly as a result of impaired transcript maturation. These findings are consistent with the hypothesis that select genes are particularly dependent upon CLP1 for proper pre-mRNA cleavage, suggesting that impaired mRNA 3' processing may contribute to pathogenesis in PCH10.


Asunto(s)
Enfermedades Cerebelosas/patología , Enfermedades Neurodegenerativas/patología , Poliadenilación , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , ARN de Transferencia/metabolismo , Proteínas de Unión al ARN/fisiología , Factores de Transcripción/fisiología , Animales , Enfermedades Cerebelosas/genética , Enfermedades Cerebelosas/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Mensajero/genética , ARN de Transferencia/genética
7.
Cell ; 133(6): 949-51, 2008 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-18555770

RESUMEN

Dominantly inherited mutations in an endoplasmic reticulum protein called VAPB have been found in a subset of patients with a rare familial form of amyotrophic lateral sclerosis (ALS). In this issue, Tsuda et al. (2008) identify a secreted form of VAPB that binds directly to Eph receptors inducing their activation and signaling, providing fresh insights into ALS pathogenesis, including non-neuronal aspects of this disorder.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Receptores de la Familia Eph/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Humanos , Transducción de Señal
8.
Nucleic Acids Res ; 49(7): 3603-3616, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33341895

RESUMEN

During mRNA translation, tRNAs are charged by aminoacyl-tRNA synthetases and subsequently used by ribosomes. A multi-enzyme aminoacyl-tRNA synthetase complex (MSC) has been proposed to increase protein synthesis efficiency by passing charged tRNAs to ribosomes. An alternative function is that the MSC repurposes specific synthetases that are released from the MSC upon cues for functions independent of translation. To explore this, we generated mammalian cells in which arginyl-tRNA synthetase and/or glutaminyl-tRNA synthetase were absent from the MSC. Protein synthesis, under a variety of stress conditions, was unchanged. Most strikingly, levels of charged tRNAArg and tRNAGln remained unchanged and no ribosome pausing was observed at codons for arginine and glutamine. Thus, increasing or regulating protein synthesis efficiency is not dependent on arginyl-tRNA synthetase and glutaminyl-tRNA synthetase in the MSC. Alternatively, and consistent with previously reported ex-translational roles requiring changes in synthetase cellular localizations, our manipulations of the MSC visibly changed localization.


Asunto(s)
Aminoacil-ARNt Sintetasas/metabolismo , Biosíntesis de Proteínas , ARN de Transferencia de Arginina/metabolismo , ARN de Transferencia de Glutamina/metabolismo , Ribosomas/metabolismo , Animales , Fibroblastos , Células HEK293 , Humanos , Ratones
9.
Trends Genet ; 34(3): 218-231, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29352613

RESUMEN

Errors during mRNA translation can lead to a reduction in the levels of functional proteins and an increase in deleterious molecules. Advances in next-generation sequencing have led to the discovery of rare genetic disorders, many caused by mutations in genes encoding the mRNA translation machinery, as well as to a better understanding of translational dynamics through ribosome profiling. We discuss here multiple neurological disorders that are linked to errors in tRNA aminoacylation and ribosome decoding. We draw on studies from genetic models, including yeast and mice, to enhance our understanding of the translational defects observed in these diseases. Finally, we emphasize the importance of tRNA, their associated enzymes, and the inextricable link between accuracy and efficiency in the maintenance of translational fidelity.


Asunto(s)
Mutación , Enfermedades del Sistema Nervioso/genética , Biosíntesis de Proteínas/genética , ARN Mensajero/genética , Animales , Humanos , Modelos Genéticos , Saccharomyces cerevisiae/genética , Aminoacilación de ARN de Transferencia/genética
10.
Proc Natl Acad Sci U S A ; 111(49): 17570-5, 2014 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-25422440

RESUMEN

Misfolded proteins are an emerging hallmark of cardiac diseases. Although some misfolded proteins, such as desmin, are associated with mutations in the genes encoding these disease-associated proteins, little is known regarding more general mechanisms that contribute to the generation of misfolded proteins in the heart. Reduced translational fidelity, caused by a hypomorphic mutation in the editing domain of alanyl-tRNA synthetase (AlaRS), resulted in accumulation of misfolded proteins in specific mouse neurons. By further genetic modulation of the editing activity of AlaRS, we generated mouse models with broader phenotypes, the severity of which was directly related to the degree of compromised editing. Severe disruption of the editing activity of AlaRS caused embryonic lethality, whereas an intermediate reduction in AlaRS editing efficacy resulted in ubiquitinated protein aggregates and mitochondrial defects in cardiomyocytes that were accompanied by progressive cardiac fibrosis and dysfunction. In addition, autophagic vacuoles accumulated in mutant cardiomyocytes, suggesting that autophagy is insufficient to eliminate misfolded proteins. These findings demonstrate that the pathological consequences of diminished tRNA synthetase editing activity, and thus translational infidelity, are dependent on the cell type and the extent of editing disruption, and provide a previously unidentified mechanism underlying cardiac proteinopathy.


Asunto(s)
Alanina-ARNt Ligasa/deficiencia , Alanina-ARNt Ligasa/genética , Cardiopatías/genética , Deficiencias en la Proteostasis/genética , Edición de ARN , Alelos , Animales , Proteínas Bacterianas/genética , Ecocardiografía , Homeostasis , Humanos , Hidrólisis , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Modelos Moleculares , Mutación , Miocitos Cardíacos/ultraestructura , Parafina/química , Unión Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína
11.
J Neurosci ; 35(11): 4587-98, 2015 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-25788676

RESUMEN

The structural microtubule-associated proteins (MAPs) are critical for the organization of neuronal microtubules (MTs). Microtubule-associated protein 1A (MAP1A) is one of the most abundantly expressed MAPs in the mammalian brain. However, its in vivo function remains largely unknown. Here we describe a spontaneous mouse mutation, nm2719, which causes tremors, ataxia, and loss of cerebellar Purkinje neurons in aged homozygous mice. The nm2719 mutation disrupts the Map1a gene. We show that targeted deletion of mouse Map1a gene leads to similar neurodegenerative defects. Before neuron death, Map1a mutant Purkinje cells exhibited abnormal focal swellings of dendritic shafts and disruptions in axon initial segment (AIS) morphology. Furthermore, the MT network was reduced in the somatodendritic and AIS compartments, and both the heavy and light chains of MAP1B, another brain-enriched MAP, was aberrantly distributed in the soma and dendrites of mutant Purkinje cells. MAP1A has been reported to bind to the membrane-associated guanylate kinase (MAGUK) scaffolding proteins, as well as to MTs. Indeed, PSD-93, the MAGUK specifically enriched in Purkinje cells, was reduced in Map1a(-/-) Purkinje cells. These results demonstrate that MAP1A functions to maintain both the neuronal MT network and the level of PSD-93 in neurons of the mammalian brain.


Asunto(s)
Proteínas Asociadas a Microtúbulos/genética , Mutación/genética , Degeneración Nerviosa/genética , Degeneración Nerviosa/patología , Células de Purkinje/patología , Animales , Encéfalo/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Red Nerviosa/patología
12.
J Neurosci ; 35(7): 3001-9, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25698737

RESUMEN

Folding of transmembrane and secretory proteins occurs in the lumen of the endoplasmic reticulum (ER) before transportation to the cell surface and is monitored by the unfolded protein response (UPR) signaling pathway. The accumulation of unfolded proteins in the ER activates the UPR that restores ER homeostasis by regulating gene expression that leads to an increase in the protein-folding capacity of the ER and a decrease in the ER protein-folding load. However, prolonged UPR activity has been associated with cell death in multiple pathological conditions, including neurodegeneration. Here, we report a spontaneous recessive mouse mutation that causes progressive cerebellar granule cell death and peripheral motor axon degeneration. By positional cloning, we identify the mutation in this strain as a retrotransposon insertion in the Clcc1 gene, which encodes a putative chloride channel localized to the ER. Furthermore, we demonstrate that the C3H/HeSnJ inbred strain has late onset cerebellar degeneration due to this mutation. Interestingly, acute knockdown of Clcc1 expression in cultured cells increases sensitivity to ER stress. In agreement, GRP78, the major HSP70 family chaperone in the ER, is upregulated in Clcc1-deficient granule cells in vivo, and ubiquitinated proteins accumulate in these neurons before their degeneration. These data suggest that disruption of chloride homeostasis in the ER disrupts the protein-folding capacity of the ER, leading to eventual neuron death.


Asunto(s)
Canales de Cloruro/deficiencia , Estrés del Retículo Endoplásmico/genética , Enfermedades Neurodegenerativas , Pliegue de Proteína , Animales , Cerebelo/patología , Canales de Cloruro/genética , Modelos Animales de Enfermedad , Chaperón BiP del Retículo Endoplásmico , Células HEK293 , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mutación/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Neuronas/metabolismo , Neuronas/patología , Enfermedades del Sistema Nervioso Periférico/metabolismo , Enfermedades del Sistema Nervioso Periférico/patología , ARN Mensajero/metabolismo , Transfección
13.
J Neurosci ; 33(31): 12656-69, 2013 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-23904602

RESUMEN

Axon degeneration is a critical pathological feature of many neurodegenerative diseases. Misregulation of local axonal ion homeostasis has been recognized as an important yet understudied mechanism for axon degeneration. Here we report a chemically induced, recessive mouse mutation, vacillator (vac), which causes ataxia and concomitant axon degeneration of cerebellar Purkinje cells. By positional cloning, we identified vac as a point mutation in the calcineurin-like EF hand protein 1 (Chp1) gene that resulted in the production of mutant CHP1 isoforms with an amino acid substitution in a functional EF-hand domain or a truncation of this motif by aberrant splicing and significantly reduced protein levels. CHP1 has been previously shown to interact with the sodium hydrogen exchanger 1 (NHE1), a major regulator of intracellular pH. We demonstrated that CHP1 assists in the full glycosylation of NHE1 that is necessary for the membrane localization of this transporter and that truncated isoforms of CHP1 were defective in stimulating NHE1 biosynthetic maturation. Consistent with this, membrane localization of NHE1 at axon terminals was greatly reduced in Chp1-deficient Purkinje cells before axon degeneration. Furthermore, genetic ablation of Nhe1 also resulted in Purkinje cell axon degeneration, pinpointing the functional convergence of the two proteins. Our findings clearly demonstrate that the polarized presynaptic localization of NHE/CHP1 is an important feature of neuronal axons and that selective disruption of NHE1-mediated proton homeostasis in axons can lead to degeneration, suggesting that local regulation of pH is pivotal for axon survival.


Asunto(s)
Axones/fisiología , Proteínas de Unión al Calcio/genética , Proteínas de Transporte de Catión/biosíntesis , Homeostasis/genética , Degeneración Nerviosa/patología , Células de Purkinje/citología , Intercambiadores de Sodio-Hidrógeno/biosíntesis , Factores de Edad , Alquilantes/farmacología , Animales , Ataxia/genética , Ataxia/patología , Axones/efectos de los fármacos , Axones/patología , Axones/ultraestructura , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/metabolismo , Células Cultivadas , Cerebelo/citología , Cerebelo/patología , Cricetinae , Modelos Animales de Enfermedad , Etilnitrosourea/farmacología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Homeostasis/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Degeneración Nerviosa/genética , Proteínas del Tejido Nervioso/metabolismo , Mutación Puntual , Células de Purkinje/efectos de los fármacos , Intercambiador 1 de Sodio-Hidrógeno
14.
PLoS Genet ; 7(5): e1002063, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21625621

RESUMEN

Sphingolipids, lipids with a common sphingoid base (also termed long chain base) backbone, play essential cellular structural and signaling functions. Alterations of sphingolipid levels have been implicated in many diseases, including neurodegenerative disorders. However, it remains largely unclear whether sphingolipid changes in these diseases are pathological events or homeostatic responses. Furthermore, how changes in sphingolipid homeostasis shape the progression of aging and neurodegeneration remains to be clarified. We identified two mouse strains, flincher (fln) and toppler (to), with spontaneous recessive mutations that cause cerebellar ataxia and Purkinje cell degeneration. Positional cloning demonstrated that these mutations reside in the Lass1 gene. Lass1 encodes (dihydro)ceramide synthase 1 (CerS1), which is highly expressed in neurons. Both fln and to mutations caused complete loss of CerS1 catalytic activity, which resulted in a reduction in sphingolipid biosynthesis in the brain and dramatic changes in steady-state levels of sphingolipids and sphingoid bases. In addition to Purkinje cell death, deficiency of CerS1 function also induced accumulation of lipofuscin with ubiquitylated proteins in many brain regions. Our results demonstrate clearly that ceramide biosynthesis deficiency can cause neurodegeneration and suggest a novel mechanism of lipofuscin formation, a common phenomenon that occurs during normal aging and in some neurodegenerative diseases.


Asunto(s)
Ceramidas/biosíntesis , Lipofuscina/metabolismo , Células de Purkinje/metabolismo , Animales , Secuencia de Bases , Células COS , Diferenciación Celular , Ceramidas/deficiencia , Chlorocebus aethiops , Homeostasis , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Mutación , Células de Purkinje/citología , Esfingosina N-Aciltransferasa/genética , Esfingosina N-Aciltransferasa/metabolismo
15.
Nat Genet ; 37(9): 974-9, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16116427

RESUMEN

Endoplasmic reticulum (ER) chaperones and ER stress have been implicated in the pathogenesis of neurodegenerative disorders, such as Alzheimer and Parkinson diseases, but their contribution to neuron death remains uncertain. In this study, we establish a direct in vivo link between ER dysfunction and neurodegeneration. Mice homozygous with respect to the woozy (wz) mutation develop adult-onset ataxia with cerebellar Purkinje cell loss. Affected cells have intracellular protein accumulations reminiscent of protein inclusions in both the ER and the nucleus. In addition, upregulation of the unfolded protein response, suggestive of ER stress, occurs in mutant Purkinje cells. We report that the wz mutation disrupts the gene Sil1 that encodes an adenine nucleotide exchange factor of BiP, a crucial ER chaperone. These findings provide evidence that perturbation of ER chaperone function in terminally differentiated neurons leads to protein accumulation, ER stress and subsequent neurodegeneration.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/fisiología , Proteínas de Choque Térmico/fisiología , Chaperonas Moleculares/fisiología , Mutación , Degeneración Nerviosa , Animales , Ataxia/etiología , Autoantígenos/metabolismo , Núcleo Celular/metabolismo , Cerebelo/patología , Retículo Endoplásmico , Chaperón BiP del Retículo Endoplásmico , Femenino , Proteínas de Choque Térmico/genética , Homocigoto , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Chaperonas Moleculares/genética , Datos de Secuencia Molecular , Células de Purkinje/metabolismo , Células de Purkinje/patología
16.
Neuron ; 112(9): 1397-1415.e6, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38377989

RESUMEN

Defects in tRNA biogenesis are associated with multiple neurological disorders, yet our understanding of these diseases has been hampered by an inability to determine tRNA expression in individual cell types within a complex tissue. Here, we developed a mouse model in which RNA polymerase III is conditionally epitope tagged in a Cre-dependent manner, allowing us to accurately profile tRNA expression in any cell type in vivo. We investigated tRNA expression in diverse nervous system cell types, revealing dramatic heterogeneity in the expression of tRNA genes between populations. We found that while maintenance of levels of tRNA isoacceptor families is critical for cellular homeostasis, neurons are differentially vulnerable to insults to distinct tRNA isoacceptor families. Cell-type-specific translatome analysis suggests that the balance between tRNA availability and codon demand may underlie such differential resilience. Our work provides a platform for investigating the complexities of mRNA translation and tRNA biology in the brain.


Asunto(s)
Encéfalo , Homeostasis , Neuronas , ARN de Transferencia , Animales , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Homeostasis/fisiología , Ratones , Encéfalo/metabolismo , Neuronas/metabolismo , ARN Polimerasa III/metabolismo , ARN Polimerasa III/genética , Ratones Transgénicos
17.
Sci Adv ; 10(2): eadh3929, 2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38198538

RESUMEN

Transcription factors play vital roles in neuron development; however, little is known about the role of these proteins in maintaining neuronal homeostasis. Here, we show that the transcription factor RREB1 (Ras-responsive element-binding protein 1) is essential for neuron survival in the mammalian brain. A spontaneous mouse mutation causing loss of a nervous system-enriched Rreb1 transcript is associated with progressive loss of cerebellar Purkinje cells and ataxia. Analysis of chromatin immunoprecipitation and sequencing, along with RNA sequencing data revealed dysregulation of RREB1 targets associated with the microtubule cytoskeleton. In agreement with the known role of microtubules in dendritic development, dendritic complexity was disrupted in Rreb1-deficient neurons. Analysis of sequencing data also suggested that RREB1 plays a role in the endomembrane system. Mutant Purkinje cells had fewer numbers of autophagosomes and lysosomes and contained P62- and ubiquitin-positive inclusions. Together, these studies demonstrate that RREB1 functions to maintain the microtubule network and proteostasis in mammalian neurons.


Asunto(s)
Proteostasis , Factores de Transcripción , Animales , Ratones , Mamíferos , Microtúbulos , Neuronas , Células de Purkinje
18.
Sci Rep ; 14(1): 13603, 2024 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-38866944

RESUMEN

Notch signaling guides vascular development and function by regulating diverse endothelial cell behaviors, including migration, proliferation, vascular density, endothelial junctions, and polarization in response to flow. Notch proteins form transcriptional activation complexes that regulate endothelial gene expression, but few of the downstream effectors that enable these phenotypic changes have been characterized in endothelial cells, limiting our understanding of vascular Notch activities. Using an unbiased screen of translated mRNA rapidly regulated by Notch signaling, we identified novel in vivo targets of Notch signaling in neonatal mouse brain endothelium, including UNC5B, a member of the netrin family of angiogenic-regulatory receptors. Endothelial Notch signaling rapidly upregulates UNC5B in multiple endothelial cell types. Loss or gain of UNC5B recapitulated specific Notch-regulated phenotypes. UNC5B expression inhibited endothelial migration and proliferation and was required for stabilization of endothelial junctions in response to shear stress. Loss of UNC5B partially or wholly blocked the ability of Notch activation to regulate these endothelial cell behaviors. In the developing mouse retina, endothelial-specific loss of UNC5B led to excessive vascularization, including increased vascular outgrowth, density, and branchpoint count. These data indicate that Notch signaling upregulates UNC5B as an effector protein to control specific endothelial cell behaviors and inhibit angiogenic growth.


Asunto(s)
Movimiento Celular , Proliferación Celular , Células Endoteliales , Receptores de Netrina , Receptores Notch , Retina , Transducción de Señal , Animales , Receptores de Netrina/metabolismo , Receptores Notch/metabolismo , Ratones , Células Endoteliales/metabolismo , Retina/metabolismo , Humanos , Vasos Retinianos/metabolismo , Neovascularización Fisiológica
19.
Curr Opin Cell Biol ; 18(4): 444-52, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16781856

RESUMEN

Unfolded proteins and other conditions affecting endoplasmic reticulum (ER) homeostasis cause ER stress. The cell reacts to ER stress by activation of the unfolded protein response (UPR), which induces profound changes in cellular metabolism including general translation attenuation, transcriptional upregulation of molecular chaperone genes, and activation of ER-associated degradation. However, prolonged or acute ER stress results in cell death. Recent progress suggests that ER stress and UPR play key roles in the immune response, diabetes, tumor growth under hypoxic conditions, and in some neurodegenerative diseases. Further research on ER stress and UPR will greatly enhance the understanding of these physiological and pathological processes, and provide novel avenues to potential therapies.


Asunto(s)
Diabetes Mellitus/metabolismo , Retículo Endoplásmico/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Animales , Humanos , Modelos Inmunológicos , Pliegue de Proteína
20.
Nat Genet ; 31(3): 279-84, 2002 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12089526

RESUMEN

Mice homozygous for the cerebellar deficient folia (cdf) mutation are ataxic and have cerebellar hypoplasia and abnormal lobulation of the cerebellum. In the cerebella of cdf/cdf homozygous mice, approximately 40% of Purkinje cells are located ectopically in the white matter and inner granule-cell layer. Many hippocampal pyramidal cells are scattered in the plexiform layers, and those that are correctly positioned are less densely packed than are cells in wild-type mice. We show that fear conditioning and prepulse inhibition of the startle response are also disrupted in cdf/cdf mice. We identify a deletion on chromosome 6 that removes approximately 150 kb in the cdf critical region. The deletion includes part of Catna2, encoding alpha N-catenin, a protein that links the classical cadherins to the neuronal cytoskeleton. Expression of a Catna2 transgene in cdf/cdf mice restored normal cerebellar and hippocampal morphology, prepulse inhibition and fear conditioning. The findings suggest that catenin cadherin cell-adhesion complexes are important in cerebellar and hippocampal lamination and in the control of startle modulation.


Asunto(s)
Proteínas del Citoesqueleto/genética , Factores de Crecimiento Nervioso/genética , Proteínas del Tejido Nervioso/genética , Reflejo de Sobresalto/fisiología , Animales , Secuencia de Bases , Cadherinas/genética , Cerebelo/patología , Miedo , Eliminación de Gen , Marcadores Genéticos , Genotipo , Hipocampo/patología , Hipocampo/ultraestructura , Homocigoto , Ratones , Ratones Endogámicos C3H , Ratones Mutantes , Repeticiones de Microsatélite , Factores de Crecimiento Nervioso/fisiología , Células de Purkinje/patología , ARN Mensajero/análisis , Reflejo de Sobresalto/genética , Lugares Marcados de Secuencia , Transgenes , alfa Catenina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA