Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nucleic Acids Res ; 52(5): 2290-2305, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38113270

RESUMEN

Phase separation regulates fundamental processes in gene expression and is mediated by the local concentration of proteins and nucleic acids, as well as nucleic acid secondary structures such as G-quadruplexes (G4s). These structures play fundamental roles in both host gene expression and in viral replication due to their peculiar localisation in regulatory sequences. Hepatitis B virus (HBV) covalently closed circular DNA (cccDNA) is an episomal minichromosome whose persistence is at the basis of chronic infection. Identifying the mechanisms controlling its transcriptional activity is indispensable to develop new therapeutic strategies against chronic hepatitis B. The aim of this study was to determine whether G4s are formed in cccDNA and regulate viral replication. Combining biochemistry and functional studies, we demonstrate that cccDNA indeed contains ten G4s structures. Furthermore, mutations disrupting two G4s located in the enhancer I HBV regulatory region altered cccDNA transcription and viral replication. Finally, we showed for the first time that cccDNA undergoes phase separation in a G4-dependent manner to promote its transcription in infected hepatocytes. Altogether, our data give new insight in the transcriptional regulation of the HBV minichromosome that might pave the way for the identification of novel targets to destabilize or silence cccDNA.


Asunto(s)
G-Cuádruplex , Hepatitis B Crónica , Humanos , Virus de la Hepatitis B/genética , ADN Circular/genética , ADN Circular/metabolismo , Separación de Fases , ADN Viral/genética , ADN Viral/metabolismo , Hepatitis B Crónica/genética , Hepatitis B Crónica/metabolismo , Hepatocitos/metabolismo , Replicación Viral/genética
2.
Brain Behav Immun ; 114: 414-429, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37716378

RESUMEN

The purinoceptor P2X7R is a promising therapeutic target for tauopathies, including Alzheimer's disease (AD). Pharmacological inhibition or genetic knockdown of P2X7R ameliorates cognitive deficits and reduces pathological tau burden in mice that model aspects of tauopathy, including mice expressing mutant human frontotemporal dementia (FTD)-causing forms of tau. However, disagreements remain over which glial cell types express P2X7R and therefore the mechanism of action is unresolved. Here, we show that P2X7R protein levels increase in human AD post-mortem brain, in agreement with an upregulation of P2RX7 mRNA observed in transcriptome profiles from the AMP-AD consortium. P2X7R protein increases mirror advancing Braak stage and coincide with synapse loss. Using RNAScope we detect P2RX7 mRNA in microglia and astrocytes in human AD brain, including in the vicinity of senile plaques. In cultured microglia, P2X7R activation modulates the NLRP3 inflammasome pathway by promoting the formation of active complexes and release of IL-1ß. In astrocytes, P2X7R activates NFκB signalling and increases production of the cytokines CCL2, CXCL1 and IL-6 together with the acute phase protein Lcn2. To further explore the role of P2X7R in a disease-relevant context, we expressed wild-type or FTD-causing mutant forms of tau in mouse organotypic brain slice cultures. Inhibition of P2X7R reduces insoluble tau levels without altering soluble tau phosphorylation or synaptic localisation, suggesting a non-cell autonomous role of glial P2X7R on pathological tau aggregation. These findings support further investigations into the cell-type specific effects of P2X7R-targeting therapies in tauopathies.


Asunto(s)
Enfermedad de Alzheimer , Demencia Frontotemporal , Tauopatías , Animales , Humanos , Ratones , Enfermedad de Alzheimer/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Demencia Frontotemporal/genética , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/patología , Microglía/metabolismo , ARN Mensajero/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Tauopatías/metabolismo
3.
Nat Commun ; 14(1): 5366, 2023 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-37666821

RESUMEN

Pharmacologic depletion of RNA-binding motif 39 (RBM39) using aryl sulfonamides represents a promising anti-cancer therapy but requires high levels of the adaptor protein DCAF15. Consequently, novel approaches to deplete RBM39 in an DCAF15-independent manner are required. Here, we uncover that RBM39 autoregulates via the inclusion of a poison exon into its own pre-mRNA and identify the cis-acting elements that govern this regulation. We also determine the NMR solution structures of RBM39's tandem RNA recognition motifs (RRM1 and RRM2) bound to their respective RNA targets, revealing how RRM1 recognises RNA stem loops whereas RRM2 binds specifically to single-stranded N(G/U)NUUUG. Our results support a model where RRM2 selects the 3'-splice site of a poison exon and the RRM3 and RS domain stabilise the U2 snRNP at the branchpoint. Our work provides molecular insights into RBM39-dependent 3'-splice site selection and constitutes a solid basis to design alternative anti-cancer therapies.


Asunto(s)
Neoplasias , Empalme del ARN , Empalme del ARN/genética , Motivos de Unión al ARN , Sitios de Empalme de ARN , Homeostasis , Factores de Empalme de ARN/genética , Neoplasias/genética
4.
Methods Mol Biol ; 2537: 1-19, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35895255

RESUMEN

Alternative pre-mRNA splicing allows for the production of multiple mRNAs from an individual gene, which not only expands the protein-coding potential of the genome but also enables complex mechanisms for the post-transcriptional control of gene expression. Regulation of alternative splicing entails a combinatorial interplay between an abundance of trans-acting splicing factors, cis-acting regulatory sequence elements and their concerted effects on the core splicing machinery. Given the extent and biological significance of alternative splicing in humans, it is not surprising that aberrant splicing patterns can cause or contribute to a wide range of diseases. In this introductory chapter, we outline the mechanisms that govern alternative pre-mRNA splicing and its regulation and discuss how dysregulated splicing contributes to human diseases affecting the motor system and the brain.


Asunto(s)
Empalme Alternativo , Precursores del ARN , Enfermedad/genética , Humanos , Precursores del ARN/genética , Precursores del ARN/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Transactivadores/metabolismo
5.
Nucleic Acids Res ; 49(13): 7713-7731, 2021 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-34233002

RESUMEN

Liquid-liquid phase separation (LLPS) of proteins and RNAs has emerged as the driving force underlying the formation of membrane-less organelles. Such biomolecular condensates have various biological functions and have been linked to disease. The protein Fused in Sarcoma (FUS) undergoes LLPS and mutations in FUS have been causally linked to the motor neuron disease Amyotrophic Lateral Sclerosis (ALS-FUS). LLPS followed by aggregation of cytoplasmic FUS has been proposed to be a crucial disease mechanism. However, it is currently unclear how LLPS impacts the behaviour of FUS in cells, e.g. its interactome. Hence, we developed a method allowing for the purification of LLPS FUS-containing droplets from cell lysates. We observe substantial alterations in the interactome, depending on its biophysical state. While non-LLPS FUS interacts mainly with factors involved in pre-mRNA processing, LLPS FUS predominantly binds to proteins involved in chromatin remodelling and DNA damage repair. Interestingly, also mitochondrial factors are strongly enriched with LLPS FUS, providing a potential explanation for the observed changes in mitochondrial gene expression in mouse models of ALS-FUS. In summary, we present a methodology to investigate the interactomes of phase separating proteins and provide evidence that LLPS shapes the FUS interactome with implications for function and disease.


Asunto(s)
Proteína FUS de Unión a ARN/metabolismo , Núcleo Celular/metabolismo , Cromatina/metabolismo , Citoplasma/metabolismo , Gránulos Citoplasmáticos/metabolismo , Células HEK293 , Células HeLa , Humanos , Mapeo de Interacción de Proteínas , ARN Mensajero/metabolismo , ARN Nuclear Pequeño/metabolismo , Proteína FUS de Unión a ARN/química , Proteína FUS de Unión a ARN/aislamiento & purificación
6.
Nat Commun ; 11(1): 6341, 2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33311468

RESUMEN

Mutations in the RNA-binding protein Fused in Sarcoma (FUS) cause early-onset amyotrophic lateral sclerosis (ALS). However, a detailed understanding of central RNA targets of FUS and their implications for disease remain elusive. Here, we use a unique blend of crosslinking and immunoprecipitation (CLIP) and NMR spectroscopy to identify and characterise physiological and pathological RNA targets of FUS. We find that U1 snRNA is the primary RNA target of FUS via its interaction with stem-loop 3 and provide atomic details of this RNA-mediated mode of interaction with the U1 snRNP. Furthermore, we show that ALS-associated FUS aberrantly contacts U1 snRNA at the Sm site with its zinc finger and traps snRNP biogenesis intermediates in human and murine motor neurons. Altogether, we present molecular insights into a FUS toxic gain-of-function involving direct and aberrant RNA-binding and strengthen the link between two motor neuron diseases, ALS and spinal muscular atrophy (SMA).


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , ARN Nuclear Pequeño/metabolismo , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Línea Celular , Predisposición Genética a la Enfermedad/genética , Humanos , Ratones , Ratones Noqueados , Modelos Moleculares , Neuronas Motoras/metabolismo , Mutación , Dominios y Motivos de Interacción de Proteínas , ARN Nuclear Pequeño/química , Proteína FUS de Unión a ARN/química , Ribonucleoproteína Nuclear Pequeña U1/química
7.
Cell Stress ; 2(3): 40-54, 2018 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-31225466

RESUMEN

Pre-mRNA splicing is an essential step in eukaryotic gene expression. Mutations in cis-acting sequence elements within pre-mRNA molecules or trans-acting factors involved in pre-mRNA processing have both been linked to splicing dysfunction that give rise to a large number of human diseases. These mutations typically affect the major splicing pathway, which excises more than 99% of all introns in humans. However, approximately 700-800 human introns feature divergent intron consensus sequences at their 5' and 3' ends and are recognized by a separate pre-mRNA processing machinery denoted as the minor spliceosome. This spliceosome has been studied less than its major counterpart, but has received increasing attention during the last few years as a novel pathomechanistic player on the stage in neurodevelopmental and neurodegenerative diseases. Here, we review the current knowledge on minor spliceosome function and discuss its potential pathomechanistic role and impact in neurodegeneration.

8.
Mol Biol Cell ; 29(2): 75-83, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29167381

RESUMEN

CRISPR/Cas9-based genome editing offers the possibility to knock out almost any gene of interest in an affordable and simple manner. The most common strategy is the introduction of a frameshift into the open reading frame (ORF) of the target gene which truncates the coding sequence (CDS) and targets the corresponding transcript for degradation by nonsense-mediated mRNA decay (NMD). However, we show that transcripts containing premature termination codons (PTCs) are not always degraded efficiently and can generate C-terminally truncated proteins which might have residual or dominant negative functions. Therefore, we recommend an alternative approach for knocking out genes, which combines CRISPR/Cas9 with gene traps (CRISPR-Trap) and is applicable to ∼50% of all spliced human protein-coding genes and a large subset of lncRNAs. CRISPR-Trap completely prevents the expression of the ORF and avoids expression of C-terminal truncated proteins. We demonstrate the feasibility of CRISPR-Trap by utilizing it to knock out several genes in different human cell lines. Finally, we also show that this approach can be used to efficiently generate gene replacements allowing for modulation of protein levels for otherwise lethal knockouts (KOs). Thus, CRISPR-Trap offers several advantages over conventional KO approaches and allows for generation of clean CRISPR/Cas9-based KOs.


Asunto(s)
Sistemas CRISPR-Cas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Técnicas de Inactivación de Genes/métodos , Células HEK293 , Células HeLa , Humanos
9.
EMBO J ; 35(14): 1504-21, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27252488

RESUMEN

Fused in sarcoma (FUS) is a ubiquitously expressed RNA-binding protein proposed to function in various RNA metabolic pathways, including transcription regulation, pre-mRNA splicing, RNA transport and microRNA processing. Mutations in the FUS gene were identified in patients with amyotrophic lateral sclerosis (ALS), but the pathomechanisms by which these mutations cause ALS are not known. Here, we show that FUS interacts with the minor spliceosome constituent U11 snRNP, binds preferentially to minor introns and directly regulates their removal. Furthermore, a FUS knockout in neuroblastoma cells strongly disturbs the splicing of minor intron-containing mRNAs, among them mRNAs required for action potential transmission and for functional spinal motor units. Moreover, an ALS-associated FUS mutant that forms cytoplasmic aggregates inhibits splicing of minor introns by trapping U11 and U12 snRNAs in these aggregates. Collectively, our findings suggest a possible pathomechanism for ALS in which mutated FUS inhibits correct splicing of minor introns in mRNAs encoding proteins required for motor neuron survival.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Intrones , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Empalme del ARN , Proteína FUS de Unión a ARN/genética , Proteína FUS de Unión a ARN/metabolismo , Línea Celular , Humanos
10.
Acta Neuropathol ; 131(4): 587-604, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26895297

RESUMEN

Deposition of the nuclear DNA/RNA-binding protein Fused in sarcoma (FUS) in cytosolic inclusions is a common hallmark of some cases of frontotemporal lobar degeneration (FTLD-FUS) and amyotrophic lateral sclerosis (ALS-FUS). Whether both diseases also share common pathological mechanisms is currently unclear. Based on our previous finding that FUS deposits are hypomethylated in FTLD-FUS but not in ALS-FUS, we have now investigated whether genetic or pharmacological inactivation of Protein arginine methyltransferase 1 (PRMT1) activity results in unmethylated FUS or in alternatively methylated forms of FUS. To do so, we generated FUS-specific monoclonal antibodies that specifically recognize unmethylated arginine (UMA), monomethylated arginine (MMA) or asymmetrically dimethylated arginine (ADMA). Loss of PRMT1 indeed not only results in an increase of UMA FUS and a decrease of ADMA FUS, but also in a significant increase of MMA FUS. Compared to ADMA FUS, UMA and MMA FUS exhibit much higher binding affinities to Transportin-1, the nuclear import receptor of FUS, as measured by pull-down assays and isothermal titration calorimetry. Moreover, we show that MMA FUS occurs exclusively in FTLD-FUS, but not in ALS-FUS. Our findings therefore provide additional evidence that FTLD-FUS and ALS-FUS are caused by distinct disease mechanisms although both share FUS deposits as a common denominator.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Degeneración Lobar Frontotemporal/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína FUS de Unión a ARN/metabolismo , beta Carioferinas/metabolismo , Esclerosis Amiotrófica Lateral/genética , Animales , Anticuerpos/farmacología , Arginina/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Embrión de Mamíferos , Células Madre Embrionarias , Inhibidores Enzimáticos/farmacología , Femenino , Degeneración Lobar Frontotemporal/genética , Humanos , Cuerpos de Inclusión/efectos de los fármacos , Cuerpos de Inclusión/metabolismo , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Proteína-Arginina N-Metiltransferasas/genética , Proteína FUS de Unión a ARN/inmunología , Ratas , beta Carioferinas/inmunología
11.
J Air Waste Manag Assoc ; 57(8): 907-18, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17824281

RESUMEN

In many locations in Eastern Canada, ambient levels of fine particulate matter (PM,25) and surface ozone (O3) depend on airflow direction and synoptic scale meteorological conditions. In this study, a cluster analysis was performed on 10 yr (1994-2003) of back-trajectory data for 11 locations in Eastern Canada, resulting in the identification of 10 unique back-trajectory clusters (or airflows) for each location. The airflows were then used to characterize and identify spatial and temporal trends in the daily maximum 8-hr average O3 (dmax 8-hr O3) and the daily average PM2.5 levels. Results showed that airflows from the southwest passing over Michigan and Southern Ontario were associated, on average, with the highest O3 levels at most locations in Eastern Canada. For PM2.5, the highest levels occurred with airflows from the Eastern Ohio River Valley. At major urban locations in Ontario and Quebec, the warm season mean (May to September) dmax 8-hr O3 and the annual mean PM2.5 were, on average, 12 parts per billion and 7.6 microg/m3 higher, respectively, than airflows from the north. Elevated levels of O3 and PM2.5 also occurred under light airflows, and, on average, the levels under light airflows were higher than their nonlight counterparts. At several locations in Canada, including Toronto, Montreal, Quebec City, and Kejimkujik, the annual warm season mean dmax 8-hr O3 experienced a statistically significant (95% confidence) increasing trend over the 10-yr period. When airflow direction was considered, a number of locations experienced statistically significant upward trends in O3 for airflow from the north and northwest. Several locations also showed significant upward trends associated with airflow from the southwest passing over Michigan and Southwestern Ontario. Although there are no statistically significant downward trends, airflows from the southwest have shown a reduction in O3 levels in Southwestern Ontario in more recent years.


Asunto(s)
Ozono/análisis , Material Particulado/análisis , Movimientos del Aire , Canadá , Análisis por Conglomerados , Geografía , Estaciones del Año , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...