Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Neurobiol Dis ; 156: 105421, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34118419

RESUMEN

Neurodegenerative disorders like frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are pathologically characterized by toxic protein deposition in the cytoplasm or nucleus of affected neurons and glial cells. Many of these aggregated proteins belong to the class of RNA binding proteins (RBP), and, when mutated, account for a significant subset of familial ALS and FTD cases. Here, we present first genetic evidence for the RBP gene RBM45 in the FTD-ALS spectrum. RBM45 shows many parallels with other FTD-ALS associated genes and proteins. Multiple lines of evidence have demonstrated that RBM45 is an RBP that, upon mutation, redistributes to the cytoplasm where it co-aggregates with other RBPs into cytoplasmic stress granules (SG), evolving to persistent toxic TDP-43 immunoreactive inclusions. Exome sequencing in two affected first cousins of a heavily affected early-onset dementia family listed a number of candidate genes. The gene with the highest pathogenicity score was the RBP gene RBM45. In the family, the RBM45 Arg183* nonsense mutation co-segregated in both affected cousins. Validation in an unrelated patient (n = 548) / control (n = 734) cohort identified an additional RBM45 Arg183* carrier with bvFTD on a shared 4 Mb haplotype. Transcript and protein expression analysis demonstrated loss of nuclear RBM45, suggestive of a loss-of-function disease mechanism. Further, two more ultra-rare VUS, one in the nuclear localization signal (NLS, p.Lys456Arg) in an ALS patient and one in the intrinsically disordered homo-oligomer assembly (HOA) domain (p.Arg314Gln) in a patient with nfvPPA were detected. Our findings suggest that the pathomechanisms linking RBM45 with FTD and ALS may be related to its loss of nuclear function as a mediator of mRNA splicing, cytoplasmic retention or its inability to form homo-oligomers, leading to aggregate formation with trapping of other RBPs including TDP-43, which may accumulate into persisted TDP-43 inclusions.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Secuenciación del Exoma/métodos , Demencia Frontotemporal/genética , Estudios de Asociación Genética/métodos , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/genética , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Esclerosis Amiotrófica Lateral/diagnóstico , Esclerosis Amiotrófica Lateral/epidemiología , Bélgica/epidemiología , Estudios de Cohortes , Femenino , Demencia Frontotemporal/diagnóstico , Demencia Frontotemporal/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Linaje
2.
Cancer Lett ; 596: 216988, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38797234

RESUMEN

Type I interferons exhibit anti-proliferative and anti-cancer activities, but their detailed regulatory mechanisms in cancer have not been fully elucidated yet. RNA binding proteins are master orchestrators of gene regulation, which are closely related to tumor progression. Here we show that the upregulated RNA binding protein RBM45 correlates with poor prognosis in breast cancer. Depletion of RBM45 suppresses breast cancer progression both in cultured cells and xenograft mouse models. Mechanistically, RBM45 ablation inhibits breast cancer progression through regulating type I interferon signaling, particularly by elevating IFN-ß production. Importantly, RBM45 recruits TRIM28 to IRF7 and stimulates its SUMOylation, thereby repressing IFNB1 transcription. Loss of RBM45 reduced the SUMOylation of IRF7 by reducing the interaction between TRIM28 and IRF7 to promote IFNB1 transcription, leading to the inhibition of breast cancer progression. Taken together, our finding uncovers a vital role of RBM45 in modulating type I interferon signaling and cancer aggressive progression, implicating RBM45 as a potential therapeutic target in breast cancer.


Asunto(s)
Neoplasias de la Mama , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Factor 7 Regulador del Interferón , Proteínas de Unión al ARN , Sumoilación , Humanos , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Femenino , Animales , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/metabolismo , Ratones , Transcripción Genética , Línea Celular Tumoral , Interferón beta/metabolismo , Interferón beta/genética , Transducción de Señal , Ratones Desnudos , Proliferación Celular , Proteína 28 que Contiene Motivos Tripartito/metabolismo , Proteína 28 que Contiene Motivos Tripartito/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Endogámicos BALB C
3.
J Mol Graph Model ; 118: 108377, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36435031

RESUMEN

Neurodegenerative diseases (NDD) are a group of cognitive and behavioral disorders characterized by progressive loss of neuronal structure and function. As the population ages, the incidence is getting higher and higher, but there is currently no effective treatment. The details of RNA/DNA recognition by the RNA-binding protein RBM45 closely related to neurodegenerative diseases through its two tandem RNA-recognition domains at its N-terminus have important implications for structure-based drug discovery against degenerative diseases. To explore the key characteristics of interaction between ssDNA and RBM45, we performed multiple molecular dynamics (MD) simulations along with MM-PBSA energy prediction on the complexes of wild type (WT) and three mutant RBM45s (K100A, F124A/Y165A, and F29A/F70A/F124A/Y165A) with ssDNA, respectively. The findings suggest that these mutated residues of RBM45 modify the interaction of their surrounding residues with ssDNA, thereby affecting RBM45 protein binding to ssDNA. In contrast with WT RBM45 protein, variations in van der Waals and electrostatic interactions with ssDNA caused by these three RBM45 mutants are critical to affect binding between them. In addition, energy analysis showed that RBM45 is a specific ssDNA-binding protein. The results of our work provide valuable theoretical guidance for the design effective drugs of NDD.


Asunto(s)
Simulación de Dinámica Molecular , Enfermedades Neurodegenerativas , Humanos , Enfermedades Neurodegenerativas/genética , ADN de Cadena Simple/genética , Electricidad Estática , ARN , Proteínas del Tejido Nervioso , Proteínas de Unión al ARN/genética
4.
Cell Rep ; 40(9): 111293, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-36044854

RESUMEN

N6-methyladenosine (m6A) is deposited co-transcriptionally on thousands of cellular mRNAs and plays important roles in mRNA processing and cellular function. m6A is particularly abundant within the brain and is critical for neurodevelopment. However, the mechanisms through which m6A contributes to brain development are incompletely understood. RBM45 acts as an m6A-binding protein that is highly expressed during neurodevelopment. We find that RBM45 binds to thousands of cellular RNAs, predominantly within intronic regions. Rbm45 depletion disrupts the constitutive splicing of a subset of target pre-mRNAs, leading to altered mRNA and protein levels through both m6A-dependent and m6A-independent mechanisms. Finally, we find that RBM45 is necessary for neuroblastoma cell differentiation and that its depletion impacts the expression of genes involved in several neurodevelopmental signaling pathways. Altogether, our findings show a role for RBM45 in controlling mRNA processing and neuronal differentiation, mediated in part by the recognition of methylated RNA.


Asunto(s)
Proteínas Portadoras , Proteínas de Unión al ARN , Proteínas Portadoras/metabolismo , Unión Proteica , ARN/metabolismo , Empalme del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
5.
mBio ; 11(2)2020 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-32156816

RESUMEN

During infection of human parvovirus B19 (B19V), one viral precursor mRNA (pre-mRNA) is transcribed by a single promoter and is alternatively spliced and alternatively polyadenylated. Here, we identified a novel cis-acting sequence (5'-GUA AAG CUA CGG GAC GGU-3'), intronic splicing enhancer 3 (ISE3), which lies 72 nucleotides upstream of the second splice acceptor (A2-2) site of the second intron that defines the exon of the mRNA encoding the 11-kDa viral nonstructural protein. RNA binding motif protein 45 (RBM45) specifically binds to ISE3 with high affinity (equilibrium dissociation constant [KD ] = 33 nM) mediated by its RNA recognition domain and 2-homo-oligomer assembly domain (RRM2-HOA). Knockdown of RBM45 expression or ectopic overexpression of RRM2-HOA in human erythroid progenitor cells (EPCs) expanded ex vivo significantly decreased the level of viral mRNA spliced at the A2-2 acceptor but not that of the mRNA spliced at A2-1 that encodes VP2. Moreover, silent mutations of ISE3 in an infectious DNA of B19V significantly reduced 11-kDa expression. Notably, RBM45 also specifically interacts in vitro with ISE2, which shares the octanucleotide (GGGACGGU) with ISE3. Taken together, our results suggest that RBM45, through binding to both ISE2 and ISE3, is an essential host factor for maturation of 11-kDa-encoding mRNA.IMPORTANCE Human parvovirus B19 (B19V) is a human pathogen that causes severe hematological disorders in immunocompromised individuals. B19V infection has a remarkable tropism with respect to human erythroid progenitor cells (EPCs) in human bone marrow and fetal liver. During B19V infection, only one viral precursor mRNA (pre-mRNA) is transcribed by a single promoter of the viral genome and is alternatively spliced and alternatively polyadenylated, a process which plays a key role in expression of viral proteins. Our studies revealed that a cellular RNA binding protein, RBM45, binds to two intron splicing enhancers and is essential for the maturation of the small nonstructural protein 11-kDa-encoding mRNA. The 11-kDa protein plays an important role not only in B19V infection-induced apoptosis but also in viral DNA replication. Thus, the identification of the RBM45 protein and its cognate binding site in B19V pre-mRNA provides a novel target for antiviral development to combat B19V infection-caused severe hematological disorders.


Asunto(s)
Regulación Viral de la Expresión Génica , Intrones , Proteínas del Tejido Nervioso/metabolismo , Parvovirus B19 Humano/genética , Empalme del ARN , Proteínas de Unión al ARN/metabolismo , Proteínas no Estructurales Virales/genética , Células Cultivadas , Células Precursoras Eritroides/virología , Genoma Viral , Células Madre Hematopoyéticas , Humanos , Proteínas del Tejido Nervioso/genética , Parvovirus B19 Humano/metabolismo , Unión Proteica , Precursores del ARN/genética , Precursores del ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas no Estructurales Virales/metabolismo
6.
Acta Neuropathol Commun ; 8(1): 91, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32586379

RESUMEN

The RNA binding protein (RBP) RBM45 forms nuclear and cytoplasmic inclusions in neurons and glia in amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration with TDP-43 proteinopathy (FTLD-TDP), and Alzheimer's disease (AD). The normal functions of RBM45 are poorly understood, as are the mechanisms by which it forms inclusions in disease. To better understand the normal and pathological functions of RBM45, we evaluated whether the protein functions via association with several membraneless organelles and whether such an association could promote the formation of nuclear RBM45 inclusions. Under basal conditions, RBM45 is diffusely distributed throughout the nucleus and does not localize to membraneless organelles, including nuclear speckles, Cajal bodies, or nuclear gems. During cellular stress, however, nuclear RBM45 undergoes a reversible, RNA-binding dependent incorporation into nuclear stress bodies (NSBs). Chronic stress leads to the persistent association of RBM45 with NSBs and the irreversible accumulation of nuclear RBM45 inclusions. We also quantified the cell type- and disease-specific patterns of RBM45 pathology in ALS, FTLD-TDP, and AD. RBM45 nuclear and cytoplasmic inclusions are found in both neurons and glia in ALS, FTLD-TDP, and AD but are absent in non-neurologic disease controls. Across neurodegenerative diseases, RBM45 nuclear inclusion pathology occurs more frequently than cytoplasmic RBM45 inclusion pathology and exhibits cell type-specific variation. Collectively, our results define new stress-associated functions of RBM45, a mechanism for nuclear RBM45 inclusion formation, a role for NSBs in the pathogenesis of ALS, FTLD-TDP, and AD, and further underscore the importance of protein self-association to both the normal and pathological functions of RBPs in these diseases.


Asunto(s)
Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/patología , Neuroglía/patología , Neuronas/patología , Proteínas de Unión al ARN/metabolismo , Anciano , Anciano de 80 o más Años , Femenino , Células HEK293 , Humanos , Cuerpos de Inclusión Intranucleares/metabolismo , Cuerpos de Inclusión Intranucleares/patología , Masculino , Persona de Mediana Edad , Enfermedades Neurodegenerativas/metabolismo , Estrés Fisiológico/fisiología
7.
Brain Res ; 1647: 79-93, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-26979993

RESUMEN

The pathological accumulation of RNA-binding proteins (RBPs) within inclusion bodies is a hallmark of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). RBP aggregation results in both toxic gain and loss of normal function. Determining the protein binding partners and normal functions of disease-associated RBPs is necessary to fully understand molecular mechanisms of RBPs in disease. Herein, we characterized the protein-protein interactions (PPIs) of RBM45, a RBP that localizes to inclusions in ALS/FTLD. Using immunoprecipitation coupled to mass spectrometry (IP-MS), we identified 132 proteins that specifically interact with RBM45 within HEK293 cells. Select PPIs were validated by immunoblot and immunocytochemistry, demonstrating that RBM45 associates with a number of other RBPs primarily via RNA-dependent interactions in the nucleus. Analysis of the biological processes and pathways associated with RBM45-interacting proteins indicates enrichment for nuclear RNA processing/splicing via association with hnRNP proteins and cytoplasmic RNA translation via eiF2 and eiF4 pathways. Moreover, several other ALS-linked RBPs, including TDP-43, FUS, Matrin-3, and hnRNP-A1, interact with RBM45, consistent with prior observations of these proteins within intracellular inclusions in ALS/FTLD. Taken together, our results define a PPI network for RBM45, suggest novel functions for this protein, and provide new insights into the contributions of RBM45 to neurodegeneration in ALS/FTLD. This article is part of a Special Issue entitled SI:RNA Metabolism in Disease.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Degeneración Lobar Frontotemporal/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Unión al ARN/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Células HEK293 , Humanos , Inmunoprecipitación , Cuerpos de Inclusión/metabolismo , Espectrometría de Masas , Factores de Iniciación de Péptidos/metabolismo , Mapas de Interacción de Proteínas , Empalme del ARN , Transducción de Señal
8.
Genes Brain Behav ; 15(2): 260-70, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26482417

RESUMEN

Genetic association mapping in structured populations of model organisms can offer a fruitful complement to human genetic studies by generating new biological hypotheses about complex traits. Here we investigated prepulse inhibition (PPI), a measure of sensorimotor gating that is disrupted in a number of psychiatric disorders. To identify genes that influence PPI, we constructed a panel of half-sibs by crossing 30 females from common inbred mouse strains with inbred C57BL/6J males to create male and female F1 offspring. We used publicly available single nucleotide polymorphism (SNP) genotype data from these inbred strains to perform a genome-wide association scan using a dense panel of over 150,000 SNPs in a combined sample of 604 mice representing 30 distinct F1 genotypes. We identified two independent PPI-associated loci on Chromosomes 2 and 7, each of which explained 12-14% of the variance in PPI. Searches of available databases did not identify any plausible causative coding polymorphisms within these loci. However, previously collected expression quantitative trait locus (eQTL) data from hippocampus and striatum indicated that the SNPs on Chromosomes 2 and 7 that showed the strongest association with PPI were also strongly associated with expression of several transcripts, some of which have been implicated in human psychiatric disorders. This integrative approach successfully identified a focused set of genes which can be prioritized for follow-up studies. More broadly, our results show that F1 crosses among common inbred strains can be used in combination with other informatics and expression datasets to identify candidate genes for complex behavioral traits.


Asunto(s)
Mapeo Cromosómico , Polimorfismo de Nucleótido Simple/genética , Inhibición Prepulso/fisiología , Sitios de Carácter Cuantitativo/genética , Animales , Mapeo Cromosómico/métodos , Femenino , Estudio de Asociación del Genoma Completo/métodos , Genotipo , Masculino , Ratones Endogámicos C57BL , Fenotipo , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA