Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Brain Res Bull ; 216: 111049, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39142444

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder distinguished by gradual depletion of motor neurons. RNA binding motif protein 5 (RBM5), an abundantly expressed RNA-binding protein, plays a critical role in the process of cellular death. However, little is known about the role of RBM5 in the pathogenesis of ALS. Here, we found that RBM5 was upregulated in ALS hSOD1G93A-NSC34 cell models and hSOD1G93A mice due to a reduction of miR-141-5p. The upregulation of RBM5 increased the apoptosis of motor neurons by inhibiting Rac1-mediated neuroprotection. In contrast, genetic knockdown of RBM5 rescued motor neurons from hSOD1G93A-induced degeneration by activating Rac1 signaling. The neuroprotective effect of RBM5-knockdown was significantly inhibited by the Rac1 inhibitor, NSC23766. These findings suggest that RBM5 could potentially serve as a therapeutic target in ALS by activating the Rac1 signalling.


Asunto(s)
Esclerosis Amiotrófica Lateral , Apoptosis , Neuronas Motoras , Proteínas Proto-Oncogénicas c-akt , Proteínas de Unión al ARN , Transducción de Señal , Proteína de Unión al GTP rac1 , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Proteína de Unión al GTP rac1/metabolismo , Proteína de Unión al GTP rac1/genética , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Apoptosis/fisiología , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Ratones , Humanos , Transducción de Señal/fisiología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratones Transgénicos , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa/genética , Masculino , Proteínas de Unión al ADN , Proteínas de Ciclo Celular , Proteínas Supresoras de Tumor
2.
Mol Cell ; 84(8): 1496-1511.e7, 2024 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-38537639

RESUMEN

Understanding the mechanisms of pre-mRNA splicing is limited by the technical challenges to examining spliceosomes in vivo. Here, we report the isolation of RNP complexes derived from precatalytic A or B-like spliceosomes solubilized from the chromatin pellet of mammalian cell nuclei. We found that these complexes contain U2 snRNP proteins and a portion of the U2 snRNA bound with protected RNA fragments that precisely map to intronic branch sites across the transcriptome. These U2 complexes also contained the splicing regulators RBM5 and RBM10. We found RBM5 and RBM10 bound to nearly all branch site complexes and not simply those at regulated exons. The deletion of a conserved RBM5/RBM10 peptide sequence, including a zinc finger motif, disrupted U2 interaction and rendered the proteins inactive for the repression of many alternative exons. We propose a model where RBM5 and RBM10 regulate splicing as components of the U2 snRNP complex following branch site base pairing.


Asunto(s)
Ribonucleoproteína Nuclear Pequeña U2 , Empalmosomas , Animales , Empalmosomas/genética , Empalmosomas/metabolismo , Ribonucleoproteína Nuclear Pequeña U2/genética , Ribonucleoproteína Nuclear Pequeña U2/metabolismo , Intrones/genética , Cromatina/genética , Cromatina/metabolismo , Empalme del ARN , Precursores del ARN/metabolismo , Mamíferos/metabolismo
3.
Biochem Biophys Res Commun ; 704: 149703, 2024 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-38402723

RESUMEN

PEI is a cationic polymer, serving as a non-viral transfection carrier grounded in nanotechnology that enhances transfection efficiency via the proton sponge effect. RBM5 is an RNA-binding protein that can inhibit tumor development. This study involved the transfection of RBM5 in prostate cancer cells with PEI, Lipo2000, and their combination. Transwell and wound healing assays were used to observe invasion and migration of prostate cancer cells and flow cytometry was used to observe the apoptosis. Detect the expression of invasion and migration-related protein MMP9 through western blotting experiment. An activity detection kit was used to detect the activity of apoptotic protein caspase-3. We found that there was no significant difference in transfection efficiency when PEI and Lipo2000 were used alone but it significantly improved when they are combined. RBM5 reduced invasion, migration, and proliferation of prostate cancer and enhanced apoptosis. MMP9 expression was reduced, and the activity of caspase-3 was increased. PEI transfection could improve the inhibition of RBM5 on tumors more than Lipo2000. The inhibitory effect is more obvious when the two are used together. RBM5 transfected with PEI can amplify its inhibitory effect on prostate cancer, and this effect is more evident when combined with Lipo2000.


Asunto(s)
Proteínas de Unión al ADN , Neoplasias de la Próstata , Proteínas de Unión al ARN , Transfección , Humanos , Masculino , Apoptosis , Caspasa 3/genética , Caspasa 3/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Proteínas de Unión al ADN/farmacología , Proteínas de Unión al ADN/uso terapéutico , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Neoplasias de la Próstata/terapia , Proteínas de Unión al ARN/farmacología , Proteínas de Unión al ARN/uso terapéutico , Transfección/métodos , Proteínas Supresoras de Tumor/metabolismo
4.
Exp Neurol ; 374: 114690, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38218585

RESUMEN

RNA binding motif 5 (RBM5) is a tumor suppressor in cancer but its role in the brain is unclear. We used conditional gene knockout (KO) mice to test if RBM5 inhibition in the brain affects chronic cortical brain tissue survival or function after a controlled cortical impact (CCI) traumatic brain injury (TBI). RBM5 KO decreased baseline contralateral hemispheric volume (p < 0.0001) and exacerbated ipsilateral tissue loss at 21 d after CCI in male mice vs. wild type (WT) (p = 0.0019). CCI injury, but not RBM5 KO, impaired beam balance performance (0-5d post-injury) and swim speed on the Morris Water Maze (MWM) (19-20d) (p < 0.0001). RBM5 KO was associated with mild learning impairment in female mice (p = 0.0426), reflected as a modest increase in escape latency early in training (14-18d post-injury). However, KO did not affect spatial memory at 19d post-injury in male or in female mice but it was impaired by CCI in females (p = 0.0061). RBM5 KO was associated with impaired visual function in male mice on the visible platform test at 20d post-injury (p = 0.0256). To explore signaling disturbances in KOs related to behavior, we first cross-referenced known brain-specific RBM5-regulated gene targets with genes in the curated RetNet database that impact vision. We then performed a secondary literature search on RBM5-regulated genes with a putative role in hippocampal function. Regulating synaptic membrane exocytosis 2 (RIMS) 2 was identified as a gene of interest because it regulates both vision and hippocampal function. Immunoprecipitation and western blot confirmed protein expression of a novel ~170 kDa RIMS2 variant in the cerebellum, and in the hippocampus, it was significantly increased in KO vs WT (p < 0.0001), and in a sex-dependent manner (p = 0.0390). Furthermore, male KOs had decreased total canonical RIMS2 levels in the cerebellum (p = 0.0027) and hippocampus (p < 0.0001), whereas female KOs had increased total RIMS1 levels in the cerebellum (p = 0.0389). In summary, RBM5 modulates brain function in mammals. Future work is needed to test if RBM5 dependent regulation of RIMS2 splicing effects vision and cognition, and to verify potential sex differences on behavior in a larger cohort of mice.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Enfermedades del Sistema Nervioso , Proteínas Supresoras de Tumor , Animales , Femenino , Masculino , Ratones , Encéfalo/metabolismo , Lesiones Encefálicas/patología , Lesiones Traumáticas del Encéfalo/patología , Proteínas de Ciclo Celular/metabolismo , Cerebelo/patología , Proteínas de Unión al ADN/metabolismo , Técnicas de Inactivación de Genes , Hipocampo/metabolismo , Aprendizaje por Laberinto/fisiología , Ratones Noqueados , Enfermedades del Sistema Nervioso/patología , Proteostasis , Proteínas de Unión al ARN/metabolismo
5.
Genome Biol ; 25(1): 16, 2024 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-38216972

RESUMEN

BACKGROUND: The oncogenic protein HOXA9 plays a critical role in leukemia transformation and maintenance, and its aberrant expression is a hallmark of most aggressive acute leukemia. Although inhibiting the upstream regulators of HOXA9 has been proven as a significant therapeutic intervention, the comprehensive regulation network controlling HOXA9 expression in leukemia has not been systematically investigated. RESULTS: Here, we perform genome-wide CRISPR/Cas9 screening in the HOXA9-driven reporter acute leukemia cells. We identify a poorly characterized RNA-binding protein, RBM5, as the top candidate gene required to maintain leukemia cell fitness. RBM5 is highly overexpressed in acute myeloid leukemia (AML) patients compared to healthy individuals. RBM5 loss triggered by CRISPR knockout and shRNA knockdown significantly impairs leukemia maintenance in vitro and in vivo. Through domain CRISPR screening, we reveal that RBM5 functions through a noncanonical transcriptional regulation circuitry rather than RNA splicing, such an effect depending on DNA-binding domains. By integrative analysis and functional assays, we identify HOXA9 as the downstream target of RBM5. Ectopic expression of HOXA9 rescues impaired leukemia cell proliferation upon RBM5 loss. Importantly, acute protein degradation of RBM5 through auxin-inducible degron system immediately reduces HOXA9 transcription. CONCLUSIONS: We identify RBM5 as a new upstream regulator of HOXA9 and reveal its essential role in controlling the survival of AML. These functional and molecular mechanisms further support RBM5 as a promising therapeutic target for myeloid leukemia treatment.


Asunto(s)
Proteínas de Homeodominio , Leucemia Mieloide Aguda , Humanos , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas de Unión al ARN/genética , Proteínas Supresoras de Tumor/metabolismo
6.
J Biol Chem ; 300(3): 105673, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38272235

RESUMEN

The protein kinase RNA-like endoplasmic reticulum kinase (PERK)-eukaryotic translation initiation factor 2 subunit α (eIF2α) pathway plays an essential role in endoplasmic reticulum (ER) stress. When the PERK-eIF2α pathway is activated, PERK phosphorylates eIF2α (p-eIF2α) at Ser51 and quenches global protein synthesis. In this study, we verified eIF2α as a bona fide substrate of the E3 ubiquitin ligase carboxyl terminus of the HSC70-interaction protein (CHIP) both in vitro and in cells. CHIP mediated the ubiquitination and degradation of nonphosphorylated eIF2α in a chaperone-independent manner and promoted the upregulation of the cyclic AMP-dependent transcription factor under endoplasmic reticulum stress conditions. Cyclic AMP-dependent transcription factor induced the transcriptional enhancement of the tumor suppressor genes PTEN and RBM5. Although transcription was enhanced, the PTEN protein was subsequently degraded by CHIP, but the expression of the RBM5 protein was upregulated, thereby suppressing the proliferation and migration of A549 cells. Overall, our study established a new mechanism that deepened the understanding of the PERK-eIF2α pathway through the ubiquitination and degradation of eIF2α. The crosstalk between the phosphorylation and ubiquitination of eIF2α shed light on a new perspective for tumor progression.


Asunto(s)
Factor 2 Eucariótico de Iniciación , Genes Supresores de Tumor , Ubiquitina-Proteína Ligasas , Ubiquitinación , Regulación hacia Arriba , Humanos , Células A549 , Proliferación Celular/genética , AMP Cíclico/metabolismo , Estrés del Retículo Endoplásmico/genética , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Fosforilación , Factores de Transcripción/metabolismo , Ubiquitinación/genética , Regulación hacia Arriba/genética , Movimiento Celular/genética , Ubiquitina-Proteína Ligasas/metabolismo
7.
BMC Musculoskelet Disord ; 24(1): 921, 2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-38031049

RESUMEN

Osteoporosis is a prevalent bone disease with multigene involved, and the molecular mechanisms of its pathogenesis are not entirely understood. This study aims to identify novel key genes involved in osteoporosis to discover potential pharmacological targets. We analyzed three microarray datasets and identified four differentially expressed genes. The LASSO model indicated that RNA-binding motif protein 5 (RBM5) is associated with osteoporosis and is a potential drug target. We conducted the Spearman correlation analysis and found 52 genes that were significantly related to RBM5. Enrichment analysis showed that these genes were primarily involved in RNA splicing and osteoclast differentiation pathways. By using lentivirus-based shRNA, we successfully knocked down RBM5 expression in RAW264.7 cell line, which showed that RBM5 knockdown significantly impaired their differentiation potential to mature osteoclasts and significantly inhibited bone-resorbing activity. RT-qPCR analyses revealed the expression of osteoclastogenesis marker genes was downregulated along with RBM5 expression. These findings suggest that RBM5 plays a crucial role in the pathogenesis of osteoporosis and provides a new potential pharmacological target.


Asunto(s)
Osteogénesis , Osteoporosis , Humanos , Osteogénesis/genética , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Osteoclastos , Diferenciación Celular/genética , Osteoporosis/tratamiento farmacológico , Osteoporosis/genética , Osteoporosis/metabolismo , Proteínas de Unión al ADN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Ciclo Celular , Proteínas Supresoras de Tumor/metabolismo
8.
Cancers (Basel) ; 16(1)2023 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-38201567

RESUMEN

Downregulated RNA-binding motif protein 5 (RBM5) promotes the development and progression of various tumors, including bladder cancer (BC). Alternative splicing (AS) plays a crucial role in the progression of cancer by producing protein isomers with different functions or by promoting nonsense-mediated mRNA decay (NMD). However, whether RBM5 modulates the progression of BC through AS-NMD remains unexplored. In this study, we revealed that the downregulation of RBM5 expression promoted the expression of coactivator-associated arginine methyltransferase 1 (CARM1) in BC cells and tissues. Increased expression of CARM1 facilitated the activation of the Wnt/ß-catenin axis and cell proliferation, which then contributed to the poor prognosis of patients with BC. Interestingly, RBM5 bound directly to CARM1 mRNA and participated in AS-NMD, downregulating the expression of CARM1. In addition, we revealed that protein kinase catalytic subunit alpha (PRKACA) functioned as a phosphorylated kinase of GSK3ß, was regulated by CARM1 at the transcription level, and promoted the growth and progression of BC cells. Furthermore, in this study, we demonstrated a regulatory mechanism of Wnt/ß-catenin activation through the RBM5/CARM1/PRKACA axis and identified a novel potential target for treating BC.

9.
J Neurotrauma ; 39(7-8): 577-589, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35152732

RESUMEN

RNA-binding motif 5 (RBM5) is a pro-death tumor suppressor gene in cancer cells. It remains to be determined if it is neurotoxic in the brain or rather if it plays a fundamentally different role in the central nervous system (CNS). Brain-specific RBM5 knockout (KO) mice were given a controlled cortical impact (CCI) traumatic brain injury (TBI). Markers of acute cellular damage and repair were measured in hippocampal homogenates 48 h post-CCI. Hippocampal CA1/CA3 cell counts were assessed 7 days post-CCI to determine if early changes in injury markers were associated with histological outcome. No genotype-dependent differences were found in the levels of apoptotic markers (caspase 3, caspase 6, and caspase 9). However, KO females had a paradoxical increase in markers of pro-death calpain activation (145/150-spectrin and breakdown products [SBDP]) and in DNA repair/survival markers. (pH2A.x and pCREB). CCI-injured male KOs had a significant increase in phosphorylated calcium/calmodulin-dependent protein kinase II (pCaMKII). Despite sex/genotype-dependent differences in KOs in the levels of acute cell signaling targets involved in cell death pathways, 7 day hippocampal neuronal survival did not differ from that of wild types (WTs). Similarly, no differences in astrogliosis were observed. Finally, gene analysis revealed increased estrogen receptor α (ERα) levels in the KO hippocampus in females and may suggest a novel mechanism to explain sex-dimorphic effects on cell signaling. In summary, RBM5 inhibition did not affect hippocampal survival after a TBI in vivo but did modify targets involved in neural signal transduction/Ca2+ signaling pathways. Findings here support the view that RBM5 may serve a purpose in the CNS that is dissimilar from its traditional pro-death role in cancer.


Asunto(s)
Hipocampo , Transducción de Señal , Animales , Muerte Celular , Femenino , Eliminación de Gen , Hipocampo/metabolismo , Masculino , Ratones , Motivos de Unión al ARN
10.
Acta Neuropathol Commun ; 9(1): 123, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34225779

RESUMEN

Cancer stem cells (CSCs) contribute to radioresistance in medulloblastoma. Thus, identification of key regulators of medulloblastoma stemness is critical for improving radiotherapy for medulloblastoma. In the present study, we profiled CSC-related long non-coding RNAs (lncRNAs) between radioresistant and parental medulloblastoma cells. The roles of the lncRNA RBM5-AS1 in the stemness and radiosensitivity of medulloblastoma cells were investigated. We found that RBM5-AS1, a novel inducer of medulloblastoma stemness, was significantly upregulated in radioresistant medulloblastoma cells compared to parental cells. Knockdown of RBM5-AS1 diminished the viability and clonogenic survival of both radioresistant and parental medulloblastoma cells after radiation. Silencing of RBM5-AS1 significantly enhanced radiation-induced apoptosis and DNA damage. In vivo studies confirmed that depletion of RBM5-AS1 inhibited tumor growth and increased radiosensitivity in a medulloblastoma xenograft model. In contrast, overexpression of RBM5-AS1 reduced radiation-induced apoptosis and DNA damage in medulloblastoma cells. Mechanistically, RBM5-AS1 interacted with and stabilized sirtuin 6 (SIRT6) protein. Silencing of SIRT6 reduced the stemness and reinforced radiation-induced DNA damage in medulloblastoma cells. Overexpression of SIRT6 rescued medulloblastoma cells from RBM5-AS1 depletion-induced radiosensitization and DNA damage. Overall, we identify RBM5-AS1 as an inducer of stemness and radioresistance in medulloblastoma. Targeting RBM5-AS1 may represent a potential strategy to overcome the resistance to radiotherapy in this malignancy.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Neoplasias Cerebelosas/patología , Proteínas de Unión al ADN/metabolismo , Meduloblastoma/patología , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/metabolismo , Sirtuinas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Línea Celular Tumoral , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/metabolismo , Xenoinjertos , Humanos , Masculino , Meduloblastoma/genética , Meduloblastoma/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Tolerancia a Radiación/fisiología
11.
Virchows Arch ; 479(4): 795-802, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34089379

RESUMEN

Aneurysmal (ABC) and simple bone cysts (SBC) have been traditionally distinguished by radiological and histopathological features. However, there is some radiological and histopathological overlap between ABC and SBC. ABC is characterised by USP6 fusions while, recently, NFATC2 fusions have been found in a large proportion of SBC. Identifying these fusions may assist in confirming the diagnosis of either ABC or SBC. To elaborate the potential benefit of molecular testing, we report a prospective series of 19 consecutive bone cysts with comprehensive radiological, histopathological and molecular diagnostics. Integrating radiological, histopathological and molecular findings, 11 cysts were diagnosed as SBC and 8 as ABC. Radiologically, 6 of 11 SBC and 6 of 8 ABC were diagnosed as ABC. Fibrin-like collagen deposits were identified in 8 of 11 (73%) SBC and 3 of 8 (38%) ABC. Nodular fasciitis-like areas were identified in 6 of 8 (75%) ABC and in 7 of 11 (64%) SBC. A USP6 fusion was identified in all 8 ABC, including a novel RBM5-USP6 fusion. An NFATC2 fusion was found in 7 of 11 SBC (FUS-NFATC2 fusion in 5 and EWSR1-NFATC2 in 2 cases). There is radiological and histopathological overlap between SBC and ABC in a significant proportion of cases. A diagnosis of ABC is frequently suggested radiologically in SBC, and fibrin-like deposits, thought to be specific for SBC, may be found in some ABC. Molecular testing may significantly improve diagnostic accuracy in bone cysts.


Asunto(s)
Quistes Óseos Aneurismáticos/diagnóstico , Quistes Óseos/diagnóstico , Adolescente , Adulto , Quistes Óseos/metabolismo , Quistes Óseos Aneurismáticos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Niño , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fascitis/patología , Femenino , Fusión Génica/fisiología , Humanos , Hibridación Fluorescente in Situ/métodos , Masculino , Persona de Mediana Edad , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Patología Molecular/métodos , Regiones Promotoras Genéticas/genética , Estudios Prospectivos , Proteínas de Unión al ARN/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo
12.
Cell Biol Int ; 45(11): 2201-2210, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34019714

RESUMEN

Hepatocellular carcinoma (HCC) is regarded as one of the most common malignancies worldwide leading to cancer-related death. Long noncoding RNAs (lncRNAs) are a critical modulator affecting HCC progression. Whereas, the pathogenesis of lncRNA RBM5-AS1 in the development of HCC remains unclear. Quantitative RT-PCR or western blot assays were applied to detect the expression of genes and proteins, respectively. The proliferation and metastasis abilities were assessed using Cell counting kit-8 (CCK-8), EdU and transwell assays. RNA immunoprecipitation (RIP) experiment was employed to validate the molecular interactions. RBM5-AS1 is highly expressed in HCC tissues and cell lines, especially in Hep3B and HepG2 cells. RBM5-AS1 knockdown dramatically restrains cell proliferation, invasion and migration of HCC cells. Importantly, RBM5-AS1 acts as an epigenetic regulator to elevate the H3K27me3 level of miR-132/212 promoter regions via recruiting PRC2 (EZH2, SUZ12, EED), and eventually reducing miR-132/212 expressions. The recovery experiments demonstrated that downregulation of miR-132/212 markedly eliminate the antitumor effects mediated by RBM5-AS1 silencing in HCC cells. The data of this work illustrate that RBM5-AS1 acts as an epigenetic regulator to promote the HCC progression by repressing miR-132/212 expressions, which would provide a new insight for understanding the action mechanism of RBM5-AS1 in HCC development.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , MicroARNs/genética , Proteínas de Unión al ARN/genética , Proteínas Supresoras de Tumor/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética/genética , Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/genética , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , MicroARNs/metabolismo , Invasividad Neoplásica/genética , ARN sin Sentido/genética , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/genética , Proteínas Supresoras de Tumor/metabolismo
13.
Front Oncol ; 11: 603932, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33718153

RESUMEN

The RNA-binding motif protein 10 (RBM10) is involved in alternative splicing and modifies mRNA post-transcriptionally. RBM10 is abnormally expressed in the lung, breast, and colorectal cancer, female genital tumors, osteosarcoma, and other malignant tumors. It can inhibit proliferation, promote apoptosis, and inhibit invasion and metastasis. RBM10 has long been considered a tumor suppressor because it promotes apoptosis through the regulation of the MDM2-p53 negative feedback loop, Bcl-2, Bax, and other apoptotic proteins and inhibits proliferation through the Notch signaling and rap1a/Akt/CREB pathways. However, it has been recently demonstrated that RBM10 can also promote cancer. Given these different views, it is necessary to summarize the research progress of RBM10 in various fields to reasonably analyze the underlying molecular mechanisms, and provide new ideas and directions for the clinical research of RBM10 in various cancer types. In this review, we provide a new perspective on the reasons for these opposing effects on cancer biology, molecular mechanisms, research progress, and clinical value of RBM10.

14.
Gene ; 783: 145463, 2021 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-33515724

RESUMEN

RBM10 is a nuclear RNA-binding protein (RBP) that regulates the alternative splicing of primary transcripts. Recently, research on RBM10 has become increasingly active owing to its clinical importance, as indicated by studies on RBM0 mutations that cause TARP syndrome, an X-linked congenital pleiotropic developmental anomaly, and various cancers such as lung adenocarcinoma in adults. Herein, the molecular biology of RBM10 and its significance in medicine are reviewed, focusing on the gene and protein structures of RBM10, its cell biology, molecular functions and regulation, relationship with the paralogous protein RBM5, and the mutations of RBM10 and their associated diseases. Finally, the challenges in future studies of RBM10 are discussed in the concluding remarks.


Asunto(s)
Predisposición Genética a la Enfermedad , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/fisiología , Animales , Proteínas de Ciclo Celular/fisiología , Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica , Humanos , Mutación , Neoplasias/genética , Empalme del ARN , Proteínas Supresoras de Tumor/fisiología
15.
Front Mol Neurosci ; 13: 126, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32765218

RESUMEN

Gene splicing modulates the potency of cell death effectors, alters neuropathological disease processes, influences neuronal recovery, but may also direct distinct mechanisms of secondary brain injury. Therapeutic targeting of RNA splicing is a promising avenue for next-generation CNS treatments. RNA-binding proteins (RBPs) regulate a variety of RNA species and are prime candidates in the hunt for druggable targets to manipulate and tailor gene-splicing responses in the brain. RBPs preferentially recognize unique consensus sequences in targeted mRNAs. Also, RBPs often contain multiple RNA-binding domains (RBDs)-each having a unique consensus sequence-suggesting the possibility that drugs could be developed to block individual functional domains, increasing the precision of RBP-targeting therapies. Empirical characterization of most RBPs is lacking and represents a major barrier to advance this emerging therapeutic area. There is a paucity of data on the role of RBPs in the brain including, identification of their unique mRNA targets, defining how CNS insults affect their levels and elucidating which RBPs (and individual domains within) to target to improve neurological outcomes. This review focuses on the state-of-the-art of the RBP tumor suppressor RNA binding motif 5 (RBM5) in the CNS. We discuss its potent pro-death roles in cancer, which motivated our interest to study it in the brain. We review recent studies showing that RBM5 levels are increased after CNS trauma and that it promotes neuronal death in vitro. Finally, we conclude with recent reports on the first set of RBM5 regulated genes identified in the intact brain, and discuss how those findings provide new clues germane to its potential function(s) in the CNS, and pose new questions on its therapeutic utility to mitigate CNS injury.

16.
Eur Neurol ; 83(3): 242-250, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32610314

RESUMEN

INTRODUCTION: RBM5 acts as a tumor suppressor gene in lung and breast cancers; however, its role in the pathogenesis of medulloblastoma (MB) remains unclear. We previously identified 4 RBM5 mutations in whole exome sequencing analysis of 40 MB patients. This study examined the role of RBM5 in MB progression. METHODS: The expression patterns of RBM5 in tissues of 40 MB patients were analyzed using immunohistochemistry. Associations between RBM5 expression and overall survival (OS) were evaluated using Kaplan-Meier analysis. The RBM5 role in Daoy cells' proliferation, migration, and Wnt/ß-catenin signaling was analyzed after RBM5 knockdown and overexpression. RESULTS: The expression level of RBM5 mRNA and protein was significantly lower in MB than that in adjacent normal control tissues, and low RBM5 expression was significantly associated with reduced OS (p = 0.034). RBM5 knockdown induced Daoy and ONS-76 cells proliferation, while RBM5 overexpression repressed cell proliferation and migration in vitro (all p < 0.05). ß-Catenin, LEF1, and cyclin D1 mRNA levels were upregulated, while DKK1 expression was downregulated in Daoy cells following RBM5 knockdown. CONCLUSION: RBM5 may function as a tumor suppressor in MB by regulating Wnt/ß-catenin signaling, and its reduced expression is associated with lower OS.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteínas de Ciclo Celular/metabolismo , Neoplasias Cerebelosas/patología , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Meduloblastoma/patología , Proteínas de Unión al ARN/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Vía de Señalización Wnt , Biomarcadores de Tumor/genética , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/metabolismo , Proteínas de Unión al ADN/genética , Genes Supresores de Tumor , Humanos , Meduloblastoma/genética , Meduloblastoma/metabolismo , Proteínas de Unión al ARN/genética , Proteínas Supresoras de Tumor/genética , Vía de Señalización Wnt/fisiología
17.
FEBS J ; 287(17): 3719-3732, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32352217

RESUMEN

Hepatitis delta virus (HDV) is the agent responsible for the most severe form of human viral hepatitis. The HDV genome consists of a single-stranded circular RNA molecule that encodes for one single protein, the delta antigen. Given its simplicity, HDV must make use of several host cellular proteins to accomplish its life cycle processes, including transcription, replication, post-transcriptional, and post-translational modifications. Consequently, identification of the interactions established between HDV components and host proteins assumes a pivotal interest in the search of novel therapeutic targets. Here, we used the yeast three-hybrid system to screen a human liver cDNA library to identify host proteins that interact with the HDV genomic RNA. One of the identified proteins corresponded to the splicing factor SF3B155, a component of the U2snRNP complex that is essential for the early recognition of 3' splice sites in the pre-mRNAs of human genes. We show that the interaction between the HDV genomic RNA and SF3B155 occurs in vivo and that the expression of HDV promotes changes in splicing of human genes whose alternative splicing is SF3B155-dependent. We further show that expression of HDV triggers alterations in several constitutive and alternative splicing events in the tumor suppressor RBM5 transcript, with consequent reduction of its protein levels. This is the first description that HDV expression promotes changes in the splicing of human genes, and we suggest that the HDV-induced alternative splicing changes, through SF3B155 sequester, may contribute for the early progression to hepatocellular carcinoma characteristic of HDV-infected patients.


Asunto(s)
Ciclo Celular/genética , Genes cdc , Hepatitis D/genética , Virus de la Hepatitis Delta/fisiología , Fosfoproteínas/genética , Precursores del ARN/genética , Factores de Empalme de ARN/genética , Empalme del ARN/genética , Carcinoma Hepatocelular/virología , Transformación Celular Neoplásica/genética , Cocarcinogénesis/genética , Coinfección/genética , Humanos , Neoplasias Hepáticas/virología , Factor de Transcripción STAT3/biosíntesis , Factor de Transcripción STAT3/genética
18.
J Mol Biol ; 432(14): 4127-4138, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32450081

RESUMEN

The multi-domain RNA binding protein RBM5 is a molecular signature of metastasis. RBM5 regulates alternative splicing of apoptotic genes including the cell death receptor Fas and the initiator Caspase-2. The RBM5 RanBP2-type zinc finger (Zf1) is known to specifically recognize single-stranded RNAs with high affinity. Here, we study the structure and conformational dynamics of the Zf1 zinc finger of human RBM5 using NMR. We show that the presence of a non-canonical cysteine in Zf1 kinetically destabilizes the protein. Metal-exchange kinetics show that mutation of the cysteine establishes high-affinity coordination of the zinc. Our data indicate that selection of such a structurally destabilizing mutation during the course of evolution could present an opportunity for functional adaptation of the protein.


Asunto(s)
Proteínas de Ciclo Celular/ultraestructura , Proteínas de Unión al ADN/ultraestructura , Chaperonas Moleculares/ultraestructura , Proteínas de Complejo Poro Nuclear/ultraestructura , Proteínas de Unión al ARN/ultraestructura , Proteínas Supresoras de Tumor/ultraestructura , Dedos de Zinc/genética , Empalme Alternativo/genética , Secuencia de Aminoácidos/genética , Apoptosis/genética , Caspasa 2/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Proteínas de Complejo Poro Nuclear/química , Proteínas de Complejo Poro Nuclear/genética , Unión Proteica/genética , Conformación Proteica , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética , Zinc/química
19.
Neuroscience ; 440: 299-315, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32335213

RESUMEN

The tumor suppressor RNA-binding motif 5 (RBM5) regulates the expression levels and cassette exon-definition (i.e. splicing) of a select set of mRNAs in a tissue-specific manner. Most RBM5-regulated targets were identified in oncological investigations and frequently involve genes which mediate apoptotic cell death. Little is known about the role of RBM5 in the brain. Also, it is unclear if a brain injury may be required to detect RBM5 mediated effects on pro-apoptotic genes due to their low expression levels in the healthy adult CNS at baseline. Conditional/floxed (brain-specific) gene deleter mice were generated to elucidate CNS-specific RBM5 mRNA targets. Male/female mice were subjected to a severe controlled cortical impact (CCI) traumatic brain injury (TBI) in order to increase the background expression of pro-death mRNAs and facilitate testing of the hypothesis that RBM5 inhibition decreases post-injury upregulation of caspases/FAS in the CNS. As expected, a CCI increased caspases/FAS mRNA in the injured cortex. RBM5 KO did not affect their levels or splicing. Surprisingly, KO increased the mRNA levels of novel targets including casein kinase 2 alpha prime interacting protein (Csnka2ip/CKT2) - a gene not thought to be expressed in the brain, contrary to findings here. Twenty-two unique splicing events were also detected in KOs including increased block-inclusion of cassette exons 20-22 in regulating synaptic membrane exocytosis 2 (Rims2). In conclusion, here we used genome-wide transcriptomic analysis on healthy and injured RBM5 KO mouse brain tissue to elucidate the first known gene targets of this enigmatic RBP in this CNS.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas Supresoras de Tumor , Animales , Encéfalo/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Masculino , Ratones , Motivos de Unión al ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Supresoras de Tumor/metabolismo
20.
Cell Biol Int ; 44(1): 295-305, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31498514

RESUMEN

A growing body of research suggests that microRNAs (miRNAs) may play a key part in the progression of various cancers, including lung adenocarcinoma (LUAD). However, the expression and mechanism of miR-938 (microRNA-938) in LUAD have not been defined. Compared with adjacent tissues, the level of miR-938 was up-regulated in LUAD tissues. miR-938 expression was significantly associated with tumor size. In vitro assays indicated that miR-938 expression was also increased in the LUAD cell lines. Overexpression of miR-938 promoted LUAD cell proliferation, whereas down-regulation of miR-938 had the opposite effect. We identified RNA-binding protein 5 (RBM5) as a potential target gene of miR-938 in LUAD. Expression of RBM5 was down-regulated in LUAD tumor tissues and negatively correlated with expression of miR-938. Up-regulation of RBM5 reversed cell proliferation by inhibition of miR-938 expression in LUAD cells. These results showed that miR-938 may act as an oncogenic miRNA by targeting RBM5 in LUAD, indicating that miR-938 could be used as a potential therapeutic target for LUAD patients.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA