Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Biol ; 21(3): e3002028, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36930682

RESUMO

A major function of TAR DNA-binding protein-43 (TDP-43) is to repress the inclusion of cryptic exons during RNA splicing. One of these cryptic exons is in UNC13A, a genetic risk factor for amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). The accumulation of cryptic UNC13A in disease is heightened by the presence of a risk haplotype located within the cryptic exon itself. Here, we revealed that TDP-43 extreme N-terminus is important to repress UNC13A cryptic exon inclusion. Further, we found hnRNP L, hnRNP A1, and hnRNP A2B1 bind UNC13A RNA and repress cryptic exon inclusion, independently of TDP-43. Finally, higher levels of hnRNP L protein associate with lower burden of UNC13A cryptic RNA in ALS/FTD brains. Our findings suggest that while TDP-43 is the main repressor of UNC13A cryptic exon inclusion, other hnRNPs contribute to its regulation and may potentially function as disease modifiers.


Assuntos
Esclerose Lateral Amiotrófica , Demência Frontotemporal , Ribonucleoproteínas Nucleares Heterogêneas Grupo L , Humanos , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Éxons/genética , Demência Frontotemporal/genética , Ribonucleoproteínas Nucleares Heterogêneas/genética , RNA , Proteínas do Tecido Nervoso/metabolismo
2.
PLoS Biol ; 19(9): e3001378, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34543262

RESUMO

Stratified epithelia such as the epidermis require coordinated regulation of stem and progenitor cell proliferation, survival, and differentiation to maintain homeostasis. Integrin-mediated anchorage of the basal layer stem cells of the epidermis to the underlying dermis through extracellular matrix (ECM) proteins is crucial for this process. It is currently unknown how the expression of these integrins and ECM genes are regulated. Here, we show that the RNA-binding protein (RBP) heterogeneous nuclear ribonucleoprotein L (HNRNPL) binds to these genes on chromatin to promote their expression. HNRNPL recruits RNA polymerase II (Pol II) to integrin/ECM genes and is required for stabilizing Pol II transcription through those genes. In the absence of HNRNPL, the basal layer of the epidermis where the stem cells reside prematurely differentiates and detaches from the underlying dermis due to diminished integrin/ECM expression. Our results demonstrate a critical role for RBPs on chromatin to maintain stem and progenitor cell fate by dictating the expression of specific classes of genes.


Assuntos
Células Epidérmicas/metabolismo , Matriz Extracelular/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Integrinas/metabolismo , Diferenciação Celular , Células Cultivadas , Cromatina , Epiderme/crescimento & desenvolvimento , Matriz Extracelular/genética , Humanos , Integrinas/genética , Células-Tronco
3.
J Nanobiotechnology ; 22(1): 62, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38360615

RESUMO

BACKGROUND: A large number of Fusobacterium nucleatum (Fn) are present in colorectal cancer (CRC) tissues of patients who relapse after chemotherapy, and Fn has been reported to promote oxaliplatin and 5-FU chemoresistance in CRC. Pathogens such as bacteria and parasites stimulate exosome production in tumor cells, and the regulatory mechanism of exosomal circRNA in the transmission of oxaliplatin and 5-FU chemotherapy resistance in Fn-infected CRC remains unclear. METHODS: Hsa_circ_0004085 was screened by second-generation sequencing of CRC tissues. The correlation between hsa_circ_0004085 and patient clinical response to oxaliplatin/5-FU was analyzed. Exosome tracing experiments and live imaging systems were used to test the effect of Fn infection in CRC on the distribution of hsa_circ_0004085. Colony formation, ER tracking analysis and immunofluorescence were carried out to verify the regulatory effect of exosomes produced by Fn-infected CRC cells on chemotherapeutic resistance and ER stress. RNA pulldown, LC-MS/MS analysis and RIP were used to explore the regulatory mechanism of downstream target genes by hsa_circ_0004085. RESULTS: First, we screened out hsa_circ_0004085 with abnormally high expression in CRC clinical samples infected with Fn and found that patients with high expression of hsa_circ_0004085 in plasma had a poor clinical response to oxaliplatin/5-FU. Subsequently, the circular structure of hsa_circ_0004085 was identified. Fn infection promoted hsa_circ_0004085 formation by hnRNP L and packaged hsa_circ_0004085 into exosomes by hnRNP A1. Exosomes produced by Fn-infected CRC cells transferred hsa_circ_0004085 between cells and delivered oxaliplatin/5-FU resistance to recipient cells by relieving ER stress. Hsa_circ_0004085 enhanced the stability of GRP78 mRNA by binding to RRBP1 and promoted the nuclear translocation of ATF6p50 to relieve ER stress. CONCLUSIONS: Plasma levels of hsa_circ_0004085 are increased in colon cancer patients with intracellular Fn and are associated with a poor response to oxaliplatin/5-FU. Fn infection promoted hsa_circ_0004085 formation by hnRNP L and packaged hsa_circ_0004085 into exosomes by hnRNP A1. Exosomes secreted by Fn-infected CRC cells deliver hsa_circ_0004085 between cells. Hsa_circ_0004085 relieves ER stress in recipient cells by regulating GRP78 and ATF6p50, thereby delivering resistance to oxaliplatin and 5-FU.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Exossomos , Ribonucleoproteínas Nucleares Heterogêneas Grupo L , MicroRNAs , Humanos , Oxaliplatina/farmacologia , Oxaliplatina/uso terapêutico , Oxaliplatina/metabolismo , Fusobacterium nucleatum/genética , Fusobacterium nucleatum/metabolismo , Ribonucleoproteína Nuclear Heterogênea A1/metabolismo , Neoplasias Colorretais/metabolismo , Exossomos/metabolismo , Cromatografia Líquida , Chaperona BiP do Retículo Endoplasmático , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Espectrometria de Massas em Tandem , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , MicroRNAs/metabolismo , Proliferação de Células
4.
Nucleic Acids Res ; 50(22): 13026-13044, 2022 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-36533518

RESUMO

The mammalian transcriptome comprises a vast family of long noncoding (lnc)RNAs implicated in physiologic processes such as myogenesis, through which muscle forms during embryonic development and regenerates in the adult. However, the specific molecular mechanisms by which lncRNAs regulate human myogenesis are poorly understood. Here, we identified a novel muscle-specific lncRNA, lncFAM71E1-2:2 (lncFAM), which increased robustly during early human myogenesis. Overexpression of lncFAM promoted differentiation of human myoblasts into myotubes, while silencing lncFAM suppressed this process. As lncFAM resides in the nucleus, chromatin isolation by RNA purification followed by mass spectrometry (ChIRP-MS) analysis was employed to identify the molecular mechanisms whereby it might promote myogenesis. Analysis of lncFAM-interacting proteins revealed that lncFAM recruited the RNA-binding protein HNRNPL to the promoter of MYBPC2, in turn increasing MYBPC2 mRNA transcription and enhancing production of the myogenic protein MYBPC2. These results highlight a mechanism whereby a novel ribonucleoprotein complex, lncFAM-HNRNPL, elevates MYBPC2 expression transcriptionally to promote myogenesis.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas Grupo L , Desenvolvimento Muscular , Regiões Promotoras Genéticas , RNA Longo não Codificante , Transcrição Gênica , Humanos , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Desenvolvimento Muscular/genética , Fibras Musculares Esqueléticas/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Transcrição Gênica/genética , Inativação Gênica , Transporte Proteico/genética
5.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34607953

RESUMO

Chemokine production by epithelial cells is important for neutrophil recruitment during viral infection, the appropriate regulation of which is critical for restraining inflammation and attenuating subsequent tissue damage. Epithelial cell expression of long noncoding RNAs (lncRNAs), RNA-binding proteins, and their functional interactions during viral infection and inflammation remain to be fully understood. Here, we identified an inducible lncRNA in the Cxcl2 gene locus, lnc-Cxcl2, which could selectively inhibit Cxcl2 expression in mouse lung epithelial cells but not in macrophages. lnc-Cxcl2-deficient mice exhibited increased Cxcl2 expression, enhanced neutrophils recruitment, and more severe inflammation in the lung after influenza virus infection. Mechanistically, nucleus-localized lnc-Cxcl2 bound to Cxcl2 promoter, recruited a ribonucleoprotein La, which inhibited the chromatin accessibility of chemokine promoters, and consequently inhibited Cxcl2 transcription in cis However, unlike mouse lnc-Cxcl2, human lnc-CXCL2-4-1 inhibited multiple immune cytokine expressions including chemokines in human lung epithelial cells. Together, our results demonstrate a self-protecting mechanism within epithelial cells to restrain chemokine and neutrophil-mediated inflammation, providing clues for better understanding chemokine regulation and epithelial cell function in lung viral infection.


Assuntos
Quimiocina CXCL2/genética , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , RNA Longo não Codificante/genética , Células A549 , Animais , Linhagem Celular Tumoral , Quimiocina CXCL2/metabolismo , Cromatina/metabolismo , Células Epiteliais/metabolismo , Células HEK293 , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Humanos , Inflamação/prevenção & controle , Mediadores da Inflamação , Vírus da Influenza A/imunologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Pneumonia/imunologia , Pneumonia/patologia , Pneumonia/virologia , Regiões Promotoras Genéticas/genética , Células RAW 264.7 , Estomatite Vesicular/imunologia , Estomatite Vesicular/patologia , Vírus da Estomatite Vesicular Indiana/imunologia
6.
Neurobiol Dis ; 181: 106080, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-36925052

RESUMO

BACKGROUND: Ischemic stroke (IS) is the primary cause of mortality and disability worldwide. Circular RNAs (circRNAs) have been proposed as crucial regulators in IS. This study focused on the role of circPDS5B in IS and its underlying mechanism. METHOD: Transient middle cerebral artery occlusion (tMCAO) mice and glucose deprivation/reoxygenation (OGD/R)-exposed human brain microvascular endothelial cells (BMECs) were used as IS models. Expression levels of circPDS5B, heterogenous nuclear ribonucleoprotein L (hnRNPL), runt-related transcription factor-1 (Runx1), and Zinc finger protein 24 (ZNF24) were quantified by qRT-PCR. MTT, wound healing, transwell and tube formation assays were employed to evaluate the cell proliferation, migration, and angiogenesis, respectively. Moreover, RNA pull-down, and RIP assay were performed to investigate the interaction among circPDS5B, hnRNPL and vascular endothelial growth factor-A (VEGF-A). RESULTS: circPDS5B was significantly up-regulated in IS patients and tMCAO mice. Deficiency of circPDS5B relieved brain infarction and neuronal injury of tMCAO mice. OGD/R-induced apoptosis, inhibition in viability, migration, and angiogenesis in BMECs were dramatically abrogated by circPDS5B knockdown. Mechanistically, circPDS5B stabilized Runx1 and ZNF24 via recruiting hnRNPL, thereby suppressing the transcription and expression of VEGFA. hnRNPL silencing strengthened circPDS5B knockdown-mediated beneficial effect on IS. CONCLUSION: Altogether, our study showed that high expression of circPDS5B exacerbated IS through recruitment of hnRNPL to stabilize Runx1/ZNF24 and subsequently inactivate VEGFA. Our findings suggest circPDS5B may be a novel therapeutic target for IS.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas Grupo L , AVC Isquêmico , MicroRNAs , Acidente Vascular Cerebral , Fator A de Crescimento do Endotélio Vascular , Animais , Humanos , Camundongos , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/farmacologia , Células Endoteliais/metabolismo , Glucose/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/farmacologia , Infarto da Artéria Cerebral Média/metabolismo , AVC Isquêmico/genética , AVC Isquêmico/metabolismo , MicroRNAs/metabolismo , Neovascularização Fisiológica , RNA Circular/genética , RNA Circular/metabolismo , RNA Circular/farmacologia , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
EMBO J ; 38(3)2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30530525

RESUMO

The human nonsense-mediated mRNA decay pathway (NMD) performs quality control and regulatory functions within complex post-transcriptional regulatory networks. In addition to degradation-promoting factors, efficient and accurate detection of NMD substrates involves proteins that safeguard normal mRNAs. Here, we identify hnRNP L as a factor that protects mRNAs with NMD-inducing features including long 3'UTRs. Using biochemical and transcriptome-wide approaches, we provide evidence that the susceptibility of a given transcript to NMD can be modulated by its 3'UTR length and ability to recruit hnRNP L. Integrating these findings with the previously defined role of polypyrimidine tract binding protein 1 in NMD evasion enables enhanced prediction of transcript susceptibility to NMD. Unexpectedly, this system is subverted in B cell lymphomas harboring translocations that produce BCL2:IGH fusion mRNAs. CRISPR/Cas9 deletion of hnRNP L binding sites near the BCL2 stop codon reduces expression of the fusion mRNAs and induces apoptosis. Together, our data indicate that protection by hnRNP L overrides the presence of multiple 3'UTR introns, allowing these aberrant mRNAs to evade NMD and promoting BCL2 overexpression and neoplasia.


Assuntos
Regiões 3' não Traduzidas/genética , Regulação Neoplásica da Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Linfoma de Células B/patologia , Degradação do RNAm Mediada por Códon sem Sentido , RNA Mensageiro/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , Humanos , Linfoma de Células B/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transcriptoma
8.
Nucleic Acids Res ; 48(21): 12326-12335, 2020 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-33231682

RESUMO

Circular RNAs (circRNAs) are a class of noncoding RNAs, generated from pre-mRNAs by circular splicing of exons and functionally largely uncharacterized. Here we report on the design, expression, and characterization of artificial circRNAs that act as protein sponges, specifically binding and functionally inactivating hnRNP (heterogeneous nuclear ribonucleoprotein) L. HnRNP L regulates alternative splicing, depending on short CA-rich RNA elements. We demonstrate that designer hnRNP L-sponge circRNAs with CA-repeat or CA-rich sequence clusters can efficiently and specifically modulate splicing-regulatory networks in mammalian cells, including alternative splicing patterns and the cellular distribution of a splicing factor. This new strategy can in principle be applied to any RNA-binding protein, opening up new therapeutic strategies in molecular medicine.


Assuntos
Processamento Alternativo , Éxons , Engenharia Genética/métodos , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Precursores de RNA/genética , RNA Circular/genética , Pareamento de Bases , Inativação Gênica , Células HEK293 , Células HeLa , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/antagonistas & inibidores , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Humanos , Imunoprecipitação/métodos , Íntrons , Conformação de Ácido Nucleico , Motivos de Nucleotídeos , Plasmídeos/química , Plasmídeos/metabolismo , Ligação Proteica , Precursores de RNA/metabolismo , RNA Catalítico/genética , RNA Catalítico/metabolismo , RNA Circular/biossíntese , RNA Circular/química
9.
Molecules ; 27(16)2022 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-36014574

RESUMO

Pulmonary fibrosis is characterized by the destruction of alveolar architecture and the irreversible scarring of lung parenchyma, with few therapeutic options and effective therapeutic drugs. Here, we demonstrate the anti-pulmonary fibrosis of 3-(4-methoxyphenyl)-4-oxo-4H-1-benzopyran-7-yl(αS)-α,3,4-trihydroxybenzenepropanoate (MOBT) in mice and a cell model induced by bleomycin and transforming growth factor-ß1. The anti-pulmonary fibrosis of MOBT was evaluated using a MicroCT imaging system for small animals, lung function analysis and H&E and Masson staining. The results of RNA fluorescence in situ hybridization, chromatin immunoprecipitation (ChIP)-PCR, RNA immunoprecipitation, ChIP-seq, RNA-seq, and half-life experiments demonstrated the anti-pulmonary fibrotic mechanism. Mechanistic dissection showed that MOBT inhibited lncITPF transcription by preventing p-Smad2/3 translocation from the cytoplasm to the nucleus, resulting in a reduction in the amount of the lncITPF-hnRNP L complex. The decreased lncITPF-hnRNP L complex reduced MEF2c expression by blocking its alternative splicing, which in turn inhibited the expression of MEF2c target genes, such as TAGLN2 and FMN1. Briefly, MOBT alleviated pulmonary fibrosis through the lncITPF-hnRNP-l-complex-targeted MEF2c signaling pathway. We hope that this study will provide not only a new drug candidate but also a novel therapeutic drug target, which will bring new treatment strategies for pulmonary fibrosis.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas Grupo L , Fibrose Pulmonar , Animais , Bleomicina/farmacologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/farmacologia , Hibridização in Situ Fluorescente , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fibrose Pulmonar/induzido quimicamente , RNA/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo
10.
J Cell Physiol ; 236(3): 2023-2035, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32730662

RESUMO

The downregulation of melatonin receptor 1A (MTNR1A) is associated with a range of pathological conditions, including membranous nephropathy. Knowledge of the mechanism underlying MTNR1A expression has been limited to the transcriptional regulation level. Here, RNA interference screening in human kidney cells revealed that heterogeneous nuclear ribonucleoprotein L (hnRNPL) upregulated MTNR1A RNA post-transcriptionally. hnRNPL knockdown or overexpression led to increased or decreased levels of cyclic adenosine monophosphate-responsive element-binding protein phosphorylation, respectively. Molecular studies showed that cytoplasmic hnRNPL exerts a stabilizing effect on the MTNR1A transcript through CA-repeat elements in its coding region. Further studies revealed that the interaction between hnRNPL and MTNR1A serves to protect MNTR1A RNA degradation by the exosome component 10 protein. MTNR1A, but not hnRNPL, displays a diurnal rhythm in mouse kidneys. Enhanced levels of MTNR1A recorded at midnight correlated with robust binding activity between cytoplasmic hnRNPL and the MTNR1A transcript. Both hnRNPL and MTNR1A were decreased in the cytoplasm of tubular epithelial cells from experimental membranous nephropathy kidneys, supporting their clinical relevance. Collectively, our data identified cytoplasmic hnRNPL as a novel player in the upregulation of MTNR1A expression in renal tubular epithelial cells, and as a potential therapeutic target.


Assuntos
Citoplasma/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Túbulos Renais/metabolismo , Receptor MT1 de Melatonina/genética , Animais , Linhagem Celular , Ritmo Circadiano/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Células Epiteliais/metabolismo , Exorribonucleases/metabolismo , Complexo Multienzimático de Ribonucleases do Exossomo/metabolismo , Glomerulonefrite Membranosa/genética , Glomerulonefrite Membranosa/patologia , Humanos , Túbulos Renais/patologia , Camundongos Endogâmicos BALB C , Modelos Biológicos , Fases de Leitura Aberta/genética , Fosforilação , Estabilidade de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor MT1 de Melatonina/metabolismo , Sequências Repetitivas de Ácido Nucleico/genética , Regulação para Cima/genética
11.
J Virol ; 94(10)2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32161169

RESUMO

Upon infection, the highly structured 5' untranslated region (5' UTR) of picornavirus is involved in viral protein translation and RNA synthesis. As a critical element in the 5' UTR, the internal ribosome entry site (IRES) binds to various cellular proteins to function in the processes of picornavirus replication. Foot-and-mouth disease virus (FMDV) is an important member in the family Picornaviridae, and its 5' UTR contains a functional IRES element. In this study, the cellular heterogeneous nuclear ribonucleoprotein L (hnRNP L) was identified as an IRES-binding protein for FMDV by biotinylated RNA pulldown assays, mass spectrometry (MS) analysis, and determination of hnRNP L-IRES interaction regions. Further, we found that hnRNP L inhibited the growth of FMDV through binding to the viral IRES and that the inhibitory effect of hnRNP L on FMDV growth was not due to FMDV IRES-mediated translation, but to influence on viral RNA synthesis. Finally, hnRNP L was demonstrated to coimmunoprecipitate with RNA-dependent RNA polymerase (3Dpol) in an FMDV RNA-dependent manner in the infected cells. Thus, our results suggest that hnRNP L, as a critical IRES-binding protein, negatively regulates FMDV replication by inhibiting viral RNA synthesis, possibly by remaining in the replication complex.IMPORTANCE Picornaviruses, as a large family of human and animal pathogens, cause a bewildering array of disease syndromes. Many host factors are implicated in the pathogenesis of these viruses, and some proteins interact with the viral IRES elements to affect function. Here, we report for the first time that cellular hnRNP L specifically interacts with the IRES of the picornavirus FMDV and negatively regulates FMDV replication through inhibiting viral RNA synthesis. Further, our results showed that hnRNP L coimmunoprecipitates with FMDV 3Dpol in a viral RNA-dependent manner, suggesting that it may remain in the replication complex to function. The data presented here would facilitate further understanding of virus-host interactions and the pathogenesis of picornavirus infections.


Assuntos
Regiões 5' não Traduzidas , Vírus da Febre Aftosa/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Sítios Internos de Entrada Ribossomal/fisiologia , RNA Viral/biossíntese , Replicação Viral/fisiologia , Animais , Linhagem Celular , Vírus da Febre Aftosa/genética , Regulação Viral da Expressão Gênica , Técnicas de Inativação de Genes , Células HEK293 , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Imunoprecipitação , Ligação Proteica , RNA Viral/genética , Transcriptoma
12.
Mol Ther ; 28(10): 2220-2236, 2020 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-32592691

RESUMO

T cell receptor signaling, together with cytokine-induced signals, can differentially regulate RNA processing to influence T helper versus regulatory T cell fate. Protein kinase C family members have been shown to function in alternative splicing and RNA processing in various cell types. T cell-specific protein kinase C theta, a molecular regulator of T cell receptor downstream signaling, has been shown to phosphorylate splicing factors and affect post-transcriptional control of T cell gene expression. In this study, we explored how using a synthetic cell-penetrating peptide mimic for intracellular anti-protein kinase C theta delivery fine-tunes differentiation of induced regulatory T cells through its differential effects on RNA processing. We identified protein kinase C theta signaling as a critical modulator of two key RNA regulatory factors, heterogeneous nuclear ribonucleoprotein L (hnRNPL) and protein-l-isoaspartate O-methyltransferase-1 (PCMT1), and loss of protein kinase C theta function initiated a "switch" in post-transcriptional organization in induced regulatory T cells. More interestingly, we discovered that protein-l-isoaspartate O- methyltransferase-1 acts as an instability factor in induced regulatory T cells, by methylating the forkhead box P3 (FOXP3) promoter. Targeting protein-l-isoaspartate O-methyltransferase-1 using a cell-penetrating antibody revealed an efficient means of modulating RNA processing to confer a stable regulatory T cell phenotype.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Proteína D-Aspartato-L-Isoaspartato Metiltransferase/genética , Proteína Quinase C-theta/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/metabolismo , Peptídeos Penetradores de Células/farmacologia , Fatores de Transcrição Forkhead/genética , Regiões Promotoras Genéticas , Ligação Proteica , Proteína D-Aspartato-L-Isoaspartato Metiltransferase/metabolismo , Estabilidade Proteica , Transdução de Sinais
13.
Mol Cell ; 49(5): 972-82, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-23394998

RESUMO

Pre-mRNA splicing is catalyzed through the activity of the spliceosome, a dynamic enzymatic complex. Forcing aberrant interactions within the spliceosome can reduce splicing efficiency and alter splice site choice; however, it is unknown whether such alterations are naturally exploited mechanisms of splicing regulation. Here, we demonstrate that hnRNP L represses CD45 exon 4 by recruiting hnRNP A1 to a sequence upstream of the 5' splice site. Together, hnRNP L and A1 induce extended contacts between the 5' splice site-bound U1 snRNA and neighboring exonic sequences that, in turn, inhibit stable association of U6 snRNA and subsequent catalysis. Importantly, analysis of several exons regulated by hnRNP L shows a clear relationship between the potential for binding of hnRNP A1 and U1 snRNA and the effect of hnRNP L on splicing. Together, our results demonstrate that conformational perturbations within the spliceosome are a naturally occurring and generalizable mechanism for controlling alternative splicing decisions.


Assuntos
Éxons , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , RNA Nuclear Pequeno/metabolismo , Spliceossomos/genética , Spliceossomos/metabolismo , Processamento Alternativo , Sequência de Bases , Sítios de Ligação , Células HEK293 , Ribonucleoproteína Nuclear Heterogênea A1 , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Humanos , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/metabolismo , Interferência de RNA , Splicing de RNA , RNA Mensageiro/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
14.
RNA ; 24(6): 761-768, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29581412

RESUMO

The fidelity of RNA splicing is regulated by a network of splicing enhancers and repressors, although the rules that govern this process are not yet fully understood. One mechanism that contributes to splicing fidelity is the repression of nonconserved cryptic exons by splicing factors that recognize dinucleotide repeats. We previously identified that TDP-43 and PTBP1/PTBP2 are capable of repressing cryptic exons utilizing UG and CU repeats, respectively. Here we demonstrate that hnRNP L (HNRNPL) also represses cryptic exons by utilizing exonic CA repeats, particularly near the 5'SS. We hypothesize that hnRNP L regulates CA repeat repression for both cryptic exon repression and developmental processes such as T cell differentiation.


Assuntos
Éxons , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Precursores de RNA/genética , Splicing de RNA , Proteínas Repressoras/metabolismo , Animais , Sequência de Bases , Genoma , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Células Jurkat , Camundongos , Proteínas Repressoras/genética
15.
RNA ; 24(3): 324-331, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29263134

RESUMO

HnRNP D, better known as AUF1, is an extensively studied protein that controls a variety of cellular pathways. Consequently, its expression has to be tightly regulated to prevent the onset of pathologies. In contrast, the cellular functions and regulation of its ubiquitously expressed paralog hnRNP DL are barely explored. Here, we present an intricate crosstalk between these two proteins. Both hnRNP D and DL are able to control their own expression by alternative splicing of cassette exons in their 3'UTRs. Exon inclusion produces mRNAs degraded by nonsense-mediated decay. Moreover, hnRNP D and DL control the expression of one another by the same mechanism. Thus, we identified two novel ways of how hnRNP D expression is controlled. The tight interconnection of expression control directly links hnRNP DL to hnRNP D-related diseases and emphasizes the importance of a systematic analysis of its cellular functions.


Assuntos
Processamento Alternativo , Regulação da Expressão Gênica/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo D/genética , Ribonucleoproteínas Nucleares Heterogêneas/genética , Degradação do RNAm Mediada por Códon sem Sentido/genética , Ribonucleoproteínas/genética , Regiões 3' não Traduzidas/genética , Éxons , Genes Reporter , Células HeLa , Ribonucleoproteína Nuclear Heterogênea D0 , Ribonucleoproteínas Nucleares Heterogêneas Grupo D/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Homeostase , Humanos , RNA Mensageiro/genética , Ribonucleoproteínas/fisiologia
16.
Mol Ther ; 27(2): 380-393, 2019 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-30528088

RESUMO

The role of long non-coding RNA (lncRNA) in idiopathic pulmonary fibrosis (IPF) is poorly understood. We found a novel lncRNA-ITPF that was upregulated in IPF. Bioinformatics and in vitro translation verified that lncITPF is an actual lncRNA, and its conservation is in evolution. Northern blot and rapid amplification of complementary DNA ends were used to analyze the full-length sequence of lncITPF. RNA fluorescence in situ hybridization and nucleocytoplasmic separation demonstrated that lncITPF was mainly located in the nucleus. RNA sequencing, chromatin immunoprecipitation (ChIP)-qPCR, CRISPR-Cas9 technology, and promoter activity analysis showed that the fibrotic function of lncITPF depends on its host gene integrin ß-like 1 (ITGBL1), but they did not share the same promoter and were not co-transcribed. Luciferase activity, pathway inhibitors, and ChIP-qPCR showed that smad2/3 binds to the lncITPF promoter, and TGF-ß1-smad2/3 was the upstream inducer of the fibrotic pathway. Furthermore, RNA-protein pull-down, liquid chromatography-mass spectrometry (LC-MS), and protein-RNA immunoprecipitation showed that lncITPF regulated H3 and H4 histone acetylation in the ITGBL1 promoter by targeting heterogeneous nuclear ribonucleoprotein L. Finally, sh-lncITPF was used to evaluate the therapeutic effect of lncITPF. Clinical analysis showed that lncITPF is associated with the clinicopathological features of IPF patients. Our findings provide a therapeutic target or diagnostic biomarker for IPF.


Assuntos
Sistemas CRISPR-Cas/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , RNA Longo não Codificante/metabolismo , Idoso , Animais , Northern Blotting , Western Blotting , Sistemas CRISPR-Cas/genética , Linhagem Celular , Movimento Celular/genética , Movimento Celular/fisiologia , Imunoprecipitação da Cromatina , Cromatografia Líquida , Feminino , Imunofluorescência , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Humanos , Fibrose Pulmonar Idiopática/genética , Imunoprecipitação , Hibridização In Situ , Masculino , Camundongos , Pessoa de Meia-Idade , Modelos Biológicos , Miofibroblastos/citologia , Miofibroblastos/metabolismo , RNA Longo não Codificante/genética , Ratos , Reação em Cadeia da Polimerase em Tempo Real
17.
Mol Cell ; 45(4): 459-69, 2012 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-22264826

RESUMO

Mediator complex is an integrative hub for transcriptional regulation. Here we show that Mediator regulates alternative mRNA processing via its MED23 subunit. Combining tandem affinity purification and mass spectrometry, we identified a number of mRNA processing factors that bind to a soluble recombinant Mediator subunit, MED23, but not to several other Mediator components. One of these factors, hnRNP L, specifically interacts with MED23 in vitro and in vivo. Consistently, Mediator partially colocalizes with hnRNP L and the splicing machinery in the cell. Functionally, MED23 regulates a subset of hnRNP L-targeted alternative splicing (AS) and alternative cleavage and polyadenylation (APA) events, as shown by minigene reporters and exon array analysis. ChIP-seq analysis revealed that MED23 can regulate hnRNP L occupancy at their coregulated genes. Taken together, these results demonstrate a crosstalk between Mediator and the splicing machinery, providing a molecular basis for coupling mRNA processing to transcription.


Assuntos
Processamento Alternativo , Complexo Mediador/fisiologia , RNA Mensageiro/metabolismo , Animais , Células Cultivadas , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/fisiologia , Camundongos , Modelos Genéticos , Poliadenilação
18.
J Cell Mol Med ; 23(4): 2667-2677, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30746871

RESUMO

Osteoporosis has been shown to intensify bone loss caused by periodontitis and both share common risk factors. One strategy utilized to manage the disease has been via the release of Sr ions by Strontium Ranelate having a direct effect on preventing osteoclast activation and promoting osteoblast differentiation. Previously we have developed and characterized porous Sr-mesoporous bioactive glass (Sr-MBG) scaffolds and demonstrated their ability to promote periodontal regeneration when compared to MBG alone. Our group further discovered a splicing factor, heterogeneous nuclear ribonucleoprotein L (hnRNPL), was drastically down-regulated in periodontal ligament stem cells (PDLCs) stimulated by Sr through the activation of AKT pathway. Furthermore, hnRNPL restrained the osteogenic differentiation of PDLCs through down-regulating H3K36me3-specific methyltransferase Setd2. The goal of the present study was to investigate the mechanism of periodontal regeneration stimulated by Sr It was first found that the epigenetic mechanism of splicing factor hnRNPL participated in the osteogenesis processing of PDLCs stimulated by SrCl2 . Meanwhile, the different role of hnRNPL and SET domain containing 2 (Setd2) may provide some implication of the treatment of periodontitis patients simultaneously suffering from osteoporosis.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Histona-Lisina N-Metiltransferase/genética , Osteogênese/efeitos dos fármacos , Osteoporose/tratamento farmacológico , Periodontite/tratamento farmacológico , Células-Tronco/efeitos dos fármacos , Estrôncio/farmacologia , Animais , Materiais Biocompatíveis/química , Diferenciação Celular/efeitos dos fármacos , Preparações de Ação Retardada/química , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Vidro , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/metabolismo , Osteogênese/genética , Osteoporose/genética , Osteoporose/metabolismo , Osteoporose/patologia , Ovariectomia , Ligamento Periodontal/efeitos dos fármacos , Ligamento Periodontal/metabolismo , Ligamento Periodontal/patologia , Periodontite/genética , Periodontite/metabolismo , Periodontite/patologia , Poliuretanos/química , Ratos , Ratos Wistar , Regeneração/efeitos dos fármacos , Regeneração/genética , Transdução de Sinais , Células-Tronco/metabolismo , Células-Tronco/patologia , Alicerces Teciduais
19.
J Biol Chem ; 293(24): 9277-9291, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29720400

RESUMO

The adhesion protein carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is widely expressed in epithelial cells as a short cytoplasmic isoform (S-iso) and in leukocytes as a long cytoplasmic isoform (L-iso) and is frequently silenced in cancer by unknown mechanisms. Previously, we reported that interferon response factor 1 (IRF1) biases alternative splicing (AS) to include the variable exon 7 (E7) in CEACAM1, generating long cytoplasmic isoforms. We now show that IRF1 and a variant of heterogeneous nuclear ribonucleoprotein L (Lv1) coordinately silence the CEACAM1 gene. RNAi-mediated Lv1 depletion in IRF1-treated HeLa and melanoma cells induced significant CEACAM1 protein expression, reversed by ectopic Lv1 expression. The Lv1-mediated CEACAM1 repression resided in residues Gly71-Gly89 and Ala38-Gly89 in Lv1's N-terminal extension. ChIP analysis of IRF1- and FLAG-tagged Lv1-treated HeLa cells and global treatment with the global epigenetic modifiers 5-aza-2'-deoxycytidine and trichostatin A indicated that IRF1 and Lv1 together induce chromatin remodeling, restricting IRF1 access to the CEACAM1 promoter. In interferon γ-treated HeLa cells, the transcription factor SP1 did not associate with the CEACAM1 promoter, but binding by upstream transcription factor 1 (USF1), a known CEACAM1 regulator, was greatly enhanced. ChIP-sequencing revealed that Lv1 overexpression in IRF1-treated cells induces transcriptional silencing across many genes, including DCC (deleted in colorectal carcinoma), associated with CEACAM5 in colon cancer. Notably, IRF1, but not IRF3 and IRF7, affected CEACAM1 expression via translational repression. We conclude that IRF1 and Lv1 coordinately regulate CEACAM1 transcription, alternative splicing, and translation and may significantly contribute to CEACAM1 silencing in cancer.


Assuntos
Antígenos CD/genética , Moléculas de Adesão Celular/genética , Regulação para Baixo , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Fator Regulador 1 de Interferon/metabolismo , Processamento Alternativo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Células HeLa , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Regiões Promotoras Genéticas , Biossíntese de Proteínas , Isoformas de Proteínas/metabolismo , Ativação Transcricional
20.
Biochem Biophys Res Commun ; 516(3): 753-759, 2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31255281

RESUMO

The GAIT (gamma-interferon-activated inhibitor of translation) complex or miR-297-RISC (RNA-induced silencing complex), together with hnRNP L or hnRNP L-bearing complex, operates an RNA switch in myeloid cells that regulates stress-dependent expression of vascular endothelial growth factor-A (VEGFA). Here, we have shown that hnRNP L directs multiple hypoxia-inducible RNA switches simultaneously and regulates expression of these oncogenic genes in addition to VEGFA. Bioinformatic and polysome profiling-microarray screens have identified DNM1L (Dynamin 1-like) and PHF21A (PHD finger protein 21A) mRNAs as regulated at the translational level by GAIT-dependent, hnRNP L-directed RNA switches. We have also uncovered CDK6 (Cyclin dependent kinase 6), MKLN1 (Muskelin 1) and EIF5 (Eukaryotic initiation factor 5) as novel miR-297-dependent, hnRNP L-directed RNA switch transcripts. Src Kinase is required for the phosphorylation of hnRNP L and activation of the RNA switch pathway. Knockdown of hnRNP L sensitizes the human U937 monocytic cells under hypoxia stress but not in normoxia via inducing cell apoptosis partially due to the reduced translation of hnRNP L target mRNAs. Collectively, our findings suggest that commonly controlled genes by the hnRNP L-directed RNA switches form a translational regulon that promotes hypoxia resistance and cell survival.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Biossíntese de Proteínas , RNA/metabolismo , Regulon , Fator A de Crescimento do Endotélio Vascular/metabolismo , Apoptose/genética , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Hipóxia Celular , Dinaminas/genética , Dinaminas/metabolismo , Fator de Iniciação 5 em Eucariotos/genética , Fator de Iniciação 5 em Eucariotos/metabolismo , Perfilação da Expressão Gênica/métodos , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , RNA/genética , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células U937 , Fator A de Crescimento do Endotélio Vascular/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA