Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
1.
PLoS Biol ; 19(9): e3001378, 2021 09.
Article in English | MEDLINE | ID: mdl-34543262

ABSTRACT

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.


Subject(s)
Epidermal Cells/metabolism , Extracellular Matrix/metabolism , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Integrins/metabolism , Cell Differentiation , Cells, Cultured , Chromatin , Epidermis/growth & development , Extracellular Matrix/genetics , Humans , Integrins/genetics , Stem Cells
2.
J Nanobiotechnology ; 22(1): 62, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38360615

ABSTRACT

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.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Exosomes , Heterogeneous-Nuclear Ribonucleoprotein L , MicroRNAs , Humans , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Oxaliplatin/metabolism , Fusobacterium nucleatum/genetics , Fusobacterium nucleatum/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Colorectal Neoplasms/metabolism , Exosomes/metabolism , Chromatography, Liquid , Endoplasmic Reticulum Chaperone BiP , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Tandem Mass Spectrometry , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , MicroRNAs/metabolism , Cell Proliferation
3.
Nucleic Acids Res ; 50(22): 13026-13044, 2022 12 09.
Article in English | MEDLINE | ID: mdl-36533518

ABSTRACT

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.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein L , Muscle Development , Promoter Regions, Genetic , RNA, Long Noncoding , Transcription, Genetic , Humans , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transcription, Genetic/genetics , Gene Silencing , Protein Transport/genetics
4.
Neurobiol Dis ; 181: 106080, 2023 06 01.
Article in English | MEDLINE | ID: mdl-36925052

ABSTRACT

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.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein L , Ischemic Stroke , MicroRNAs , Stroke , Vascular Endothelial Growth Factor A , Animals , Humans , Mice , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor Alpha 2 Subunit/pharmacology , Endothelial Cells/metabolism , Glucose/metabolism , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Heterogeneous-Nuclear Ribonucleoprotein L/pharmacology , Infarction, Middle Cerebral Artery/metabolism , Ischemic Stroke/genetics , Ischemic Stroke/metabolism , MicroRNAs/metabolism , Neovascularization, Physiologic , RNA, Circular/genetics , RNA, Circular/metabolism , RNA, Circular/pharmacology , Stroke/genetics , Stroke/metabolism , Vascular Endothelial Growth Factor A/metabolism
5.
EMBO J ; 38(3)2019 02 01.
Article in English | MEDLINE | ID: mdl-30530525

ABSTRACT

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.


Subject(s)
3' Untranslated Regions/genetics , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Lymphoma, B-Cell/pathology , Nonsense Mediated mRNA Decay , RNA, Messenger/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Lymphoma, B-Cell/genetics , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcriptome
6.
Nucleic Acids Res ; 48(21): 12326-12335, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33231682

ABSTRACT

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.


Subject(s)
Alternative Splicing , Exons , Genetic Engineering/methods , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , RNA Precursors/genetics , RNA, Circular/genetics , Base Pairing , Gene Silencing , HEK293 Cells , HeLa Cells , Heterogeneous-Nuclear Ribonucleoprotein L/antagonists & inhibitors , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Humans , Immunoprecipitation/methods , Introns , Nucleic Acid Conformation , Nucleotide Motifs , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , RNA Precursors/metabolism , RNA, Catalytic/genetics , RNA, Catalytic/metabolism , RNA, Circular/biosynthesis , RNA, Circular/chemistry
7.
Molecules ; 27(16)2022 Aug 22.
Article in English | MEDLINE | ID: mdl-36014574

ABSTRACT

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.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein L , Pulmonary Fibrosis , Animals , Bleomycin/pharmacology , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Heterogeneous-Nuclear Ribonucleoprotein L/pharmacology , In Situ Hybridization, Fluorescence , Lung/metabolism , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , RNA/metabolism , Signal Transduction , Transforming Growth Factor beta1/metabolism
8.
J Cell Physiol ; 236(3): 2023-2035, 2021 03.
Article in English | MEDLINE | ID: mdl-32730662

ABSTRACT

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.


Subject(s)
Cytoplasm/metabolism , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Kidney Tubules/metabolism , Receptor, Melatonin, MT1/genetics , Animals , Cell Line , Circadian Rhythm/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Epithelial Cells/metabolism , Exoribonucleases/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Glomerulonephritis, Membranous/genetics , Glomerulonephritis, Membranous/pathology , Humans , Kidney Tubules/pathology , Mice, Inbred BALB C , Models, Biological , Open Reading Frames/genetics , Phosphorylation , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Melatonin, MT1/metabolism , Repetitive Sequences, Nucleic Acid/genetics , Up-Regulation/genetics
9.
J Virol ; 94(10)2020 05 04.
Article in English | MEDLINE | ID: mdl-32161169

ABSTRACT

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.


Subject(s)
5' Untranslated Regions , Foot-and-Mouth Disease Virus/physiology , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Internal Ribosome Entry Sites/physiology , RNA, Viral/biosynthesis , Virus Replication/physiology , Animals , Cell Line , Foot-and-Mouth Disease Virus/genetics , Gene Expression Regulation, Viral , Gene Knockout Techniques , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Host Microbial Interactions/physiology , Humans , Immunoprecipitation , Protein Binding , RNA, Viral/genetics , Transcriptome
10.
Mol Ther ; 28(10): 2220-2236, 2020 10 07.
Article in English | MEDLINE | ID: mdl-32592691

ABSTRACT

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.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Protein D-Aspartate-L-Isoaspartate Methyltransferase/genetics , Protein Kinase C-theta/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/metabolism , Cell-Penetrating Peptides/pharmacology , Forkhead Transcription Factors/genetics , Promoter Regions, Genetic , Protein Binding , Protein D-Aspartate-L-Isoaspartate Methyltransferase/metabolism , Protein Stability , Signal Transduction
11.
Mol Cell ; 49(5): 972-82, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-23394998

ABSTRACT

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.


Subject(s)
Exons , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , RNA, Small Nuclear/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism , Alternative Splicing , Base Sequence , Binding Sites , HEK293 Cells , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Humans , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , RNA Interference , RNA Splicing , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
RNA ; 24(6): 761-768, 2018 06.
Article in English | MEDLINE | ID: mdl-29581412

ABSTRACT

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.


Subject(s)
Exons , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , RNA Precursors/genetics , RNA Splicing , Repressor Proteins/metabolism , Animals , Base Sequence , Genome , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , High-Throughput Nucleotide Sequencing , Humans , Jurkat Cells , Mice , Repressor Proteins/genetics
13.
Mol Ther ; 27(2): 380-393, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30528088

ABSTRACT

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.


Subject(s)
CRISPR-Cas Systems/physiology , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , RNA, Long Noncoding/metabolism , Aged , Animals , Blotting, Northern , Blotting, Western , CRISPR-Cas Systems/genetics , Cell Line , Cell Movement/genetics , Cell Movement/physiology , Chromatin Immunoprecipitation , Chromatography, Liquid , Female , Fluorescent Antibody Technique , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Humans , Idiopathic Pulmonary Fibrosis/genetics , Immunoprecipitation , In Situ Hybridization , Male , Mice , Middle Aged , Models, Biological , Myofibroblasts/cytology , Myofibroblasts/metabolism , RNA, Long Noncoding/genetics , Rats , Real-Time Polymerase Chain Reaction
14.
Mol Cell ; 45(4): 459-69, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22264826

ABSTRACT

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.


Subject(s)
Alternative Splicing , Mediator Complex/physiology , RNA, Messenger/metabolism , Animals , Cells, Cultured , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Heterogeneous-Nuclear Ribonucleoprotein L/physiology , Mice , Models, Genetic , Polyadenylation
15.
J Cell Mol Med ; 23(4): 2667-2677, 2019 04.
Article in English | MEDLINE | ID: mdl-30746871

ABSTRACT

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.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Histone-Lysine N-Methyltransferase/genetics , Osteogenesis/drug effects , Osteoporosis/drug therapy , Periodontitis/drug therapy , Stem Cells/drug effects , Strontium/pharmacology , Animals , Biocompatible Materials/chemistry , Cell Differentiation/drug effects , Delayed-Action Preparations/chemistry , Disease Models, Animal , Female , Gene Expression Regulation , Glass , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Osteogenesis/genetics , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , Ovariectomy , Periodontal Ligament/drug effects , Periodontal Ligament/metabolism , Periodontal Ligament/pathology , Periodontitis/genetics , Periodontitis/metabolism , Periodontitis/pathology , Polyurethanes/chemistry , Rats , Rats, Wistar , Regeneration/drug effects , Regeneration/genetics , Signal Transduction , Stem Cells/metabolism , Stem Cells/pathology , Tissue Scaffolds
16.
J Biol Chem ; 293(24): 9277-9291, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29720400

ABSTRACT

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.


Subject(s)
Antigens, CD/genetics , Cell Adhesion Molecules/genetics , Down-Regulation , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Interferon Regulatory Factor-1/metabolism , Alternative Splicing , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Neoplasms/genetics , Neoplasms/metabolism , Promoter Regions, Genetic , Protein Biosynthesis , Protein Isoforms/metabolism , Transcriptional Activation
17.
Biochem Biophys Res Commun ; 516(3): 753-759, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31255281

ABSTRACT

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.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Protein Biosynthesis , RNA/metabolism , Regulon , Vascular Endothelial Growth Factor A/metabolism , Apoptosis/genetics , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Hypoxia , Dynamins/genetics , Dynamins/metabolism , Eukaryotic Initiation Factor-5/genetics , Eukaryotic Initiation Factor-5/metabolism , Gene Expression Profiling/methods , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , RNA/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , U937 Cells , Vascular Endothelial Growth Factor A/genetics
18.
Hepatology ; 68(5): 1817-1832, 2018 11.
Article in English | MEDLINE | ID: mdl-29790588

ABSTRACT

The identification of viability-associated long noncoding RNAs (lncRNAs) might be a promising rationale for new therapeutic approaches in liver cancer. Here, we applied an RNA interference screening approach in hepatocellular carcinoma (HCC) cell lines to find viability-associated lncRNAs. Among the multiple identified lncRNAs with a significant impact on HCC cell viability, we selected cancer susceptibility 9 (CASC9) due to the strength of its phenotype, expression, and up-regulation in HCC versus normal liver. CASC9 regulated viability across multiple HCC cell lines as shown by clustered regularly interspaced short palindromic repeats interference and single small interfering RNA (siRNA)-mediated and siRNA pool-mediated depletion of CASC9. Further, CASC9 depletion caused an increase in apoptosis and a decrease of proliferation. We identified the RNA binding protein heterogeneous nuclear ribonucleoprotein L (HNRNPL) as a CASC9 interacting protein by RNA affinity purification and validated it by native RNA immunoprecipitation. Knockdown of HNRNPL mimicked the loss-of-viability phenotype observed upon CASC9 depletion. Analysis of the proteome (stable isotope labeling with amino acids in cell culture) of CASC9-depleted and HNRNPL-depleted cells revealed a set of coregulated genes which implied a role of the CASC9:HNRNPL complex in AKT signaling and DNA damage sensing. CASC9 expression levels were elevated in patient-derived tumor samples compared to normal control tissue and had a significant association with overall survival of HCC patients. In a xenograft chicken chorioallantoic membrane model, we measured decreased tumor size after knockdown of CASC9. Conclusion: Taken together, we provide a comprehensive list of viability-associated lncRNAs in HCC; we identified the CASC9:HNRNPL complex as a clinically relevant viability-associated lncRNA/protein complex which affects AKT signaling and DNA damage sensing in HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Liver Neoplasms/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Chickens , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , RNA, Small Interfering , Signal Transduction
19.
Nucleic Acids Res ; 45(16): 9654-9678, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28934469

ABSTRACT

Inhibition of the Akt kinase activates HPV16 late gene expression by reducing HPV16 early polyadenylation and by activating HPV16 late L1 mRNA splicing. We identified 'hot spots' for RNA binding proteins at the early polyA signal and at splice sites on HPV16 late mRNAs. We observed that hnRNP L was associated with sequences at all HPV16 late splice sites and at the early polyA signal. Akt kinase inhibition resulted in hnRNP L dephosphorylation and reduced association of hnRNP L with HPV16 mRNAs. This was accompanied by an increased binding of U2AF65 and Sam68 to HPV16 mRNAs. Furthermore, siRNA knock-down of hnRNP L or Akt induced HPV16 gene expression. Treatment of HPV16 immortalized keratinocytes with Akt kinase inhibitor reduced hnRNP L binding to HPV16 mRNAs and induced HPV16 L1 mRNA production. Finally, deletion of the hnRNP L binding sites in HPV16 subgenomic expression plasmids resulted in activation of HPV16 late gene expression. In conclusion, the Akt kinase inhibits HPV16 late gene expression at the level of RNA processing by controlling the RNA-binding protein hnRNP L. We speculate that Akt kinase activity upholds an intracellular milieu that favours HPV16 early gene expression and suppresses HPV16 late gene expression.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Human papillomavirus 16/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Splicing , RNA, Viral/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Binding Sites , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Human papillomavirus 16/pathogenicity , Humans , Phosphorylation , Piperazines/pharmacology , Polyadenylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Pyrimidines/pharmacology , RNA Splice Sites , RNA, Messenger , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
20.
Nucleic Acids Res ; 45(13): 7950-7964, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28520992

ABSTRACT

MicroRNAs (miRNAs) and heterogeneous nuclear ribonucleoproteins (hnRNPs) are families of sequence-specific, posttranscriptional modulators of gene expression. Despite extensive mechanistic and functional studies on both regulatory classes, the interactions and crosstalk between them are largely unexplored. We have reported that competition between miR-297 and hnRNP L to bind a 3΄UTR-localized CA-rich element (CARE) of VEGFA mRNA regulates its translation. Here, we show that translation of VEGFA mRNA in human myeloid cells is dictated by a bi-directional interaction between miR-574-3p, a CA-rich microRNA, and hnRNP L. In normoxia, miR-574-3p, acting as a decoy, binds cytoplasmic hnRNP L and prevents its binding to the CARE and stimulation of VEGFA mRNA translation, simultaneously permitting miR-297-mediated translational silencing. However, in hypoxia, cytoplasmic accumulation of Tyr359-phosphorylated hnRNP L sequesters miR-574-3p, overcoming its decoy activity and seed sequence-dependent gene silencing activity. Ectopically expressed miR-574-3p binds multiple RNA recognition motif (RRM) domains of hnRNP L, synergizes with miR-297, reduces VEGFA mRNA translation, and triggers apoptosis, thereby suppressing tumorigenesis. Our studies establish a novel condition-dependent interplay between a miRNA and an hnRNP that regulates their functions in a bidirectional manner.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , 3' Untranslated Regions , Carboxypeptidases/antagonists & inhibitors , Carboxypeptidases/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Hypoxia/genetics , Cell Hypoxia/physiology , Cell Transformation, Neoplastic/genetics , Humans , Mutagenesis, Site-Directed , Myeloid Cells/cytology , Myeloid Cells/metabolism , Protein Biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL