Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Stem Cell Rev Rep ; 19(6): 1981-1993, 2023 08.
Article in English | MEDLINE | ID: mdl-37243830

ABSTRACT

Osteogeinc differentiation from mesenchymal stem cells (MSCs) into osteoblasts is a key step for bone tissue engineering in regenerative medicine. The insight into regulatory mechanism of osteogenesis of MSCs facilitates achieving better recovery effect. Long non-coding RNAs are regarded as a family of important moderators in osteogenesis. In this study, we found a novel lncRNA, lnc-PPP2R1B was up-regulated during osteogenesis of MSCs by Illumina HiSeq transcritome sequencing. We demonstrated lnc-PPP2R1B overexpression promoted osteogenesis and knockdown of lnc-PPP2R1B inhibited osteogenesis of MSCs. Mechanically, it physically interacted with and up-regulated heterogeneous nuclear ribonucleoprotein L Like (HNRNPLL), which is a master regulator of activation-induced alternative splicing in T cells. We found lnc-PPP2R1B knockdown or HNRNPLL knockdown decreased transcript-201 of Protein Phosphatase 2A, Regulatory Subunit A, Beta Isoform (PPP2R1B) while increased transcript-203 of PPP2R1B, and did not affect transcript-202/204/206. PPP2R1B is a constant regulatory subunit of protein phosphatase 2 (PP2A), which activates Wnt/ß-catenin pathway by removing phosphorylation and stabilization of ß-catenin and translocation into nucleus. The transcript-201 retained exon 2 and 3, compared to transcript-203. And it was reported the exon 2 and 3 of PPP2R1B were one part of B subunit binding domain on A subunit in PP2A trimer, and therefore retaining exon 2 and 3 promised formation and enzyme function of PP2A. Finally, lnc-PPP2R1B promoted ectopic osteogenesis in vivo. Conclusively, lnc-PPP2R1B mediated alternative splicing of PPP2R1B through retaining exon 2 and 3 by interacting with HNRNPLL and then promoted osteogenesis, which may facilitate an in-depth understanding of function and mechanism of lncRNAs in osteogenesis. Lnc-PPP2R1B interacted with HNRNPLL, and regulated alternative splicing of PPP2R1B through retaining exon 2 and 3, which preserved enzyme function of PP2A and enhanced dephosphorylation and nuclear translocation of ß-catenin, thereby promoting Runx2 and OSX expression and then osteogenesis. And it provided experimental data and potential target for promoting bone formation and bone regeneration.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein L , Mesenchymal Stem Cells , Alternative Splicing/genetics , beta Catenin/genetics , beta Catenin/metabolism , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Heterogeneous-Nuclear Ribonucleoprotein L/pharmacology , Mesenchymal Stem Cells/metabolism , Osteogenesis , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Protein Phosphatase 2/pharmacology , Humans
3.
J Exp Clin Cancer Res ; 42(1): 41, 2023 Feb 06.
Article in English | MEDLINE | ID: mdl-36747239

ABSTRACT

BACKGROUND: The response rate to immunotherapy in patients with bladder cancer (BCa) remains relatively low. Considering the stable existence and important functions in tumour metabolism, the role of circRNAs in regulating immune escape and immunotherapy sensitivity is receiving increasing attention. METHODS: Circular RNA (circRNA) sequencing was performed on five pairs of BCa samples, and circFAM13B (hsa_circ_0001535) was screened out because of its remarkably low expression in BCa. Further mRNA sequencing was conducted, and the association of circFAM13B with glycolysis process and CD8+ T cell activation was confirmed. The functions of circFAM13B were verified by proliferation assays, glycolysis assays, BCa cells-CD8+ T cell co-culture assays and tumorigenesis experiment among human immune reconstitution NOG mice. Bioinformatic analysis, RNA-protein pull down, mass spectrometry, RNA immunoprecipitation, luciferase reporter assay and fluorescence in situ hybridization were performed to validate the HNRNPL/circFAM13B/IGF2BP1/PKM2 cascade. RESULTS: Low expression of circFAM13B was observed in BCa, and it was positively associated with lower tumour stage and better prognosis among patients with BCa. The function of CD8+ T cells was promoted by circFAM13B, and it could attenuate the glycolysis of BCa cells and reverse the acidic tumour microenvironment (TME). The production of granzyme B and IFN-γ was improved, and the immunotherapy (PD-1 antibodies) sensitivity was facilitated by the inhibition of acidic TME. Mechanistically, circFAM13B was competitively bound to the KH3-4 domains of IGF2BP1 and subsequently reduced the binding of IGF2BP1 and PKM2 3'UTR. Thus, the stability of the PKM2 mRNA decreased, and glycolysis-induced acidic TME was inhibited. The generation of circFAM13B was explored by confirming whether heterogeneous nuclear ribonucleoprotein L (HNRNPL) could promote circFAM13B formation via pre-mRNA back-splicing. CONCLUSIONS: HNRNPL-induced circFAM13B could repress immune evasion and enhance immunotherapy sensitivity by inhibiting glycolysis and acidic TME in BCa through the novel circFAM13B/IGF2BP1/PKM2 cascade. Therefore, circFAM13B can be used as a biomarker for guiding the immunotherapy among patients with BCa.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein L , MicroRNAs , Urinary Bladder Neoplasms , Humans , Animals , Mice , MicroRNAs/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , In Situ Hybridization, Fluorescence , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/therapy , Urinary Bladder Neoplasms/metabolism , RNA, Circular/genetics , Glycolysis , RNA, Messenger/metabolism , Immunotherapy , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Tumor Microenvironment
4.
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
5.
Sci Adv ; 8(31): eabp9153, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35921415

ABSTRACT

Alternative splicing plays key roles for cell type-specific regulation of protein function. It is controlled by cis-regulatory RNA elements that are recognized by RNA binding proteins (RBPs). The MALT1 paracaspase is a key factor of signaling pathways that mediate innate and adaptive immune responses. Alternative splicing of MALT1 is critical for controlling optimal T cell activation. We demonstrate that MALT1 splicing depends on RNA structural elements that sequester the splice sites of the alternatively spliced exon7. The RBPs hnRNP U and hnRNP L bind competitively to stem-loop RNA structures that involve the 5' and 3' splice sites flanking exon7. While hnRNP U stabilizes RNA stem-loop conformations that maintain exon7 skipping, hnRNP L disrupts these RNA elements to facilitate recruitment of the essential splicing factor U2AF2, thereby promoting exon7 inclusion. Our data represent a paradigm for the control of splice site selection by differential RBP binding and modulation of pre-mRNA structure.


Subject(s)
Heterogeneous-Nuclear Ribonucleoprotein L , RNA Precursors , Alternative Splicing , Binding Sites , Exons , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Heterogeneous-Nuclear Ribonucleoprotein U/genetics , Heterogeneous-Nuclear Ribonucleoprotein U/metabolism , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/genetics , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , RNA Precursors/genetics , RNA Splice Sites , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
6.
Int J Biol Sci ; 18(13): 4824-4836, 2022.
Article in English | MEDLINE | ID: mdl-35982900

ABSTRACT

Long noncoding RNAs (lncRNAs) are dysregulated in many cancers. Here, we identified the molecular mechanisms of lncRNA Cancer Susceptibility Candidate 8 (CASC8) in promoting the malignancy of esophageal squamous cell carcinoma (ESCC). CASC8 was highly overexpressed in ESCC tissues and upregulation of CASC8 predicted poor prognosis in ESCC patients. Moreover, CASC8 decreased the cisplatin sensitivity of ESCC cells and promoted ESCC tumor growth in vivo. Mechanistically, CASC8 interacted with heterogeneous nuclear ribonucleoprotein L (hnRNPL) and inhibited its polyubiquitination and proteasomal degradation, thus stabilizing hnRNPL protein levels and activating the Bcl2/caspase3 pathway. Additionally, AlkB Homolog 5, RNA demethylase (ALKBH5)-mediated m6A demethylation stabilized the CASC8 transcript, resulting in CASC8 upregulation. Taken together, these findings identified an oncogenic function of CASC8 in the progression of ESCC, which suggest that CASC8 might become a potential prognostic biomarker in ESCC.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Heterogeneous-Nuclear Ribonucleoprotein L , RNA, Long Noncoding , Ribonucleoproteins , Cell Line, Tumor , Cell Proliferation/genetics , Drug Resistance, Neoplasm/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Gene Expression Regulation, Neoplastic/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Up-Regulation/genetics
7.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Article in English | MEDLINE | ID: mdl-34607953

ABSTRACT

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.


Subject(s)
Chemokine CXCL2/genetics , Neutrophil Infiltration/immunology , Neutrophils/immunology , RNA, Long Noncoding/genetics , A549 Cells , Animals , Cell Line, Tumor , Chemokine CXCL2/metabolism , Chromatin/metabolism , Epithelial Cells/metabolism , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Humans , Inflammation/prevention & control , Inflammation Mediators , Influenza A virus/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology , Pneumonia/immunology , Pneumonia/pathology , Pneumonia/virology , Promoter Regions, Genetic/genetics , RAW 264.7 Cells , Vesicular Stomatitis/immunology , Vesicular Stomatitis/pathology , Vesicular stomatitis Indiana virus/immunology
8.
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
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.
Nat Commun ; 10(1): 5787, 2019 12 19.
Article in English | MEDLINE | ID: mdl-31857580

ABSTRACT

Emerging evidence supports roles of enhancer RNAs (eRNAs) in regulating target gene. Here, we study eRNA regulation and function during skeletal myoblast differentiation. We provide a panoramic view of enhancer transcription and categorization of eRNAs. Master transcription factor MyoD is crucial in activating eRNA production. Super enhancer (se) generated seRNA-1 and -2 promote myogenic differentiation in vitro and in vivo. seRNA-1 regulates expression levels of two nearby genes, myoglobin (Mb) and apolipoprotein L6 (Apol6), by binding to heterogeneous nuclear ribonucleoprotein L (hnRNPL). A CAAA tract on seRNA-1 is essential in mediating seRNA-1/hnRNPL binding and function. Disruption of seRNA-1-hnRNPL interaction attenuates Pol II and H3K36me3 deposition at the Mb locus, in coincidence with the reduction of its transcription. Furthermore, analyses of hnRNPL binding transcriptome-wide reveal its association with eRNAs is a general phenomenon in multiple cells. Collectively, we propose that eRNA-hnRNPL interaction represents a mechanism contributing to target mRNA activation.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Muscle Development/genetics , MyoD Protein/metabolism , RNA, Messenger/genetics , Animals , Cell Differentiation/genetics , Cell Line , Male , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/growth & development , Myoblasts/physiology , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , Transcription, Genetic
11.
Biomed Res Int ; 2019: 9612425, 2019.
Article in English | MEDLINE | ID: mdl-31828152

ABSTRACT

BACKGROUND AND OBJECTIVES: Alternative splicing is increasingly associated with cancers. HnRNP L is a splicing factor that promotes carcinogenesis in head and neck squamous cell carcinoma (HNSCC) and other cancers. Alternative exon 7 of hnRNP L contains an in-frame stop codon. Exon 7-included transcripts can be degraded via nonsense-mediated decay or encode a truncated hnRNP L protein. Exon 7-excluded transcripts can encode full-length functional hnRNP L protein. HnRNP L has an autoregulation mechanism by promoting the inclusion of its own exon 7. This study aimed to understand the relationship between the alternative splicing of exon 7 and HNSCC. Oncogenic splicing factor SRSF3 has an alternative exon 4 and similar autoregulation mechanism. HnRNP L promotes SRSF3 exon 4 inclusion and then inhibits SRSF3 autoregulation. MATERIALS AND METHODS: The relationship between alternative splicing of hnRNP L exon 7 and clinical characteristics of HNSCC in a TCGA dataset was analyzed and confirmed by RT-PCR in a cohort of 61 oral squamous cell carcinoma (OSCC) patients. The regulators of exon 7 splicing were screened in 29 splicing factors and confirmed by overexpression or silencing assay in HEK 293, CAL 27, and SCC-9 cell lines. RESULTS: The inclusion of hnRNP L exon 7 was significantly negatively associated with the progression and prognosis of HNSCC, which was confirmed in the cohort of 61 OSCC patients. SRSF3 inhibited exon 7 inclusion and hnRNP L autoregulation and then promoted the expression of full-length functional hnRNP L protein. SRSF3 exon 4 inclusion was correlated with hnRNP L exon 7 inclusion in both HNSCC and breast cancer. HNSCC patients with both low hnRNP L exon 7 and SRSF3 exon 4 inclusion show poor overall survival. CONCLUSIONS: Inclusion of hnRNP L alternative exon 7 is associated with good prognosis and inhibited by oncogene SRSF3 in HNSCC.


Subject(s)
Head and Neck Neoplasms , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Serine-Arginine Splicing Factors/genetics , Squamous Cell Carcinoma of Head and Neck , Alternative Splicing/genetics , Disease Progression , Exons/genetics , HEK293 Cells , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/mortality , Heterogeneous-Nuclear Ribonucleoprotein L/antagonists & inhibitors , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Humans , Mouth Neoplasms/diagnosis , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/mortality , Prognosis , Serine-Arginine Splicing Factors/metabolism , Squamous Cell Carcinoma of Head and Neck/diagnosis , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/mortality
12.
Cell Death Dis ; 10(8): 550, 2019 07 18.
Article in English | MEDLINE | ID: mdl-31320608

ABSTRACT

Although oxaliplatin is an effective chemotherapeutic drug for treatment of colorectal cancer (CRC), tumor cells can develop mechanisms to evade oxaliplatin-induced cell death and show high tolerance and acquired resistance to this drug. Heterogeneous nuclear ribonucleoprotein L (hnRNP L) has been proved to play a critical role in DNA repair during IgH class switch recombination (CSR) in B lymphocytes, while, its role in CRC and chemotherapeutic resistance remain unknown. Our study aims to uncover an unidentified mechanism of regulating DNA double-strand breaks (DSBs) by hnRNP L in CRC cells treated by oxaliplatin. In present study, we observed that knockdown of hnRNP L enhanced the level of DNA breakage and sensitivity of CRC cells to oxaliplatin. The expression of key DNA repair factors (BRCA1, 53BP1, and ATM) was unaffected by hnRNP L knockdown, thereby excluding the likelihood of hnRNP L mediation via mRNA regulation. Moreover, we observed that phosphorylation level of ATM changed oppositely to 53BP1 and BRCA1 in the CRC cells (SW620 and HCT116) which exhibit synergistic effect by oxaliplatin plus hnRNP L impairment. And similar phenomenon was observed in the foci formation of these critical repair factors. We also found that hnRNP L binds directly with these DNA repair factors through its RNA-recognition motifs (RRMs). Analysis of cell death indicated that the RRMs of hnRNP L are required for cell survival under incubation with oxaliplatin. In conclusion, hnRNP L is critical for the recruitment of the DNA repair factors in oxaliplatin-induced DSBs. Targeting hnRNP L is a promising new clinical approach that could enhance the effectiveness of current chemotherapeutic treatment in patients with resistance to oxaliplatin.


Subject(s)
Antineoplastic Agents/pharmacology , BRCA1 Protein/metabolism , Colorectal Neoplasms/metabolism , DNA Breaks, Double-Stranded , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Oxaliplatin/pharmacology , Tumor Suppressor p53-Binding Protein 1/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Ataxia Telangiectasia Mutated Proteins/chemistry , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/chemistry , BRCA1 Protein/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , DNA Repair/genetics , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , HCT116 Cells , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Humans , Phosphorylation , Protein Binding , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Tumor Suppressor p53-Binding Protein 1/chemistry , Tumor Suppressor p53-Binding Protein 1/genetics
13.
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
14.
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
15.
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
16.
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
17.
Mol Cell Biol ; 38(12)2018 06 15.
Article in English | MEDLINE | ID: mdl-29610151

ABSTRACT

The pituitary-derived somatolactotrophe GH3 cells secrete both growth hormone (GH) and prolactin (PRL). We have found that the hnRNP L and L-like (LL) paralogs differentially regulate alternative splicing of genes in these cells. Here, we show that hnRNP L is essential for PRL only, but LL is essential for both PRL and GH production. Transcriptome-wide RNA sequencing (RNA-Seq) analysis indicates that they differentially control groups of hormone or hormone-related genes involved in hormone production/regulation at total transcript and alternative exon levels. Interestingly, hnRNP L also specifically binds and prevents the aberrant usage of a nonconserved CA-rich intron piece of Prl pre-mRNA transcripts, and many others involved in endocrine functions, to prevent mostly cryptic last exons and mRNA truncation. Essential for the full hnRNP L effect on specific exons is a proline-rich region that emerged during evolution in vertebrate hnRNP L only but not LL. Together, our data demonstrate that the hnRNP L and its paralog, LL, differentially control hormone gene expression programs at multiple levels, and hnRNP L in particular is critical for protecting the transcriptome from aberrant usage of intronic sequences. The multilevel differential control by hnRNPs likely tailors the transcriptome to help refine and safeguard the different gene expression programs for different hormones.


Subject(s)
Gene Expression Regulation/genetics , Growth Hormone/biosynthesis , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Lactotrophs/metabolism , Prolactin/biosynthesis , Somatotrophs/metabolism , Alternative Splicing/genetics , Amino Acid Sequence/genetics , Cell Differentiation/genetics , Cell Line , HEK293 Cells , HeLa Cells , Humans , Pituitary Gland/cytology , Pituitary Gland/metabolism , RNA Interference , RNA, Small Interfering/genetics , Transcriptome/genetics
18.
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
19.
RNA ; 24(3): 324-331, 2018 03.
Article in English | MEDLINE | ID: mdl-29263134

ABSTRACT

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.


Subject(s)
Alternative Splicing , Gene Expression Regulation/genetics , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Nonsense Mediated mRNA Decay/genetics , Ribonucleoproteins/genetics , 3' Untranslated Regions/genetics , Exons , Genes, Reporter , HeLa Cells , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/physiology , Heterogeneous-Nuclear Ribonucleoprotein L/genetics , Homeostasis , Humans , RNA, Messenger/genetics , Ribonucleoproteins/physiology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...