Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cells ; 13(8)2024 Apr 14.
Article in English | MEDLINE | ID: mdl-38667295

ABSTRACT

Decorin (DCN), a member of the small leucine-rich proteoglycan gene family, is secreted from stromal fibroblasts with non-cell-autonomous anti-breast-cancer effects. Therefore, in the present study, we sought to elucidate the function of decorin in breast stromal fibroblasts (BSFs). We first showed DCN downregulation in active cancer-associated fibroblasts (CAFs) compared to their adjacent tumor counterpart fibroblasts at both the mRNA and protein levels. Interestingly, breast cancer cells and the recombinant IL-6 protein, both known to activate fibroblasts in vitro, downregulated DCN in BSFs. Moreover, specific DCN knockdown in breast fibroblasts modulated the expression/secretion of several CAF biomarkers and cancer-promoting proteins (α-SMA, FAP- α, SDF-1 and IL-6) and enhanced the invasion/proliferation abilities of these cells through activation of the STAT3/AUF1 signaling. Furthermore, DCN-deficient fibroblasts promoted the epithelial-to-mesenchymal transition and stemness processes in BC cells in a paracrine manner, which increased their resistance to cisplatin. These DCN-deficient fibroblasts also enhanced angiogenesis and orthotopic tumor growth in mice in a paracrine manner. On the other hand, ectopic expression of DCN in CAFs suppressed their active features and their paracrine pro-carcinogenic effects. Together, the present findings indicate that endogenous DCN suppresses the pro-carcinogenic and pro-metastatic effects of breast stromal fibroblasts.


Subject(s)
Breast Neoplasms , Cancer-Associated Fibroblasts , Decorin , Down-Regulation , Interleukin-6 , STAT3 Transcription Factor , Signal Transduction , Decorin/metabolism , Decorin/genetics , Humans , STAT3 Transcription Factor/metabolism , Female , Interleukin-6/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/genetics , Mice , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Down-Regulation/genetics , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Fibroblasts/metabolism , Stromal Cells/metabolism , Cell Line, Tumor , Carcinogenesis/pathology , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Proliferation , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Breast/pathology , Breast/metabolism
2.
Mol Carcinog ; 63(5): 977-990, 2024 May.
Article in English | MEDLINE | ID: mdl-38376344

ABSTRACT

Nickel pollution is a recognized factor contributing to lung cancer. Understanding the molecular mechanisms of its carcinogenic effects is crucial for lung cancer prevention and treatment. Our previous research identified the downregulation of a long noncoding RNA, maternally expressed gene 3 (MEG3), as a key factor in transforming human bronchial epithelial cells (HBECs) into malignant cells following nickel exposure. In our study, we found that deletion of MEG3 also reduced the expression of RhoGDIß. Notably, artificially increasing RhoGDIß levels counteracted the malignant transformation caused by MEG3 deletion in HBECs. This indicates that the reduction in RhoGDIß contributes to the transformation of HBECs due to MEG3 deletion. Further exploration revealed that MEG3 downregulation led to enhanced c-Jun activity, which in turn promoted miR-200c transcription. High levels of miR-200c subsequently increased the translation of AUF1 protein, stabilizing SOX2 messenger RNA (mRNA). This stabilization affected the regulation of miR-137, SP-1 protein translation, and the suppression of RhoGDIß mRNA transcription and protein expression, leading to cell transformation. Our study underscores the co-regulation of RhoGDIß expression by long noncoding RNA MEG3, multiple microRNAs (miR-200c and miR-137), and RNA-regulated transcription factors (c-Jun, SOX2, and SP1). This intricate network of molecular events sheds light on the nature of lung tumorigenesis. These novel findings pave the way for developing targeted strategies for the prevention and treatment of human lung cancer based on the MEG3/RhoGDIß pathway.


Subject(s)
Lung Neoplasms , MicroRNAs , RNA, Long Noncoding , Humans , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Down-Regulation , Epithelial Cells/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Nickel , rho Guanine Nucleotide Dissociation Inhibitor beta/antagonists & inhibitors , rho Guanine Nucleotide Dissociation Inhibitor beta/genetics , rho Guanine Nucleotide Dissociation Inhibitor beta/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger , SOXB1 Transcription Factors/genetics , Heterogeneous Nuclear Ribonucleoprotein D0/genetics , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism
4.
Open Biol ; 13(11): 230158, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37989221

ABSTRACT

Alpha-synuclein (SNCA) accumulation plays a central role in the pathogenesis of Parkinson's disease. Determining and interfering with the mechanisms that control SNCA expression is one approach to limiting disease progression. Currently, most of our understanding of SNCA regulation is protein-based. Post-transcriptional mechanisms directly regulating SNCA mRNA expression via its 3' untranslated region (3'UTR) were investigated here. Mass spectrometry of proteins pulled down from murine brain lysates using a biotinylated SNCA 3'UTR revealed multiple RNA-binding proteins, of which HNRNPD/AUF1 was chosen for further analysis. AUF1 bound both proximal and distal regions of the SNCA 3'UTR, but not the 5'UTR or CDS. In the nucleus, AUF1 attenuated SNCA pre-mRNA maturation and was indispensable for the export of SNCA transcripts. AUF1 destabilized SNCA transcripts in the cytosol, primarily those with shorter 3'UTRs, independently of microRNAs by recruiting the CNOT1-CNOT7 deadenylase complex to trim the polyA tail. Furthermore, AUF1 inhibited SNCA mRNA binding to ribosomes. These data identify AUF1 as a multi-tasking protein regulating maturation, nucleocytoplasmic shuttling, stability and translation of SNCA transcripts.


Subject(s)
RNA-Binding Proteins , Mice , Animals , 3' Untranslated Regions , Active Transport, Cell Nucleus , Heterogeneous Nuclear Ribonucleoprotein D0/genetics , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Protein Binding
5.
Cancer Sci ; 114(4): 1582-1595, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36632741

ABSTRACT

Cervical squamous cell carcinoma (CSCC) is one of the leading causes of cancer death in women worldwide. Patients with advanced cervical carcinoma always have a poor prognosis once resistant to cisplatin due to the lack of effective treatment. It is urgent to investigate the molecular mechanisms of cisplatin resistance. Circular RNAs (circRNAs) are known to exert their regulatory functions in a series of malignancies. However, their effects on CSCC remain to be elucidated. Here, we found that cytoplasmic circARHGAP5, derived from second and third exons of the ARHGAP5 gene, was downregulated in cisplatin-resistant tissues compared with normal cervix tissues and untreated cervical cancer tissues. In addition, experiments from overexpression/knockdown cell lines revealed that circARHGAP5 could inhibit cisplatin-mediated cell apoptosis in CSCC cells both in vitro and in vivo. Mechanistically, circARHGAP5 interacted with AU-rich element RNA-binding protein (AUF1) directly. Overexpression of AUF1 could also inhibit cell apoptosis mediated by cisplatin. Furthermore, we detected the potential targets of AUF1 related to the apoptotic pathway and found that bcl-2-like protein 11 (BIM) was not only negatively regulated by AUF1 but positively regulated by circARHGAP5, which indicated that BIM mRNA might be degraded by AUF1 and thereby inhibited tumor cell apoptosis. Collectively, our data indicated that circARHGAP5 directly bound to AUF1 and prevented AUF1 from interacting with BIM mRNA, thereby playing a pivotal role in cisplatin resistance in CSCC. Our study provides insights into overcoming cancer resistance to cisplatin treatment.


Subject(s)
Carcinoma, Squamous Cell , Heterogeneous Nuclear Ribonucleoprotein D0 , RNA, Circular , Uterine Cervical Neoplasms , Female , Humans , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Cell Proliferation , Cisplatin/pharmacology , GTPase-Activating Proteins/genetics , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , RNA, Circular/genetics , RNA, Messenger/metabolism , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
6.
Biomed Res Int ; 2022: 8610467, 2022.
Article in English | MEDLINE | ID: mdl-36246972

ABSTRACT

Coxsackievirus B (CVB) 3C protease (3Cpro) plays a specific cleavage role on AU-rich binding factor (AUF1, also called hnRNP D), which consequently disputes the regulation of AUF1 on downstream molecules. In our study, the iTRAQ approach was first used to quantify the differentially expressed cellular proteins in AUF1-overexpressing HeLa cells, which provides straightforward insight into the role of AUF1 during viral infection. A total of 1,290 differentially expressed proteins (DEPs), including 882 upregulated and 408 downregulated proteins, were identified. The DEPs are involved in a variety of cellular processes via GO terms, protein-protein interactions, and a series of further bioinformatics analyses. Among the DEPs, some demonstrated important roles in cellular metabolism. In particular, DDX5 was further verified to be negatively regulated by AUF1 and increased in CVB-infected cells, which in turn promoted CVB replication. These findings provide potential novel ideas for exploring new antiviral therapy targets.


Subject(s)
DEAD-box RNA Helicases , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D , Proteomics , Antiviral Agents , DEAD-box RNA Helicases/metabolism , Enterovirus B, Human/metabolism , HeLa Cells , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Heterogeneous-Nuclear Ribonucleoprotein D/genetics , Humans , Virus Replication
7.
J Biol Chem ; 298(10): 102437, 2022 10.
Article in English | MEDLINE | ID: mdl-36041631

ABSTRACT

Mammalian target of rapamycin (mTOR), which is part of mTOR complex 1 (mTORC1) and mTORC2, controls cellular metabolism in response to levels of nutrients and other growth signals. A hallmark of mTORC2 activation is the phosphorylation of Akt, which becomes upregulated in cancer. How mTORC2 modulates Akt phosphorylation remains poorly understood. Here, we found that the RNA-binding protein, AUF1 (ARE/poly(U)-binding/degradation factor 1), modulates mTORC2/Akt signaling. We determined that AUF1 is required for phosphorylation of Akt at Thr308, Thr450, and Ser473 and that AUF1 also mediates phosphorylation of the mTORC2-modulated metabolic enzyme glutamine fructose-6-phosphate amidotransferase 1 at Ser243. In addition, AUF1 immunoprecipitation followed by quantitative RT-PCR revealed that the mRNAs of Akt, glutamine fructose-6-phosphate amidotransferase 1, and the mTORC2 component SIN1 associate with AUF1. Furthermore, expression of the p40 and p45, but not the p37 or p42, isoforms of AUF1 specifically mediate Akt phosphorylation. In the absence of AUF1, subcellular fractionation indicated that Akt fails to localize to the membrane. However, ectopic expression of a membrane-targeted allele of Akt is sufficient to allow Akt-Ser473 phosphorylation despite AUF1 depletion. Finally, conditions that enhance mTORC2 signaling, such as acute glutamine withdrawal, augment AUF1 phosphorylation, whereas mTOR inhibition abolishes AUF1 phosphorylation. Our findings unravel a role for AUF1 in promoting membrane localization of Akt to facilitate its phosphorylation on this cellular compartment. Targeting AUF1 could have therapeutic benefit for cancers with upregulated mTORC2/Akt signaling.


Subject(s)
Heterogeneous Nuclear Ribonucleoprotein D0 , Proto-Oncogene Proteins c-akt , RNA-Binding Proteins , Cell Proliferation , Glutamine/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Humans , Heterogeneous Nuclear Ribonucleoprotein D0/genetics , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Cell Membrane/metabolism , Glutamine-Fructose-6-Phosphate Transaminase (Isomerizing)/metabolism
8.
Cell Mol Life Sci ; 79(5): 228, 2022 Apr 07.
Article in English | MEDLINE | ID: mdl-35391558

ABSTRACT

BACKGROUND: The AU-rich element (ARE)-binding factor 1 (AUF1) acts as a switch for septic shock, although its underlying mechanisms remain largely unknown. In this study, we examined the biological significance and potential molecular mechanism of AUF1 in regulating ferroptosis in sepsis-induced acute lung injury (ALI). METHODS: Alveolar epithelial cells (AECs) challenged with ferroptosis-inducing compounds and cecum ligation and puncture (CLP)-induced ALI were used as the in vitro and in vivo model, respectively. The stability of AUF1 and its degradation by ubiquitin-proteasome pathway were examined by cycloheximide chase analysis and co-immunoprecipitation assay. The regulation of AUF1 on nuclear factor E2-related factor 2 (NRF2) and activation transcription factor 3 (ATF3) was explored by RNA immunoprecipitation (RIP), RNA pull-down, and mRNA stability assays. Functionally, the effects of altering AUF1, NRF2 or ATF3 on ferroptosis in AECs or ALI mice were evaluated by measuring cell viability, lipid peroxidation, iron accumulation, and total glutathione level. RESULTS: AUF1 was down-regulated in AECs challenged with ferroptosis-inducing compounds, both on mRNA and protein levels. The E3 ubiquitin ligase FBXW7 was responsible for protein degradation of AUF1 during ferroptosis. By up-regulating NRF2 and down-regulating ATF3, AUF1 antagonized ferroptosis in AECs in vitro. In the CLP-induced ALI model, the survival rate of AUF1 knockout mice was significantly reduced and the lung injuries were aggravated, which were related to the enhancement of lung ferroptosis. CONCLUSIONS: FBXW7 mediates the ubiquitination and degradation of AUF1 in ferroptosis. AUF1 antagonizes ferroptosis by regulating NRF2 and ATF3 oppositely. Activating AUF1 pathway may be beneficial to the treatment of sepsis-induced ALI.


Subject(s)
Activating Transcription Factor 3 , Acute Lung Injury , Ferroptosis , Heterogeneous Nuclear Ribonucleoprotein D0 , NF-E2-Related Factor 2 , Sepsis , Activating Transcription Factor 3/metabolism , Acute Lung Injury/chemically induced , Acute Lung Injury/prevention & control , Animals , F-Box-WD Repeat-Containing Protein 7/metabolism , Heterogeneous Nuclear Ribonucleoprotein D0/genetics , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Mice , NF-E2-Related Factor 2/metabolism , RNA , Sepsis/complications
9.
Carcinogenesis ; 42(12): 1439-1448, 2021 12 31.
Article in English | MEDLINE | ID: mdl-34718441

ABSTRACT

Active breast cancer-associated fibroblasts (CAFs), the most influential cells in breast tumor microenvironment, express/secrete high levels of the proinvasive/metastatic interleukin-6 (IL-6). Therefore, we have tested here the effect of the IL-6 receptor (IL-6R) inhibitor tocilizumab (TCZ; Actemra) on different active breast CAFs. We have shown that TCZ potently and persistently suppresses the expression of various CAF biomarkers, namely α-SMA, SDF-1 as well as the STAT3 pathway and its downstream target AUF1. TCZ also inhibited the proliferation, migration and invasion abilities of active breast CAF cells. Additionally, TCZ repressed the ability of CAF cells in promoting epithelial-to-mesenchymal transition, and enhancing the migratory/invasive and proliferative capacities of breast cancer cells in vitro. Importantly, these findings were confirmed in orthotopic humanized breast tumors in mice. Furthermore, TCZ suppressed the expression of the pro-angiogenic factor VEGF-A and its transactivator HIF-1α in CAF cells, and consequently inhibited the angiogenic-promoting effect of active CAFs both in vitro and in orthotopic tumor xenografts. These results indicate that inhibition of the IL-6/STAT3/AUF1 pathway by TCZ can normalize active breast CAFs and suppress their paracrine pro-carcinogenic effects, which paves the way toward development of specific CAF-targeting therapy, badly needed for more efficient breast cancer treatments.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Heterogeneous Nuclear Ribonucleoprotein D0/genetics , Humans , Interleukin-6/metabolism , Receptors, Interleukin-6/antagonists & inhibitors , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
10.
Am J Drug Alcohol Abuse ; 47(5): 581-589, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34407719

ABSTRACT

BACKGROUND: The functional mechanism is unknown for many genetic variants associated with substance use disorder phenotypes. Rs678849, an intronic variant in the delta-opioid receptor gene (OPRD1), has been found to predict regional brain volume, addiction risk, and the efficacy of buprenorphine/naloxone in treating opioid use disorder. The variant has also been implicated as an expression quantitative trait locus (eQTL) for several genes. OBJECTIVES: The objective of this study was to identify functional differences between the two alleles of rs678849 in vitro. We hypothesized that the two alleles of rs678849 would have different effects on transcriptional activity due to differential interactions with transcription factors. METHODS: 15bp regions containing the C or T alleles of rs678849 were cloned into luciferase constructs and transfected into BE(2)C neuroblastoma cells to test the effect on transcription. Electrophoretic mobility shift assays (EMSA) using nuclear lysates from BE(2)C cell or human postmortem medial prefrontal cortex were used to identify proteins that differentially bound the two alleles. RESULTS: At 24 hours post-transfection, the C allele construct had significantly lower luciferase expression than the T allele construct and empty vector control (ANOVA p < .001). Proteomic analysis and supershift assays identified XRCC6 as a transcription factor specifically binding the C allele, whereas hnRNP D0 was found to specifically bind the T allele. CONCLUSION: These functional differences between the C and T alleles may help explain the psychiatric and neurological phenotype differences predicted by rs678849 genotype and the potential role of the variant as an eQTL.


Subject(s)
Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Ku Autoantigen/metabolism , Pharmacogenomic Variants , Receptors, Opioid, delta/genetics , Transcription Factors/metabolism , Alleles , Electrophoretic Mobility Shift Assay , Genotype , Humans , Luciferases, Firefly , Protein Binding/genetics , Quantitative Trait Loci/genetics
11.
J Mol Cell Cardiol ; 160: 111-120, 2021 11.
Article in English | MEDLINE | ID: mdl-34302813

ABSTRACT

Long non-coding RNAs (lncRNAs) have emerged as critical regulators in human disease including atherosclerosis. However, the mechanisms involved in the post-transcriptional regulation of the expression of disease-associated lncRNAs are not fully understood. Gene expression studies revealed that Nuclear Paraspeckle Assembly Transcript 1 (NEAT1) lncRNA expression was increased by >2-fold in peripheral blood mononuclear cells (PBMCs) derived from patients with coronary artery disease (CAD) or in carotid artery atherosclerotic plaques. We observed a linear association between NEAT1 lncRNA expression and prevalence of CAD which was independent of age, sex, cardiovascular traditional risk factors and renal function. NEAT1 expression was induced by TNF-α, while silencing of NEAT1 profoundly attenuated the TNF-α-induced vascular endothelial cell pro-inflammatory response as defined by the expression of CXCL8, CCL2, VCAM1 and ICAM1. Overexpression of the RNA editing enzyme adenosine deaminase acting on RNA-1 (ADAR1), but not of its editing-deficient mutant, upregulated NEAT1 levels. Conversely, silencing of ADAR1 suppressed the basal levels and the TNF-α-induced increase of NEAT1. NEAT1 lncRNA expression was strongly associated with ADAR1 in CAD and peripheral arterial vascular disease. RNA editing mapping studies revealed the presence of several inosines in close proximity to AU-rich elements within the AluSx3+/AluJo- double-stranded RNA complex. Silencing of the stabilizing RNA-binding protein AUF1 reduced NEAT1 levels while silencing of ADAR1 profoundly affected the binding capacity of AUF1 to NEAT1. Together, our findings propose a mechanism by which ADAR1-catalyzed A-to-I RNA editing controls NEAT1 lncRNA stability in ASCVD.


Subject(s)
Adenosine/metabolism , Alu Elements/genetics , Atherosclerosis/blood , Coronary Artery Disease/blood , Inosine/metabolism , Plaque, Atherosclerotic/blood , RNA Editing/genetics , RNA Stability/genetics , RNA, Long Noncoding/metabolism , Signal Transduction/genetics , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Adult , Aged , Aged, 80 and over , Atherosclerosis/genetics , Binding Sites , Cells, Cultured , Cohort Studies , Coronary Artery Disease/genetics , Female , Gene Silencing , Heterogeneous Nuclear Ribonucleoprotein D0/genetics , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Plaque, Atherosclerotic/genetics , RNA, Long Noncoding/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Transfection
12.
Cell Death Dis ; 12(4): 321, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33767152

ABSTRACT

Long noncoding RNAs (lncRNAs) play an essential role in tumor progression. Few researches focused on the clinical and biological relevance of lncRNAs in peripheral T cell lymphoma (PTCL). In this research, a novel lncRNA (ENST00000503502) was identified overexpressed in the main subtypes of PTCL, and designated as T cell lymphoma-associated lncRNA1 (TCLlnc1). Serum TCLlnc1 was associated with extranodal involvement, high-risk International Prognostic Index, and poor prognosis of the patients. Both in vitro and in vivo, overexpression of TCLlnc1 promoted T-lymphoma cell proliferation and migration, both of which were counteracted by the knockdown of TCLlnc1 using small interfering RNAs. As the mechanism of action, TCLlnc1 directly interacted with transcription activator heterogeneous nuclear ribonucleoprotein D (HNRNPD) and Y-box binding protein-1 (YBX1) by acting as a modular scaffold. TCLlnc1/HNRNPD/YBX1 complex upregulated transcription of TGFB2 and TGFBR1 genes, activated the tumor growth factor-ß signaling pathway, resulting in lymphoma progression, and might be a potential target in PTCL.


Subject(s)
Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Lymphoma, T-Cell, Peripheral/metabolism , RNA, Long Noncoding/metabolism , Y-Box-Binding Protein 1/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Disease Progression , Female , HEK293 Cells , Heterogeneous Nuclear Ribonucleoprotein D0/genetics , Humans , Jurkat Cells , Lymphoma, T-Cell, Peripheral/blood , Lymphoma, T-Cell, Peripheral/genetics , Lymphoma, T-Cell, Peripheral/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , RNA, Long Noncoding/blood , RNA, Long Noncoding/genetics , Y-Box-Binding Protein 1/genetics
13.
Cell Death Dis ; 12(2): 153, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542193

ABSTRACT

Studies have found that RNA-binding proteins (RBPs) are dysfunctional and play a significant regulatory role in the development of glioma. Based on The Cancer Genome Atlas database and the previous studies, we selected heterogeneous nuclear ribonucleoprotein (HNRNPD) as the research candidate and sought its downstream targeted genes. In the present study, HNRNPD, linc00707, and specific protein 2 (SP2) were highly expressed, while zinc fingers and homeboxes 2 (ZHX2) and miR-651-3p were remarkedly downregulated in glioma tissues and cells. HNRNPD, linc00707, and SP2 knockdown or ZHX2 and miR-651-3p overexpression suppressed glioma cells proliferation, migration, and invasion and vasculogenic mimicry (VM) formation. Knockdown of HNRNPD increased the stability of ZHX2 mRNA. ZHX2 bound to the promoter region of linc00707 and negatively regulate its expression. Linc00707 could bind with miR-651-3p, while miR-651-3p bound to the 3' untranslated region (3'UTR) of SP2 mRNA to negatively regulate its expression. The transcription factor SP2 directly bound to the promoter regions of the VM formation-related proteins MMP2, MMP9, and VE-cadherin, playing a role in promoting transcription in order to regulate the VM formation ability of glioma cells.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Homeodomain Proteins/metabolism , MicroRNAs/metabolism , Molecular Mimicry , Neovascularization, Pathologic , RNA, Long Noncoding/metabolism , Sp2 Transcription Factor/metabolism , Transcription Factors/metabolism , 3' Untranslated Regions , Animals , Binding Sites , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , HEK293 Cells , Heterogeneous Nuclear Ribonucleoprotein D0/genetics , Homeodomain Proteins/genetics , Humans , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , RNA, Long Noncoding/genetics , Signal Transduction , Sp2 Transcription Factor/genetics , Transcription Factors/genetics
14.
Nucleic Acids Res ; 49(3): 1631-1646, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33444453

ABSTRACT

Mammalian circRNAs can influence different cellular processes by interacting with proteins and other nucleic acids. Here, we used ribonucleoprotein immunoprecipitation (RIP) analysis to identify systematically the circRNAs associated with the cancer-related protein AUF1. Among the circRNAs interacting with AUF1 in HeLa (human cervical carcinoma) cells, we focused on hsa_circ_0032434 (circPCNX), an abundant target of AUF1. Overexpression of circPCNX specifically interfered with the binding of AUF1 to p21 (CDKN1A) mRNA, thereby promoting p21 mRNA stability and elevating the production of p21, a major inhibitor of cell proliferation. Conversely, silencing circPCNX increased AUF1 binding to p21 mRNA, reducing p21 production and promoting cell division. Importantly, eliminating the AUF1-binding region of circPCNX abrogated the rise in p21 levels and rescued proliferation. Therefore, we propose that the interaction of circPCNX with AUF1 selectively prevents AUF1 binding to p21 mRNA, leading to enhanced p21 mRNA stability and p21 protein production, thereby suppressing cell growth.


Subject(s)
Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , RNA, Circular/metabolism , 3' Untranslated Regions , Binding Sites , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/metabolism , HeLa Cells , Humans , RNA, Circular/chemistry , RNA, Messenger/metabolism
15.
Biochem Biophys Res Commun ; 534: 491-497, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33220927

ABSTRACT

Cytoplasmic polyadenylation element binding protein 1 (CPEB1) regulates polyadenylation and subsequent translation of CPE-containing mRNAs involved in various physiological and pathological phenomena. Although the significance of CPEB1-mediated translational regulation has recently been reported, the detailed regulatory mechanism of Cpeb1 expression remains unclear. To elucidate the post-transcriptional regulatory mechanisms of Cpeb1 expression, we constructed reporter plasmids containing various deletions or mutations in the Cpeb1 mRNA 3' untranslated region (3'UTR). We investigated their expression levels in Neuro2a neuroblastoma cells. We found that Cpeb1 expression is regulated through an AU-rich element in its 3'UTR. Furthermore, the mRNA decay factor AU-rich binding factor 1 (AUF1) regulates Cpeb1 expression, and knockdown of AUF1 upregulates Cpeb1 mRNA expression but results in a decrease in CPEB1 protein levels. These findings indicate that AUF1 has a discordant role in the expression of Cpeb1.


Subject(s)
Heterogeneous Nuclear Ribonucleoprotein D0/genetics , RNA, Messenger/genetics , Transcription Factors/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , 3' Untranslated Regions , Animals , Cell Line , Gene Expression Regulation , Gene Knockdown Techniques , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Mice , Mice, Inbred C57BL , RNA Stability
16.
RNA Biol ; 18(6): 843-853, 2021 06.
Article in English | MEDLINE | ID: mdl-32924750

ABSTRACT

Proper base-pairing of a miRNA with its target mRNA is a key step in miRNA-mediated mRNA repression. RNA remodelling by RNA-binding proteins (RBPs) can improve access of miRNAs to their target mRNAs. The largest isoform p45 of the RBP AUF1 has previously been shown to remodel viral or AU-rich RNA elements. Here, we show that AUF1 is capable of directly promoting the binding of the miRNA let-7b to its target site within the 3'UTR of the POLR2D mRNA. Our data suggest this occurs in two ways. First, the helix-destabilizing RNA chaperone activity of AUF1 disrupts a stem-loop structure of the target mRNA and thus exposes the miRNA target site. Second, the RNA annealing activity of AUF1 drives hybridization of the miRNA and its target site within the mRNA. Interestingly, the RNA remodelling activities of AUF1 were found to be isoform-specific. AUF1 isoforms containing a YGG motif are competent RNA chaperones, whereas isoforms lacking the YGG motif are not. Overall, our study demonstrates that AUF1 has the ability to modulate a miRNA-target site interaction, thus revealing a new regulatory function for AUF1 proteins during post-transcriptional control of gene expression. Moreover, tests with other RBPs suggest the YGG motif acts as a key element of RNA chaperone activity.


Subject(s)
Alternative Splicing , Heterogeneous Nuclear Ribonucleoprotein D0/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , RNA-Binding Motifs/genetics , 3' Untranslated Regions/genetics , Algorithms , Amino Acid Sequence , Gene Expression Regulation , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Humans , Kinetics , MicroRNAs/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Messenger/metabolism
17.
Cancer Med ; 9(22): 8612-8623, 2020 11.
Article in English | MEDLINE | ID: mdl-33016643

ABSTRACT

BACKGROUND: AUF1 is one of the AU-rich binding proteins, which promotes rapid ARE-mRNA degradation. Recently, it has been reported that AUF1 is involved in regulating the antioxidant system because of its capacity to bind specifically to RNA containing oxidized bases and degrade oxidized RNA. Many antioxidant proteins have been reported to be overexpressed in colorectal cancer (CRC), however, the role of AUF1 in the progression of CRC has not been explored. METHODS: The expression level of AUF1 protein in human CRC cell lines and CRC tissues was detected by western blotting and immunohistochemistry (IHC. The effects of AUF1 knockdown on CRC cell proliferation, migration, invasion and changes in the signaling pathways were evaluated using a cell counting kit-8 (CCK-8), Transwell assays and western blotting. Subcutaneous xenograft tumor model was employed to further substantiate the role of AUF1 in CRC. RESULTS: AUF1 protein was upregulated in CRC tissues and CRC cells, and high expression of AUF1 was significantly associated with advanced AJCC stage (P = .001), lymph node metastasis (P = .007), distant metastasis (P = .038) and differentiation (P = .009) of CRC specimens. CRC patients with the high expression of AUF1 had an extremely poor prognosis. The knockdown of AUF1 suppressed CRC cell line proliferation, migration and invasion, inhibited CRC cells tumorigenesis and growth in nude mice, and reduced phosphorylated-ERK1/2 and phosphorylated AKT in CRC cells. CONCLUSION: Our findings demonstrate that AUF1 is probably involved in the progression of CRC via the activation of the ERK1/2 and AKT pathways. AU-rich RNA-binding factor 1 could be used as a novel prognostic biomarker and a potential therapeutic target for CRC.


Subject(s)
Colorectal Neoplasms/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Aged , Cell Movement , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Progression , Enzyme Activation , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Heterogeneous Nuclear Ribonucleoprotein D0/genetics , Humans , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Phosphorylation , Signal Transduction , Tumor Burden , Up-Regulation
18.
Nat Commun ; 11(1): 4775, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32963221

ABSTRACT

Enterovirus 71 (EV71) poses serious threats to human health, particularly in Southeast Asia, and no drugs or vaccines are available. Previous work identified the stem loop II structure of the EV71 internal ribosomal entry site as vital to viral translation and a potential target. After screening an RNA-biased library using a peptide-displacement assay, we identify DMA-135 as a dose-dependent inhibitor of viral translation and replication with no significant toxicity in cell-based studies. Structural, biophysical, and biochemical characterization support an allosteric mechanism in which DMA-135 induces a conformational change in the RNA structure that stabilizes a ternary complex with the AUF1 protein, thus repressing translation. This mechanism is supported by pull-down experiments in cell culture. These detailed studies establish enterovirus RNA structures as promising drug targets while revealing an approach and mechanism of action that should be broadly applicable to functional RNA targeting.


Subject(s)
Enterovirus A, Human/genetics , Enterovirus A, Human/physiology , Enterovirus Infections/metabolism , Host-Pathogen Interactions/physiology , Internal Ribosome Entry Sites/physiology , Virus Replication/physiology , 5' Untranslated Regions , Cell Line , Enterovirus Infections/virology , Gene Expression Regulation, Viral , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Humans , Ligands , Models, Molecular , Protein Binding , RNA, Viral/chemistry , Viral Proteins/metabolism
19.
Mol Cell Biol ; 40(20)2020 09 28.
Article in English | MEDLINE | ID: mdl-32778571

ABSTRACT

Many proteins, including DICER1 and hAgo2, are involved in the biogenesis of microRNAs (miRNAs). Whether hAgo2 regulates DICER1 expression is unknown. Exogenously overexpressed hAgo2 suppressed DICER1 expression at the levels of both protein and mRNA, and the reduction in hAgo2 expression enhanced DICER1 expression. Precursor miRNA processing mediated by DICER1 was also modulated by hAgo2. However, hAgo2 protein did not suppress DICER1 promoter activity. Therefore, hAgo2 protein probably regulates DICER1 expression at the posttranscriptional level. Indeed, hAgo2 protein inhibited the reporter assay of the DICER1 mRNA 3' untranslated region (3'-UTR). Previous reports have demonstrated that miRNAs (e.g., let-7 and miR-103/107) inhibited DICER1 expression posttranscriptionally. However, hAgo2 still suppressed DICER1 expression in the cells depleted of these miRNAs. Moreover, the reporter activities of the DICER1 mRNA 3'-UTR without these miRNA binding sites were still suppressed by hAgo2. Therefore, in addition to an miRNA-dependent pathway, hAgo2 can also modulate DICER1 expression through an miRNA-independent mechanism. Downregulation of DICER1 expression was further proven to be dependent on both hAgo2 and AUF1 proteins. Interactions of hAgo2 and AUF1 proteins were demonstrated by the coimmunoprecipitation assay. As expected, hAgo2 could not suppress the DICER1 mRNA 3'-UTR reporter with a mutation in the potential AUF1-binding site. Thus, downregulation of DICER1 expression through the 3'-UTR requires both hAgo2 and AUF1.


Subject(s)
Argonaute Proteins/genetics , DEAD-box RNA Helicases/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Neoplasms/genetics , Ribonuclease III/genetics , 3' Untranslated Regions/genetics , A549 Cells , Argonaute Proteins/metabolism , Binding Sites/genetics , CRISPR-Cas Systems , Cell Line, Tumor , DEAD-box RNA Helicases/metabolism , HEK293 Cells , HeLa Cells , Heterogeneous Nuclear Ribonucleoprotein D0/genetics , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , Humans , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Ribonuclease III/metabolism
20.
Mol Cancer ; 19(1): 26, 2020 02 05.
Article in English | MEDLINE | ID: mdl-32020881

ABSTRACT

BACKGROUND: Although trastuzumab provides significant clinical benefit for HER2-positive breast cancers, responses are limited by the emergence of resistance. Recent evidence suggests that long noncoding RNAs (lncRNAs) play important roles in tumorigenesis and chemoresistance. However, the regulatory mechanism of lncRNAs in trastuzumab resistance is not well established to date. In this research, we identified the differentially expressed lncRNA and investigated its regulatory role in trastuzumab resistance of breast cancer. METHODS: LncRNA microarray and qRT-PCR were performed to identify the dysregulated lncRNAs. Transmission electron microscopy, differential ultracentrifugation and qRT-PCR were used to verify the existence of exosomal AFAP1-AS1 (actin filament associated protein 1 antisense RNA 1). Bioinformatics prediction, RNA fluorescence in situ hybridization (RNA-FISH) and immunoprecipitation assays were performed to identify the direct interactions between AFAP1-AS1 and other associated targets, such as AU-binding factor 1 (AUF1) and ERBB2. Finally, a series gain- or loss-functional assays were done to prove the precise role of AFAP1-AS1 in trastuzumab resistance. RESULTS: AFAP1-AS1 was screened out due to its higher expression in trastuzumab-resistant cells compared to sensitive cells. Increased expression of AFAP1-AS1was associate with poorer response and shorter survival time of breast cancer patients. AFAP1-AS1 was upregulated by H3K27ac modification at promoter region, and knockdown of AFAP1-AS1 reversed trastuzumab resistance. Moreover, extracellular AFAP1-AS1 secreted from trastuzumab resistant cells was packaged into exosomes and then disseminated trastuzumab resistance of receipt cells. Mechanically, AFAP1-AS1 was associated with AUF1 protein, which further promoted the translation of ERBB2 without influencing the mRNA level. CONCLUSION: Exosomal AFAP1-AS1 could induce trastuzumab resistance through associating with AUF1 and promoting ERBB2 translation. Therefore, AFAP1-AS1 level may be useful for prediction of trastuzumab resistance and breast cancer treatment.


Subject(s)
Breast Neoplasms/pathology , Drug Resistance, Neoplasm , Exosomes/genetics , Heterogeneous Nuclear Ribonucleoprotein D0/metabolism , RNA, Long Noncoding/genetics , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology , Animals , Antineoplastic Agents, Immunological/pharmacology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Proliferation , Exosomes/metabolism , Female , Gene Expression Regulation, Neoplastic , Heterogeneous Nuclear Ribonucleoprotein D0/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Biosynthesis , Receptor, ErbB-2/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...