Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 859
Filter
1.
Cell Biol Toxicol ; 40(1): 41, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38833095

ABSTRACT

Hippocampal neurons maintain the ability of proliferation throughout life to support neurogenesis. Deoxynivalenol (DON) is a mycotoxin that exhibits brain toxicity, yet whether and how DON affects hippocampal neurogenesis remains unknown. Here, we use mouse hippocampal neuron cells (HT-22) as a model to illustrate the effects of DON on neuron proliferation and to explore underlying mechanisms. DON exposure significantly inhibits the proliferation of HT-22 cells, which is associated with an up-regulation of cell cycle inhibitor p21 at both mRNA and protein levels. Global and site-specific m6A methylation levels on the 3'UTR of p21 mRNA are significantly increased in response to DON treatment, whereas inhibition of m6A hypermethylation significantly alleviates DON-induced cell cycle arrest. Further mechanistic studies indicate that the m6A readers YTHDF1 and IGF2BP1 are responsible for m6A-mediated increase in p21 mRNA stability. Meanwhile, 3'UTR of E3 ubiquitin ligase TRIM21 mRNA is also m6A hypermethylated, and another m6A reader YTHDF2 binds to the m6A sites, leading to decreased TRIM21 mRNA stability. Consequently, TRIM21 suppression impairs ubiquitin-mediated p21 protein degradation. Taken together, m6A-mediated upregulation of p21, at both post-transcriptional and post-translational levels, contributes to DON-induced inhibition of hippocampal neuron proliferation. These results may provide new insights for epigenetic therapy of neurodegenerative diseases.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21 , Hippocampus , Neurons , Trichothecenes , Up-Regulation , Animals , Trichothecenes/toxicity , Trichothecenes/pharmacology , Hippocampus/metabolism , Hippocampus/drug effects , Hippocampus/cytology , Mice , Neurons/drug effects , Neurons/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Up-Regulation/drug effects , Cell Proliferation/drug effects , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Cell Line , 3' Untranslated Regions/genetics , Neurogenesis/drug effects , RNA, Messenger/metabolism , RNA, Messenger/genetics , RNA Stability/drug effects , Cell Cycle Checkpoints/drug effects , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Methylation/drug effects
2.
Endocrinology ; 165(6)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38717933

ABSTRACT

CYP19A1 encodes aromatase, which converts testosterone to estrogen, and is induced during placental maturation. To elucidate the molecular mechanism underlying this function, histone methylation was analyzed using the placental cytotrophoblast cell line, JEG3. Treatment of JEG3 cells with 3-deazaneplanocin A, an inhibitor of several methyltransferases, resulted in increased CYP19A1 expression, accompanied by removal of the repressive mark H3K27me3 from the CYP19A1 promoter. However, this increase was not observed in cells treated with GSK126, another specific inhibitor for H3K27me3 methylation. Expression of TFAP2C, which encodes AP-2γ, a transcription factor that regulates CYP19A1, was also elevated on 3-deazaneplanocin A treatment. Interestingly, TFAP2C messenger RNA (mRNA) was readily degraded in JEG3 cells but protected from degradation in the presence of 3-deazaneplanocin A. TFAP2C mRNA contained N6-methyladenosines, which were reduced on drug treatment. These observations indicate that the TFAP2C mRNA undergoes adenosine methylation and rapid degradation, whereas 3-deazaneplanocin A suppresses methylation, resulting in an increase in AP-2γ levels. We conclude that the increase in AP-2γ expression via stabilization of the TFAP2C mRNA is likely to underlie the increased CYP19A1 expression.


Subject(s)
Aromatase , Placenta , RNA Stability , Transcription Factor AP-2 , Humans , Transcription Factor AP-2/metabolism , Transcription Factor AP-2/genetics , Aromatase/genetics , Aromatase/metabolism , Female , Placenta/metabolism , Placenta/drug effects , Pregnancy , RNA Stability/drug effects , Adenosine/analogs & derivatives , Adenosine/pharmacology , RNA, Messenger/metabolism , RNA, Messenger/genetics , Cell Line, Tumor , Histones/metabolism
3.
Cancer Biol Ther ; 25(1): 2349429, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38738555

ABSTRACT

Cervical cancer (CC) is a prevalent malignancy among women worldwide. This study was designed to investigate the role of METTL14 in sorafenib-induced ferroptosis in CC. METTL14 expression and m6A methylation were determined in CC tissues, followed by analyzes correlating these factors with clinical features. Subsequently, METTL14 was knocked down in CC cell lines, and the effects on cell proliferation, mitochondrial morphology and ferroptosis were assessed using CCK-8, microscopy, and markers associated with ferroptosis, respectively. The regulatory relationship between METTL14 and FTH1 was verified using qRT-PCR and luciferase reporter assays. The functional significance of this interaction was further investigated both in vitro and in vivo by co-transfecting cells with overexpression vectors or shRNAs targeting METTL14 and FTH1 after sorafenib treatment. METTL14 expression and m6A methylation were significantly reduced in CC tissues, and lower METTL14 expression levels were associated with a poorer CC patients' prognosis. Notably, METTL14 expression increased during sorafenib-induced ferroptosis, and METTL14 knockdown attenuated the ferroptotic response induced by sorafenib in CC cells. FTH1 was identified as a direct target of METTL14, with METTL14 overexpression leading to increased m6A methylation of FTH1 mRNA, resulting in reduced stability and expression of FTH1 in CC. Furthermore, FTH1 overexpression or treatment with LY294002 partially counteracted the promotion of sorafenib-induced ferroptosis by METTL14. In vivo xenograft experiments demonstrated that inhibiting METTL14 reduced the anticancer effects of sorafenib, whereas suppression of FTH1 significantly enhanced sorafenib-induced ferroptosis and increased its anticancer efficacy. METTL14 reduces FTH1 mRNA stability through m6A methylation, thereby enhancing sorafenib-induced ferroptosis, which contributes to suppressing CC progression via the PI3K/Akt signaling pathway.


Subject(s)
Ferroptosis , Methyltransferases , RNA Stability , Sorafenib , Uterine Cervical Neoplasms , Humans , Sorafenib/pharmacology , Sorafenib/therapeutic use , Female , Ferroptosis/drug effects , Ferroptosis/genetics , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/genetics , Mice , Animals , Methyltransferases/metabolism , Methyltransferases/genetics , RNA Stability/drug effects , Mice, Nude , Gene Expression Regulation, Neoplastic/drug effects , Methylation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Xenograft Model Antitumor Assays , RNA, Messenger/genetics , RNA, Messenger/metabolism , Prognosis , Ferritins , Oxidoreductases
4.
Bioorg Med Chem Lett ; 108: 129815, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38795737

ABSTRACT

We report the use of trimethylsilyl azide and Selectfluor to implement a standard protocol targeted at the prenylated nucleic acid known as i6A-RNA. After optimizing the conditions, we applied this method to regulate a wide range of i6A-RNA species using synthetic imidazole-based probes (I-IV). We observed that prenylated nucleic acid plays a crucial role in the cell hemostasis in A549 cell lines.


Subject(s)
Azides , Click Chemistry , Halogenation , Imidazoles , Humans , Imidazoles/chemistry , Imidazoles/chemical synthesis , Azides/chemistry , A549 Cells , RNA/chemistry , RNA/metabolism , Molecular Structure , RNA Stability/drug effects
5.
Biochem Biophys Res Commun ; 715: 149994, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38692139

ABSTRACT

Many virus lysis/transport buffers used in molecular diagnostics, including the detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA, contain guanidine-based chaotropic salts, primarily guanidine hydrochloride (GuHCl) or guanidine isothiocyanate (GITC). Although the virucidal effects of GuHCl and GITC alone against some enveloped viruses have been established, standardized data on their optimum virucidal concentrations against SARS-CoV-2 and effects on viral RNA stability are scarce. Thus, we aimed to determine the optimum virucidal concentrations of GuHCl and GITC against SARS-CoV-2 compared to influenza A virus (IAV), another enveloped respiratory virus. We also evaluated the effectiveness of viral RNA stabilization at the determined optimum virucidal concentrations under high-temperature conditions (35°C) using virus-specific real-time reverse transcription polymerase chain reaction. Both viruses were potently inactivated by 1.0 M GITC and 2.5 M GuHCl, but the GuHCl concentration for efficient SARS-CoV-2 inactivation was slightly higher than that for IAV inactivation. GITC showed better viral RNA stability than GuHCl at the optimum virucidal concentrations. An increased concentration of GuHCl or GITC increased viral RNA degradation at 35°C. Our findings highlight the need to standardize GuHCl and GITC concentrations in virus lysis/transport buffers and the potential application of these guanidine-based salts alone as virus inactivation solutions in SARS-CoV-2 and IAV molecular diagnostics.


Subject(s)
Guanidine , Influenza A virus , RNA, Viral , SARS-CoV-2 , Specimen Handling , SARS-CoV-2/drug effects , SARS-CoV-2/genetics , Influenza A virus/drug effects , Influenza A virus/genetics , Guanidine/pharmacology , Guanidine/chemistry , RNA, Viral/genetics , Humans , Specimen Handling/methods , Genome, Viral , COVID-19/virology , COVID-19/diagnosis , Chlorocebus aethiops , Vero Cells , Virus Inactivation/drug effects , Animals , RNA Stability/drug effects , Containment of Biohazards , Guanidines/pharmacology , Guanidines/chemistry , Salts/pharmacology , Salts/chemistry
6.
Prostaglandins Other Lipid Mediat ; 172: 106832, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38460759

ABSTRACT

Atherosclerosis (AS) represents a prevalent initiating factor for cardiovascular events. Insulin-like growth factor 2 mRNA binding protein 3 (IGF2BP3) is an oncofetal RNA-binding protein that participates in cardiovascular diseases. This work aimed to elaborate the effects of IGF2BP3 on AS and the probable mechanism by using an oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cells (HUVECs) model. Results indicated that IGF2BP3 expression was declined in the blood of AS patients and ox-LDL-induced HUVECs. IGF2BP3 elevation alleviated ox-LDL-provoked viability loss, apoptosis, oxidative DNA damage and endothelial dysfunction in HUVECs. Moreover, IGF2BP3 bound SESN1 and stabilized SESN1 mRNA. Furthermore, SESN1 interference reversed the impacts of IGF2BP3 overexpression on the apoptosis, oxidative DNA damage and endothelial dysfunction of ox-LDL-challenged HUVECs. Additionally, the activation of Nrf2 signaling mediated by IGF2BP3 up-regulation in ox-LDL-treated HUVECs was blocked by SESN1 absence. Collectively, SESN1 stabilized by IGF2BP3 might protect against AS by activating Nrf2 signaling.


Subject(s)
Human Umbilical Vein Endothelial Cells , Lipoproteins, LDL , NF-E2-Related Factor 2 , Oxidative Stress , RNA, Messenger , RNA-Binding Proteins , Signal Transduction , Humans , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Lipoproteins, LDL/pharmacology , Lipoproteins, LDL/metabolism , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Oxidative Stress/drug effects , Signal Transduction/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Apoptosis/drug effects , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , RNA Stability/drug effects , DNA Damage , Sestrins
7.
PLoS One ; 16(12): e0257503, 2021.
Article in English | MEDLINE | ID: mdl-34898625

ABSTRACT

Metabolic labeling is a widely used tool to investigate different aspects of pre-mRNA splicing and RNA turnover. The labeling technology takes advantage of native cellular machineries where a nucleotide analog is readily taken up and incorporated into nascent RNA. One such analog is 4-thiouridine (4sU). Previous studies demonstrated that the uptake of 4sU at elevated concentrations (>50µM) and extended exposure led to inhibition of rRNA synthesis and processing, presumably induced by changes in RNA secondary structure. Thus, it is possible that 4sU incorporation may also interfere with splicing efficiency. To test this hypothesis, we carried out splicing analyses of pre-mRNA substrates with varying levels of 4sU incorporation (0-100%). We demonstrate that increased incorporation of 4sU into pre-mRNAs decreased splicing efficiency. The overall impact of 4sU labeling on pre-mRNA splicing efficiency negatively correlates with the strength of splice site signals such as the 3' and the 5' splice sites. Introns with weaker splice sites are more affected by the presence of 4sU. We also show that transcription by T7 polymerase and pre-mRNA degradation kinetics were impacted at the highest levels of 4sU incorporation. Increased incorporation of 4sU caused elevated levels of abortive transcripts, and fully labeled pre-mRNA is more stable than its uridine-only counterpart. Cell culture experiments show that a small number of alternative splicing events were modestly, but statistically significantly influenced by metabolic labeling with 4sU at concentrations considered to be tolerable (40 µM). We conclude that at high 4sU incorporation rates small, but noticeable changes in pre-mRNA splicing can be detected when splice sites deviate from consensus. Given these potential 4sU artifacts, we suggest that appropriate controls for metabolic labeling experiments need to be included in future labeling experiments.


Subject(s)
Alternative Splicing/drug effects , RNA Precursors/metabolism , RNA Splice Sites , Thiouridine/pharmacology , HEK293 Cells , Humans , Nucleic Acid Conformation , RNA Precursors/genetics , RNA Stability/drug effects , Staining and Labeling
8.
Nat Commun ; 12(1): 7299, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911927

ABSTRACT

Huntington's disease (HD) is a hereditary neurodegenerative disorder caused by expansion of cytosine-adenine-guanine (CAG) trinucleotide repeats in the huntingtin (HTT) gene. Consequently, the mutant protein is ubiquitously expressed and drives pathogenesis of HD through a toxic gain-of-function mechanism. Animal models of HD have demonstrated that reducing huntingtin (HTT) protein levels alleviates motor and neuropathological abnormalities. Investigational drugs aim to reduce HTT levels by repressing HTT transcription, stability or translation. These drugs require invasive procedures to reach the central nervous system (CNS) and do not achieve broad CNS distribution. Here, we describe the identification of orally bioavailable small molecules with broad distribution throughout the CNS, which lower HTT expression consistently throughout the CNS and periphery through selective modulation of pre-messenger RNA splicing. These compounds act by promoting the inclusion of a pseudoexon containing a premature termination codon (stop-codon psiExon), leading to HTT mRNA degradation and reduction of HTT levels.


Subject(s)
Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/drug therapy , Huntington Disease/genetics , RNA Splicing , Small Molecule Libraries/administration & dosage , Animals , Central Nervous System/drug effects , Central Nervous System/metabolism , Disease Models, Animal , Humans , Huntington Disease/metabolism , Mice , RNA Splicing/drug effects , RNA Stability/drug effects , Trinucleotide Repeat Expansion/drug effects
9.
Int J Mol Sci ; 22(22)2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34830143

ABSTRACT

Small regulatory RNAs play a major role in bacterial gene regulation by binding their target mRNAs, which mostly influences the stability or translation of the target. Expression levels of sRNAs are often regulated by their own promoters, but recent reports have highlighted the presence and importance of sRNAs that are derived from mRNA 3' untranslated regions (UTRs). In this study, we investigated the maturation of 5' and 3' UTR-derived sRNAs on a global scale in the facultative phototrophic alphaproteobacterium Rhodobacter sphaeroides. Including some already known UTR-derived sRNAs like UpsM or CcsR1-4, 14 sRNAs are predicted to be located in 5 UTRs and 16 in 3' UTRs. The involvement of different ribonucleases during maturation was predicted by a differential RNA 5'/3' end analysis based on RNA next generation sequencing (NGS) data from the respective deletion strains. The results were validated in vivo and underline the importance of polynucleotide phosphorylase (PNPase) and ribonuclease E (RNase E) during processing and maturation. The abundances of some UTR-derived sRNAs changed when cultures were exposed to external stress conditions, such as oxidative stress and also during different growth phases. Promoter fusions revealed that this effect cannot be solely attributed to an altered transcription rate. Moreover, the RNase E dependent cleavage of several UTR-derived sRNAs varied significantly during the early stationary phase and under iron depletion conditions. We conclude that an alteration of ribonucleolytic processing influences the levels of UTR-derived sRNAs, and may thus indirectly affect their mRNA targets.


Subject(s)
3' Untranslated Regions/genetics , 5' Untranslated Regions/genetics , RNA, Bacterial/genetics , RNA, Small Untranslated/genetics , Rhodobacter sphaeroides/genetics , Adaptation, Physiological/drug effects , Adaptation, Physiological/genetics , Endoribonucleases/metabolism , Gene Expression Regulation, Bacterial/drug effects , Gene Knockout Techniques , High-Throughput Nucleotide Sequencing/methods , Hydrogen Peroxide/pharmacology , Oxidants/pharmacology , Polyribonucleotide Nucleotidyltransferase/metabolism , RNA Stability/drug effects , RNA, Bacterial/metabolism , RNA, Small Untranslated/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rhodobacter sphaeroides/growth & development
11.
Cell Death Dis ; 12(11): 952, 2021 10 16.
Article in English | MEDLINE | ID: mdl-34657141

ABSTRACT

Drug resistance strikingly limits the therapeutic effect of temozolomide (TMZ) (a common drug for glioma). Long non-coding RNA (lncRNA) RMRP has been found to be implicated in glioma progression. However, the effect of RMRP on TMZ resistance along with related molecular mechanisms is poorly defined in glioma. In the present study, RMRP, ZNRF3, and IGF2BP3 were screened out by bioinformatics analysis. The expression levels of lncRNAs and mRNAs were measured by RT-qPCR assay. Protein levels of genes were detected by western blot and immunofluorescence assays. ZNRF3 mRNA stability was analyzed using Actinomycin D assay. Cell proliferative ability and survival rate were determined by CCK-8 assay. Cell apoptotic pattern was estimated by flow cytometry. The effect of RMRP knockdown on the growth of TMZ-treated glioma xenograft tumors was explored in vivo. The relationships of IGF2BP3, RMRP, and ZNRF3 were explored by bioinformatics prediction analysis, RNA immunoprecipitation, luciferase, and RNA pull-down, and chromatin immunoprecipitation assays. The results showed that RMRP was highly expressed in glioma. RMRP knockdown curbed cell proliferation, facilitated cell apoptosis and reduced TMZ resistance in glioma cells, and hindered the growth of TMZ-treated glioma xenograft tumors. RMRP exerted its functions by down-regulating ZNRF3 in glioma cells. IGF2BP3 interacted with RMRP and ZNRF3 mRNA. IGF2BP3 knockdown weakened the interaction of Argonaute 2 (Ago2) and ZNRF3. RMRP reduced ZNRF3 expression and mRNA stability by IGF2BP3. RMRP knockdown inhibited ß-catenin expression by up-regulating ZNRF3. The inhibition of Wnt/ß-catenin signaling pathway by XAV-939 weakened RMRP-mediated TMZ resistance in glioma cells. ß-catenin promoted RMRP expression by TCF4 in glioma cells. In conclusion, RMRP/ZNRF3 axis and Wnt/ß-catenin signaling formed a positive feedback loop to regulate TMZ resistance in glioma. The sustained activation of Wnt/ß-catenin signaling by RMRP might contribute to the better management of cancers.


Subject(s)
Disease Progression , Drug Resistance, Neoplasm , Feedback, Physiological , Glioma/drug therapy , Glioma/genetics , RNA, Long Noncoding/metabolism , Temozolomide/therapeutic use , Ubiquitin-Protein Ligases/metabolism , Wnt Signaling Pathway , Adult , Animals , Apoptosis/drug effects , Apoptosis/genetics , Brain/pathology , Cell Line, Tumor , Down-Regulation/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Glioma/pathology , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , RNA Stability/drug effects , RNA Stability/genetics , RNA, Long Noncoding/genetics , RNA-Binding Proteins/metabolism , Temozolomide/pharmacology , Transcription Factor 4/metabolism , Transcription, Genetic , Tumor Burden , Up-Regulation/drug effects , Up-Regulation/genetics , Xenograft Model Antitumor Assays , Young Adult , beta Catenin/metabolism
12.
Cell Mol Life Sci ; 78(17-18): 6337-6349, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34398253

ABSTRACT

Signaling via the B-cell receptor (BCR) is a key driver and therapeutic target in chronic lymphocytic leukemia (CLL). BCR stimulation of CLL cells induces expression of eIF4A, an initiation factor important for translation of multiple oncoproteins, and reduces expression of PDCD4, a natural inhibitor of eIF4A, suggesting that eIF4A may be a critical nexus controlling protein expression downstream of the BCR in these cells. We, therefore, investigated the effect of eIF4A inhibitors (eIF4Ai) on BCR-induced responses. We demonstrated that eIF4Ai (silvestrol and rocaglamide A) reduced anti-IgM-induced global mRNA translation in CLL cells and also inhibited accumulation of MYC and MCL1, key drivers of proliferation and survival, respectively, without effects on upstream signaling responses (ERK1/2 and AKT phosphorylation). Analysis of normal naïve and non-switched memory B cells, likely counterparts of the two main subsets of CLL, demonstrated that basal RNA translation was higher in memory B cells, but was similarly increased and susceptible to eIF4Ai-mediated inhibition in both. We probed the fate of MYC mRNA in eIF4Ai-treated CLL cells and found that eIF4Ai caused a profound accumulation of MYC mRNA in anti-IgM treated cells. This was mediated by MYC mRNA stabilization and was not observed for MCL1 mRNA. Following drug wash-out, MYC mRNA levels declined but without substantial MYC protein accumulation, indicating that stabilized MYC mRNA remained blocked from translation. In conclusion, BCR-induced regulation of eIF4A may be a critical signal-dependent nexus for therapeutic attack in CLL and other B-cell malignancies, especially those dependent on MYC and/or MCL1.


Subject(s)
Eukaryotic Initiation Factor-4A/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptors, Antigen, B-Cell/metabolism , Antibodies, Anti-Idiotypic/pharmacology , Benzofurans/pharmacology , Cells, Cultured , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Protein Biosynthesis/drug effects , Proto-Oncogene Proteins c-myc/genetics , RNA Stability/drug effects , RNA, Messenger/metabolism , Signal Transduction/drug effects , Triterpenes/pharmacology
13.
Aging (Albany NY) ; 13(14): 18238-18256, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34310344

ABSTRACT

BACKGROUND: N6-methyladenosine (m6A) modification is one of the most common chemical modifications of eukaryotic mRNAs, which play an important role in tumors and cardiovascular disease through regulating mRNA stability, splicing and translation. However, the changes of m6A mRNA and m6A-related enzymes in pulmonary arterial hypertension (PAH) remain largely unexplored. METHODS: MeRIP-seq was used to identify m6A methylation in lung tissues from control and MCT-PAH rats. Western blot and immunofluorescence were used to evaluate expression of m6A-related enzymes. RESULTS: Compared with control group, m6A methylation was mainly increased in lung tissues from MCT-PAH rats. The up-methylated coding genes in MCT-PAH rats were primarily enriched in processes associated with inflammation, glycolysis, ECM-receptor interaction and PDGF signal pathway, while genes with down-methylation were enriched in processes associated with TGF-ß family receptor members. The expression of FTO and ALKBH5 downregulated, METTL3 and YTHDF1 increased and other methylation modification-related proteins was not significantly changed in MCT-PAH rats lung tissues. Immunofluorescence indicated that expression of FTO decreased and YTHDF1 increased in small pulmonary arteries of MCT-PAH rats. CONCLUSION: m6A levels and the expression of methylation-related enzymes were altered in PAH rats, in which FTO and YTHDF1 may play a crucial role in m6A modification.


Subject(s)
Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Methyltransferases/metabolism , Pulmonary Arterial Hypertension/metabolism , RNA-Binding Proteins/metabolism , Animals , Down-Regulation , Fluorescent Antibody Technique , Male , Methylation , Monocrotaline/toxicity , Pulmonary Arterial Hypertension/chemically induced , RNA Stability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
14.
Biochimie ; 187: 67-74, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34022290

ABSTRACT

The RNA Degradosome (RNAD) is a multi-enzyme complex, which performs important functions in post-transcriptional regulation in Escherichia coli with the assistance of regulatory sRNAs and the RNA chaperone Hfq. Although the interaction of the canonical RNAD components with RNase E has been extensively studied, the dynamic nature of the interactions in vivo remains largely unknown. In this work, we explored the rearrangements upon glucose stress using fluorescence energy transfer (hetero-FRET). Results revealed differences in the proximity of the canonical components with 1% (55.5 mM) glucose concentration, with the helicase RhlB and the glycolytic enzyme Enolase exhibiting the largest changes to the C-terminus of RNase E, followed by PNPase. We quantified ptsG mRNA decay and SgrS sRNA synthesis as they mediate bacterial adaptation to glucose stress conditions. We propose that once the mRNA degradation is completed, the RhlB, Enolase and PNPase decrease their proximity to the C-terminus of RNase E. Based on the results, we present a model where the canonical components of the RNAD coalesce when the bacteria is under glucose-6-phosphate stress and associate it with RNA decay. Our results demonstrate that FRET is a helpful tool to study conformational rearrangements in enzymatic complexes in bacteria in vivo.


Subject(s)
Escherichia coli/metabolism , Glucose/pharmacology , RNA Stability/drug effects , RNA, Bacterial/metabolism , RNA, Messenger/metabolism , Stress, Physiological/drug effects , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Host Factor 1 Protein/genetics , Host Factor 1 Protein/metabolism , RNA Stability/genetics , RNA, Bacterial/genetics , RNA, Messenger/genetics , Stress, Physiological/genetics
15.
Life Sci ; 279: 119645, 2021 Aug 15.
Article in English | MEDLINE | ID: mdl-34043991

ABSTRACT

AIMS: Norcantharidin (NCTD) exhibits antitumor, anti-inflammatory, and anti-fibrosis properties, which makes NCTD an attractive candidate for the treatment of pathological scars. This study was designed to investigate the potential effects of NCTD on fibroblast proliferation and explore the underlying mechanisms. MATERIALS AND METHODS: First, cell viability and cell apoptosis were evaluated to determine the effects of NCTD on human skin fibroblasts, at 10, 50, and 100 µM. To explore the mechanism, bioinformatics analyses, chromatin immunoprecipitation, RNA immunoprecipitation, and RNA pulldown assays, and luciferase reporter assays were performed to verify the relationships among NCTD, signal transducer and activator of transcription 3 (STAT3), annexin A2 pseudogene 2 (ANXA2P2), and ubiquitin-associated protein 2-like (UBAP2L) mRNA in fibroblasts. Loss-of-function experiments were performed to investigate the roles played by STAT3, ANXA2P2, and UBAP2L in the proliferation and apoptosis of fibroblasts. KEY FINDINGS: We found that NCTD administration induced fibroblast apoptosis and inhibited fibroblast proliferation in a dose-dependent manner. Mechanistically, NCTD inhibited ANXA2P2 transcription through the inhibition of STAT3 phosphorylation. Subsequently, ANXA2P2 was found to enhance the physical interaction between UBAP2L mRNA and lin-28 homolog B (LIN28B), which increased the stability and levels of UBAP2L mRNA. Loss-of-function assays demonstrated that ANXA2P2 and UBAP2L knockdown induced fibroblast apoptosis and suppressed fibroblast proliferation. SIGNIFICANCE: In conclusion, we confirmed that NCTD inhibits fibroblast proliferation by inhibiting the STAT3/ANXA2P2/UBAP2L axis, which suggested that NCTD could represent a new candidate for the treatment of pathological scars.


Subject(s)
Annexin A2/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Carrier Proteins/metabolism , Cell Proliferation , Fibroblasts/cytology , Gene Expression Regulation/drug effects , RNA Stability/drug effects , RNA-Binding Proteins/metabolism , Annexin A2/genetics , Antineoplastic Agents/pharmacology , Apoptosis , Carrier Proteins/genetics , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Pseudogenes , RNA-Binding Proteins/genetics
16.
PLoS One ; 16(5): e0251290, 2021.
Article in English | MEDLINE | ID: mdl-33983964

ABSTRACT

Extracellular vesicles (EVs) have emerged as promising candidates in biomarker discovery and diagnostics. Protected by the lipid bilayer, the molecular content of EVs in diverse biofluids are protected from RNases and proteases in the surrounding environment that may rapidly degrade targets of interests. Nonetheless, cryopreservation of EV-containing samples to -80°C may expose the lipid bilayer to physical and biological stressors which may result in cryoinjury and contribute to changes in EV yield, function, or molecular cargo. In the present work, we systematically evaluate the effect of cryopreservation at -80°C for a relatively short duration of storage (up to 12 days) on plasma- and media-derived EV particle count and/or RNA yield/quality, as compared to paired fresh controls. On average, we found that the plasma-derived EV concentration of stored samples decreased to 23% of fresh samples. Further, this significant decrease in EV particle count was matched with a corresponding significant decrease in RNA yield whereby plasma-derived stored samples contained only 47-52% of the total RNA from fresh samples, depending on the extraction method used. Similarly, media-derived EVs showed a statistically significant decrease in RNA yield whereby stored samples were 58% of the total RNA from fresh samples. In contrast, we did not obtain clear evidence of decreased RNA quality through analysis of RNA traces. These results suggest that samples stored for up to 12 days can indeed produce high-quality RNA; however, we note that when directly comparing fresh versus cryopreserved samples without cryoprotective agents there are significant losses in total RNA. Finally, we demonstrate that the addition of the commonly used cryoprotectant agent, DMSO, alongside greater control of the rate of cooling/warming, can rescue EVs from damaging ice formation and improve RNA yield.


Subject(s)
Extracellular Vesicles/metabolism , RNA/isolation & purification , Specimen Handling/methods , Cryopreservation/methods , Culture Media/chemistry , Healthy Volunteers , Humans , Plasma/chemistry , RNA/metabolism , RNA Stability/drug effects , RNA Stability/physiology
17.
Cell Mol Gastroenterol Hepatol ; 12(2): 633-651, 2021.
Article in English | MEDLINE | ID: mdl-33848642

ABSTRACT

BACKGROUND & AIMS: N6-methyladenosine (m6A), the most prevalent and dynamic posttranscriptional methylation modification of mammalian mRNA, is involved in various biological processes, but its role in liver regeneration has not been characterized. METHODS: We first conducted transcriptome-wide m6A mRNA sequencing and characterized the expression pattern of m6A in regenerating mouse liver. Next, we generated hepatocyte-specific Mettl3- or Mettl14-deficient mice and investigated their role in liver regeneration. A series of biochemical experiments in vitro and in vivo was further performed to investigate potential mechanisms. RESULTS: We identified an overwhelming proportion of m6A-modified genes with initially up-regulated and subsequently down-regulated m6A levels as liver regeneration progressed. Loss of Mettl14 but not of Mettl3 resulted in markedly disrupted liver regeneration, and Mettl14-ablated hepatocytes were arrested in the G1 phase of the cell cycle. Most strikingly, the Mettl14-ablated regenerating liver exhibited extensive parenchymal necrosis. mRNA transcripts, such as Hsp90b1, Erp29, Stt3a, P4hb, and Lman1, encoding proteins involved in polypeptide processing and the endoplasmic reticulum (ER) stress response, were m6A-hypomethylated, and their mRNA and protein levels were subsequently decreased, resulting in unresolved ER stress, hepatocyte death, and inhibited proliferation. CONCLUSIONS: We demonstrate the essential role of Mettl14 in facilitating liver regeneration by modulating polypeptide-processing proteins in the ER in an m6A-dependent manner.


Subject(s)
Adenosine/analogs & derivatives , Endoplasmic Reticulum/metabolism , Homeostasis , Liver Regeneration , Methyltransferases/metabolism , Adenosine/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Proliferation/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Deletion , Hepatectomy , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/ultrastructure , Homeostasis/drug effects , Homeostasis/genetics , Liver/metabolism , Liver/surgery , Liver Regeneration/drug effects , Liver Regeneration/genetics , Male , Methyltransferases/deficiency , Mice, Knockout , Necrosis , Organ Specificity/drug effects , Organ Specificity/genetics , Peptides/genetics , Peptides/metabolism , RNA Stability/drug effects , RNA Stability/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Taurochenodeoxycholic Acid/pharmacology , Transcriptome/genetics
18.
Nat Commun ; 12(1): 2183, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33846348

ABSTRACT

Here we show that FTO as an N6-methyladenosine (m6A) RNA demethylase is degraded by selective autophagy, which is impaired by low-level arsenic exposure to promote tumorigenesis. We found that in arsenic-associated human skin lesions, FTO is upregulated, while m6A RNA methylation is downregulated. In keratinocytes, chronic relevant low-level arsenic exposure upregulated FTO, downregulated m6A RNA methylation, and induced malignant transformation and tumorigenesis. FTO deletion inhibited arsenic-induced tumorigenesis. Moreover, in mice, epidermis-specific FTO deletion prevented skin tumorigenesis induced by arsenic and UVB irradiation. Targeting FTO genetically or pharmacologically inhibits the tumorigenicity of arsenic-transformed tumor cells. We identified NEDD4L as the m6A-modified gene target of FTO. Finally, arsenic stabilizes FTO protein through inhibiting p62-mediated selective autophagy. FTO upregulation can in turn inhibit autophagy, leading to a positive feedback loop to maintain FTO accumulation. Our study reveals FTO-mediated dysregulation of mRNA m6A methylation as an epitranscriptomic mechanism to promote arsenic tumorigenicity.


Subject(s)
Adenosine/analogs & derivatives , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/metabolism , Arsenic/toxicity , Autophagy , Carcinogenesis/genetics , Adenosine/metabolism , Alpha-Ketoglutarate-Dependent Dioxygenase FTO/genetics , Animals , Autophagy/drug effects , Autophagy/genetics , Base Sequence , Carcinogenesis/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/pathology , Down-Regulation/drug effects , Down-Regulation/genetics , Epidermis/metabolism , Gene Ontology , HEK293 Cells , HaCaT Cells , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Mice , NF-kappa B/metabolism , Nedd4 Ubiquitin Protein Ligases/metabolism , Protein Stability/drug effects , RNA Stability/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequestosome-1 Protein/metabolism , Transcriptome/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics , Vacuoles/drug effects , Vacuoles/metabolism , Vacuoles/ultrastructure
19.
Nucleic Acids Res ; 49(8): 4705-4724, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33849070

ABSTRACT

Antisense peptide nucleic acids (PNAs) inhibiting mRNAs of essential genes provide a straight-forward way to repurpose our knowledge of bacterial regulatory RNAs for development of programmable species-specific antibiotics. While there is ample proof of PNA efficacy, their target selectivity and impact on bacterial physiology are poorly understood. Moreover, while antibacterial PNAs are typically designed to block mRNA translation, effects on target mRNA levels are not well-investigated. Here, we pioneer the use of global RNA-seq analysis to decipher PNA activity in a transcriptome-wide manner. We find that PNA-based antisense oligomer conjugates robustly decrease mRNA levels of the widely-used target gene, acpP, in Salmonella enterica, with limited off-target effects. Systematic analysis of several different PNA-carrier peptides attached not only shows different bactericidal efficiency, but also activation of stress pathways. In particular, KFF-, RXR- and Tat-PNA conjugates especially induce the PhoP/Q response, whereas the latter two additionally trigger several distinct pathways. We show that constitutive activation of the PhoP/Q response can lead to Tat-PNA resistance, illustrating the utility of RNA-seq for understanding PNA antibacterial activity. In sum, our study establishes an experimental framework for the design and assessment of PNA antimicrobials in the long-term quest to use these for precision editing of microbiota.


Subject(s)
Oligonucleotides, Antisense/pharmacology , Peptide Nucleic Acids/pharmacology , Peptides/chemistry , RNA, Messenger/metabolism , Salmonella enterica/drug effects , Stress, Physiological/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Proliferation/drug effects , Cell Proliferation/genetics , Drug Resistance, Bacterial/genetics , Microbial Sensitivity Tests , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Peptide Nucleic Acids/metabolism , Peptides/metabolism , Peptides/pharmacology , RNA Stability/drug effects , RNA, Messenger/genetics , RNA-Seq , Salmonella enterica/genetics , Salmonella enterica/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , tat Gene Products, Human Immunodeficiency Virus/pharmacology
20.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article in English | MEDLINE | ID: mdl-33762305

ABSTRACT

DNA-methyltransferase inhibitors (DNMTis), such as azacitidine and decitabine, are used clinically to treat myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Decitabine activates the transcription of endogenous retroviruses (ERVs), which can induce immune response by acting as cellular double-stranded RNAs (dsRNAs). Yet, the posttranscriptional regulation of ERV dsRNAs remains uninvestigated. Here, we find that the viral mimicry and subsequent cell death in response to decitabine require the dsRNA-binding protein Staufen1 (Stau1). We show that Stau1 directly binds to ERV RNAs and stabilizes them in a genome-wide manner. Furthermore, Stau1-mediated stabilization requires a long noncoding RNA TINCR, which enhances the interaction between Stau1 and ERV RNAs. Analysis of a clinical patient cohort reveals that MDS and AML patients with lower Stau1 and TINCR expressions exhibit inferior treatment outcomes to DNMTi therapy. Overall, our study reveals the posttranscriptional regulatory mechanism of ERVs and identifies the Stau1-TINCR complex as a potential target for predicting the efficacy of DNMTis and other drugs that rely on dsRNAs.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cytoskeletal Proteins/metabolism , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Adult , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/pharmacology , Azacitidine/therapeutic use , Cohort Studies , Cytoskeletal Proteins/genetics , DNA Methylation/drug effects , DNA Methylation/immunology , Decitabine/pharmacology , Decitabine/therapeutic use , Drug Resistance, Neoplasm/genetics , Endogenous Retroviruses/genetics , Female , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/immunology , Gene Knockout Techniques , HCT116 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/mortality , Progression-Free Survival , RNA Stability/drug effects , RNA Stability/immunology , RNA, Double-Stranded/metabolism , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/genetics , RNA-Seq
SELECTION OF CITATIONS
SEARCH DETAIL
...