Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 175(6): 1492-1506.e19, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30449617

ABSTRACT

Approximately half of human genes generate mRNAs with alternative 3' untranslated regions (3'UTRs). Through 3'UTR-mediated protein-protein interactions, alternative 3'UTRs enable multi-functionality of proteins with identical amino acid sequence. While studying how information on protein features is transferred from 3'UTRs to proteins, we discovered that the broadly expressed RNA-binding protein TIS11B forms a membraneless organelle, called TIS granule, that enriches membrane protein-encoding mRNAs with multiple AU-rich elements. TIS granules form a reticular meshwork intertwined with the endoplasmic reticulum (ER). The association between TIS granules and the ER creates a subcellular compartment-the TIGER domain-with a biophysically and biochemically distinct environment from the cytoplasm. This compartment promotes 3'UTR-mediated interaction of SET with membrane proteins, thus allowing increased surface expression and functional diversity of proteins, including CD47 and PD-L1. The TIGER domain is a subcellular compartment that enables formation of specific and functionally relevant protein-protein interactions that cannot be established outside.


Subject(s)
3' Untranslated Regions , Cytoplasmic Granules/metabolism , Endoplasmic Reticulum/metabolism , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Animals , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Butyrate Response Factor 1 , CD47 Antigen/genetics , CD47 Antigen/metabolism , Cytoplasmic Granules/genetics , Drosophila melanogaster , Endoplasmic Reticulum/genetics , HEK293 Cells , Humans , MCF-7 Cells , Mice , NIH 3T3 Cells , Nuclear Proteins/genetics , Protein Domains , RNA-Binding Proteins/genetics
2.
Nat Immunol ; 18(6): 683-693, 2017 06.
Article in English | MEDLINE | ID: mdl-28394372

ABSTRACT

RNA-binding proteins of the ZFP36 family are best known for inhibiting the expression of cytokines through binding to AU-rich elements in the 3' untranslated region and promoting mRNA decay. Here we identified an indispensable role for ZFP36L1 as the regulator of a post-transcriptional hub that determined the identity of marginal-zone B cells by promoting their proper localization and survival. ZFP36L1 controlled a gene-expression program related to signaling, cell adhesion and locomotion; it achieved this in part by limiting expression of the transcription factors KLF2 and IRF8, which are known to enforce the follicular B cell phenotype. These mechanisms emphasize the importance of integrating transcriptional and post-transcriptional processes by RNA-binding proteins for maintaining cellular identity among closely related cell types.


Subject(s)
B-Lymphocytes/immunology , Cell Adhesion/genetics , Cell Movement/genetics , Nuclear Proteins/genetics , RNA-Binding Proteins/genetics , Animals , Butyrate Response Factor 1 , Cell Adhesion/immunology , Cell Movement/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Regulation/genetics , High-Throughput Nucleotide Sequencing , Interferon Regulatory Factors/genetics , Kruppel-Like Transcription Factors/genetics , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Mice , Nuclear Proteins/immunology , Phenotype , RNA-Binding Proteins/immunology , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Signal Transduction
3.
J Virol ; 98(9): e0111424, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39194213

ABSTRACT

Zinc finger protein 36 (ZFP36) is a key regulator of inflammatory and cytokine production. However, the interplay between swine zinc-finger protein 36 (sZFP36) and foot-and-mouth disease virus (FMDV) has not yet been reported. Here, we demonstrate that overexpression of sZFP36 restricted FMDV replication, while the knockdown of sZFP36 facilitated FMDV replication. To subvert the antagonism of sZFP36, FMDV decreased sZFP36 protein expression through its non-structural protein 3C protease (3Cpro). Our results also suggested that 3Cpro-mediated sZFP36 degradation was dependent on its protease activity. Further investigation revealed that both N-terminal and C-terminal-sZFP36 could be degraded by FMDV and FMDV 3Cpro. In addition, both N-terminal and C-terminal-sZFP36 decreased FMDV replication. Moreover, sZFP36 promotes the degradation of FMDV structural proteins VP3 and VP4 via the CCCH-type zinc finger and NES domains of sZFP36. Together, our results confirm that sZFP36 is a host restriction factor that negatively regulates FMDV replication.IMPORTANCEFoot-and-mouth disease (FMD) is an infectious disease of animals caused by the pathogen foot-and-mouth disease virus (FMDV). FMD is difficult to prevent and control because there is no cross-protection between its serotypes. Thus, we designed this study to investigate virus-host interactions. We first demonstrate that swine zinc-finger protein 36 (sZFP36) impaired FMDV structural proteins VP3 and VP4 to suppress viral replication. To subvert the antagonism of sZFP36, FMDV and FMDV 3Cpro downregulate sZFP36 expression to facilitate FMDV replication. Taken together, the present study reveals a previously unrecognized antiviral mechanism for ZFP36 and elucidates the role of FMDV in counteracting host antiviral activity.


Subject(s)
Foot-and-Mouth Disease Virus , Foot-and-Mouth Disease , Virus Replication , Foot-and-Mouth Disease Virus/genetics , Foot-and-Mouth Disease Virus/metabolism , Animals , Swine , Foot-and-Mouth Disease/virology , Foot-and-Mouth Disease/metabolism , Viral Proteins/metabolism , Viral Proteins/genetics , 3C Viral Proteases/metabolism , Cell Line , Host-Pathogen Interactions , HEK293 Cells , Proteolysis , Butyrate Response Factor 1/metabolism , Cysteine Endopeptidases/metabolism , Cysteine Endopeptidases/genetics
4.
J Mol Cell Cardiol ; 192: 13-25, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38653384

ABSTRACT

The RNA-binding zinc finger protein 36 (ZFP36) family participates in numerous physiological processes including transition and differentiation through post-transcriptional regulation. ZFP36L1 is a member of the ZFP36 family. This study aimed to evaluate the role of ZFP36L1 in restenosis. We found that the expression of ZFP36L1 was inhibited in VSMC-phenotypic transformation induced by TGF-ß, PDGF-BB, and FBS and also in the rat carotid injury model. In addition, we found that the overexpression of ZFP36L1 inhibited the proliferation and migration of VSMCs and promoted the expression of VSMC contractile genes; whereas ZFP36L1 interference promoted the proliferation and migration of VSMCs and suppressed the expression of contractile genes. Furthermore, the RNA binding protein immunoprecipitation and double luciferase reporter gene experiments shows that ZFP36L1 regulates the phenotypic transformation of VSMCs through the posttranscriptional regulation of KLF16. Finally, our research results in the rat carotid balloon injury animal model further confirmed that ZFP36L1 regulates the phenotypic transformation of VSMCs through the posttranscriptional regulation of KLF16 and further plays a role in vascular injury and restenosis in vivo.


Subject(s)
Butyrate Response Factor 1 , Cell Proliferation , Kruppel-Like Transcription Factors , Muscle, Smooth, Vascular , Vascular System Injuries , Animals , Humans , Male , Rats , Butyrate Response Factor 1/metabolism , Butyrate Response Factor 1/genetics , Cell Movement/genetics , Disease Models, Animal , Gene Expression Regulation , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Rats, Sprague-Dawley , RNA Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vascular System Injuries/metabolism , Vascular System Injuries/genetics , Vascular System Injuries/pathology
5.
Cell Immunol ; 403-404: 104859, 2024.
Article in English | MEDLINE | ID: mdl-39067169

ABSTRACT

Asthma is a chronic inflammatory airway disease, in which inflammatory cytokines play a pivotal role. The zinc finger binding protein 36 (ZFP36) family includes ZFP36, ZFP36L1, and ZFP36L2 and is among the RNA-binding proteins (RBPs) reported to cause inflammation. The present study aimed to clarify the roles of the ZFP36 family in asthma, particularly highlighting the relationship between the ZFP36 family and Th2 cells, which are key players in type 2 inflammation in asthma. Real-time PCR analysis revealed the preferential expression of ZFP36 family mRNAs in human white blood cells. Gene expression analysis using public datasets from the GEO database (https://www.ncbi.nlm.nih.gov/gds) showed significantly suppressed expression of ZFP36 family mRNAs in patients with asthma compared to that in healthy controls. Using multiple cytokine assays, Th2 cell transfection with ZFP36 family siRNAs enhanced the expression of inflammatory cytokines IL-8, IFN-γ, CCL3/MIP-1α, CCL4/MIP-1ß, and TNF-α and cell surface molecules CCR4 (CD194) and PSGL-1 (CD162). Treatment with IL-2, 4, and 15 significantly suppressed, and corticosteroid significantly enhanced the expressions of ZFP36 family mRNAs by Th2 cells. In conclusion, the ZFP36 family expressed by Th2 cells was suppressed in patients with asthma, leading to the enhanced expression of cytokines and cell surface molecules. Suppressed ZFP36 expression in asthma may be involved in the enhancement of airway inflammation, and the ZFP36 family may be a therapeutic target for inflammatory diseases, including asthma.


Subject(s)
Asthma , Cytokines , Th2 Cells , Tristetraprolin , Humans , Asthma/immunology , Asthma/metabolism , Tristetraprolin/metabolism , Tristetraprolin/genetics , Cytokines/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Inflammation/immunology , Female , Male , Adult , Gene Expression Regulation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors , Butyrate Response Factor 1
6.
Respir Res ; 25(1): 67, 2024 Feb 05.
Article in English | MEDLINE | ID: mdl-38317146

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a leading aging related cause of global mortality. Small airway narrowing is recognized as an early and significant factor for COPD development. Senescent fibroblasts were observed to accumulate in lung of COPD patients and promote COPD progression through aberrant extracellular matrix (ECM) deposition and senescence-associated secretory phenotype (SASP). On the basis of our previous study, we further investigated the the causes for the increased levels of miR-377-3p in the blood of COPD patients, as well as its regulatory function in the pathological progression of COPD. We found that the majority of up-regulated miR-377-3p was localized in lung fibroblasts. Inhibition of miR-377-3p improved chronic smoking-induced COPD in mice. Mechanistically, miR-377-3p promoted senescence of lung fibroblasts, while knockdown of miR-377-3p attenuated bleomycin-induced senescence in lung fibroblasts. We also identified ZFP36L1 as a direct target for miR-377-3p that likely mediated its pro senescence activity in lung fibroblasts. Our data reveal that miR-377-3p is crucial for COPD pathogenesis, and may serve as a potential target for COPD therapy.


Subject(s)
Butyrate Response Factor 1 , MicroRNAs , Pulmonary Disease, Chronic Obstructive , Animals , Humans , Mice , Aging , Butyrate Response Factor 1/metabolism , Cellular Senescence/genetics , Fibroblasts/metabolism , Lung/metabolism , MicroRNAs/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism
7.
Int J Mol Sci ; 25(6)2024 Mar 07.
Article in English | MEDLINE | ID: mdl-38542074

ABSTRACT

Lethal toxin (LT) is the critical virulence factor of Bacillus anthracis, the causative agent of anthrax. One common symptom observed in patients with anthrax is thrombocytopenia, which has also been observed in mice injected with LT. Our previous study demonstrated that LT induces thrombocytopenia by suppressing megakaryopoiesis, but the precise molecular mechanisms behind this phenomenon remain unknown. In this study, we utilized 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced megakaryocytic differentiation in human erythroleukemia (HEL) cells to identify genes involved in LT-induced megakaryocytic suppression. Through cDNA microarray analysis, we identified Dachshund homolog 1 (DACH1) as a gene that was upregulated upon TPA treatment but downregulated in the presence of TPA and LT, purified from the culture supernatants of B. anthracis. To investigate the function of DACH1 in megakaryocytic differentiation, we employed short hairpin RNA technology to knock down DACH1 expression in HEL cells and assessed its effect on differentiation. Our data revealed that the knockdown of DACH1 expression suppressed megakaryocytic differentiation, particularly in polyploidization. We demonstrated that one mechanism by which B. anthracis LT induces suppression of polyploidization in HEL cells is through the cleavage of MEK1/2. This cleavage results in the downregulation of the ERK signaling pathway, thereby suppressing DACH1 gene expression and inhibiting polyploidization. Additionally, we found that known megakaryopoiesis-related genes, such as FOSB, ZFP36L1, RUNX1, FLI1, AHR, and GFI1B genes may be positively regulated by DACH1. Furthermore, we observed an upregulation of DACH1 during in vitro differentiation of CD34-megakaryocytes and downregulation of DACH1 in patients with thrombocytopenia. In summary, our findings shed light on one of the molecular mechanisms behind LT-induced thrombocytopenia and unveil a previously unknown role for DACH1 in megakaryopoiesis.


Subject(s)
Anthrax , Bacillus anthracis , Leukemia, Erythroblastic, Acute , Thrombocytopenia , Animals , Humans , Mice , Antigens, Bacterial/metabolism , Bacillus anthracis/metabolism , Butyrate Response Factor 1/metabolism , Cell Differentiation , Thrombocytopenia/chemically induced , Thrombocytopenia/genetics
8.
J Virol ; 96(1): e0166521, 2022 01 12.
Article in English | MEDLINE | ID: mdl-34643435

ABSTRACT

Zinc-finger protein 36, CCCH type-like 1 (ZFP36L1), containing tandem CCCH-type zinc-finger motifs with an RNA-binding property, plays an important role in cellular RNA metabolism mainly by RNA decay pathways. Recently, we demonstrated that human ZFP36L1 has potent antiviral activity against influenza A virus infection. However, its role in the host defense response against flaviviruses has not been addressed. Here, we demonstrate that ZFP36L1 functions as a host innate defender against flaviviruses, including Japanese encephalitis virus (JEV) and dengue virus (DENV). Overexpression of ZFP36L1 reduced JEV and DENV infection, and ZFP36L1 knockdown enhanced viral replication. ZFP36L1 destabilized the JEV genome by targeting and degrading viral RNA mediated by both 5'-3' XRN1 and 3'-5' RNA-exosome RNA decay pathways. Mutation in both zinc-finger motifs of ZFP36L1 disrupted RNA-binding and antiviral activity. Furthermore, the viral RNA sequences specifically recognized by ZFP36L1 were mapped to the 3'-untranslated region of the JEV genome with the AU-rich element (AUUUA) motif. We extend the function of ZFP36L1 to host antiviral defense by directly binding and destabilizing the viral genome via recruiting cellular mRNA decay machineries. IMPORTANCE Cellular RNA-binding proteins are among the first lines of defense against various viruses, particularly RNA viruses. ZFP36L1 belongs to the CCCH-type zinc-finger protein family and has RNA-binding activity; it has been reported to bind directly to the AU-rich elements (AREs) of a subset of cellular mRNAs and then lead to mRNA decay by recruiting mRNA-degrading enzymes. However, the antiviral potential of ZFP36L1 against flaviviruses has not yet been fully demonstrated. Here, we reveal the antiviral potential of human ZFP36L1 against Japanese encephalitis virus (JEV) and dengue virus (DENV). ZFP36L1 specifically targeted the ARE motif within viral RNA and triggered the degradation of viral RNA transcripts via cellular degrading enzymes 5'-3' XRN1 and 3'-5' RNA exosome. These findings provide mechanistic insights into how human ZFP36L1 serves as a host antiviral factor to restrict flavivirus replication.


Subject(s)
Butyrate Response Factor 1/metabolism , Exoribonucleases/metabolism , Exosome Multienzyme Ribonuclease Complex/metabolism , Flavivirus Infections/metabolism , Flavivirus Infections/virology , Flavivirus/physiology , Microtubule-Associated Proteins/metabolism , RNA Stability , Virus Replication , 3' Untranslated Regions , Amino Acid Motifs , Butyrate Response Factor 1/chemistry , Dengue Virus/physiology , Encephalitis Virus, Japanese/physiology , Host-Pathogen Interactions , Humans , Protein Binding , Protein Interaction Domains and Motifs , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Binding Proteins
9.
Am J Pathol ; 192(2): 208-225, 2022 02.
Article in English | MEDLINE | ID: mdl-34774847

ABSTRACT

Zinc finger protein 36 like 1 (ZFP36L1) enhances the turnover of mRNAs containing AU-rich elements (AREs) in their 3'-untranslated regions (3'UTR). The physiological and pathological functions of ZFP36L1 in liver, however, remain largely unknown. Liver-specific ZFP36L1-deficient (Zfp36l1flox/flox/Cre+; L1LKO) mice were generated to investigate the role of ZFP36L1 in liver physiology and pathology. Under normal conditions, the L1LKO mice and their littermate controls (Zfp36l1flox/flox/Cre-; L1FLX) appeared normal. When fed a Lieber-DeCarli liquid diet containing alcohol, L1LKO mice were significantly protected from developing alcohol-induced hepatic steatosis, injury, and inflammation compared with L1FLX mice. Most importantly, fibroblast growth factor 21 (Fgf21) mRNA was significantly increased in the livers of alcohol diet-fed L1LKO mice compared with the alcohol diet-fed L1FLX group. The Fgf21 mRNA contains three AREs in its 3'UTR, and Fgf21 3'UTR was directly regulated by ZFP36L1 in luciferase reporter assays. Steady-state levels of Fgf21 mRNA were significantly decreased by wild-type ZFP36L1, but not by a non-binding zinc finger ZFP36L1 mutant. Finally, wild-type ZFP36L1, but not the ZFP36L1 mutant, bound to the Fgf21 3'UTR ARE RNA probe. These results demonstrate that ZFP36L1 inactivation protects against alcohol-induced hepatic steatosis and liver injury and inflammation, possibly by stabilizing Fgf21 mRNA. These findings suggest that the modulation of ZFP36L1 may be beneficial in the prevention or treatment of human alcoholic liver disease.


Subject(s)
3' Untranslated Regions , Butyrate Response Factor 1/metabolism , Fatty Liver, Alcoholic/metabolism , Fibroblast Growth Factors/metabolism , Liver/metabolism , RNA Stability , Animals , Butyrate Response Factor 1/genetics , Fatty Liver, Alcoholic/genetics , Fatty Liver, Alcoholic/pathology , Fibroblast Growth Factors/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Liver/pathology , Mice , Mice, Knockout , Mutation
10.
Neurol Sci ; 44(6): 2137-2148, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36658410

ABSTRACT

The majority of the biomarkers were associated with the diagnosis of epilepsy and few of them can be applied to predict the response to antiseizure medications (ASMs). In this study, we identified 26 significantly up-regulated genes and 32 down-regulated genes by comparing the gene expression profiles of patients with epilepsy that responded to valproate with those without applying any ASM. The results of gene set enrichment analysis indicated that the ferroptosis pathway was significantly impacted (p = 0.0087) in patients who responded to valproate. Interestingly, the gene NCOA4 in this pathway exhibited significantly different expression levels between the two groups, indicating that NCOA4 could serve as a potential biomarker to better understand the mechanism of valproate resistance. In addition, six up-regulated genes SF3A2, HMGN2, PABPN1, SSBP3, EFTUD2, and CREB3L2 as well as six down-regulated genes ZFP36L1, ACRC, SUB1, CALM2, TLK1, and STX2 also showed significantly different expression patterns between the two groups. Moreover, based on the gene expression profiles of the patients with the treatment of valproate, carbamazepine, and phenytoin, we proposed a strategy for predicting the response to the ASMs by using the Connectivity Map scoring method. Our findings could be helpful for better understanding the mechanisms of drug resistance of ASMs and improving the clinical treatment of epilepsy.


Subject(s)
Carbamazepine , Valproic Acid , Humans , Pilot Projects , Valproic Acid/pharmacology , Valproic Acid/therapeutic use , Phenytoin , Research Design , Transcription Factors , Anticonvulsants/pharmacology , Anticonvulsants/therapeutic use , Butyrate Response Factor 1 , Protein Serine-Threonine Kinases , Poly(A)-Binding Protein I , Peptide Elongation Factors
11.
Int J Mol Sci ; 24(6)2023 Mar 12.
Article in English | MEDLINE | ID: mdl-36982508

ABSTRACT

The association between Porphyromonas gingivalis (P. gingivalis) and Alzheimer's disease (AD) remains unclear. The major aim of this study was to elucidate the role of genes and molecular targets in P. gingivalis-associated AD. Two Gene Expression Omnibus (GEO) datasets, GSE5281 for AD (n = 84 Alzheimer's, n = 74 control) and GSE9723 (n = 4 P. gingivalis, n = 4 control), were downloaded from the GEO database. Differentially expressed genes (DEGs) were obtained, and genes common to both diseases were drawn. Additionally, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analysis was performed from the top 100 genes (50 upregulated and 50 downregulated genes). We then proceeded with CMap analysis to screen for possible small drug molecules targeting these genes. Subsequently, we performed molecular dynamics simulations. A total of 10 common genes (CALD1, HES1, ID3, PLK2, PPP2R2D, RASGRF1, SUN1, VPS33B, WTH3DI/RAB6A, and ZFP36L1) were identified with a p-value < 0.05. The PPI network of the top 100 genes showed UCHL1, SST, CHGB, CALY, and INA to be common in the MCC, DMNC, and MNC domains. Out of the 10 common genes identified, only 1 was mapped in CMap. We found three candidate small drug molecules to be a fit for PLK2, namely PubChem ID: 24971422, 11364421, and 49792852. We then performed molecular docking of PLK2 with PubChem ID: 24971422, 11364421, and 49792852. The best target, 11364421, was used to conduct the molecular dynamics simulations. The results of this study unravel novel genes to P. gingivalis-associated AD that warrant further validation.


Subject(s)
Alzheimer Disease , Gene Expression Profiling , Humans , Alzheimer Disease/genetics , Porphyromonas gingivalis/genetics , Molecular Docking Simulation , Models, Molecular , Computational Biology/methods , Vesicular Transport Proteins/genetics , Butyrate Response Factor 1/genetics , Protein Phosphatase 2/genetics
12.
Nat Immunol ; 11(8): 717-24, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20622884

ABSTRACT

ZFP36L1 and ZFP36L2 are RNA-binding proteins (RBPs) that interact with AU-rich elements in the 3' untranslated region of mRNA, which leads to mRNA degradation and translational repression. Here we show that mice that lacked ZFP36L1 and ZFP36L2 during thymopoiesis developed a T cell acute lymphoblastic leukemia (T-ALL) dependent on the oncogenic transcription factor Notch1. Before the onset of T-ALL, thymic development was perturbed, with accumulation of cells that had passed through the beta-selection checkpoint without first expressing the T cell antigen receptor beta-chain (TCRbeta). Notch1 expression was higher in untransformed thymocytes in the absence of ZFP36L1 and ZFP36L2. Both RBPs interacted with evolutionarily conserved AU-rich elements in the 3' untranslated region of Notch1 and suppressed its expression. Our data establish a role for ZFP36L1 and ZFP36L2 during thymocyte development and in the prevention of malignant transformation.


Subject(s)
Nuclear Proteins/deficiency , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Tristetraprolin/deficiency , Amino Acid Sequence , Animals , Butyrate Response Factor 1 , Conserved Sequence , Humans , Immunophenotyping , Kaplan-Meier Estimate , Mice , Mice, Knockout , Molecular Sequence Data , Nuclear Proteins/genetics , Nuclear Proteins/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/immunology , Receptor, Notch1/genetics , Receptor, Notch1/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Sequence Alignment , Thymus Gland/growth & development , Transcription, Genetic , Tristetraprolin/genetics , Tristetraprolin/immunology
13.
Nucleic Acids Res ; 48(13): 7371-7384, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32556261

ABSTRACT

ZFP36L1, a CCCH-type zinc finger protein, is an RNA-binding protein that participates in controlling cellular mRNA abundance and turnover by posttranscriptional regulation. Here, we demonstrated that ZFP36L1 has an important role in host defense against influenza A virus (IAV) infection. Overexpression of ZFP36L1 reduced IAV replication via translational repression of HA, M and NS RNA segment transcripts. IAV infection upregulated cellular ZFP36L1 expression, and endogenous ZFP36L1 knockdown significantly enhanced IAV replication. ZFP36L1 directly binds to IAV NS1 mRNA in the cytoplasm and blocks the expression and function of NS1 protein. Mutation of CCCH-type zinc finger domains of ZFP36L1 lost its antiviral potential and NS1 mRNA binding. Thus, ZFP36L1 can act as a host innate defense by targeting HA, M and NS mRNA transcripts to suppress viral protein translation.


Subject(s)
Butyrate Response Factor 1/metabolism , Viral Matrix Proteins/genetics , Viral Nonstructural Proteins/genetics , A549 Cells , Animals , Binding Sites , Butyrate Response Factor 1/chemistry , Butyrate Response Factor 1/genetics , Dogs , HEK293 Cells , Humans , Influenza A virus/metabolism , Influenza A virus/physiology , Madin Darby Canine Kidney Cells , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Viral Matrix Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication
14.
Molecules ; 25(19)2020 Oct 01.
Article in English | MEDLINE | ID: mdl-33019656

ABSTRACT

An increased understanding of low-density lipoprotein receptor (LDLR) and its regulation may facilitate drug development for the treatment of hypercholesterolemia. Triciribine (TCN), which is a highly selective AKT inhibitor, increases the stability of LDLR mRNA downstream of extracellular signal-regulated kinase (ERK) in human hepatoma cells (HepG2). Here, a candidate approach was used in order to determine whether the RNA-binding proteins (RBPs) ZFP36 ring finger protein like 1 (ZFP36L1) and Hu antigen R (HuR) play a role in TCN-mediated stabilization of LDLR mRNA. The depletion of HuR led to a reduction of LDLR mRNA stability, an event that was more pronounced in TCN-treated cells. TCN was found to induce the translocation of nuclear HuR to cytoplasm in an ERK-dependent manner. ZFP36L1 depletion increased the stability of LDLR mRNA consistent with its destabilizing role. However, in contrast to HuR, TCN had no effect on LDLR mRNA turnover in ZFP36L1-depleted cells. TCN induced the phosphorylation of ZFP36L1 in an ERK/RSK-dependent manner and promoted its dissociation from the CCR4-NOT complex. In sum, these data suggest that TCN utilizes ERK signaling to increase the activity of HuR and inhibit ZFP36L1 to stabilize LDLR mRNA in HepG2 cells.


Subject(s)
ELAV-Like Protein 1/metabolism , RNA Stability/genetics , Receptors, LDL/genetics , Ribonucleosides/pharmacology , Butyrate Response Factor 1/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Exoribonucleases/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Hep G2 Cells , Humans , Phosphorylation/drug effects , Protein Transport/drug effects , RNA Stability/drug effects , Repressor Proteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism
15.
Haematologica ; 104(8): 1572-1579, 2019 08.
Article in English | MEDLINE | ID: mdl-30655376

ABSTRACT

In this study we interrogated the DNA methylome of myelofibrosis patients using high-density DNA methylation arrays. We detected 35,215 differentially methylated CpG, corresponding to 10,253 genes, between myelofibrosis patients and healthy controls. These changes were present both in primary and secondary myelofibrosis, which showed no differences between them. Remarkably, most differentially methylated CpG were located outside gene promoter regions and showed significant association with enhancer regions. This aberrant enhancer hypermethylation was negatively correlated with the expression of 27 genes in the myelofibrosis cohort. Of these, we focused on the ZFP36L1 gene and validated its decreased expression and enhancer DNA hypermethylation in an independent cohort of patients and myeloid cell-lines. In vitro reporter assay and 5'-azacitidine treatment confirmed the functional relevance of hyper-methylation of ZFP36L1 enhancer. Furthermore, in vitro rescue of ZFP36L1 expression had an impact on cell proliferation and induced apoptosis in SET-2 cell line indicating a possible role of ZFP36L1 as a tumor suppressor gene in myelofibrosis. Collectively, we describe the DNA methylation profile of myelofibrosis, identifying extensive changes in enhancer elements and revealing ZFP36L1 as a novel candidate tumor suppressor gene.


Subject(s)
Butyrate Response Factor 1/genetics , DNA Methylation , Enhancer Elements, Genetic/genetics , Epigenomics/methods , Primary Myelofibrosis/genetics , Apoptosis/drug effects , Butyrate Response Factor 1/metabolism , Butyrate Response Factor 1/pharmacology , Case-Control Studies , Cell Line , Cell Proliferation/drug effects , Epigenesis, Genetic , Genes, Tumor Suppressor , Humans
16.
Zygote ; 27(3): 143-152, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31182178

ABSTRACT

SummaryMuch effort has been devoted to improving the efficiency of animal cloning. The aim of this study was to investigate the effect of BRG1 contained in Xenopus egg extracts on the development of cloned mouse embryos. The results showed that mouse NIH/3T3 cells were able to express pluripotent genes after treatment with egg extracts, indicating that the egg extracts contained reprogramming factors. After co-injection of Xenopus egg extracts and single mouse cumulus cells into enucleated mouse oocytes, statistically higher pronucleus formation and development rates were observed in the egg Extract- co-injected group compared with those in the no egg extract-injected (NT) group (38-66% vs 18-34%, P<0.001). Removal of BRG1 protein from Xenopus egg extracts was conducted, and the BRG1-depleted extracts were co-injected with single donor cells into recipient oocytes. The results showed that the percentages of pronucleus formation were significantly higher in both BRG1-depleted and BRG1-intact groups than that in the nuclear transfer (NT) group (94, 64% vs 50%, P<0.05). Furthermore, percentages in the BRG1-depleted group were even higher than in the BRG1-intact group (94% vs 64%). More confined expression of Oct4 in the inner cell mass (ICM) was observed in the blastocyst derived from the egg extract-injected groups. However, Nanog expression was more contracted in the ICM of cloned blastocysts in the BRG1-depleted group than in the BGR1-intact group. Based on the present study, BRG1 might not play an essential role in reprogramming, but the factors enhancing pronucleus formation and development of cloned mouse embryos are contained in Xenopus egg extracts.


Subject(s)
Blastocyst/cytology , Cell Extracts/chemistry , Oocytes/metabolism , RNA-Binding Proteins/metabolism , Xenopus laevis/metabolism , Animals , Blastocyst/metabolism , Butyrate Response Factor 1 , Cloning, Organism/methods , Cumulus Cells/metabolism , Embryonic Development , Female , Gene Expression Regulation, Developmental , Mice , NIH 3T3 Cells , Nuclear Transfer Techniques , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Oocytes/cytology , RNA-Binding Proteins/genetics
17.
J Cell Mol Med ; 22(9): 4496-4506, 2018 09.
Article in English | MEDLINE | ID: mdl-29993187

ABSTRACT

Aplastic anaemia (AA) is a life-threatening hematopoietic disorder characterized by hypoplasia and pancytopenia with increasing fat cells in the bone marrow (BM). The BM-derived mesenchymal stem cells (MSCs) from AA are more susceptible to be induced into adipogenic differentiation compared with that from control, which may be causatively associated with the fatty BM and defective hematopoiesis of AA. Here in this study, we first demonstrated that levamisole displayed a significant suppressive effect on the in vitro adipogenic differentiation of AA BM-MSCs. Mechanistic investigation revealed that levamisole could increase the expression of ZFP36L1 which was subsequently demonstrated to function as a negative regulator of adipogenic differentiation of AA BM-MSCs through lentivirus-mediated ZFP36L1 knock-down and overexpression assay. Peroxisome proliferator-activated receptor gamma coactivator 1 beta (PPARGC1B) whose 3'-untranslated region bears adenine-uridine-rich elements was verified as a direct downstream target of ZFP36L1, and knock-down of PPARGC1B impaired the adipogenesis of AA BM-MSCs. Collectively, our work demonstrated that ZFP36L1-mediated post-transcriptional control of PPARGC1B expression underlies the suppressive effect of levamisole on the adipogenic differentiation of AA BM-MSCs, which not only provides novel therapeutic targets for alleviating the BM fatty phenomenon of AA patients, but also lays the theoretical and experimental foundation for the clinical application of levamisole in AA therapy.


Subject(s)
Adipocytes/drug effects , Adipogenesis/drug effects , Anemia, Aplastic/genetics , Butyrate Response Factor 1/genetics , Carrier Proteins/genetics , Levamisole/pharmacology , Mesenchymal Stem Cells/drug effects , Adipocytes/metabolism , Adipocytes/pathology , Adipogenesis/genetics , Adolescent , Adult , Anemia, Aplastic/metabolism , Anemia, Aplastic/pathology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Butyrate Response Factor 1/agonists , Butyrate Response Factor 1/metabolism , Carrier Proteins/metabolism , Case-Control Studies , Cell Differentiation , Female , Gene Expression Regulation , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Middle Aged , Primary Cell Culture , RNA-Binding Proteins , Signal Transduction
18.
J Cell Physiol ; 233(5): 4068-4076, 2018 05.
Article in English | MEDLINE | ID: mdl-28885689

ABSTRACT

Since the past 30 years, the prevalence of diabetes has more than doubled, making it an urgent challenge globally. We carried out systematic analysis with the public data of mRNA expression profiles in skeletal muscle to study the pathogenesis, since insulin resistance in the skeletal muscle is an early feature. We utilized three GEO datasets, containing total 60 cases and 63 normal samples. After the background removal, R package QC was utilized to finish the preprocessing of datasets. We obtained a dataset containing 2481 genes and 123 samples after the preprocessing. Quantitative quality control measures were calculated to represent the quality of these datasets. MetaDE package provides functions for conducting different systematic analysis methods for differential expression analysis. The GO term enrichment was carried out using PANTHER. Protein-protein interactions, drug-gene interactions, and genetic association of the identified differentially expressed genes were analyzed using STRING v10.0 online tool, DGIdb, and the Genetic Association Database, respectively. The datasets had good performances on IQC and EQC, which suggested that the datasets had good internal and external quality. Totally 96 differentially expressed genes were detected using 0.01 as cutoff of AW. The enriched GO terms were mainly associated with the response to glucocorticoid. There were seven genes involving in the gluconeogenesis were differentially expressed, which might be the potential treatment target for this disease. The closely connected networks and potential targets of existed drugs suggested that some of the drugs might be applied to the treatment of diabetes as well.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Gene Expression Regulation/genetics , Muscle, Skeletal/metabolism , RNA, Messenger/genetics , Butyrate Response Factor 1/genetics , Computational Biology , Cyclin-Dependent Kinase Inhibitor p21 , Databases, Genetic , Diabetes Mellitus, Type 2/pathology , Gene Expression Profiling/methods , Gene Regulatory Networks/genetics , Humans , Insulin Resistance/genetics , MicroRNAs , Muscle, Skeletal/pathology , Plasma Membrane Calcium-Transporting ATPases/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Transcriptome/genetics , Ubiquitin-Conjugating Enzymes/genetics , Uncoupling Protein 3/genetics
19.
Biochem Biophys Res Commun ; 501(2): 387-393, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29709483

ABSTRACT

The ZFP36 family is a prototypical member of a highly conserved group of proteins with CCCH-type RNA-binding domains, whose functional role and regulatory mechanism in mitotic cells remain obscure. In this study, we provide the first evidence that ZFP36L1 phosphorylation is modulated in a cell cycle-dependent manner. The C-terminal region of ZFP36L1 is critical for its cell cycle-dependent phosphorylation of this protein. We also suggest that the phosphorelay-dependent regulation of ZFP36L1 influences mitotic spindle organization. Thus, our data demonstrate a new class of regulatory mechanism for CCCH-type zinc-finger proteins in cell cycle control.


Subject(s)
Butyrate Response Factor 1/metabolism , Cell Cycle Proteins/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals , Butyrate Response Factor 1/genetics , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Chromosome Segregation , Embryo, Nonmammalian/cytology , HeLa Cells , Humans , Phosphorylation , Serine/metabolism , Spindle Apparatus/physiology , Xenopus Proteins/genetics
20.
Clin Genet ; 94(2): 259-263, 2018 08.
Article in English | MEDLINE | ID: mdl-29722023

ABSTRACT

Seven new risk coding variants have been identified through an exome-wide association study (EWAS), which studied the contributions of protein-coding variants to leprosy susceptibility. But some potential susceptibility loci were not studied in the previous EWAS study because of the project consideration. Seventeen unstudied potential susceptibility loci of the previous EWAS were validated in 3169 cases and 9814 controls in this study. Four disease-associated exonic loci were identified: rs671 in ALDH2 (P = 2.0 × 10-20 , odds ratio [OR] = 1.35), rs13259978 in SLC7A2 (P = 1.74 × 10-8 , OR = 1.28), rs925368 in GIT2 (P = 9.18 × 10-17 , OR = 1.44), and rs75680863 in TCN2 (P = 8.37 × 10-21 , OR = 0.74). Potentially implicating ZFP36L1 as a new susceptibility gene, 1 intergenic single nucleotide polymorphism (SNP), rs1465788 (P = 7.81 × 10-6 , OR = 0.88), was also suggested to be associated with leprosy. A luciferase reporter assay showed that the rs1465788 risk allele notably decreased the transcription activity of the flanking sequence. These findings suggest the possible involvement of lipid metabolism, NF-κB homeostasis and macrophage antimicrobial pathways in leprosy pathogenesis.


Subject(s)
Genetic Predisposition to Disease , Genome-Wide Association Study , Leprosy/genetics , Aldehyde Dehydrogenase, Mitochondrial/genetics , Asian People/genetics , Butyrate Response Factor 1/genetics , Cationic Amino Acid Transporter 2/genetics , DNA, Intergenic/genetics , Exome/genetics , Exons/genetics , Female , GTPase-Activating Proteins/genetics , Humans , Leprosy/physiopathology , Male , NF-kappa B/genetics , Polymorphism, Single Nucleotide/genetics , Transcobalamins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL