Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Basic Res Cardiol ; 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38834767

ABSTRACT

Nuclear factor of activated T cells 5 (NFAT5) is an osmosensitive transcription factor that is well-studied in renal but rarely explored in cardiac diseases. Although the association of Coxsackievirus B3 (CVB3) with viral myocarditis is well-established, the role of NFAT5 in this disease remains largely unexplored. Previous research has demonstrated that NFAT5 restricts CVB3 replication yet is susceptible to cleavage by CVB3 proteases. Using an inducible cardiac-specific Nfat5-knockout mouse model, we uncovered that NFAT5-deficiency exacerbates cardiac pathology, worsens cardiac function, elevates viral load, and reduces survival rates. RNA-seq analysis of CVB3-infected mouse hearts revealed the significant impact of NFAT5-deficiency on gene pathways associated with cytokine signaling and inflammation. Subsequent in vitro and in vivo investigation validated the disruption of the cytokine signaling pathway in response to CVB3 infection, evidenced by reduced expression of key cytokines such as interferon ß1 (IFNß1), C-X-C motif chemokine ligand 10 (CXCL10), interleukin 6 (IL6), among others. Furthermore, NFAT5-deficiency hindered the formation of stress granules, leading to a reduction of important stress granule components, including plakophilin-2, a pivotal protein within the intercalated disc, thereby impacting cardiomyocyte structure and function. These findings unveil a novel mechanism by which NFAT5 inhibits CVB3 replication and pathogenesis through the promotion of antiviral type I interferon signaling and the formation of cytoplasmic stress granules, collectively identifying NFAT5 as a new cardio protective protein.

2.
Int J Mol Sci ; 22(9)2021 May 04.
Article in English | MEDLINE | ID: mdl-34064510

ABSTRACT

The nuclear factor of activated T cells 5 (NFAT5) is well known for its sensitivity to cellular osmolarity changes, such as in the kidney medulla. Accumulated evidence indicates that NFAT5 is also a sensitive factor to stress signals caused by non-hypertonic stimuli such as heat shock, biomechanical stretch stress, ischaemia, infection, etc. These osmolality-related and -unrelated stimuli can induce NFAT5 upregulation, activation and nuclear accumulation, leading to its protective role against various detrimental effects. However, dysregulation of NFAT5 expression may cause pathological conditions in different tissues, leading to a variety of diseases. These protective or pathogenic effects of NFAT5 are dictated by the regulation of its target gene expression and activation of its signalling pathways. Recent studies have found a number of kinases that participate in the phosphorylation/activation of NFAT5 and related signal proteins. Thus, this review will focus on the NFAT5-mediated signal transduction pathways. As for the stimuli that upregulate NFAT5, in addition to the stresses caused by hyperosmotic and non-hyperosmotic environments, other factors such as miRNA, long non-coding RNA, epigenetic modification and viral infection also play an important role in regulating NFAT5 expression; thus, the discussion in this regard is another focus of this review. As the heart, unlike the kidneys, is not normally exposed to hypertonic environments, studies on NFAT5-mediated cardiovascular diseases are just emerging and rapidly progressing. Therefore, we have also added a review on the progress made in this field of research.


Subject(s)
Cardiovascular Diseases/genetics , Epigenesis, Genetic , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Transcription Factors/genetics , Virus Diseases/genetics , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , DNA Methylation , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Response/genetics , Histones/genetics , Histones/metabolism , Humans , Kidney Medulla/metabolism , Kidney Medulla/pathology , MicroRNAs/metabolism , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Osmolar Concentration , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction , Transcription Factors/metabolism , Virus Diseases/metabolism , Virus Diseases/pathology , Virus Diseases/virology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Front Microbiol ; 11: 767, 2020.
Article in English | MEDLINE | ID: mdl-32457708

ABSTRACT

Coxsackievirus B3 (CVB3) is the primary etiologic agent of viral myocarditis, a major heart disease that occurs predominantly in children and young adolescents. In the heart, intercalated disks (ICD) are important structural formations that connect adjacent cardiomyocytes to maintain cardiac architecture and mediate signal communication. Deficiency in ICD components, such as desmosome proteins, leads to heart dysfunction. γ-catenin, a component protein of desmosomes, normally binds directly to desmocollin-2 and desmoglein-2. In this study, we found that CVB3 infection downregulated γ-catenin at the protein level but not the mRNA level in mouse HL-1 cardiomyocytes. We further found that this reduction of γ-catenin protein is a result of ubiquitin proteasome-mediated degradation, since the addition of proteasome inhibitor MG132 inhibited γ-catenin downregulation. In addition, we found that desmocollin-2 and desmoglein-2 were cleaved by both viral protease 3C and virus-activated cellular caspase, respectively. These cleavages led to the release of bound γ-catenin from the desmosome into the cytosol, resulting in rapid degradation of γ-catenin. Since γ-catenin shares high sequence homology with ß-catenin in binding the TCF/LEF transcription factor, we further studied the effect of γ-catenin degradation on Wnt/ß-catenin signaling. Luciferase assay showed that γ-catenin expression inhibited Wnt/ß-catenin signaling. This finding was substantiated by qPCR to show that overexpression of γ-catenin downregulated transcription of Wnt signal target genes, c-myc and MMP9, while silencing γ-catenin upregulated these target genes. Finally, we demonstrated that γ-catenin expression inhibited CVB3 replication. In search for the underlying mechanism, we found that silencing γ-catenin caused down-regulation of interferon-ß and its stimulated antiviral genes MDA5, MAVS, and ISG15. Taken together, our results indicate, for the first time, that CVB3 infection causes cardiomyocyte death through, at least in part, direct damage to the desmosome structure and reduction of γ-catenin protein, which in return promotes Wnt/ß-catenin signaling and downregulates interferon-ß stimulated immune responses.

4.
Cell Microbiol ; 22(7): e13198, 2020 07.
Article in English | MEDLINE | ID: mdl-32083795

ABSTRACT

Our previous study of coxsackievirus B3 (CVB3)-induced unfolded protein responses (UPR) found that overexpression of ATF6a enhances CVB3 VP1 capsid protein production and increases viral particle formation. These findings implicate that ATF6a signalling benefits CVB3 replication. However, the mechanism by which ATF6a signalling is transduced to promote virus replication is unclear. In this study, using a Tet-On inducible ATF6a HeLa cell line, we found that ATF6a signalling downregulated the protein expression of the endoplasmic reticulum (ER) degradation-enhancing α-mannosidase-like protein 1 (EDEM1), resulting in accumulation of CVB3 VP1 protein; in contrast, expression of a dominant negative ATF6a had the opposite effect. Furthermore, we found that EDEM1 was cleaved by both CVB3 protease 3C and virus-activated caspase and subsequently degraded via the ubiquitin-proteasome pathway. However, overexpression of EDEM1 caused VP1 degradation, likely via a glycosylation-independent and ubiquitin-lysosome pathway. Finally, we demonstrated that CRISPR/Cas9-mediated knockout of EDEM1 increased VP1 accumulation and thus CVB3 replication. This is the first study to report the ER protein quality control of non-enveloped RNA virus and reveals a novel mechanism by which CVB3 evades host ER quality control pathways through cleavage and degradation of the UPR target gene EDEM1, to ultimately benefit its own replication.


Subject(s)
Activating Transcription Factor 6/metabolism , Coxsackievirus Infections/virology , Membrane Proteins/metabolism , Signal Transduction/physiology , Unfolded Protein Response/physiology , Virus Replication/physiology , Animals , Down-Regulation , Endoplasmic Reticulum Chaperone BiP , Enterovirus , Gene Knockout Techniques , Glycosylation , HeLa Cells , Humans , Membrane Proteins/genetics , Mice , Proteolysis , alpha-Mannosidase/metabolism
5.
Article in English | MEDLINE | ID: mdl-31396490

ABSTRACT

Myocarditis, inflammation of the heart muscle, affects all demographics and is a major cause of sudden and unexpected death in young people. It is most commonly caused by viral infections of the heart, with coxsackievirus B3 (CVB3) being among the most prevalent pathogens. To understand the molecular pathogenesis of CVB3 infection and provide strategies for developing treatments, we examined the role of a key nuclear pore protein 98 (NUP98) in the setting of viral myocarditis. NUP98 was cleaved as early as 2 h post-CVB3 infection. This cleavage was further verified through both the ectopic expression of viral proteases and in vitro using purified recombinant CVB3 proteases (2A and 3C), which demonstrated that CVB3 2A but not 3C is responsible for this cleavage. By immunostaining and confocal imaging, we observed that cleavage resulted in the redistribution of NUP98 to punctate structures in the cytoplasm. Targeted siRNA knockdown of NUP98 during infection further increased viral protein expression and viral titer, and reduced cell viability, suggesting a potential antiviral role of NUP98. Moreover, we discovered that expression levels of neuregulin-1 (NRG1), a cardioprotective gene, and presenilin-1 (PSEN1), a cellular protease processing the tyrosine kinase receptor ERBB4 of NRG1, were reliant upon NUP98 and were downregulated during CVB3 infection. In addition, expression of these NUP98 target genes in myocardium tissue not only occurred at an earlier phase of infection, but also appeared in areas away from the initial inflammatory regions. Collectively, CVB3-induced cleavage of NUP98 and subsequent impairment of the cardioprotective NRG1-ERBB4/PSEN1 signaling cascade may contribute to increased myocardial damage in the context of CVB3-induced myocarditis. To our knowledge, this is the first study to demonstrate the link between NUP98 and the NRG1 signaling pathway in viral myocarditis.


Subject(s)
Coxsackievirus Infections/pathology , Cysteine Endopeptidases/metabolism , Enterovirus B, Human/growth & development , Host-Pathogen Interactions , Myocarditis/pathology , Myocardium/pathology , Nuclear Pore Complex Proteins/metabolism , Viral Proteins/metabolism , Animals , Disease Models, Animal , Gene Expression , HeLa Cells , Humans , Mice , Models, Biological , Neuregulin-1/metabolism , Presenilin-1/metabolism , Protein Transport , Proteolysis
6.
Heart Fail Rev ; 24(1): 115-132, 2019 01.
Article in English | MEDLINE | ID: mdl-30288656

ABSTRACT

Intercalated discs (ICDs) are highly orchestrated structures that connect neighboring cardiomyocytes in the heart. Three major complexes are distinguished in ICD: desmosome, adherens junction (AJ), and gap junction (GJ). Desmosomes are major cell adhesion junctions that anchor cell membrane to the intermediate filament network; AJs connect the actin cytoskeleton of adjacent cells; and gap junctions metabolically and electrically connect the cytoplasm of adjacent cardiomyocytes. All these complexes work as a single unit, the so-called area composita, interdependently rather than individually. Mutation or altered expression of ICD proteins results in various cardiac diseases, such as ARVC (arrhythmogenic right ventricular cardiomyopathy), dilated cardiomyopathy, and hypotrophy cardiomyopathy, eventually leading to heart failure. In this article, we first review the recent findings on the structural organization of ICD and their functions and then focus on the recent advances in molecular pathogenesis of the ICD-related heart diseases, which include two major areas: i) the ICD gene mutations in cardiac diseases, and ii) the involvement of ICD proteins in signal transduction pathways leading to myocardium remodeling and eventual heart failure. These major ICD-related signaling pathways include Wnt/ß-catenin pathway, p38 MAPK cascade, Rho-dependent serum response factor (SRF) signaling, calcineurin/NFAT signaling, Hippo kinase cascade, etc., which are differentially regulated in pathological conditions.


Subject(s)
Adherens Junctions/metabolism , Cell Adhesion , Desmosomes/metabolism , Gap Junctions/metabolism , Heart Diseases/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Adherens Junctions/chemistry , Adherens Junctions/genetics , Animals , Desmosomes/chemistry , Desmosomes/genetics , Gap Junctions/chemistry , Gap Junctions/genetics , Heart Diseases/genetics , Humans , Mutation, Missense , Signal Transduction
7.
Cell Microbiol ; 19(7)2017 07.
Article in English | MEDLINE | ID: mdl-28095607

ABSTRACT

We previously demonstrated that coxsackievirus B3 (CVB3) infection upregulated heat shock protein 70 (Hsp70) and promoted CVB3 multiplication. Here, we report the underlying mechanism by which Hsp70 enhances viral RNA translation. By using an Hsp70-overexpressing cell line infected with CVB3, we found that Hsp70 enhanced CVB3 VP1 translation at two stages. First, Hsp70 induced upregulation of VP1 translation at the initiation stage via upregulation of internal ribosome entry site trans-acting factor lupus autoantigen protein and activation of eIF4E binding protein 1, a cap-dependent translation suppressor. Second, we found that Hsp70 increased CVB3 VP1 translation by enhancing translation elongation. This was mediated by the Akt-mammalian target of rapamycin complex 1 signal cascade, which led to the activation of eukaryotic elongation factor 2 via p70S6K- and cell division cycle protein 2 homolog (Cdc2)-mediated phosphorylation and inactivation of eukaryotic elongation factor 2 kinase. We also determined the position of Cdc2 in this signal pathway, indicating that Cdc2 is regulated by mammalian target of rapamycin complex 1. This signal transduction pathway was validated using a number of specific pharmacological inhibitors, short interfering RNAs (siRNAs) and a dominant negative Akt plasmid. Because Hsp70 is a central component of the cellular network of molecular chaperones enhancing viral replication, these data may provide new strategies to limit this viral infection.


Subject(s)
CDC2 Protein Kinase/metabolism , Enterovirus B, Human/metabolism , HSP70 Heat-Shock Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Peptide Chain Elongation, Translational/physiology , Peptide Chain Initiation, Translational/physiology , Protein Biosynthesis/physiology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Cell Cycle Proteins , Cell Line, Tumor , Coxsackievirus Infections/virology , Enterovirus B, Human/growth & development , Eukaryotic Initiation Factor-2/metabolism , HSP70 Heat-Shock Proteins/biosynthesis , HeLa Cells , Humans , Phosphoproteins/biosynthesis , Phosphorylation , RNA Interference , RNA, Small Interfering/genetics , Viral Structural Proteins/biosynthesis , Virus Replication/physiology
8.
Biochem J ; 473(4): 473-85, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26621875

ABSTRACT

CVB3 (coxsackievirus 3) is a primary causal agent of viral myocarditis. Emodin is a natural compound isolated from certain plant roots. In the present study, we found that emodin inhibited CVB3 replication in vitro and in mice, and now we report an unrecognized mechanism by which emodin inhibits CVB3 replication through suppression of viral protein translation via multiple pathways. On one hand, emodin treatment inhibited Akt/mTOR (mammalian target of rapamycin) signalling and activated 4EBP1 (eukaryotic initiation factor 4R-binding protein 1), leading to suppression of translation initiation of ribosomal protein L32 encoded by a 5'-TOP (terminal oligopyrimidine) mRNA. On the other hand, emodin treatment differentially regulated multiple signal cascades, including Akt/mTORC1/p70(S6K) (p70 S6 kinase), ERK1/2 (extracellular-signal-regulated kinase 1/2)/p90(RSK) (p90 ribosomal S6 kinase) and Ca(2+)/calmodulin, leading to activation of eEF2K (eukaryotic elongation factor 2 kinase) and subsequent inactivation of eEF2 (eukaryotic elongation factor 2), resulting in inhibition of CVB3 VP1 (viral protein 1) synthesis. These data imply that eEF2K is a major factor mediating cross-talk of different arms of signalling cascades in this signal network. This notion was verified by either overexpressing eEF2K or treating the cells with siRNAs or eEF2K inhibitor A484954. We showed further that the emodin-induced decrease in p70(S6K) phosphorylation plays a dominant positive role in activation of eEF2K and in turn in conferring the antiviral effect of emodin. This finding was further solidified by expressing constitutively active and dominant-negative Akt. Collectively, our data reveal that emodin inhibits viral replication through impairing translational machinery and suppression of viral translation elongation.


Subject(s)
Emodin/pharmacology , Enterovirus B, Human/drug effects , Protein Biosynthesis/drug effects , Signal Transduction/drug effects , Virus Replication/drug effects , Adenylate Kinase/metabolism , Animals , Calcium/metabolism , Cell Line, Transformed , Elongation Factor 2 Kinase/metabolism , Enterovirus B, Human/physiology , Male , Mice , Phosphorylation , Ribosomal Protein S6 Kinases/metabolism
9.
ACS Chem Biol ; 9(4): 1015-24, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24547890

ABSTRACT

We report here an isatin derivative 45 (ID45) against coxsackievirus B3 (CVB3) replication, which was synthesized based on a high-throughput screen of a unique natural product library. ID45 showed the most potent anti-CVB3 activity among the four synthesized compounds. Treatment of cells with ID45 before or after infection significantly reduced viral particle formation, resulting in protection of cells from virus-induced apoptosis. In addition, ID45 treatment caused remarkable up-regulation of glucose-regulated protein 78 (GRP78), a hallmark of endoplasmic reticulum (ER) stress and an indicator of enhanced cell viability. In identifying the ER stress response pathway induced by ID45, we found that ID45 activated PKR-like ER protein kinase (PERK) but failed to up-regulate eIF2α phosphorylation. Instead ID45 activated transcription factor Nrf2 (NF-E2-related factor-2), which is evidenced by its nuclear translocation and upregulation of its downstream target genes NQO1 (NAD(P)H quinone-oxidoreductase 1) and GCLM (glutamate-cysteine ligase, modifier subunit). This observation was further verified by using siRNAs of GRP78 or Nrf2, which blocked both the translocation of Nrf2 and up-regulation of its target genes, leading to aggressive viral replication and enhanced cell apoptosis. Finally, we found that ID45-induced up-regulation of NQO1 protected eIF4GI, a eukaryotic cap-dependent translation initiation factor, from cleavage by CVB3 protease and degradation by proteasomes. Taken together, our findings established that a novel antiviral mechanism of isatin derivative ID45 inhibits CVB3 replication by promoting cell survival through a PERK/Nrf2-dependent ER stress pathway, which benefits host cap-dependent translation but suppresses CVB3 cap-independent translation.


Subject(s)
Antiviral Agents/pharmacology , Gene Expression Regulation, Viral/drug effects , Isatin/chemistry , NF-E2-Related Factor 2/metabolism , eIF-2 Kinase/metabolism , Blotting, Western , Cell Survival/drug effects , Dose-Response Relationship, Drug , Endoplasmic Reticulum Chaperone BiP , HeLa Cells , Humans , Isatin/chemical synthesis , Isatin/pharmacology , Models, Biological , NF-E2-Related Factor 2/chemistry , NF-E2-Related Factor 2/genetics , Protein Biosynthesis/drug effects , Virus Replication/drug effects , eIF-2 Kinase/chemistry , eIF-2 Kinase/genetics
10.
Cell Microbiol ; 16(3): 411-24, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24134518

ABSTRACT

Previously we found that prolonged endoplasmic reticulum (ER) stress caused by coxsackievirus B3 (CVB3) infection led to p58(IPK) downregulation and subsequent cell apoptosis. This finding implies that p58(IPK) expression benefits cell survival and counteracts CVB3-induced apoptosis. In testing this hypothesis, we first found that PI3K/Akt survival pathway is more sensitive than ERK1/2 in response to p58(IPK) expression. This finding was further verified by silencing p58(IPK) with specific siRNAs, which led to the significant suppression of phosphorylation of Akt (p-Akt) but not ERK1/2. Further, using CVB3-infected cell line expressing dominant negative ATF6a (DN-ATF6a), we found that expression of p58(IPK) and p-Akt was significantly reduced, which led to the decreased cell viability. However, when the DN-ATF6a cells were transiently transfected with p58(IPK) , an opposite result was obtained. Finally, by CVB3 infection of cells stably expressing p58(IPK) , we found that CVB3-induced mitochondria-mediated apoptosis was suppressed, which was evidenced by the reduced cytochrome c release and upregulation of the mitochondrial membrane protein mitofusin 2. However, silencing p58(IPK) with either specific siRNAs or DN-ATF6a sensitized cells to CVB3-induced apoptosis. These results suggest that p58(IPK) suppresses CVB3-induced apoptosis through selective activation of PI3K/Akt pathway that requires activation of ATF6a and subsequently upregulates mitofusin 2.


Subject(s)
Apoptosis/genetics , Enterovirus/pathogenicity , HSP40 Heat-Shock Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Activating Transcription Factor 6/biosynthesis , Animals , Butadienes/pharmacology , Cell Line, Tumor , Cell Survival/genetics , Chromones/pharmacology , Coxsackievirus Infections , Cytochromes c/metabolism , Down-Regulation , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress/genetics , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , GTP Phosphohydrolases , HSP40 Heat-Shock Proteins/biosynthesis , HSP40 Heat-Shock Proteins/genetics , HeLa Cells , Humans , Mice , Mitochondria , Mitochondrial Proteins , Morpholines/pharmacology , Nitriles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/biosynthesis , RNA Interference , RNA, Small Interfering , Up-Regulation
11.
Cell Mol Life Sci ; 70(2): 277-91, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22842794

ABSTRACT

Coxsackievirus B3 (CVB3) is the primary causal agent of viral myocarditis. During infection, it hijacks host genes to favour its own replication. However, the underlying mechanism is still unclear. Although the viral receptor is an important factor for viral infectivity, other factors such as microRNAs (miRNA) may also play an essential role in its replication after host cell entry. miRNAs are post-transcriptional gene regulators involved in various fundamental biological processes as well as in diseases. To identify miRNAs involved in CVB3 pathogenesis, we performed microarray analysis of miRNAs using CVB3-infected murine hearts and identified miR-203 as one of the most upregulated candidates. We found that miR-203 upregulation is through the activation of protein kinase C/transcription factor AP-1 pathway. We further identified zinc finger protein-148 (ZFP-148), a transcription factor, as a novel target of miR-203. Ectopic expression of miR-203 downregulated ZFP-148 translation, increased cell viability and subsequently enhanced CVB3 replication. Silencing of ZFP-148 by siRNA showed similar effects on CVB3 replication. Finally, analyses of the signalling cascade downstream of ZFP-148 revealed that miR-203-induced suppression of ZFP-148 differentially regulated the expression of prosurvival and proapoptotic genes of the Bcl-2 family proteins as well as the cell cycle regulators. This altered gene expression promoted cell survival and growth, which provided a favourable environment for CVB3 replication, contributing to the further damage of the infected cells. Taken together, this study identified a novel target of miR-203 and revealed, for the first time, the molecular link between miR-203/ZFP-148 and the pathogenesis of CVB3.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Enterovirus B, Human/physiology , MicroRNAs/metabolism , Myocarditis/virology , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cell Survival , Coxsackievirus Infections/genetics , Coxsackievirus Infections/pathology , Enterovirus B, Human/pathogenicity , Gene Expression Regulation , Genes, bcl-2 , Heart/virology , Mice , MicroRNAs/biosynthesis , Oligonucleotide Array Sequence Analysis , Protein Kinase C/metabolism , RNA Interference , RNA, Small Interfering , Signal Transduction/genetics , Transcription Factor AP-1/metabolism , Up-Regulation , Virus Replication/genetics
12.
Front Microbiol ; 3: 92, 2012.
Article in English | MEDLINE | ID: mdl-22461781

ABSTRACT

Many virus infections and stresses can induce endoplasmic reticulum (ER) stress response, a host self-defense mechanism against viral invasion and stress. During this event, viral and cellular gene expression is actively regulated and often encounters a switching of the translation initiation from cap-dependent to internal ribosome-entry sites (IRES)-dependent. This switching is largely dependent on the mRNA structure of the 5' untranslated region (5' UTR) and on the particular stress stimuli. Picornaviruses and some other viruses contain IRESs within their 5' UTR of viral genome and employ an IRES-driven mechanism for translation initiation. Recently, a growing number of cellular genes involved in growth control, cell cycle progression and apoptosis were also found to contain one or more IRES within their long highly structured 5' UTRs. These genes initiate translation usually by a cap-dependent mechanism under normal physiological conditions; however, in certain environments, such as infection, starvation, and heat shock they shift translation initiation to an IRES-dependent modality. Although the molecular mechanism is not entirely understood, a number of studies have revealed that several cellular biochemical processes are responsible for the switching of translation initiation to IRES-dependent. These include the cleavage of translation initiation factors by viral and/or host proteases, phosphorylation (inactivation) of host factors for translation initiation, overproduction of homologous proteins of cap-binding protein eukaryotic initiation factors (eIF)4E, suppression of cap-binding protein eIF4E expression by specific microRNA, activation of enzymes for mRNA decapping, as well as others. Here, we summarize the recent advances in our understanding of the molecular mechanisms for the switching of translation initiation, particularly for the proteins involved in cell survival and apoptosis in the ER stress pathways during viral infections.

13.
Wiley Interdiscip Rev RNA ; 3(4): 469-81, 2012.
Article in English | MEDLINE | ID: mdl-22362726

ABSTRACT

Bacteriophage 29 (Phi29) packaging RNA (pRNA) is one of the key components in the viral DNA-packaging motor. It contains two functional domains facilitating the translocation of DNA into the viral capsid by interacting with other elements in the motor and promoting adenosine triphosphates hydrolysis. Through the connection between interlocking loops in adjacent pRNA monomers, pRNA functions in the form of multimer ring in the motor. Previous studies have addressed the unique structure and conformation of pRNA. However, there are different DNA-packaging models proposed for the viral genome transportation mechanism. The DNA-packaging ability and the unique features of pRNA have been attracting efforts to study its potential applications in nanotechnology. The pRNA has been proved to be a promising tool for delivering nucleic acid-based therapeutic molecules by covalent linkage with ribozymes, small interfering RNAs, aptamers, and artificial microRNAs. The flexibility in constructing dimers, trimers, and hexamers enables the assembly of polyvalent nanoparticles to carry drug molecules for therapeutic purposes, cell ligands for target delivery, image detector for drug entry monitoring, and endosome disrupter for drug release. Besides these fascinating pharmacological advantages, pRNA-based drug delivery has also been demonstrated to prolong the drug half life with minimal induction of immune response and toxicity.


Subject(s)
Bacillus Phages/physiology , Drug Delivery Systems , RNA, Viral/genetics , Animals , DNA Packaging
14.
Cell Microbiol ; 14(1): 133-46, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21981022

ABSTRACT

The IRG protein Irgm3 preserves cell survival during coxsackievirus B3 (CVB3) infection. However, the molecular mechanisms are not clear. Here, we examined the effect of Irgm3 expression on ER stress triggered by pharmacological agents or CVB3 infection. In Tet-On/Irgm3 HeLa cells, Irgm3 expression suppressed either chemical- or CVB3-induced upregulation of glucose-regulated protein 78. Further, Irgm3 strongly inhibited the activation of both the PERK and ATF6 pathways of ER stress responses, which further led to the diminished phosphorylation of eIF2α, reduced cleavage/activation of transcription factor SREBP1 and attenuated induction of proapoptotic genes CHOP and GADD34. These data were further supported by experiments using Irgm3 knockout mouse embryonic fibroblasts, in which the ER stress induced by CVB3 was not relieved due to the lack of Irgm3 expression. In addition, the tunicamycin-triggered ER stress promoted the subsequent CVB3 infection. The effect of Irgm3 on ER stress and CVB3 infection was diminished by the PI3K inhibitor, LY294002, while inhibitors of ERK, JNK and p38 had no effect. These data were further corroborated by transfection of cells with a dominant negative Akt. Taken together, these data suggest that Irgm3 relieves the ER stress response via a PI3K/Akt dependent mechanism, which contributes to host defence against CVB3 infection.


Subject(s)
Endoplasmic Reticulum Stress , Enterovirus B, Human/pathogenicity , Enterovirus Infections/metabolism , GTP-Binding Proteins/metabolism , MAP Kinase Signaling System , Activating Transcription Factor 6/metabolism , Apoptosis , Chromones/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Enterovirus B, Human/immunology , Enterovirus B, Human/metabolism , Eukaryotic Initiation Factor-2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Phosphatase 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sterol Regulatory Element Binding Protein 1/metabolism , Stress, Physiological , Transcription Factor CHOP/metabolism , eIF-2 Kinase/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
15.
J Virol ; 84(17): 8446-59, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20554776

ABSTRACT

Cardiomyocyte apoptosis is a hallmark of coxsackievirus B3 (CVB3)-induced myocarditis. We used cardiomyocytes and HeLa cells to explore the cellular response to CVB3 infection, with a focus on pathways leading to apoptosis. CVB3 infection triggered endoplasmic reticulum (ER) stress and differentially regulated the three arms of the unfolded protein response (UPR) initiated by the proximal ER stress sensors ATF6a (activating transcription factor 6a), IRE1-XBP1 (X box binding protein 1), and PERK (PKR-like ER protein kinase). Upon CVB3 infection, glucose-regulated protein 78 expression was upregulated, and in turn ATF6a and XBP1 were activated via protein cleavage and mRNA splicing, respectively. UPR activity was further confirmed by the enhanced expression of UPR target genes ERdj4 and EDEM1. Surprisingly, another UPR-associated gene, p58(IPK), which often is upregulated during infections with other types of viruses, was downregulated at both mRNA and protein levels after CVB3 infection. These findings were observed similarly for uninfected Tet-On HeLa cells induced to overexpress ATF6a or XBP1. In exploring potential connections between the three UPR pathways, we found that the ATF6a-induced downregulation of p58(IPK) was associated with the activation of PKR (PERK) and the phosphorylation of eIF2alpha, suggesting that p58(IPK), a negative regulator of PERK and PKR, mediates cross-talk between the ATF6a/IRE1-XBP1 and PERK arms. Finally, we found that CVB3 infection eventually produced the induction of the proapoptoic transcription factor CHOP and the activation of SREBP1 and caspase-12. Taken together, these data suggest that CVB3 infection activates UPR pathways and induces ER stress-mediated apoptosis through the suppression of P58(IPK) and induction/activation of CHOP, SREBP1, and caspase-12.


Subject(s)
Apoptosis , Coxsackievirus Infections/metabolism , Down-Regulation , Enterovirus/physiology , HSP40 Heat-Shock Proteins/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Transcription Factor CHOP/metabolism , Unfolded Protein Response , Animals , Caspase 12/genetics , Caspase 12/metabolism , Coxsackievirus Infections/genetics , Coxsackievirus Infections/physiopathology , Coxsackievirus Infections/virology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , HSP40 Heat-Shock Proteins/genetics , HeLa Cells , Humans , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/virology , Phosphorylation , Sterol Regulatory Element Binding Protein 1/genetics , Transcription Factor CHOP/genetics
16.
Cell Microbiol ; 12(5): 599-614, 2010 May 01.
Article in English | MEDLINE | ID: mdl-19951366

ABSTRACT

Our previous study reported that mouse BNIP-21 (mBNIP-21) induces apoptosis through a mitochondria-dependent pathway. To map the functional domains of mBNIP-21, we performed mutational analyses and demonstrated that the BNIP-2 and Cdc42GAP homology (BCH) domain is required for apoptosis induction by mBNIP-21 targeting the mitochondria and inducing cytochrome c release. This pro-apoptotic activity was enhanced by coxsackievirus infection. However, deletion of the Bcl-2 homology 3 (BH3)-like domain, a well-known cell 'death domain' in proapoptotic Bcl-2 family proteins, did not affect the activity of mBNIP-21. These data were further supported by transfection of a mouse Bax (mBax) mutant, whose BH3 was replaced by the mBNIP-21 BH3-like domain. This replacement significantly reduced the pro-apoptotic activity of mBax. We also found that the predicted calcium binding domain has no contribution to the mBNIP-21-induced apoptosis. Further mapping of the motifs of BCH domain demonstrated that deletion of the hydrophobic motif proximal to the C-terminal of the BCH significantly reduced its proapoptotic activity. These findings suggest that mBNIP-21, as a member of the BNIP subgroup of the Bcl-2-related proteins, functions without need of BH3 but its BCH domain is critical for its activity in inducing cell elongation, membrane protrusions and apoptotic cell death.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Enterovirus B, Human/pathogenicity , Amino Acid Motifs , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Cell Line , GTPase-Activating Proteins/genetics , Humans , Mice , Protein Structure, Tertiary , Sequence Deletion , Sequence Homology, Amino Acid
17.
Antiviral Res ; 83(3): 307-16, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19616030

ABSTRACT

Coxsackievirus B3 (CVB3) is a common pathogen of myocarditis. We previously synthesized a siRNA targeting the CVB3 protease 2A (siRNA/2A) gene and achieved reduction of CVB3 replication by 92% in vitro. However, like other drugs under development, CVB3 siRNA faces a major challenge of targeted delivery. In this study, we investigated a novel approach to deliver CVB3 siRNAs to a specific cell population (e.g. HeLa cells containing folate receptor) using receptor ligand (folate)-linked packaging RNA (pRNA) from bacterial phage phi29. pRNA monomers can spontaneously form dimers and multimers under optimal conditions by base-pairing between their stem loops. By covalently linking a fluorescence-tag to folate, we delivered the conjugate specifically to HeLa cells without the need of transfection. We further demonstrated that pRNA covalently conjugated to siRNA/2A achieved an equivalent antiviral effect to that of the siRNA/2A alone. Finally, the drug targeted delivery was further evaluated by using pRNA monomers or dimers, which carried both the siRNA/2A and folate ligand and demonstrated that both of them strongly inhibited CVB3 replication. These data indicate that pRNA as a siRNA carrier can specifically deliver the drug to target cells via its ligand and specific receptor interaction and inhibit virus replication effectively.


Subject(s)
Antiviral Agents/metabolism , Drug Delivery Systems/methods , Enterovirus B, Human/drug effects , Genetic Therapy/methods , RNA, Small Interfering/metabolism , Bacillus Phages/genetics , Base Sequence , HeLa Cells , Humans , Models, Biological , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation
18.
Curr Drug Discov Technol ; 5(1): 49-58, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18537567

ABSTRACT

MicroRNAs (miRNAs) are endogenous, short, double-stranded and noncoding RNA molecules that have been identified in a variety of organisms and certain viruses. This group of new molecules is transcribed mainly from the introns and/or exons or intergenic regions and plays important regulatory roles in development and gene expression. Mature miRNAs are typically 20-24 nucleotides in length and regulate target mRNAs post transcriptionally by interactions with partially mismatched sequences in the 3'untraslated regions of these messengers. These interactions result in the suppression of translation or degradation of target mRNAs. At the present, although the biological functions of miRNAs are not completely revealed, a growing body of evidence implicates that miRNA pathway is a new mechanism of gene regulation in both normal and diseased conditions and therefore investigation of miRNA biogenesis and function may add new tools for gene functional study and drug development. In this article, we will briefly review the structure, biogenesis and basic mechanism of action of miRNAs identified in higher organisms and viruses and then focus on the recent progress in research for drug development using the miRNA pathway as a strategy. Particularly, we will discuss the advance, challenge and future directions on antiviral drug development using miRNA as a target or a gene silencing tool for the treatment of viral infections.


Subject(s)
MicroRNAs/therapeutic use , Virus Diseases/drug therapy , Animals , Gene Silencing/drug effects , Humans , MicroRNAs/biosynthesis , MicroRNAs/physiology , Oligonucleotides/therapeutic use , RNA, Viral/genetics , Virus Diseases/pathology , Viruses/genetics
19.
Cell Microbiol ; 10(9): 1787-800, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18452580

ABSTRACT

Interferon-gamma-inducible GTPase (IGTP) expression is upregulated in coxsackievirus B3 (CVB3)-infected murine heart and inhibits CVB3-induced apoptosis through activation of the PI3 kinase/Akt pathway. However, the mechanism of this pathway activation is unknown. In this study, using doxcycycline-inducible Tet-On HeLa cells that overexpress IGTP, we have demonstrated that focal adhesion kinase (FAK) is phosphorylated in response to IGTP expression and that transfection of the Tet-On HeLa cells with a dominant negative FAK (FRNK) blocks Akt activation. Furthermore, induction of IGTP also promoted the NF-kappaB activation as evidenced by its enhanced nuclear translocation, binding to transcriptional promoters and increased transcriptional activity. However, FRNK transfection and phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 both blocked the IGTP-induced translocation and NF-kappaB activation. Furthermore, silencing NF-kappaB with siRNAs significantly inhibited the phosphorylation of FAK and Akt, but not their total expression levels, indicating that NF-kappaB activation is required for the IGTP-induced activation of FAK and PI3K/Akt. Finally, blocking this survival pathway by transfection of FRNK or silencing of NF-kappaB reduced CVB3 replication and enhanced cell death during CVB3 infection. Taken together, these results suggest that FAK is a mediator upstream of PI3K/Akt and NF-kappaB functions as a downstream effector and also positively regulates the activity of upstream kinases.


Subject(s)
Coxsackievirus Infections/enzymology , Enterovirus B, Human , Focal Adhesion Protein-Tyrosine Kinases/metabolism , GTP Phosphohydrolases/metabolism , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Cell Survival , Coxsackievirus Infections/virology , Feedback, Physiological , Focal Adhesion Protein-Tyrosine Kinases/genetics , GTP Phosphohydrolases/genetics , Heart/virology , Humans , Male , Mice , Myocarditis/enzymology , Myocarditis/virology , Myocardium/enzymology , NF-kappa B/genetics , Phosphorylation , Promoter Regions, Genetic , Protein-Tyrosine Kinases/genetics , Signal Transduction , Transcriptional Activation , Transfection
20.
Am J Physiol Cell Physiol ; 290(5): C1310-20, 2006 May.
Article in English | MEDLINE | ID: mdl-16354757

ABSTRACT

Focal adhesion kinase (FAK) integrates various extracellular and intracellular signals and is implicated in a variety of biological functions, but its exact role and downstream targeting signals in the regulation of apoptosis in intestinal epithelial cells (IECs) remains unclear. The current study tested the hypothesis that FAK has an antiapoptotic role in the IEC-6 cell line by altering NF-kappaB signaling. Induced FAK expression by stable transfection with the wild-type (WT)-FAK gene increased FAK phosphorylation, which was associated with an increase in NF-kappaB activity. These stable WT-FAK-transfected IECs also exhibited increased resistance to apoptosis when they were exposed to TNF-alpha plus cycloheximide (TNF-alpha/CHX). Specific inhibition of NF-kappaB by the recombinant adenoviral vector containing the IkappaBalpha superrepressor prevented increased resistance to apoptosis in WT-FAK-transfected cells. In contrast, inactivation of FAK by ectopic expression of dominant-negative mutant of FAK (DNM-FAK) inhibited NF-kappaB activity and increased the sensitivity to TNF-alpha/CHX-induced apoptosis. Furthermore, induced expression of endogenous FAK by depletion of cellular polyamines increased NF-kappaB activity and resulted in increased resistance to TNF-alpha/CHX-induced apoptosis, both of which were prevented by overexpression of DNM-FAK. These results indicate that increased expression of FAK suppresses TNF-alpha/CHX-induced apoptosis, at least partially, through the activation of NF-kappaB signaling in IECs.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/physiology , Focal Adhesion Kinase 1/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/physiology , NF-kappa B/metabolism , Signal Transduction/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Line , Epithelial Cells/drug effects , Intestinal Mucosa/drug effects , Rats , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...