Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
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
3.
J Gen Virol ; 90(Pt 4): 854-862, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19264606

ABSTRACT

Clathrin- and caveolae-mediated endocytosis have been implicated in the productive entry of many viruses into host cells. ADP-ribosylation factor 6 (Arf6)-dependent endocytosis is another endocytosis pathway that traffics from the cell surface and it is the only Arf that traffics at the plasma membrane. However, little is known about Arf6-dependent trafficking during virus entry. This study showed that coxsackievirus type B3 (CVB3) associated with decay-accelerating factor in non-polarized HeLa cells can be redirected into non-productive compartments by Arf6-dependent internalization, thus restricting infection. Overexpression of wild-type (WT) and constitutively active (CA) Arf6 in HeLa cells resulted in a 2.3- and 3.6-fold decrease in infection, respectively. A dominant-negative inhibitor of Arf6 recovered restriction of infection by WT-Arf6 and CA-Arf6. RNA interference of endogenous Arf6 resulted in a 3.3-fold increase in CVB3 titre in HeLa cells. It was shown that coxsackie-adenovirus receptor (CAR) ligation by virus or CAR-specific antibody could activate extracellular signal-regulated kinase (ERK) of the mitogen-activated protein kinase family and lead to Arf6-mediated viral restriction. In the absence of ERK activation, CVB3 internalization into early endosomes was inhibited and subsequent infection was reduced, but Arf6-mediated restriction was also abolished. In conclusion, receptor-mediated signalling enhances CVB3 entry whilst also activating non-productive pathways of virus entry; thus, virus infection is an equilibrium of productive and non-productive pathways of entry.


Subject(s)
ADP-Ribosylation Factors/metabolism , Enterovirus B, Human/pathogenicity , Extracellular Signal-Regulated MAP Kinases/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , Cell Line , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Endocytosis , Endosomes/virology , Enterovirus B, Human/genetics , Enterovirus B, Human/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , HeLa Cells , Humans , Receptors, Virus/genetics , Receptors, Virus/metabolism , Signal Transduction
4.
Circ Res ; 104(5): 628-38, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19168435

ABSTRACT

Coxsackievirus (CV)B3 is the primary cause of viral myocarditis. We previously observed CXC chemokine ligand 10 (CXCL10) upregulation in the myocardium early in infection. However, the impact of CXCL10 in CVB3-induced myocarditis is unknown. Using isolated primary mouse cardiomyocytes we demonstrated for the first time that cardiomyocytes can express CXCL10 on interferon-gamma stimulation. To explore the role of CXCL10 in CVB3-induced myocarditis, both CXCL10 transgenic and knockout mice were used. Following CVB3 challenges, the viral titer in the hearts inversely correlated with the levels of CXCL10 at early phase of infection before visible immune infiltration. Furthermore, as compared with the control mice, the decreased virus titers in the CXCL10 transgenic mouse hearts led to less cardiac damage and better cardiac function and vice verse in the knockout mice. This antiviral ability of CXCL10 might be through recruitment of natural killer (NK) cells to the heart and increased interferon-gamma expression early in infection. At day 7 postinfection, with massive influx of mononuclear cells the expression of CXCL10 enhanced the infiltration of CXCR3(+) cells, CD4(+), and CD8(+) T cells, as well as the expression of associated inflammatory cytokines. However, the augmented accumulation of these immune cells and associated cytokines failed to alter the viral clearance and mice survival. These results suggest the protective role of CXCL10 during the early course of CVB3 infection, which is attributed to the recruitment of NK cells. Nonetheless, CXCL10-directed chemoattractant effect is not sufficient for host to clear the virus in the heart.


Subject(s)
Chemokine CXCL10/metabolism , Chemotaxis , Coxsackievirus Infections/complications , Enterovirus/pathogenicity , Killer Cells, Natural/immunology , Myocarditis/immunology , Myocardium/immunology , Virus Replication , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Chemokine CXCL10/deficiency , Chemokine CXCL10/genetics , Coxsackievirus Infections/immunology , Coxsackievirus Infections/pathology , Coxsackievirus Infections/virology , Disease Models, Animal , Inflammation/immunology , Inflammation/virology , Interferon-gamma/metabolism , Interleukin-10/metabolism , Killer Cells, Natural/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Mice, Transgenic , Myocarditis/pathology , Myocarditis/prevention & control , Myocarditis/virology , Myocardium/pathology , RNA, Messenger/metabolism , Receptors, CXCR3/metabolism , Time Factors
5.
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
6.
Front Biosci ; 13: 4707-25, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18508540

ABSTRACT

Anti-picornaviral antisense agents are part of a broader group of nucleic acid-based molecules developed for sequence-specific inhibition of translation and/or transcription of the target sequence through induced nuclease activity or physical hindrance. Three types of nucleic acid-based gene silencing molecules can be distinguished, including DNA-base antisense oligonucleotides (ASO), nucleic acid enzymes (ribozyme and DNAzyme) and double-stranded small interfering RNA (siRNA or microRNA). These antisense DNA and RNA molecules have been widely studied for gene functional studies and therapeutic purposes. In this review, we focus on drug development using ASO and siRNA strategies to inhibit picornavirus infections. The picornavirus genome organization and life cycle is described, followed by discussion of design considerations, chemical modifications and drug delivery approaches. Recent studies using antisense against picornavirus are reviewed. Finally, we compare the advantages and disadvantages of the antisense agents with those of other therapeutics, taking into consideration their limitations which need to be overcome to achieve the final goal of clinical application.


Subject(s)
Antiviral Agents/therapeutic use , DNA, Antisense/therapeutic use , Genome, Viral , Picornaviridae Infections/drug therapy , Picornaviridae/drug effects , Picornaviridae/genetics , RNA, Antisense/therapeutic use , Antiviral Agents/administration & dosage , DNA, Antisense/administration & dosage , Humans , Liposomes , Oligonucleotides, Antisense/therapeutic use , Picornaviridae/growth & development , Proteome , RNA, Antisense/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use
7.
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
8.
Int J Mol Sci ; 9(6): 978-999, 2008 Jun.
Article in English | MEDLINE | ID: mdl-19325841

ABSTRACT

MicroRNAs (miRNAs) are a class of short non-coding RNAs with posttranscriptional regulatory functions. To date, more than 600 human miRNAs have been experimentally identified, and estimated to regulate more than one third of cellular messenger RNAs. Accumulating evidence has linked the dysregulated expression patterns of miRNAs to a variety of diseases, such as cancer, neurodegenerative diseases, cardiovascular diseases and viral infections. MiRNAs provide its particular layer of network for gene regulation, thus possessing the great potential both as a novel class of therapeutic targets and as a powerful intervention tool. In this regard, synthetic RNAs that contain the binding sites of miRNA have been shown to work as a "decoy" or "miRNA sponge" to inhibit the function of specific miRNAs. On the other hand, miRNA expression vectors have been used to restore or overexpress specific miRNAs to achieve a long-term effect. Further, double-stranded miRNA mimetics for transient replacement have been experimentally validated. Endogenous precursor miRNAs have also been used as scaffolds for the induction of RNA interference. This article reviews the recent progress on this emerging technology as a powerful tool for gene regulation studies and particularly as a rationale strategy for design of therapeutics.

9.
Apoptosis ; 12(3): 513-24, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17195095

ABSTRACT

By transfection of Coxsackievirus B3 (CVB3) individual protease gene into HeLa cells, we demonstrated that 2A(pro) and 3C(pro) induced apoptosis through multiple converging pathways. Firstly, both 2A(pro) and 3C(pro) induced caspase-8-mediated activation of caspase-3 and dramatically reduced cell viability. Secondly, they both activated the intrinsic mitochondria-mediated apoptosis pathway leading to cytochrome c release from mitochondria and activation of caspase-9. However, 3C(pro) induced these events via both up-regulation of Bax and cleavage of Bid, and 2A(pro) induced these events via cleavage of Bid only. Nevertheless, neither altered Bcl-2 expression. Thirdly, both proteases induced cell death through cleavage or down regulation of cellular factors for translation and transcription: both 2A(pro) and 3C(pro) cleaved eukaryotic translation initiation factor 4GI but their cleavage products are different, indicating different cleavage sites; further, both 2A(pro) and 3C(pro) down-regulated cyclic AMP responsive element binding protein, a transcription factor, with 2A(pro) exhibiting a stronger effect than 3C(pro). Surprisingly, neither could cleave DAP5/p97/NAT1, a translation regulator, although this cleavage was observed during CVB3 infection and could not be blocked by caspase inhibitor z-VAD-fmk. Taken together, these data suggest that 2A(pro) and 3C(pro) induce apoptosis through both activation of proapoptotic mediators and suppression of translation and transcription.


Subject(s)
Apoptosis/physiology , Cysteine Endopeptidases/metabolism , Eukaryotic Initiation Factor-4G/metabolism , Mitochondria/metabolism , Viral Proteins/metabolism , 3C Viral Proteases , Adult , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspase 9/metabolism , Caspase Inhibitors , Cell Shape , Child , Cysteine Endopeptidases/genetics , Cytochromes c/metabolism , Enzyme Activation , Eukaryotic Initiation Factor-4G/genetics , HeLa Cells , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , Viral Proteins/genetics , bcl-2-Associated X Protein/metabolism
10.
Curr Opin Mol Ther ; 8(2): 104-7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16610761

ABSTRACT

Positive single-stranded RNA viruses constitute a broad and prevalent group of pathogens that threaten human health and life worldwide. While effective vaccines have been developed for some, such as poliovirus and hepatitis A, others such as coxsackievirus, severe acute respiratory syndrome coronavirus (SARS-CoV) and West Nile virus have no accredited drug treatments. Antisense technologies, which encompass small interfering RNA, antisense oligonucleotides, ribozymes and their chemically modified analogs, involve small sequence-specific nucleic-acid-based molecules that inhibit viral replication at the level of translation. Many antisense oligomers are proven antiviral agents in vitro. In this review, iwe provide an overview of the antiviral antisense field, highlighting specific studies of interest over the past several years, using our experience with coxsackievirus B3 as a reference point. Overall, both the challenges and successes of existing antisense therapies for positive single-stranded RNA viruses can be paralleled to those for other virus groups, and vice versa.


Subject(s)
Antiviral Agents/pharmacology , Nucleic Acids/chemistry , RNA Viruses/drug effects , RNA, Viral/chemistry , RNA, Viral/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Humans , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacology , RNA, Small Interfering/therapeutic use , RNA, Viral/genetics , Review Literature as Topic , Virus Replication/drug effects
11.
Oncogene ; 23(28): 4938-44, 2004 Jun 17.
Article in English | MEDLINE | ID: mdl-15064715

ABSTRACT

We previously reported that BARF1 gene has either an immortalizing effect, when expressed in primary primate epithelial cells, or a malignant transforming activity, when expressed in established and nontumoral rodent fibroblast or human B-cell lines. As predicted from sequence analysis, we found that BARF1 coded protein can be secreted from different cell lines, among them BARF1-transfected Balb/c3T3 rodent fibroblasts. Thus, as an initial step to clarify BARF1 oncogenic functions, we investigated whether the secreted form of BARF1 protein can activate the cell cycle as a growth factor. Since efficient BARF1 expression could be obtained from 293-tTA cells infected with a tetracycline-regulatable recombinant adenovirus, secreted BARF1 product could be purified from the culture medium of such cells by ammonium sulfate precipitation, ion exchange chromotography and sucrose gradient sedimentation. We describe in this paper that addition of a purified product of secreted BARF1 protein to serum-free culture medium of Balb/c3T3 rodent fibroblasts, human Louckes B-cell line and primary monkey kidney epithelial cells resulted in a cell cycle activation that was inhibited by affinity-purified anti-BARF1 antibody. Our demonstration of a specific stimulation of cell cycle in vitro by BARF1 secreted product suggests that this EBV-encoded BARF1 protein could act as a growth factor in vivo.


Subject(s)
Cell Division/drug effects , Receptor, Macrophage Colony-Stimulating Factor/physiology , Viral Proteins/pharmacology , 3T3 Cells , Animals , Cloning, Molecular , Mice , Mice, Inbred BALB C , Recombinant Proteins/pharmacology , Transfection , Viral Proteins/genetics
12.
Mol Cell Biol ; 22(4): 1218-32, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11809812

ABSTRACT

We have analyzed the expression pattern of the D1 gene and the localization of its product, the AT hook-bearing nonhistone chromosomal protein D1, during Drosophila melanogaster development. D1 mRNAs and protein are maternally contributed, and the protein localizes to discrete foci on the chromosomes of early embryos. These foci correspond to 1.672- and 1.688-g/cm(3) AT-rich satellite repeats found in the centromeric heterochromatin of the X and Y chromosomes and on chromosomes 3 and 4. D1 mRNA levels subsequently decrease throughout later development, followed by the accumulation of the D1 protein in adult gonads, where two distributions of D1 can be correlated to different states of gene activity. We show that the EP473 mutation, a P-element insertion upstream of D1 coding sequences, affects the expression of the D1 gene and results in an embryonic homozygous lethal phenotype correlated with the depletion of D1 protein during embryogenesis. Remarkably, decreased levels of D1 mRNA and protein in heterozygous flies lead to the suppression of position-effect variegation (PEV) of the white gene in the white-mottled (w(m4h)) X-chromosome inversion. Our results identify D1 as a DNA-binding protein of known sequence specificity implicated in PEV. D1 is the primary factor that binds the centromeric 1.688-g/cm(3) satellite repeats which are likely involved in white-mottled variegation. We propose that the AT-hook D1 protein nucleates heterochromatin assembly by recruiting specialized transcriptional repressors and/or proteins involved in chromosome condensation.


Subject(s)
DNA, Satellite/metabolism , DNA-Binding Proteins/metabolism , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Animals , Base Composition , DNA Transposable Elements/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Eye Color , Female , Gametogenesis , Genes, Insect , Heterochromatin/genetics , Heterochromatin/metabolism , Immunohistochemistry , In Situ Hybridization , Male , Molecular Sequence Data , Photoreceptor Cells, Invertebrate/physiology , X Chromosome
SELECTION OF CITATIONS
SEARCH DETAIL