Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Viruses ; 13(12)2021 11 30.
Article in English | MEDLINE | ID: mdl-34960671

ABSTRACT

Rhinoviruses (RV), like many other viruses, modulate programmed cell death to their own advantage. The viral protease, 3C has an integral role in the modulation, and we have shown that RVA-16 3C protease cleaves Receptor-interacting protein kinase-1 (RIPK1), a key host factor that modulates various cell death and cell survival pathways. In the current study, we have investigated whether this cleavage is conserved across selected RV strains. RIPK1 was cleaved in cells infected with strains representing diversity across phylogenetic groups (A and B) and receptor usage (major and minor groups). The cleavage was abrogated in the presence of the specific 3C protease inhibitor, Rupintrivir. Interestingly, there appears to be involvement of another protease (maybe 2A protease) in RIPK1 cleavage in strains belonging to genotype B. Our data show that 3C protease from diverse RV strains cleaves RIPK1, highlighting the importance of the cleavage to the RV lifecycle.


Subject(s)
3C Viral Proteases/metabolism , Picornaviridae Infections/enzymology , Rhinovirus/enzymology , 3C Viral Proteases/genetics , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Apoptosis/drug effects , HeLa Cells , Host-Pathogen Interactions , Humans , Isoxazoles/chemistry , Isoxazoles/pharmacology , Phenylalanine/analogs & derivatives , Phenylalanine/chemistry , Phenylalanine/pharmacology , Picornaviridae Infections/genetics , Picornaviridae Infections/virology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Pyrrolidinones/chemistry , Pyrrolidinones/pharmacology , Rhinovirus/chemistry , Rhinovirus/drug effects , Rhinovirus/genetics , Valine/analogs & derivatives , Valine/chemistry , Valine/pharmacology
2.
Cell Mol Life Sci ; 78(21-22): 6735-6744, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34459952

ABSTRACT

Kallikrein-related peptidases (KLKs) or kallikreins have been linked to diverse (patho) physiological processes, such as the epidermal desquamation and inflammation, seminal clot liquefaction, neurodegeneration, and cancer. Recent mounting evidence suggests that KLKs also represent important regulators of viral infections. It is well-established that certain enveloped viruses, including influenza and coronaviruses, require proteolytic processing of their hemagglutinin or spike proteins, respectively, to infect host cells. Similarly, the capsid protein of the non-enveloped papillomavirus L1 should be proteolytically cleaved for viral uncoating. Consequently, extracellular or membrane-bound proteases of the host cells are instrumental for viral infections and represent potential targets for drug development. Here, we summarize how extracellular proteolysis mediated by the kallikreins is implicated in the process of influenza (and potentially coronavirus and papillomavirus) entry into host cells. Besides direct proteolytic activation of viruses, KLK5 and 12 promote viral entry indirectly through proteolytic cascade events, like the activation of thrombolytic enzymes that also can process hemagglutinin, while additional functions of KLKs in infection cannot be excluded. In the light of recent evidence, KLKs represent potential host targets for the development of new antivirals. Humanized animal models to validate their key functions in viral infections will be valuable.


Subject(s)
COVID-19/enzymology , COVID-19/virology , Host Microbial Interactions/physiology , Kallikreins/metabolism , SARS-CoV-2 , Virus Diseases/enzymology , Animals , Asthma/etiology , Coronavirus/genetics , Coronavirus/pathogenicity , Coronavirus/physiology , Host Microbial Interactions/genetics , Humans , Orthomyxoviridae/genetics , Orthomyxoviridae/pathogenicity , Orthomyxoviridae/physiology , Papillomavirus Infections/enzymology , Papillomavirus Infections/virology , Picornaviridae Infections/complications , Picornaviridae Infections/enzymology , Picornaviridae Infections/virology , Protein Processing, Post-Translational , Proteolysis , Rhinovirus/pathogenicity , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology , Varicella Zoster Virus Infection/enzymology , Varicella Zoster Virus Infection/virology , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Diseases/virology , Virus Internalization
3.
Clin Sci (Lond) ; 132(15): 1711-1723, 2018 08 16.
Article in English | MEDLINE | ID: mdl-29980604

ABSTRACT

Rhinovirus (RV) infection is involved in acute exacerbations of asthma and chronic obstructive pulmonary disease (COPD). RV primarily infects upper and lower airway epithelium. Immunoproteasomes (IP) are proteolytic machineries with multiple functions including the regulation of MHC class I antigen processing during viral infection. However, the role of IP in RV infection has not been explored. We sought to investigate the expression and function of IP during airway RV infection. Primary human tracheobronchial epithelial (HTBE) cells were cultured at air-liquid interface (ALI) and treated with RV16, RV1B, or interferon (IFN)-λ in the absence or presence of an IP inhibitor (ONX-0914). IP gene (i.e. LMP2) deficient mouse tracheal epithelial cells (mTECs) were cultured for the mechanistic studies. LMP2-deficient mouse model was used to define the in vivo role of IP in RV infection. IP subunits LMP2 and LMP7, antiviral genes MX1 and OAS1 and viral load were measured. Both RV16 and RV1B significantly increased the expression of LMP2 and LMP7 mRNA and proteins, and IFN-λ mRNA in HTBE cells. ONX-0914 down-regulated MX1 and OAS1, and increased RV16 load in HTBE cells. LMP2-deficient mTECs showed a significant increase in RV1B load compared with the wild-type (WT) cells. LMP2-deficient (compared with WT) mice increased viral load and neutrophils in bronchoalveolar lavage (BAL) fluid after 24 h of RV1B infection. Mechanistically, IFN-λ induction by RV infection contributed to LMP2 and LMP7 up-regulation in HTBE cells. Our data suggest that IP are induced during airway RV infection, which in turn may serve as an antiviral and anti-inflammatory mechanism.


Subject(s)
Epithelial Cells/immunology , Picornaviridae Infections/immunology , Proteasome Endopeptidase Complex/immunology , Rhinovirus/immunology , Animals , Asthma/enzymology , Asthma/immunology , Asthma/virology , Cell Line , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/immunology , Cysteine Endopeptidases/metabolism , Epithelial Cells/enzymology , Epithelial Cells/virology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Mice, Knockout , Oligopeptides/pharmacology , Picornaviridae Infections/enzymology , Picornaviridae Infections/virology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Respiratory System/enzymology , Respiratory System/immunology , Respiratory System/virology , Rhinovirus/physiology
4.
Article in English | MEDLINE | ID: mdl-27354786

ABSTRACT

Human rhinovirus (HRV) infections target airway epithelium and are the leading cause of acute exacerbations of COPD. Cigarette smoke (CS) increases the severity of viral infections, but there is no effective therapy for HRV infection. We determined whether α1-antitrypsin (A1AT) reduces HRV-16 infection in CS-exposed primary human airway epithelial cells. Brushed bronchial epithelial cells from normal subjects and patients diagnosed with COPD were cultured at air-liquid interface to induce mucociliary differentiation. These cells were treated with A1AT or bovine serum albumin for 2 hours and then exposed to air or whole cigarette smoke (WCS) with or without HRV-16 (5×10(4) 50% Tissue Culture Infective Dose [TCID50]/transwell) infection for 24 hours. WCS exposure significantly increased viral load by an average of fivefold and decreased the expression of antiviral genes interferon-λ1, OAS1, and MX1. When A1AT was added to WCS-exposed cells, viral load significantly decreased by an average of 29-fold. HRV-16 infection significantly increased HRV-16 receptor intercellular adhesion molecule-1 messenger RNA expression in air-exposed cells, which was decreased by A1AT. A1AT-mediated reduction of viral load was not accompanied by increased epithelial antiviral gene expression or by inhibiting the activity of 3C protease involved in viral replication or maturation. Our findings demonstrate that A1AT treatment prevents a WCS-induced increase in viral load and for the first time suggest a therapeutic effect of A1AT on HRV infection.


Subject(s)
Antiviral Agents/pharmacology , Bronchi/drug effects , Epithelial Cells/drug effects , Picornaviridae Infections/prevention & control , Pulmonary Disease, Chronic Obstructive/drug therapy , Respiratory Mucosa/drug effects , Respiratory Tract Infections/prevention & control , Rhinovirus/drug effects , Smoke/adverse effects , Smoking/adverse effects , alpha 1-Antitrypsin/pharmacology , Aged , Bronchi/enzymology , Bronchi/pathology , Bronchi/virology , Case-Control Studies , Cells, Cultured , Epithelial Cells/enzymology , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Host-Pathogen Interactions , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Male , Middle Aged , Picornaviridae Infections/enzymology , Picornaviridae Infections/pathology , Picornaviridae Infections/virology , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/pathology , Respiratory Mucosa/enzymology , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Respiratory Tract Infections/enzymology , Respiratory Tract Infections/pathology , Respiratory Tract Infections/virology , Rhinovirus/pathogenicity , Viral Load
5.
Respir Res ; 16: 42, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25889951

ABSTRACT

BACKGROUND AND PURPOSE: The persistent influx of neutrophils into the lung and subsequent tissue damage are characteristics of COPD, cystic fibrosis and acute lung inflammation. VAP-1/SSAO is an endothelial bound adhesion molecule with amine oxidase activity that is reported to be involved in neutrophil egress from the microvasculature during inflammation. This study explored the role of VAP-1/SSAO in neutrophilic lung mediated diseases and examined the therapeutic potential of the selective inhibitor PXS-4728A. METHODS: Mice treated with PXS-4728A underwent intra-vital microscopy visualization of the cremaster muscle upon CXCL1/KC stimulation. LPS inflammation, Klebsiella pneumoniae infection, cecal ligation and puncture as well as rhinovirus exacerbated asthma models were also assessed using PXS-4728A. RESULTS: Selective VAP-1/SSAO inhibition by PXS-4728A diminished leukocyte rolling and adherence induced by CXCL1/KC. Inhibition of VAP-1/SSAO also dampened the migration of neutrophils to the lungs in response to LPS, Klebsiella pneumoniae lung infection and CLP induced sepsis; whilst still allowing for normal neutrophil defense function, resulting in increased survival. The functional effects of this inhibition were demonstrated in the RV exacerbated asthma model, with a reduction in cellular infiltrate correlating with a reduction in airways hyperractivity. CONCLUSIONS AND IMPLICATIONS: This study demonstrates that the endothelial cell ligand VAP-1/SSAO contributes to the migration of neutrophils during acute lung inflammation, pulmonary infection and airway hyperractivity. These results highlight the potential of inhibiting of VAP-1/SSAO enzymatic function, by PXS-4728A, as a novel therapeutic approach in lung diseases that are characterized by neutrophilic pattern of inflammation.


Subject(s)
Allylamine/analogs & derivatives , Amine Oxidase (Copper-Containing)/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Asthma/drug therapy , Benzamides/pharmacology , Cell Adhesion Molecules/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Klebsiella Infections/drug therapy , Lung/drug effects , Neutrophil Infiltration/drug effects , Picornaviridae Infections/drug therapy , Pneumonia/drug therapy , Respiratory Tract Infections/drug therapy , Allylamine/pharmacokinetics , Allylamine/pharmacology , Amine Oxidase (Copper-Containing)/metabolism , Animals , Anti-Inflammatory Agents/pharmacokinetics , Asthma/enzymology , Asthma/immunology , Asthma/physiopathology , Asthma/virology , Benzamides/pharmacokinetics , Bronchoconstriction/drug effects , Cecum/microbiology , Cecum/surgery , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/immunology , Enzyme Inhibitors/pharmacokinetics , Klebsiella Infections/enzymology , Klebsiella Infections/immunology , Klebsiella Infections/microbiology , Klebsiella pneumoniae/pathogenicity , Leukocyte Rolling/drug effects , Ligation , Lipopolysaccharides , Lung/enzymology , Lung/immunology , Lung/physiopathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Picornaviridae Infections/enzymology , Picornaviridae Infections/immunology , Picornaviridae Infections/physiopathology , Picornaviridae Infections/virology , Pneumonia/enzymology , Pneumonia/etiology , Pneumonia/immunology , Punctures , Rats, Wistar , Respiratory Tract Infections/enzymology , Respiratory Tract Infections/etiology , Respiratory Tract Infections/immunology , Rhinovirus/pathogenicity
6.
PLoS One ; 9(12): e116055, 2014.
Article in English | MEDLINE | ID: mdl-25541728

ABSTRACT

Human rhinoviruses (HRV) are a major cause of exacerbations of airways disease. Aspects of cell signalling responses to HRV infection remain unclear, particularly with regard to signalling via PI3K, and the PI3K-dependent pathway, autophagy. We investigated the roles of PI3K and autophagy in the responses of epithelial cells to major and minor group HRV infection. The PI3K inhibitor 3-MA, commonly used to inhibit autophagy, markedly reduced HRV-induced cytokine induction. Further investigation of potential targets of 3-MA and comparison of results using this inhibitor to a panel of general and class I-selective PI3K inhibitors showed that several PI3Ks cooperatively regulate responses to HRV. Targeting by siRNA of the autophagy proteins Beclin-1, Atg7, LC3, alone or in combination, or targeting of the autophagy-specific class III PI3K had at most only modest effects on HRV-induced cell signalling as judged by induction of proinflammatory cytokine production. Our data indicate that PI3K and mTOR are involved in induction of proinflammatory cytokines after HRV infection, and that autophagy has little role in the cytokine response to HRV or control of HRV replication.


Subject(s)
Autophagy , Epithelial Cells/microbiology , Phosphoinositide-3 Kinase Inhibitors , Picornaviridae Infections/enzymology , Picornaviridae Infections/physiopathology , Protein Kinase Inhibitors/pharmacology , Rhinovirus/physiology , Cell Line , Cytokines/immunology , Epithelial Cells/immunology , Epithelial Cells/pathology , Host-Pathogen Interactions , Humans , Phosphatidylinositol 3-Kinases/immunology , Picornaviridae Infections/immunology , Signal Transduction , TOR Serine-Threonine Kinases/immunology
7.
J Virol ; 88(12): 6586-98, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24672044

ABSTRACT

UNLABELLED: Phosphatidylinositol 4-kinase IIIß (PI4KB) is a host factor required for the replication of certain picornavirus genomes. We previously showed that nonstructural proteins 2B, 2BC, 2C, 3A, and 3AB of Aichi virus (AiV), a picornavirus, interact with the Golgi protein, acyl-coenzyme A binding domain containing 3 (ACBD3), which interacts with PI4KB. These five viral proteins, ACBD3, PI4KB, and the PI4KB product phosphatidylinositol 4-phosphate (PI4P) colocalize to the AiV RNA replication sites (J. Sasaki et al., EMBO J. 31:754-766, 2012). We here examined the roles of these viral and cellular molecules in the formation of AiV replication complexes. Immunofluorescence microscopy revealed that treatment of AiV polyprotein-expressing cells with a small interfering RNA targeting ACBD3 abolished colocalization of the viral 2B, 2C, and 3A proteins with PI4KB. A PI4KB-specific inhibitor also prevented their colocalization. Virus RNA replication increased the level of cellular PI4P without affecting that of PI4KB, and individual expression of 2B, 2BC, 2C, 3A, or 3AB stimulated PI4P generation. These results suggest that the viral protein/ACBD3/PI4KB complex plays an important role in forming the functional replication complex by enhancing PI4P synthesis. Of the viral proteins, 3A and 3AB were shown to stimulate the in vitro kinase activity of PI4KB through forming a 3A or 3AB/ACBD3/PI4KB complex, whereas the ACBD3-mediated PI4KB activation by 2B and 2C remains to be demonstrated. IMPORTANCE: The phosphatidylinositol 4-kinase PI4KB is a host factor required for the replication of certain picornavirus genomes. Aichi virus, a picornavirus belonging to the genus Kobuvirus, forms a complex comprising one of the viral nonstructural proteins 2B, 2BC, 2C, 3A, and 3AB, the Golgi protein ACBD3, and PI4KB to synthesize PI4P at the sites for viral RNA replication. However, the roles of this protein complex in forming the replication complex are unknown. This study showed that virus RNA replication and individual viral proteins enhance the level of cellular PI4P, and suggested that the viral protein/ACBD3/PI4KB complex plays an important role in forming a functional replication complex. Thus, the present study provides a new example of modulation of cellular lipid metabolism by viruses to support the replication of their genomes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Kobuvirus/physiology , Membrane Proteins/metabolism , Phosphatidylinositol Phosphates/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Picornaviridae Infections/enzymology , Viral Nonstructural Proteins/metabolism , Virus Replication , Adaptor Proteins, Signal Transducing/genetics , Humans , Kobuvirus/genetics , Membrane Proteins/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Picornaviridae Infections/genetics , Picornaviridae Infections/virology , Protein Binding , Protein Transport , Viral Nonstructural Proteins/genetics
8.
Vet Microbiol ; 162(1): 254-8, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23265242

ABSTRACT

The D11-JW-018 strain of duck hepatitis A virus (DHAV) was isolated from 7-day-old ducklings in Kyeonggi province, South Korea with no clinical signs of typical hepatitis. Phylogenetic analyzed of whole genome showed that D11-JW-018 strain was belonged to DHAV-3 genotype. The pathogenicity of the D11-JW-018 strain in 1-, 7-, 14-, and 21-day-old ducklings was examined. Mortality of D11-JW-018 strain was lower than DRL-62 (DHAV-1) age-dependent but incubation period was longer in 1-day-old ducklings. Unlike DRL-62 strain infection, D11-JW-018 strain induced only liver discoloration without hemorrhagic mottling and lymphocyte infiltration and bile duct hyperplasia in histological lesion. The D11-JW-018 strain was detected only in the heart, liver, spleen, gall bladder, pancreas, and kidney among 12 organs in infected 1-day-old ducklings. Serum biochemical analyses revealed a significant difference in aspartate transaminase and alanine transaminase between the D11-JW-018 strain-infected ducklings and those infected with the DRL-62 strain (P<0.05). We identified the D11-JW-018 strain in South Korean ducklings and provide the characteristics of DHAV-3.


Subject(s)
Ducks , Hepatitis Virus, Duck/classification , Hepatitis, Viral, Animal/virology , Picornaviridae Infections/veterinary , Poultry Diseases/virology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Hepatitis Virus, Duck/genetics , Hepatitis Virus, Duck/isolation & purification , Hepatitis, Viral, Animal/enzymology , Hepatitis, Viral, Animal/pathology , Phylogeny , Picornaviridae Infections/enzymology , Picornaviridae Infections/pathology , Picornaviridae Infections/virology , Poultry Diseases/enzymology , Poultry Diseases/pathology , Republic of Korea
9.
J Immunol ; 188(9): 4621-30, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22461694

ABSTRACT

Matrix metalloproteinase-9 is implicated in airway inflammation and airway remodeling in asthma. We have previously confirmed that human rhinovirus-16 (HRV-16) infection increases MMP-9 expression both in vivo and in vitro. However, the role of the AP-1 sites within the MMP-9 promoter and the effect of commonly used asthma pharmacotherapies in modulating human rhinovirus (HRV)-induced MMP-9 production have not yet been elucidated. Experiments were performed in vitro in the human bronchial epithelial (HBE) cell line BEAS-2B and in primary HBE cells obtained from non-transplanted lungs. Using site-directed mutagenesis approaches, AP-1 sites were found to be necessary for HRV-induced MMP-9 promoter drive. EMSAs and supershift assays identified complexes consisting of Fos-related Ag-1 (Fra-1) in addition to other AP-1 subunits. Small interfering RNA approaches indicated that Fra-1 was induced upon HRV-16 infection in BEAS-2B cells and was necessary for MMP-9 expression in both BEAS-2B and primary HBE cells. Inhibition of MEK1/2 activity using PD98059 and U0126 reduced Fra-1 expression, DNA binding, MMP-9 promoter drive, and MMP-9 protein production. The long-acting ß(2)-agonist formoterol and the glucocorticoid dexamethasone significantly reduced HRV-induced ERK phosphorylation, Fra-1, and MMP-9 expression in BEAS-2B cells. These data indicate that HRV-induced activation of the MEK/ERK MAPK pathway and Fra-1 expression are necessary for the upregulation of MMP-9 and can be modulated by two distinct but commonly used asthma pharmacotherapies. Together, these results offer insights into the mechanisms by which long-acting ß(2)-agonists and glucocorticoids might reduce HRV-related asthma exacerbations.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bronchodilator Agents/pharmacology , Dexamethasone/pharmacology , Ethanolamines/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Matrix Metalloproteinase 9/immunology , Picornaviridae Infections/immunology , Proto-Oncogene Proteins c-fos/immunology , Rhinovirus/immunology , Butadienes/pharmacology , Cell Line , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Formoterol Fumarate , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/immunology , Humans , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/immunology , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase 2/genetics , MAP Kinase Kinase 2/immunology , MAP Kinase Kinase 2/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Nitriles/pharmacology , Phosphorylation/drug effects , Phosphorylation/genetics , Phosphorylation/immunology , Picornaviridae Infections/drug therapy , Picornaviridae Infections/enzymology , Picornaviridae Infections/genetics , Picornaviridae Infections/pathology , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , Response Elements/genetics , Response Elements/immunology , Rhinovirus/genetics , Rhinovirus/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/immunology , Transcription Factor AP-1/metabolism
10.
J Virol ; 86(7): 3605-16, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22258260

ABSTRACT

The activity of phosphatidylinositol 4-kinase class III beta (PI4KIIIß) has been shown to be required for the replication of multiple picornaviruses; however, it is unclear whether a physical association between PI4KIIIß and the viral replication machinery exists and, if it does, whether association is necessary. We examined the ability of the 3A protein from 18 different picornaviruses to form a complex with PI4KIIIß by affinity purification of Strep-Tagged transiently transfected constructs followed by mass spectrometry and Western blotting for putative interacting targets. We found that the 3A proteins of Aichi virus, bovine kobuvirus, poliovirus, coxsackievirus B3, and human rhinovirus 14 all copurify with PI4KIIIß. Furthermore, we found that multiple picornavirus 3A proteins copurify with the Golgi adaptor protein acyl coenzyme A (acyl-CoA) binding domain protein 3 (ACBD3/GPC60), including those from Aichi virus, bovine kobuvirus, human rhinovirus 14, poliovirus, and coxsackievirus B2, B3, and B5. Affinity purification of ACBD3 confirmed interaction with multiple picornaviral 3A proteins and revealed the ability to bind PI4KIIIß in the absence of 3A. Mass-spectrometric analysis of transiently expressed Aichi virus, bovine kobuvirus, and human klassevirus 3A proteins demonstrated that the N-terminal glycines of these 3A proteins are myristoylated. Alanine-scanning mutagenesis along the entire length of Aichi virus 3A followed by transient expression and affinity purification revealed that copurification of PI4KIIIß could be eliminated by mutation of specific residues, with little or no effect on recruitment of ACBD3. One mutation at the N terminus, I5A, significantly reduced copurification of both ACBD3 and PI4KIIIß. The dependence of Aichi virus replication on the activity of PI4KIIIß was confirmed by both chemical and genetic inhibition. Knockdown of ACBD3 by small interfering RNA (siRNA) also prevented replication of both Aichi virus and poliovirus. Point mutations in 3A that eliminate PI4KIIIß association sensitized Aichi virus to PIK93, suggesting that disruption of the 3A/ACBD3/PI4KIIIß complex may represent a novel target for therapeutic intervention that would be complementary to the inhibition of the kinase activity itself.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Picornaviridae Infections/metabolism , Picornaviridae/metabolism , Viral Nonstructural Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Cell Line , Golgi Apparatus/genetics , Humans , Kobuvirus/genetics , Kobuvirus/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Minor Histocompatibility Antigens , Molecular Sequence Data , Phosphotransferases (Alcohol Group Acceptor)/genetics , Picornaviridae/chemistry , Picornaviridae/genetics , Picornaviridae Infections/enzymology , Picornaviridae Infections/genetics , Picornaviridae Infections/virology , Protein Binding , Sequence Alignment , Viral Nonstructural Proteins/genetics
11.
Expert Opin Ther Pat ; 20(1): 59-71, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20021285

ABSTRACT

IMPORTANCE OF THE FIELD: Picornaviruses are small non-enveloped RNA viruses with genomic RNA of 7500 - 8000 nucleotides, whereas coronaviruses (CoV) are RNA viruses with larger genome of 27 - 32 kb. Both types of viruses translate their genetic information into polyprotein precursors that are processed by virally encoded 3C proteases (3C(pro)) and 3C-like proteases (3CL(pro)), respectively, to generate functional viral proteins. The most studied human rhinoviruses (HRV) belonging to picornaviridae family are the main etiologic agents of the common cold. Due to lack of effective drugs, 3C(pro) has served as an excellent target for anti-viral intervention and considerable efforts have been made in the development of inhibitors. Interestingly, the inhibitors of 3C(pro) cannot inhibit 3CL(pro) potently without modification due to subtle differences in their active-site structures, but a group of common inhibitors against 3C(pro) and 3CL(pro) were found recently. AREAS COVERED IN THIS REVIEW: The inhibitors against 3C(pro) reported in the literatures and patents, with a focus on those inhibiting HRV and the dual picornaviral 3C(pro)/coronaviral 3CL(pro) inhibitors, are summarized in this review. WHAT THE READERS WILL GAIN: Readers will rapidly gain an overview of the individual and dual 3C(pro) inhibitors and the structural basis for discriminating them. TAKE HOME MESSAGE: In the future, more selective potent inhibitors against each protease and dual inhibitors against both proteases can be further developed to treat the diseases caused by picornaviruses and CoV.


Subject(s)
Antiviral Agents/pharmacology , Protease Inhibitors/pharmacology , Viral Proteins/antagonists & inhibitors , 3C Viral Proteases , Animals , Coronavirus/drug effects , Coronavirus/enzymology , Coronavirus Infections/drug therapy , Coronavirus Infections/enzymology , Cysteine Endopeptidases , Drug Delivery Systems , Drug Design , Humans , Patents as Topic , Picornaviridae/drug effects , Picornaviridae/enzymology , Picornaviridae Infections/drug therapy , Picornaviridae Infections/enzymology
12.
Int J Biochem Cell Biol ; 41(3): 498-502, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18487072

ABSTRACT

Replication of the picornavirus genome is catalysed by a viral encoded RNA-dependent RNA polymerase, termed 3D polymerase. Together with other viral and host proteins, this enzyme performs its functions in the cytoplasm of host cells. The crystal structure of 3D polymerase from a number of picornaviruses has been determined. This review discusses the structure and function of the poliovirus 3D polymerase. The high error rates of 3D polymerase result in high sequence diversity such that virus populations exist as quasispecies. This phenomenon is thought to facilitate survival of the virus population in complex environments.


Subject(s)
Host-Pathogen Interactions , Picornaviridae Infections/enzymology , Picornaviridae/enzymology , RNA-Dependent RNA Polymerase/metabolism , Virus Replication/genetics , Amino Acid Motifs , Animals , Crystallization , Humans , Mutation , Oligoribonucleotides/metabolism , Organ Specificity , Picornaviridae/genetics , Picornaviridae/growth & development , Picornaviridae/pathogenicity , Picornaviridae Infections/genetics , Picornaviridae Infections/virology , Protein Conformation , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/genetics , Virulence/genetics , Virus Assembly/genetics
13.
Int Arch Allergy Immunol ; 147(4): 299-304, 2008.
Article in English | MEDLINE | ID: mdl-18617749

ABSTRACT

BACKGROUND: Rhinovirus infection is a major cause of asthma exacerbations. While rhinovirus infection is known to generate kinins in the upper respiratory track, little is known about the effect of rhinovirus on kinin generation in the lower airway. We previously identified human tissue kallikrein (hTK) as the principal lung kininogenase during allergic airway inflammation. In this report we investigate the effect of experimental rhinovirus infection on hTK activity in the airways of atopic subjects with and without asthma. METHODS: Eight atopic subjects, 4 with asthma, underwent bronchoscopy with lavage. At least 1 month later, subjects were inoculated with rhinovirus, then underwent repeat bronchoscopy with lavage 4 and 18 days later. hTK mRNA was measured in nasal scrape samples by quantitative real-time PCR. hTK activity (chromogenic substrate assay) and IL-8 levels (ELISA) were assessed in the bronchoalveolar lavage fluid. RESULTS: At day 4 after rhinovirus inoculation, nasal hTK mRNA was modestly increased in both the rhinitis (1.7-fold) and asthmatic (2.1-fold) groups. A doubling or greater increase in hTK activity after rhinovirus infection was observed in all 4 asthmatic subjects (mean 19-fold increase) but only in 1 of 4 atopic subjects without asthma (mean 2-fold increase). Rhinovirus infection also increased the IL-8 protein level in bronchoalveolar lavage fluid, which correlated with hTK activity (R = 0.82). CONCLUSION: Experimental rhinovirus infection in allergic asthmatic subjects is accompanied by increased lower airway hTK activation, which parallels the appearance of IL-8. Rhinovirus-induced hTK activation may contribute to airway inflammation and asthmatic exacerbations.


Subject(s)
Hypersensitivity/enzymology , Picornaviridae Infections/enzymology , Rhinovirus , Tissue Kallikreins/metabolism , Adult , Enzyme Activation , Female , Humans , Interleukin-8/biosynthesis , Male , Picornaviridae Infections/immunology , RNA, Messenger/analysis , Tissue Kallikreins/genetics
14.
Cell Physiol Biochem ; 17(3-4): 159-66, 2006.
Article in English | MEDLINE | ID: mdl-16543732

ABSTRACT

Rhinoviral infections belong to the most frequent human infections characterized by common cold, chronic bronchitis, exacerbations of asthma, otitis media and sinusitis. Here, we define molecular mechanisms that mediate infections of human epithelial cells with human rhinovirus strain 14 (RV14). We demonstrate that RV14 activates p38-MAPKinase (p38-K) in a biphasic time course. Early stimulation of p38-K by RV14 was observed a few minutes after initiation of the infection, while the late increase of p38-K activity occurred 7-12 hrs upon infection. The stimulation of p38-K was mediated by the small G-protein RhoA,which was activated by RV14. Transfection of a genetic construct preventing RhoA activation blocked RV14-induced p38-K activation. Further, integrity of cholesterol and sphingolipid-enriched membrane domains was required for RV14-mediated p38-K activation, which was inhibited by destruction of membrane rafts. The data indicate that RV employs a signaling cascade from membrane rafts via the small G-protein RhoA to p38-K to infect human cells.


Subject(s)
Membrane Microdomains/metabolism , Picornaviridae Infections/virology , Rhinovirus/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Blotting, Western , Enzyme Activation , HeLa Cells , Humans , Membrane Microdomains/chemistry , Picornaviridae Infections/enzymology , Rhinovirus/classification , Serotyping , beta-Cyclodextrins/pharmacology , rhoA GTP-Binding Protein/analysis
15.
J Biomed Sci ; 11(5): 617-22, 2004.
Article in English | MEDLINE | ID: mdl-15316137

ABSTRACT

Reactive oxygen species produced by NADPH oxidase appear to play a role in the response of human lung fibroblast cells to rhinovirus infection. The purpose of the following studies was to characterize the NADPH oxidase components in these cells, to examine the effect of rhinovirus challenge on the expression of these proteins, and to confirm previous studies suggesting a role for p47-phox in the oxidant response to rhinovirus challenge. The results revealed that the NADPH oxidase components p47-phox, p67-phox, p22-phox, and NOX4 were expressed in lung fibroblast cells. In contrast, gp91-phox was not expressed in this cell line. Expression of p67-phox was upregulated by rhinovirus challenge. The functional role of NADPH oxidase in the rhinovirus-induced oxidant stress and elaboration of IL-8 was confirmed by detection of significant reductions in oxidant stress and IL-8 elaboration following transfection of the cells with antisense nucleotides to p47-phox. The lack of gp91- phox in cultured lung fibroblast cells, the induction of p67-phox by rhinovirus, and the confirmation of participation of p47-phox in rhinovirus-induced oxidant stress are significant findings of this study and form a basis for future investigations into understanding the mechanisms of the NADPH oxidase response to rhinovirus infection.


Subject(s)
Fibroblasts/enzymology , Lung/enzymology , NADPH Oxidases/metabolism , Base Sequence , Cells, Cultured , DNA Primers , Humans , Lung/cytology , NADPH Oxidases/genetics , Picornaviridae Infections/enzymology , Reverse Transcriptase Polymerase Chain Reaction
16.
Am J Respir Cell Mol Biol ; 24(5): 633-9, 2001 May.
Article in English | MEDLINE | ID: mdl-11350835

ABSTRACT

Rhinovirus (RV) is a major cause of wheezing in asthmatics and has been reported to cause beta2 adrenergic receptor hyporesponsiveness in human airway smooth muscle (HASM) via cellular secretion of interleukin (IL)-1beta. We studied the effects of IL-1beta and RV on cyclic adenosine monophosphate (cAMP) production in HASM cells. Chronic incubation with IL-1beta or RV caused a significant increase (approximately 3- and approximately 2-fold, respectively) in forskolin (FSK)-stimulated cAMP production, suggesting a sensitization of adenylyl cyclase (AC). The observed augmentation of FSK-stimulated cAMP formation by IL-1beta was completely abrogated by pretreatment with an IL-1 receptor antagonist or cycloheximide, demonstrating that the effect is mediated via the IL-1 receptor 1 (IL-1R1) and that de novo protein synthesis is required. In contrast, RV-induced AC sensitization was not mediated via the IL-1R1 but was observed to be protein kinase C-dependent. We suggest that the sensitization of AC observed after exposure to IL-1beta or RV infection is a cellular defense mechanism to promote pathways that induce relaxation in the inflamed airway.


Subject(s)
Adenylyl Cyclases/metabolism , Interleukin-1/pharmacology , Muscle, Smooth/enzymology , Rhinovirus/metabolism , Trachea , Adenylate Cyclase Toxin , Adrenergic beta-Agonists/pharmacology , Cells, Cultured , Colforsin/pharmacology , Cyclic AMP/metabolism , Cycloheximide/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Feedback , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , Humans , Interleukin 1 Receptor Antagonist Protein , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Muscle, Smooth/virology , Picornaviridae Infections/enzymology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Synthesis Inhibitors/pharmacology , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/metabolism , Receptors, Interleukin-1 Type I , Sialoglycoproteins/pharmacology , Virulence Factors, Bordetella/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...