Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 10(2): e1003914, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24550726

ABSTRACT

Vector-borne viruses are an important class of emerging and re-emerging pathogens; thus, an improved understanding of the cellular factors that modulate infection in their respective vertebrate and insect hosts may aid control efforts. In particular, cell-intrinsic antiviral pathways restrict vector-borne viruses including the type I interferon response in vertebrates and the RNA interference (RNAi) pathway in insects. However, it is likely that additional cell-intrinsic mechanisms exist to limit these viruses. Since insects rely on innate immune mechanisms to inhibit virus infections, we used Drosophila as a model insect to identify cellular factors that restrict West Nile virus (WNV), a flavivirus with a broad and expanding geographical host range. Our genome-wide RNAi screen identified 50 genes that inhibited WNV infection. Further screening revealed that 17 of these genes were antiviral against additional flaviviruses, and seven of these were antiviral against other vector-borne viruses, expanding our knowledge of invertebrate cell-intrinsic immunity. Investigation of two newly identified factors that restrict diverse viruses, dXPO1 and dRUVBL1, in the Tip60 complex, demonstrated they contributed to antiviral defense at the organismal level in adult flies, in mosquito cells, and in mammalian cells. These data suggest the existence of broadly acting and functionally conserved antiviral genes and pathways that restrict virus infections in evolutionarily divergent hosts.


Subject(s)
DNA Helicases/genetics , Drosophila Proteins/genetics , Flavivirus Infections/genetics , Host-Parasite Interactions/genetics , Karyopherins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Animals , Culicidae , Drosophila melanogaster , Flavivirus/genetics , Flavivirus Infections/immunology , Genome-Wide Association Study , Humans , RNA, Small Interfering/analysis , RNA, Small Interfering/genetics , Reverse Transcriptase Polymerase Chain Reaction , West Nile virus/genetics , West Nile virus/immunology , Exportin 1 Protein
2.
Cell Rep ; 5(6): 1737-48, 2013 Dec 26.
Article in English | MEDLINE | ID: mdl-24332855

ABSTRACT

Alphaviruses are a large class of insect-borne human pathogens and little is known about the host-factor requirements for infection. To identify such factors, we performed a genome-wide RNAi screen using model Drosophila cells and validated 94 genes that impacted infection of Sindbis virus (SINV), the prototypical alphavirus. We identified a conserved role for SEC61A and valosin-containing protein (VCP) in facilitating SINV entry in insects and mammals. SEC61A and VCP selectively regulate trafficking of the entry receptor NRAMP2, and loss or pharmacological inhibition of these proteins leads to altered NRAMP2 trafficking to lysosomal compartments and proteolytic digestion within lysosomes. NRAMP2 is the major iron transporter in cells, and loss of NRAMP2 attenuates intracellular iron transport. Thus, this study reveals genes and pathways involved in both infection and iron homeostasis that may serve as targets for antiviral therapeutics or for iron-imbalance disorders.


Subject(s)
Adenosine Triphosphatases/metabolism , Drosophila Proteins/metabolism , Genome, Insect , Membrane Proteins/metabolism , Sindbis Virus/pathogenicity , Virus Internalization , Adenosine Triphosphatases/genetics , Aedes/genetics , Aedes/metabolism , Aedes/virology , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line, Tumor , Drosophila/genetics , Drosophila/metabolism , Drosophila/virology , Drosophila Proteins/genetics , Humans , Iron/metabolism , Membrane Proteins/genetics , Protein Transport , RNA Interference , RNA, Small Interfering/genetics , SEC Translocation Channels , Valosin Containing Protein
3.
Cell Host Microbe ; 10(2): 97-104, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21843867

ABSTRACT

Alphaviruses, including several emerging human pathogens, are a large family of mosquito-borne viruses with Sindbis virus being a prototypical member of the genus. The host factor requirements and receptors for entry of this class of viruses remain obscure. Using a Drosophila system, we identified the divalent metal ion transporter natural resistance-associated macrophage protein (NRAMP) as a host cell surface molecule required for Sindbis virus binding and entry into Drosophila cells. Consequently, flies mutant for dNRAMP were protected from virus infection. NRAMP2, the ubiquitously expressed vertebrate homolog, mediated binding and infection of Sindbis virus into mammalian cells, and murine cells deficient for NRAMP2 were nonpermissive to infection. Alphavirus glycoprotein chimeras demonstrated that the requirement for NRAMP2 is at the level of Sindbis virus entry. Given the conserved structure of alphavirus glycoproteins, and the widespread use of transporters for viral entry, other alphaviruses may use conserved multipass membrane proteins for infection.


Subject(s)
Cation Transport Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/virology , Receptors, Virus/metabolism , Sindbis Virus/pathogenicity , Alphavirus Infections/metabolism , Alphavirus Infections/virology , Animals , Biotinylation/methods , Cation Transport Proteins/genetics , Cell Line , Culicidae , Drosophila Proteins/genetics , Humans , Immunoprecipitation , Iron/pharmacology , Mammals , Mice , RNA Interference , RNA Stability , Sindbis Virus/drug effects , Sindbis Virus/genetics , Transfection , Vesiculovirus/genetics , Vesiculovirus/metabolism , Vesiculovirus/pathogenicity , Virus Attachment , Virus Internalization , West Nile virus/drug effects , West Nile virus/genetics , West Nile virus/pathogenicity
4.
Cell Host Microbe ; 9(1): 70-82, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-21238948

ABSTRACT

Enteroviruses, including coxsackievirus B (CVB) and poliovirus (PV), can access the CNS through the blood brain barrier (BBB) endothelium to cause aseptic meningitis. To identify cellular components required for CVB and PV infection of human brain microvascular endothelial cells, an in vitro BBB model, we performed comparative RNAi screens and identified 117 genes that influenced infection. Whereas a large proportion of genes whose depletion enhanced infection (17 of 22) were broadly antienteroviral, only 46 of the 95 genes whose depletion inhibited infection were required by both CVB and PV and included components of cell signaling pathways such as adenylate cyclases. Downregulation of genes including Rab GTPases, Src tyrosine kinases, and tyrosine phosphatases displayed specificity in their requirement for either CVB or PV infection. These findings highlight the pathways hijacked by enteroviruses for entry and replication in the BBB endothelium, a specialized and clinically relevant cell type for these viruses.


Subject(s)
Endothelial Cells/immunology , Endothelial Cells/virology , Enterovirus B, Human/immunology , Enterovirus B, Human/pathogenicity , Host-Pathogen Interactions , Poliovirus/immunology , Poliovirus/pathogenicity , Blood-Brain Barrier , Gene Silencing , Humans , Mass Screening/methods , Models, Biological
5.
PLoS One ; 5(11): e15483, 2010 Nov 24.
Article in English | MEDLINE | ID: mdl-21124804

ABSTRACT

As an arthropod-borne human pathogen, Rift Valley fever virus (RVFV) cycles between an insect vector and mammalian hosts. Little is known about the cellular requirements for infection in either host. Here we developed a tissue culture model for RVFV infection of human and insect cells that is amenable to high-throughput screening. Using this approach we screened a library of 1280 small molecules with pharmacologically defined activities and identified 59 drugs that inhibited RVFV infection with 15 inhibiting RVFV replication in both human and insect cells. Amongst the 15 inhibitors that blocked infection in both hosts was a subset that inhibits protein kinase C. Further studies found that infection is dependent upon the novel protein kinase C isozyme epsilon (PKCε) in both human and insect cells as well as in adult flies. Altogether, these data show that inhibition of cellular factors required for early steps in the infection cycle including PKCε can block RVFV infection, and may represent a starting point for the development of anti-RVFV therapeutics.


Subject(s)
Insecta/virology , Protein Kinase C-epsilon/metabolism , Rift Valley fever virus/physiology , Acetophenones/pharmacology , Animals , Benzophenanthridines/pharmacology , Benzopyrans/pharmacology , Cell Line , Chlorocebus aethiops , Diptera/genetics , Diptera/metabolism , Diptera/virology , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , HEK293 Cells , Humans , Immunoblotting , Insecta/cytology , Insecta/metabolism , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Kinase C-epsilon/genetics , RNA Interference , Rift Valley fever virus/drug effects , Vero Cells
6.
Curr Opin Immunol ; 22(1): 4-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20137906

ABSTRACT

The study of Drosophila, and other genetically tractable insects, has expanded our understanding of innate immunity and more recently antiviral innate mechanisms. The Drosophila antiviral program includes inflammatory signaling cascades as well as antiviral RNA silencing and autophagy. This review will highlight the recent discoveries in antiviral immunity in insects and will reveal some of the lessons learned.


Subject(s)
Drosophila melanogaster/immunology , Drosophila melanogaster/virology , Immunity, Innate , Animals , Autophagy , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Host-Pathogen Interactions , Humans , RNA Interference , Signal Transduction
7.
J Infect Dis ; 197(3): 465-73, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18184091

ABSTRACT

Pseudomonas aeruginosa undergoes phase variation in the expression of the phosphorylcholine (ChoP) epitope, a structure crucial for the virulence of several respiratory pathogens. In this study, ChoP expression analysis comparing organisms from acute and chronic infections revealed that expression of ChoP at 37 degrees C was higher among strains from chronic infections. Coimmunoprecipitation experiments and mass spectrometry analysis demonstrated that ChoP was on the protein elongation factor Tu. The presence of ChoP at the surface was confirmed by immunofluorescence and flow cytometry analysis of intact bacteria. Pretreatment of bronchial epithelial cells or mice with a platelet-activating factor receptor (PAFR) antagonist reduced adhesion and invasion of the ChoP-positive P. aeruginosa isolates. Results of this study suggest that ChoP expression may represent a novel phenotype expressed by the chronic infection isolates that could mediate P. aeruginosa colonization of the epithelial airway by means of the interaction with the PAFR.


Subject(s)
Bacterial Proteins/pharmacology , Phosphorylcholine/pharmacology , Pseudomonas Infections/physiopathology , Pseudomonas aeruginosa/pathogenicity , Respiratory Mucosa/microbiology , Acute Disease , Animals , Chronic Disease , Disease Models, Animal , Humans , Mice , Mice, Inbred ICR , Pseudomonas aeruginosa/isolation & purification , Respiratory Mucosa/drug effects , Sputum/microbiology
8.
J Infect Dis ; 196 Suppl 2: S237-46, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17940955

ABSTRACT

BACKGROUND: The lectin DC-SIGN (dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin) augments Ebola virus (EBOV) infection. However, it its unclear whether DC-SIGN promotes only EBOV attachment (attachment factor function, nonessential) or actively facilitates EBOV entry (receptor function, essential). METHODS: We investigated whether DC-SIGN on B cell lines and dendritic cells acts as an EBOV attachment factor or receptor. RESULTS: Engineered DC-SIGN expression rendered some B cell lines susceptible to EBOV glycoprotein (EBOV GP)-driven infection, whereas others remained refractory, suggesting that cellular factors other than DC-SIGN are also required for susceptibility to EBOV infection. Augmentation of entry was independent of efficient DC-SIGN internalization and might not involve lectin-mediated endocytic uptake of virions. Therefore, DC-SIGN is unlikely to function as an EBOV receptor on B cell lines; instead, it might concentrate virions onto cells, thereby allowing entry into cell lines expressing low levels of endogenous receptor(s). Indeed, artificial concentration of virions onto cells mirrored DC-SIGN expression, confirming that optimization of viral attachment is sufficient for EBOV GP-driven entry into some B cell lines. Finally, EBOV infection of dendritic cells was only partially dependent on mannose-specific lectins, such as DC-SIGN, suggesting an important contribution of other factors. CONCLUSIONS: Our results indicate that DC-SIGN is not an EBOV receptor but, rather, is an attachment-promoting factor that boosts entry into B cell lines susceptible to low levels of EBOV GP-mediated infection.


Subject(s)
Cell Adhesion Molecules/physiology , Ebolavirus/physiology , Glycoproteins/physiology , Lectins, C-Type/physiology , Receptors, Cell Surface/physiology , Viral Proteins/physiology , Animals , B-Lymphocytes/physiology , Base Sequence , Cell Adhesion Molecules/genetics , Cell Line , Humans , Lectins, C-Type/genetics , Molecular Sequence Data , Plasmids , Polymerase Chain Reaction , Receptors, Cell Surface/genetics , Receptors, Virus/physiology
9.
J Biol Chem ; 282(17): 12397-409, 2007 Apr 27.
Article in English | MEDLINE | ID: mdl-17339324

ABSTRACT

The two lectin receptors, CLEC-2 and Dectin-1, have been shown to signal through a Syk-dependent pathway, despite the presence of only a single YXXL in their cytosolic tails. In this study, we show that stimulation of CLEC-2 in platelets and in two mutant cell lines is dependent on the YXXL motif and on proteins that participate in signaling by immunoreceptor tyrosine-based activation motif receptors, including Src, Syk, and Tec family kinases, and on phospholipase Cgamma. Strikingly, mutation of either Src homology (SH) 2 domain of Syk blocks signaling by CLEC-2 despite the fact that it has only a single YXXL motif. Furthermore, signaling by CLEC-2 is only partially dependent on the BLNK/SLP-76 family of adapter proteins in contrast to that of immunoreceptor tyrosine-based activation motif receptors. The C-type lectin receptor, Dectin-1, which contains a YXXL motif preceded by the same four amino acids as for CLEC-2 (DEDG), signals like CLEC-2 and also requires the two SH2 domains of Syk and is only partially dependent on the BLNK/SLP-76 family of adapters. In marked contrast, the C-type lectin receptor, DC-SIGN, which has a distinct series of amino acids preceding a single YXXL, signals independent of this motif. A mutational analysis of the DEDG sequence of CLEC-2 revealed that the glycine residue directly upstream of the YXXL tyrosine is important for CLEC-2 signaling. These results demonstrate that CLEC-2 and Dectin-1 signal through a single YXXL motif that requires the tandem SH2 domains of Syk but is only partially dependent on the SLP-76/BLNK family of adapters.


Subject(s)
Blood Platelets/metabolism , Cell Adhesion Molecules/metabolism , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Motifs/genetics , Animals , Cell Adhesion Molecules/genetics , Cell Line , Chickens , Humans , Intracellular Signaling Peptides and Proteins , Lectins, C-Type/genetics , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Mutation , Nerve Tissue Proteins/genetics , Protein-Tyrosine Kinases/metabolism , Receptors, Cell Surface/genetics , Syk Kinase , src Homology Domains/genetics , src-Family Kinases/metabolism
10.
Virology ; 359(2): 336-48, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17055550

ABSTRACT

A major neutralizing epitope (here referred to as the T332 epitope) located on the lateral surface of domain III (DIII) of the West Nile virus (WNV) envelope protein has been identified based on the analysis of murine monoclonal antibodies. However, little is known about the humoral immune response against WNV in a natural host or whether DIII in general or the T332 epitope in particular are important targets of neutralizing antibodies in vivo. To characterize the types of antibodies produced during infection with WNV, we studied a group of naturally infected horses. Using immune adsorption assays coupled with the use of virus particles bearing mutations in the T332 epitope, we found that in some animals neutralizing activity against DIII and the T332 epitope was below the limit of detection. In contrast, some animals generated a significant fraction of neutralizing activity to DIII and the T332 epitope. Thus, while antibodies to the T332 epitope did not represent a significant fraction of the total antibody response in the infected animals studied, in some horses, they comprised a significant fraction of neutralizing activity, making this an important but far from dominant neutralizing epitope. Rather, the neutralizing response to WNV generated in infected horses is both variable and polyclonal in nature, with epitopes within and outside of DIII playing important roles.


Subject(s)
Antibodies, Viral/immunology , Horse Diseases/immunology , Horse Diseases/virology , Viral Envelope Proteins/immunology , West Nile Fever/veterinary , Animals , Antibodies, Viral/blood , Antibody Affinity , Cell Line , Epitopes/chemistry , Epitopes/immunology , Horse Diseases/blood , Horses , Humans , Neutralization Tests , Protein Conformation , Viral Envelope Proteins/chemistry , West Nile Fever/blood , West Nile Fever/immunology
11.
J Virol ; 80(3): 1290-301, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16415006

ABSTRACT

The C-type lectins DC-SIGN and DC-SIGNR bind mannose-rich glycans with high affinity. In vitro, cells expressing these attachment factors efficiently capture, and are infected by, a diverse array of appropriately glycosylated pathogens, including dengue virus. In this study, we investigated whether these lectins could enhance cellular infection by West Nile virus (WNV), a mosquito-borne flavivirus related to dengue virus. We discovered that DC-SIGNR promoted WNV infection much more efficiently than did DC-SIGN, particularly when the virus was grown in human cell types. The presence of a single N-linked glycosylation site on either the prM or E glycoprotein of WNV was sufficient to allow DC-SIGNR-mediated infection, demonstrating that uncleaved prM protein present on a flavivirus virion can influence viral tropism under certain circumstances. Preferential utilization of DC-SIGNR was a specific property conferred by the WNV envelope glycoproteins. Chimeras between DC-SIGN and DC-SIGNR demonstrated that the ability of DC-SIGNR to promote WNV infection maps to its carbohydrate recognition domain. WNV virions and subviral particles bound to DC-SIGNR with much greater affinity than DC-SIGN. We believe this is the first report of a pathogen interacting more efficiently with DC-SIGNR than with DC-SIGN. Our results should lead to the discovery of new mechanisms by which these well-studied lectins discriminate among ligands.


Subject(s)
Cell Adhesion Molecules/physiology , Lectins, C-Type/physiology , Receptors, Cell Surface/physiology , West Nile virus/physiology , West Nile virus/pathogenicity , Animals , Binding Sites , Cell Adhesion , Cell Adhesion Molecules/genetics , Cell Line , Cricetinae , Glycosylation , HeLa Cells , Humans , Lectins, C-Type/genetics , Ligands , Mutation , Receptors, Cell Surface/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/physiology , Virulence , West Nile virus/genetics
12.
J Virol ; 79(21): 13262-74, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16227249

ABSTRACT

West Nile virus (WNV) encodes two envelope proteins, premembrane (prM) and envelope (E). While the prM protein of all WNV strains contains a single N-linked glycosylation site, not all strains contain an N-linked site in the E protein. The presence of N-linked glycosylation on flavivirus E proteins has been linked to virus production, pH sensitivity, and neuroinvasiveness. Therefore, we examined the impact of prM and E glycosylation on WNV assembly and infectivity. Similar to other flaviviruses, expression of WNV prM and E resulted in the release of subviral particles (SVPs). Removing the prM glycosylation site in a lineage I or II strain decreased SVP release, as did removal of the glycosylation site in a lineage I E protein. Addition of the E protein glycosylation site in a lineage II strain that lacked this site increased SVP production. Similar results were obtained in the context of either reporter virus particles (RVPs) or infectious lineage II WNV. RVPs or virions bearing combinations of glycosylated and nonglycosylated forms of prM and E could infect mammalian, avian, and mosquito cells (BHK-21, QT6, and C6/36, respectively). Those particles lacking glycosylation on the E protein were modestly more infectious per genome copy on BHK-21 and QT6 cells, while this absence greatly enhanced the infection of C6/36 cells. Thus, glycosylation of WNV prM and E proteins can affect the efficiency of virus release and infection in a manner that is cell type and perhaps species dependent. This suggests a multifaceted role for envelope N-linked glycosylation in WNV biology and tropism.


Subject(s)
Viral Envelope Proteins/metabolism , West Nile Fever/virology , West Nile virus/physiology , West Nile virus/pathogenicity , Animals , Cell Line , Endoplasmic Reticulum/metabolism , Glycosylation , Humans , Virus Assembly , West Nile virus/metabolism
13.
Virology ; 336(1): 70-82, 2005 May 25.
Article in English | MEDLINE | ID: mdl-15866072

ABSTRACT

We produced nine monoclonal antibodies (MAbs) directed against the West Nile virus E glycoprotein using three different immunization strategies: inactivated virus, naked DNA, and recombinant protein. Most of the MAbs bound to conformation dependent epitopes in domain III of the E protein. Four of the MAbs neutralized WNV infection and bound to the same region of domain III with high affinity. The neutralizing MAbs were obtained from mice immunized with inactivated virus alone or in combination with a DNA plasmid. In contrast, MAbs obtained by immunization with a soluble version of the E glycoprotein did not exhibit neutralizing activity. These non-neutralizing antibodies were cross-reactive with several other flaviviruses, including Saint Louis encephalitis, Japanese encephalitis, Yellow Fever and Powassan viruses. Interestingly, some non-neutralizing MAbs bound with high affinity to domains I or III, indicating that both affinity and the precise epitope recognized by an antibody are important determinants of WNV neutralization.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , West Nile virus/immunology , Animals , Antigens, Viral/immunology , Cross Reactions , Encephalitis Virus, Japanese/immunology , Encephalitis Virus, St. Louis/immunology , Encephalitis Viruses, Tick-Borne/immunology , Enzyme-Linked Immunosorbent Assay , Epitope Mapping , Epitopes/immunology , Glycoproteins/immunology , Mice , Neutralization Tests , Protein Structure, Tertiary , Yellow fever virus/immunology
14.
Virology ; 334(1): 28-40, 2005 Mar 30.
Article in English | MEDLINE | ID: mdl-15749120

ABSTRACT

West Nile virus is a mosquito-borne, neurotropic flavivirus that causes encephalitis in humans and animals. Since being introduced into the Western hemisphere in 1999, WNV has spread rapidly across North America, identifying this virus as an important emerging pathogen. In this study, we developed a DNA-launched infectious molecular clone of WNV that encodes a GFP reporter gene. Transfection of cells with the plasmid encoding this recombinant virus (pWNII-GFP) resulted in the production of infectious WNV capable of expressing GFP at high levels shortly after infection of a variety of cell types, including primary neurons and dendritic cells. Infection of cells with WNII-GFP virus was productive, and could be inhibited with both monoclonal antibodies and interferon-beta, highlighting the potential of this system in the development and characterization of novel inhibitors and therapeutics for WNV infection. As expected, insertion of the reporter gene into the viral genome was associated with a reduced rate of viral replication, providing the selective pressure for the development of variants that no longer encoded the full-length reporter gene cassette. We anticipate this DNA-based, infectious WNV reporter virus will allow novel approaches for the study of WNV infection and its inhibition both in vitro and in vivo.


Subject(s)
Genes, Reporter , Green Fluorescent Proteins/genetics , West Nile virus/genetics , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Viral/administration & dosage , Cell Line , Cricetinae , Gene Expression , HeLa Cells , Humans , Interferon Type I/pharmacology , Kinetics , Plasmids/genetics , Recombinant Proteins/genetics , Transfection , Virus Replication/drug effects , West Nile virus/immunology , West Nile virus/pathogenicity , West Nile virus/physiology
15.
J Virol ; 77(16): 8962-72, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12885913

ABSTRACT

We have studied the receptor-specific function of four linker-insertion mutants of herpes simplex virus type 1 glycoprotein D (gD) representing each of the functional regions of gD. We used biosensor analysis to measure binding of the gD mutants to the receptors HVEM (HveA) and nectin-1 (HveC). One of the mutants, gD(inverted Delta 34t), failed to bind HVEMt but showed essentially wild-type (WT) affinity for nectin-1t. The receptor-binding kinetics and affinities of the other three gD mutants varied over a 1,000-fold range, but each mutant had the same affinity for both receptors. All of the mutants were functionally impaired in virus entry and cell fusion, and the levels of activity were strikingly similar in these two assays. gD(inverted Delta 34)-containing virus was defective on HVEM-expressing cells but did enter nectin-1-expressing cells to about 60% of WT levels. This showed that the defect of this form of gD on HVEM-expressing cells was primarily one of binding and that this was separable from its later function in virus entry. gD(inverted Delta 243t) showed WT binding affinity for both receptors, but virus containing this form of gD had a markedly reduced rate of entry, suggesting that gD(inverted Delta 243) is impaired in a postbinding step in the entry process. There was no correlation between gD mutant activity in fusion or virus entry and receptor-binding affinity. We conclude that gD functions in virus entry and cell fusion regardless of its receptor-binding kinetics and that as long as binding to a functional receptor occurs, entry will progress.


Subject(s)
Herpesvirus 1, Human/physiology , Membrane Fusion/physiology , Receptors, Virus/physiology , Viral Envelope Proteins/physiology , Cell Line , Genetic Complementation Test , Models, Molecular , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
16.
AIDS ; 16(12): 1603-8, 2002 Aug 16.
Article in English | MEDLINE | ID: mdl-12172081

ABSTRACT

BACKGROUND: Two of the fusion inhibitors T-20 and 5-helix polypeptide have been shown to be potent inhibitors of cell-to-cell fusion and are currently under investigation as therapy for HIV-1. OBJECTIVES: To examine variability of HIV-1 gp41 heptads repeat regions (HR1 and HR2), with special emphasis on the presence of T-20 resistance mutations and 5-helix variability at critical epitopes, in treatment-naive patients infected with diverse HIV-1 subtypes from different geographic regions. METHODS: A total of 150 specimens representing HIV-1 group M subtypes (A-G) from persons naive to HIV-1 viral entry inhibitor therapy were used to amplify and sequence a 506 bp segment of transmembrane protein. RESULTS: In general, both HR1 (a.a. 540-593) and HR2 (a.a. 628-673) domains were highly conserved. Sequence analysis of the T-20 resistant domain (a.a. 547-549, GIV) revealed that 99% of the specimens (149 of 150) carried a T-20 sensitive genotype. The critical epitopes involved in the 5-helix interaction include residues at positions 628W, 631W, 635I, 638Y, 642I, 645L, 649S, 652Q, 656N, and 659E. Analysis of the 150 specimens revealed that all had identical residues at six of these positions, whereas two positions had minor variations (635 and 649) and two (645 and 659) appeared to have subtype-specific substitutions. CONCLUSIONS: This data indicates that there is limited resistance to T-20 in these worldwide populations and that the critical epitopes for effective 5-helix binding are highly conserved across all subtypes. Taken together, these data suggest that T-20 and 5-helix should provide useful additives to current antiretroviral therapy for clinical management of HIV disease.


Subject(s)
Drug Resistance, Microbial/genetics , HIV Fusion Inhibitors/pharmacology , HIV-1/drug effects , Membrane Fusion/drug effects , Amino Acid Sequence , HIV Envelope Protein gp41/chemistry , HIV Infections/blood , HIV-1/physiology , Humans , Molecular Sequence Data
17.
Microbiology (Reading) ; 146 ( Pt 10): 2495-2508, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11021925

ABSTRACT

Isolates of Pseudomonas aeruginosa from chronic lung infections in cystic fibrosis (CF) patients have phenotypes distinct from those initially infecting CF patients, as well as from other clinical or environmental isolates. To gain a better understanding of the differences in these isolates, protein expression was followed using two-dimensional (2-D) gel electrophoresis and protein identification by peptide sequencing using micro-capillary column liquid chromatography-tandem mass spectrometry (microLC/MS/MS). The isolates selected for this analysis were from the sputum of a CF patient: strain 383 had a nonmucoid phenotype typical of isolates from the environment, and strain 2192, obtained from the same patient, had a mucoid phenotype typical of isolates from chronic CF lung infections. Strains 383 and 2192 were confirmed to be genetically identical by restriction endonuclease analysis, random amplified polymorphic DNA-PCR, and pulsed-field gel electrophoresis. Conditions of protein extraction were optimized for consistent high-resolution separation of several hundred proteins from these clinical isolates as detected by Coomassie staining of 2-D gels. Fourteen proteins were selected for analysis; this group included those whose expression was common between both strains as well as unique for each strain. The proteins were identified by microLC/MS/MS of the peptides produced by an in-gel tryptic digestion and compared to translated data from the Pseudomonas Genome Project; optimization of this technique has allowed for the comparison of proteins expressed by strains 383 and 2192.


Subject(s)
Bacterial Proteins/metabolism , Cystic Fibrosis/microbiology , Pseudomonas aeruginosa/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Chronic Disease , Electrophoresis, Gel, Two-Dimensional/methods , Gas Chromatography-Mass Spectrometry/methods , Genotype , Humans , Molecular Sequence Data , Phenotype , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/classification , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/isolation & purification , Sequence Analysis, Protein/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...