Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Science ; 347(6225): 995-8, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25722412

ABSTRACT

Ebola virus causes sporadic outbreaks of lethal hemorrhagic fever in humans, but there is no currently approved therapy. Cells take up Ebola virus by macropinocytosis, followed by trafficking through endosomal vesicles. However, few factors controlling endosomal virus movement are known. Here we find that Ebola virus entry into host cells requires the endosomal calcium channels called two-pore channels (TPCs). Disrupting TPC function by gene knockout, small interfering RNAs, or small-molecule inhibitors halted virus trafficking and prevented infection. Tetrandrine, the most potent small molecule that we tested, inhibited infection of human macrophages, the primary target of Ebola virus in vivo, and also showed therapeutic efficacy in mice. Therefore, TPC proteins play a key role in Ebola virus infection and may be effective targets for antiviral therapy.


Subject(s)
Antiviral Agents/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/therapy , Molecular Targeted Therapy , Virus Internalization/drug effects , Animals , Antiviral Agents/therapeutic use , BALB 3T3 Cells , Benzylisoquinolines/pharmacology , Benzylisoquinolines/therapeutic use , Calcium Channel Blockers/therapeutic use , Calcium Channels/genetics , Ebolavirus/drug effects , Female , Gene Knockout Techniques , HeLa Cells , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/virology , Humans , Macrophages/drug effects , Macrophages/virology , Mice , NADP/analogs & derivatives , NADP/metabolism , RNA Interference , Signal Transduction , Verapamil/pharmacology , Verapamil/therapeutic use
2.
PLoS One ; 8(4): e60579, 2013.
Article in English | MEDLINE | ID: mdl-23577127

ABSTRACT

BACKGROUND: The rapid development of effective medical countermeasures against potential biological threat agents is vital. Repurposing existing drugs that may have unanticipated activities as potential countermeasures is one way to meet this important goal, since currently approved drugs already have well-established safety and pharmacokinetic profiles in patients, as well as manufacturing and distribution networks. Therefore, approved drugs could rapidly be made available for a new indication in an emergency. METHODOLOGY/PRINCIPAL FINDINGS: A large systematic effort to determine whether existing drugs can be used against high containment bacterial and viral pathogens is described. We assembled and screened 1012 FDA-approved drugs for off-label broad-spectrum efficacy against Bacillus anthracis; Francisella tularensis; Coxiella burnetii; and Ebola, Marburg, and Lassa fever viruses using in vitro cell culture assays. We found a variety of hits against two or more of these biological threat pathogens, which were validated in secondary assays. As expected, antibiotic compounds were highly active against bacterial agents, but we did not identify any non-antibiotic compounds with broad-spectrum antibacterial activity. Lomefloxacin and erythromycin were found to be the most potent compounds in vivo protecting mice against Bacillus anthracis challenge. While multiple virus-specific inhibitors were identified, the most noteworthy antiviral compound identified was chloroquine, which disrupted entry and replication of two or more viruses in vitro and protected mice against Ebola virus challenge in vivo. CONCLUSIONS/SIGNIFICANCE: The feasibility of repurposing existing drugs to face novel threats is demonstrated and this represents the first effort to apply this approach to high containment bacteria and viruses.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antiviral Agents/pharmacology , Biological Warfare Agents , Drug Approval , Drug Evaluation, Preclinical/methods , United States Food and Drug Administration , Animals , Cell Line , Female , Humans , Male , Mice , United States
3.
J Virol ; 86(14): 7473-83, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22573858

ABSTRACT

Acid sphingomyelinase (ASMase) converts the lipid sphingomyelin (SM) to phosphocholine and ceramide and has optimum activity at acidic pH. Normally, ASMase is located in lysosomes and endosomes, but membrane damage or the interaction with some bacterial and viral pathogens can trigger its recruitment to the plasma membrane. Rhinovirus and measles viruses each require ASMase activity during early stages of infection. Both sphingomyelin and ceramide are important components of lipid rafts and are potent signaling molecules. Each plays roles in mediating macropinocytosis, which has been shown to be important for ebolavirus (EBOV) infection. Here, we investigated the role of ASMase and its substrate, SM, in EBOV infection. The work was performed at biosafety level 4 with wild-type virus with specificity and mechanistic analysis performed using virus pseudotypes and virus-like particles. We found that virus particles strongly associate with the SM-rich regions of the cell membrane and depletion of SM reduces EBOV infection. ASM-specific drugs and multiple small interfering RNAs strongly inhibit the infection by EBOV and EBOV glycoprotein pseudotyped viruses but not by the pseudotypes bearing the glycoprotein of vesicular stomatitis virus. Interestingly, the binding of virus-like particles to cells is strongly associated with surface-localized ASMase as well as SM-enriched sites. Our work suggests that ASMase activity and SM presence are necessary for efficient infection of cells by EBOV. The inhibition of this pathway may provide new avenues for drug treatment.


Subject(s)
Ebolavirus/physiology , Ebolavirus/pathogenicity , Sphingomyelin Phosphodiesterase/metabolism , Sphingomyelins/metabolism , Animals , Cell Membrane/metabolism , Cell Membrane/virology , Chlorocebus aethiops , HEK293 Cells , HeLa Cells , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Lysosomes/enzymology , Membrane Microdomains/metabolism , Membrane Microdomains/virology , Membrane Proteins/biosynthesis , Membrane Proteins/metabolism , RNA Interference , RNA, Small Interfering , Sphingomyelin Phosphodiesterase/genetics , Vero Cells , Virus Internalization
4.
Arch Virol ; 157(1): 121-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21947546

ABSTRACT

Arenaviruses and filoviruses are capable of causing hemorrhagic fever syndrome in humans. Limited therapeutic and/or prophylactic options are available for humans suffering from viral hemorrhagic fever. In this report, we demonstrate that pre-treatment of host cells with the kinase inhibitors genistein and tyrphostin AG1478 leads to inhibition of infection or transduction in cells infected with Ebola virus, Marburg virus, and Lassa virus. In all, the results demonstrate that a kinase inhibitor cocktail consisting of genistein and tyrphostin AG1478 is a broad-spectrum antiviral that may be used as a therapeutic or prophylactic against arenavirus and filovirus hemorrhagic fever.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Genistein/pharmacology , Hemorrhagic Fever, Ebola/virology , Lassa Fever/virology , Lassa virus/drug effects , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Tyrphostins/pharmacology , Cell Line , Ebolavirus/genetics , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/drug therapy , Humans , Lassa Fever/drug therapy , Lassa virus/genetics , Lassa virus/physiology
5.
J Virol ; 86(3): 1421-32, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22090132

ABSTRACT

For retroviruses such as HIV-1 and murine leukemia virus (MLV), active receptor recruitment and trafficking occur during viral entry. However, the underlying mechanisms and cellular factors involved in the process are largely uncharacterized. The viral receptor for ecotropic MLV (eMLV), a classical model for retrovirus infection mechanisms and pathogenesis, is mouse cationic amino acid transporter 1 (mCAT-1). Growth factor receptor-bound protein 2 (GRB2) is an adaptor protein that has been shown to couple cell surface receptors, such as epidermal growth factor receptor (EGFR) and hepatocyte growth factor receptor, to intracellular signaling events. Here we examined if GRB2 could also play a role in controlling infection by retroviruses by affecting receptor function. The GRB2 RNA interference (RNAi)-mediated suppression of endogenous GRB2 resulted in a consistent and significant reduction of virus binding and membrane fusion. The binding between eMLV and cells promoted increased GRB2-mCAT-1 interactions, as detected by immunoprecipitation. Consistently, the increased colocalization of GRB2 and mCAT-1 signals was detected by confocal microscopy. This association was time dependent and paralleled the kinetics of cell-virus membrane fusion. Interestingly, unlike the canonical binding pattern seen for GRB2 and growth factor receptors, GRB2-mCAT-1 binding does not depend on the GRB2-SH2 domain-mediated recognition of tyrosine phosphorylation on the receptor. The inhibition of endogenous GRB2 led to a reduction in surface levels of mCAT-1, which was detected by immunoprecipitation and by a direct binding assay using a recombinant MLV envelope protein receptor binding domain (RBD). Consistent with this observation, the expression of a dominant negative GRB2 mutant (R86K) resulted in the sequestration of mCAT-1 from the cell surface into intracellular vesicles. Taken together, these findings suggest a novel role for GRB2 in ecotropic MLV entry and infection by facilitating mCAT-1 trafficking.


Subject(s)
Calcium Channels/metabolism , GRB2 Adaptor Protein/metabolism , Leukemia Virus, Murine/physiology , Membrane Fusion , TRPV Cation Channels/metabolism , Animals , Cell Line , Flow Cytometry , GRB2 Adaptor Protein/genetics , Humans , Leukemia Virus, Murine/metabolism , Mice , Phosphorylation , Protein Binding , RNA Interference , RNA, Small Interfering
6.
Gastroenterology ; 141(2): 674-85, 685.e1-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21683701

ABSTRACT

BACKGROUND & AIMS: The hepatitis C virus (HCV) serine protease NS3/4A can cleave mitochondria-associated antiviral signaling protein (MAVS) and block retinoic acid-inducible gene I-mediated interferon (IFN) responses. Although this mechanism is thought to have an important role in HCV-mediated innate immunosuppression, its significance in viral persistence is not clear. METHODS: We generated transgenic mice that express the HCV NS3/4A proteins specifically in the liver and challenged the animals with a recombinant vesicular stomatitis virus, a synthetic HCV genome, IFN alfa, or IFN beta. We evaluated the effects of HCV serine protease on the innate immune responses and their interactions. RESULTS: Expression of HCV NS3/4A resulted in cleavage of intrahepatic MAVS; challenge of transgenic mice with vesicular stomatitis virus or a synthetic HCV genome induced strong, type I IFN-mediated responses that were not significantly lower than those of control mice. Different challenge agents induced production of different ratios of IFN alfa and beta, resulting in different autophagic responses and vesicular trafficking patterns of endoplasmic reticulum- and mitochondria-associated viral proteins. IFN beta promoted degradation of the viral proteins by the autolysosome. Variant isoforms of MAVS were associated with distinct, type I IFN-mediated autophagic responses; these responses have a role in trafficking of viral components to endosomal compartments that contain Toll-like receptor-3. CONCLUSIONS: IFN beta mediates a distinct autophagic mechanism of antiviral host defense. MAVS has an important role in type I IFN-induced autophagic trafficking of viral proteins.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Hepacivirus/immunology , Interferon-alpha/immunology , Interferon-beta/immunology , Liver/metabolism , RNA, Viral/immunology , Vesiculovirus/immunology , Viral Nonstructural Proteins/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/immunology , Animals , Autophagy/immunology , Biological Transport , DEAD Box Protein 58 , DEAD-box RNA Helicases/metabolism , Endoplasmic Reticulum/immunology , Gene Expression , Immunity, Innate , Interferon Regulatory Factor-1/metabolism , Interferon Type I/immunology , Interferon-alpha/deficiency , Interferon-alpha/metabolism , Interferon-beta/deficiency , Interferon-beta/metabolism , Liver/immunology , Mice , Mice, Knockout , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Recombinant Proteins , Signal Transduction
7.
J Virol ; 85(1): 334-47, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21047970

ABSTRACT

Axl, a plasma membrane-associated Tyro3/Axl/Mer (TAM) family member, is necessary for optimal Zaire ebolavirus (ZEBOV) glycoprotein (GP)-dependent entry into some permissive cells but not others. To date, the role of Axl in virion entry is unknown. The focus of this study was to characterize entry pathways that are used for ZEBOV uptake in cells that require Axl for optimal transduction and to define the role of Axl in this process. Through the use of biochemical inhibitors, interfering RNA (RNAi), and dominant negative constructs, we demonstrate that ZEBOV-GP-dependent entry into these cells occurs through multiple uptake pathways, including both clathrin-dependent and caveola/lipid raft-mediated endocytosis. Other dynamin-dependent and -independent pathways such as macropinocytosis that mediate high-molecular-weight dextran uptake also stimulated ZEBOV-GP entry into these cells, and inhibitors that are known to block macropinocytosis inhibited both dextran uptake and ZEBOV infection. These findings provided strong evidence for the importance of this pathway in filovirus entry. Reduction of Axl expression by RNAi treatment resulted in decreased ZEBOV entry via macropinocytosis but had no effect on the clathrin-dependent or caveola/lipid raft-mediated endocytic mechanisms. Our findings demonstrate for the first time that Axl enhances macropinocytosis, thereby increasing productive ZEBOV entry.


Subject(s)
Ebolavirus/pathogenicity , Pinocytosis/physiology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Cell Line , Chlorocebus aethiops , Clathrin/metabolism , Ebolavirus/genetics , Ebolavirus/metabolism , Endocytosis , Humans , Kidney/cytology , Kidney/virology , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Vero Cells , Viral Envelope Proteins/metabolism , Virus Internalization , Axl Receptor Tyrosine Kinase
8.
Infect Immun ; 78(11): 4532-41, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20823205

ABSTRACT

The bacterial pathogen Listeria monocytogenes causes food-borne illnesses culminating in gastroenteritis, meningitis, or abortion. Listeria induces its internalization into some mammalian cells through binding of the bacterial surface protein InlB to the host receptor tyrosine kinase Met. Interaction of InlB with the Met receptor elicits host downstream signaling pathways that promote F-actin cytoskeletal changes responsible for pathogen engulfment. Here we show that the mammalian signaling protein ARAP2 plays a critical role in cytoskeletal remodeling and internalization of Listeria. Depletion of ARAP2 through RNA interference (RNAi) caused a marked inhibition of InlB-mediated F-actin rearrangements and bacterial entry. ARAP2 contains multiple functional domains, including a GTPase-activating protein (GAP) domain that antagonizes the GTPase Arf6 and a domain capable of binding the GTPase RhoA. Genetic data indicated roles for both the Arf GAP and RhoA binding domains in Listeria entry. Experiments involving Arf6 RNAi or a constitutively activated allele of Arf6 demonstrated that one of the ways in which ARAP2 promotes bacterial uptake is by restraining the activity of Arf6. Conversely, Rho activity was dispensable for Listeria internalization, suggesting that the RhoA binding domain in ARAP2 acts by engaging a host ligand other than Rho proteins. Collectively, our findings indicate that ARAP2 promotes InlB-mediated entry of Listeria, in part, by antagonizing the host GTPase Arf6.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , GTPase-Activating Proteins/metabolism , Listeria monocytogenes/pathogenicity , Membrane Proteins/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Bacterial Proteins/genetics , Carrier Proteins/genetics , Epithelial Cells/microbiology , GTPase-Activating Proteins/genetics , HeLa Cells , Host-Pathogen Interactions , Humans , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Membrane Proteins/genetics , Phosphatidylinositol 3-Kinases/metabolism
9.
PLoS Pathog ; 6(9): e1001110, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20862315

ABSTRACT

Zaire ebolavirus (ZEBOV), a highly pathogenic zoonotic virus, poses serious public health, ecological and potential bioterrorism threats. Currently no specific therapy or vaccine is available. Virus entry is an attractive target for therapeutic intervention. However, current knowledge of the ZEBOV entry mechanism is limited. While it is known that ZEBOV enters cells through endocytosis, which of the cellular endocytic mechanisms used remains unclear. Previous studies have produced differing outcomes, indicating potential involvement of multiple routes but many of these studies were performed using noninfectious surrogate systems such as pseudotyped retroviral particles, which may not accurately recapitulate the entry characteristics of the morphologically distinct wild type virus. Here we used replication-competent infectious ZEBOV as well as morphologically similar virus-like particles in specific infection and entry assays to demonstrate that in HEK293T and Vero cells internalization of ZEBOV is independent of clathrin, caveolae, and dynamin. Instead the uptake mechanism has features of macropinocytosis. The binding of virus to cells appears to directly stimulate fluid phase uptake as well as localized actin polymerization. Inhibition of key regulators of macropinocytosis including Pak1 and CtBP/BARS as well as treatment with the drug EIPA, which affects macropinosome formation, resulted in significant reduction in ZEBOV entry and infection. It is also shown that following internalization, the virus enters the endolysosomal pathway and is trafficked through early and late endosomes, but the exact site of membrane fusion and nucleocapsid penetration in the cytoplasm remains unclear. This study identifies the route for ZEBOV entry and identifies the key cellular factors required for the uptake of this filamentous virus. The findings greatly expand our understanding of the ZEBOV entry mechanism that can be applied to development of new therapeutics as well as provide potential insight into the trafficking and entry mechanism of other filoviruses.


Subject(s)
Ebolavirus/physiology , Endosomes/metabolism , Endosomes/virology , Hemorrhagic Fever, Ebola/prevention & control , Pinocytosis/physiology , Virus Internalization/drug effects , Alcohol Oxidoreductases/genetics , Alcohol Oxidoreductases/metabolism , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Blotting, Western , Caveolae/metabolism , Caveolae/virology , Cells, Cultured , Chlorocebus aethiops , Clathrin/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endocytosis/physiology , Hemorrhagic Fever, Ebola/metabolism , Hemorrhagic Fever, Ebola/virology , Humans , Kidney/cytology , Kidney/drug effects , Kidney/metabolism , Membrane Fusion , Membrane Microdomains/physiology , Phosphorylation , Pinocytosis/drug effects , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Vero Cells , Virus Replication/drug effects , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
10.
Int J Cancer ; 126(4): 864-75, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-19697327

ABSTRACT

Expression of gastrin and cholecystokinin 2 (CCK(2)) receptor splice variants (CCK(2)R and CCK(2i4sv)R) are upregulated in human colonic adenomas where they are thought to contribute to tumor growth and progression. To determine the effects of ectopic CCK(2) receptor variant expression on colonic epithelial cell growth in vitro and in vivo, we employed the non-tumorigenic colonic epithelial cell line, NCM356. Receptor expression was induced using a retroviral expression vector containing cDNAs for either CCK(2i4sv)R or CCK(2)R. RT-PCR and intracellular Ca(2+) ([Ca(2+)](i)) imaging of RIE/CCK(2)R cells treated with conditioned media (CM) from NCM356 revealed that NCM356 cells express gastrin mRNA and secrete endogenous, biologically active peptide. NCM356 cells expressing either CCK(2)R or CCK(2i4sv)R (71 and 81 fmol/mg, respectively) grew faster in vitro, and exhibited an increase in basal levels of phosphorylated ERK (pERK), compared with vector. CCK(2) receptor selective antagonist, YM022, partially inhibited the growth of both receptor-expressing NCM356 cells, but not the control cells. Inhibitors of mitogen activated protein kinase pathway (MEK/ERK) or protein kinase C (PKC) isozymes partially inhibited the elevated levels of basal pERK and in vitro growth of receptor-expressing cells. Vector-NCM356 cells did not form tumors in nude mice, whereas, either CCK(2) receptor-expressing cells formed large tumors. Autocrine activation CCK(2) receptor variants are sufficient to increase in vitro growth and tumorigenicity of non-transformed NCM356 colon epithelial cells through a pathway involving PKC and the MEK/ERK axis. These findings support the hypothesis that expression of gastrin and its receptors in human colonic adenomas contributes to tumor growth and progression.


Subject(s)
Colon/physiology , Colorectal Neoplasms/pathology , Intestinal Mucosa/physiology , Receptor, Cholecystokinin B/genetics , Adenoma/pathology , Animals , Calcium/metabolism , Carcinoma/genetics , Cell Culture Techniques/methods , Cell Division/genetics , Colon/metabolism , Colon/pathology , Colorectal Neoplasms/genetics , DNA Primers , Disease Progression , Gastrins/genetics , Gastrins/metabolism , Genetic Variation , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mutation , Neoplasm Staging , Reverse Transcriptase Polymerase Chain Reaction
11.
Drug Dev Res ; 70(4): 255-265, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-20930947

ABSTRACT

While much progress has been made in developing drugs against a few prominent viruses such as HIV, few examples exist for emerging infectious agents. In some cases broad spectrum anti-viral drugs, such as ribavirin, are effective, but for some groups of viruses, these show little efficacy in animal models. Traditional methods focus on screening small molecule libraries to identify drugs that target virus factors, with the intention that side-effects to the host can be minimized. However, this greatly limits potential drug targets and virus genes can rapidly mutate to avoid drug action. Recent advances in siRNA gene targeting technologies have provided a powerful tool to specifically target and suppress the expression of cell genes. Since viruses are completely dependent upon host cell proteins for propagation, siRNA screening promises to reveal novel cell proteins and signaling pathways that may be viable targets for drug therapy regimens. Here we used an siRNA screening approach to identify gene products that play critical roles in Ebola virus infection. By gene cluster analysis, proteins in phosphatidylinositol-3-kinase and calcium/calmodulin kinase related networks were identified as important for Zaire Ebola virus infection and prioritized for further evaluation. Key roles of each were confirmed by testing available drugs specific for members of each pathway. Interestingly, both sets of proteins are also important in cancer and subject to intense investigation. Thus development of new drugs against these cancer targets may also prove useful in combating Ebola virus.

12.
PLoS Pathog ; 4(8): e1000141, 2008 Aug 29.
Article in English | MEDLINE | ID: mdl-18769720

ABSTRACT

The phosphoinositide-3 kinase (PI3K) pathway regulates diverse cellular activities related to cell growth, migration, survival, and vesicular trafficking. It is known that Ebola virus requires endocytosis to establish an infection. However, the cellular signals that mediate this uptake were unknown for Ebola virus as well as many other viruses. Here, the involvement of PI3K in Ebola virus entry was studied. A novel and critical role of the PI3K signaling pathway was demonstrated in cell entry of Zaire Ebola virus (ZEBOV). Inhibitors of PI3K and Akt significantly reduced infection by ZEBOV at an early step during the replication cycle. Furthermore, phosphorylation of Akt-1 was induced shortly after exposure of cells to radiation-inactivated ZEBOV, indicating that the virus actively induces the PI3K pathway and that replication was not required for this induction. Subsequent use of pseudotyped Ebola virus and/or Ebola virus-like particles, in a novel virus entry assay, provided evidence that activity of PI3K/Akt is required at the virus entry step. Class 1A PI3Ks appear to play a predominant role in regulating ZEBOV entry, and Rac1 is a key downstream effector in this regulatory cascade. Confocal imaging of fluorescently labeled ZEBOV indicated that inhibition of PI3K, Akt, or Rac1 disrupted normal uptake of virus particles into cells and resulted in aberrant accumulation of virus into a cytosolic compartment that was non-permissive for membrane fusion. We conclude that PI3K-mediated signaling plays an important role in regulating vesicular trafficking of ZEBOV necessary for cell entry. Disruption of this signaling leads to inappropriate trafficking within the cell and a block in steps leading to membrane fusion. These findings extend our current understanding of Ebola virus entry mechanism and may help in devising useful new strategies for treatment of Ebola virus infection.


Subject(s)
Ebolavirus/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Virus Internalization , Virus Replication/physiology , Animals , Chlorocebus aethiops , Endocytosis , Hemorrhagic Fever, Ebola/enzymology , Hemorrhagic Fever, Ebola/therapy , Humans , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Vero Cells , rac1 GTP-Binding Protein/antagonists & inhibitors , rac1 GTP-Binding Protein/metabolism
13.
Virology ; 377(1): 133-42, 2008 Jul 20.
Article in English | MEDLINE | ID: mdl-18485440

ABSTRACT

Virus-host interactions essential for alphavirus pathogenesis are poorly understood. To address this shortcoming, we coupled retrovirus insertional mutagenesis and a cell survival selection strategy to generate clonal cell lines broadly resistant to Sindbis virus (SINV) and other alphaviruses. Resistant cells had significantly impaired SINV production relative to wild-type (WT) cells, although virus binding and fusion events were similar in both sets of cells. Analysis of the retroviral integration sites identified the neurofibromin 1 (NF1) gene as disrupted in alphavirus-resistant cell lines. Subsequent analysis indicated that expression of NF1 was significantly reduced in alphavirus-resistant cells. Importantly, independent down-regulation of NF1 expression in WT HEK 293 cells decreased virus production and increased cell viability during SINV infection, relative to infected WT cells. Additionally, we observed hyperactive RAS signalling in the resistant HEK 293 cells, which was anticipated because NF1 is a negative regulator of RAS. Expression of constitutively active RAS (HRAS-G12V) in a WT HEK 293 cell line resulted in a marked delay in virus production, compared with infected cells transfected with parental plasmid or dominant-negative RAS (HRAS-S17N). This work highlights novel host cell determinants required for alphavirus pathogenesis and suggests that RAS signalling may play an important role in neuronal susceptibility to SINV infection.


Subject(s)
Neurofibromin 1/deficiency , Sindbis Virus/physiology , Sindbis Virus/pathogenicity , Alphavirus Infections/etiology , Alphavirus Infections/genetics , Alphavirus Infections/virology , Base Sequence , Cell Line , DNA Primers/genetics , DNA, Viral/genetics , Down-Regulation , Genes, ras , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Mutagenesis, Insertional , Neurofibromin 1/genetics , Neurons/virology , RNA, Small Interfering/genetics , Signal Transduction , Virus Integration , Virus Replication
14.
Virology ; 369(1): 78-91, 2007 Dec 05.
Article in English | MEDLINE | ID: mdl-17707875

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is a New World alphavirus that can cause fatal encephalitis in humans. It remains a naturally emerging disease as well as a highly developed biological weapon. VEEV is transmitted to humans in nature by mosquito vectors. Little is known about VEEV entry, especially in mosquito cells. Here, a novel luciferase-based virus entry assay is used to show that the entry of VEEV into mosquito cells requires acidification. Furthermore, mosquito homologs of key human proteins (Rab5 and Rab7) involved in endocytosis were isolated and characterized. Rab5 is found on early endosomes and Rab7 on late endosomes and both are important for VEEV entry in mammalian cells. Each was shown to have analogous function in mosquito cells to that seen in mammalian cells. The wild-type, dominant negative and constitutively active mutants were then used to demonstrate that VEEV requires passage through early and late endosomes before infection can take place. This work indicates that the infection mechanism in mosquitoes and mammals is through a common and ancient evolutionarily conserved pathway.


Subject(s)
Culicidae/virology , Encephalitis Virus, Venezuelan Equine/growth & development , Endosomes/virology , Virus Internalization , rab GTP-Binding Proteins/physiology , rab5 GTP-Binding Proteins/physiology , Amino Acid Sequence , Animals , Cell Line , Culicidae/cytology , Endosomes/chemistry , Genes, Reporter , Humans , Luciferases/biosynthesis , Luciferases/genetics , Microscopy, Confocal , Molecular Sequence Data , Sequence Alignment , rab GTP-Binding Proteins/analysis , rab GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/analysis , rab5 GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins
15.
J Virol ; 81(14): 7786-800, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17494077

ABSTRACT

Respiratory syncytial virus (RSV) is a common cause of respiratory tract infections in infants and the elderly. Like many other pH-independent enveloped viruses, RSV is thought to enter at the cell surface, independently of common endocytic pathways. We have used a targeted small interfering RNA (siRNA) library to identify key cellular genes involved in cytoskeletal dynamics and endosome trafficking that are important for RSV infection. Surprisingly, RSV infection was potently inhibited by siRNAs targeting genes associated with clathrin-mediated endocytosis, including clathrin light chain. The important role of clathrin-mediated endocytosis was confirmed by the expression of well-characterized dominant-negative mutants of genes in this pathway and by using the clathrin endocytosis inhibitor chlorpromazine. We conclude that, while RSV may be competent to enter at the cell surface, clathrin function and endocytosis are a necessary and important part of a productive RSV infection, even though infection is strictly independent of pH. These findings raise the possibility that other pH-independent viruses may share a similar dependence on endocytosis for infection and provide a new potential avenue for treatment of infection.


Subject(s)
Clathrin/physiology , Endocytosis , Endosomes , Gene Expression Profiling , RNA, Small Interfering/genetics , Respiratory Syncytial Virus Infections/genetics , Cell Line , Humans , Hydrogen-Ion Concentration , Respiratory Syncytial Virus Infections/physiopathology
16.
Nano Lett ; 6(11): 2414-21, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17090066

ABSTRACT

One of the most important steps in a productive viral infection is when the virus fuses to a cell membrane and delivers its genome into the cell cytosol. This dynamic event is mediated by interactions between specific virus envelope proteins with their cell-bound receptors. This process is exemplified by Moloney murine leukemia virus (Mo-MLV) where envelope protein interaction with its receptor, mCAT-1, leads to virus-cell membrane fusion and infection of cells. Here, fluorescent nanoparticles (NPs) were coated with Mo-MLV derived membranes (Mo-NPs) by extrusion. Electron microscopy and biochemical analysis showed tight association of the virus membranes and NPs. The coated NPs mimic native virus by binding and entering only cells expressing the virus receptor. Confocal microscopy revealed that the coated NPs were taken up into endocytic compartments containing receptor and were also seen associated with caveolin, a marker of caveolae. To demonstrate that the Mo-NPs could escape endosomes and deliver a protein cargo into the cell cytosol, beta-lactamase (betalac) was covalently coupled to the Mo-NP cores and incubated with cells. betalac activity was only detected in the cytosol of mCAT-1-expressing cells. This is the first time that virus proteins have been used to specifically target NPs to receptor-bearing cells as well as penetration into the cell cytosol. Extrusion provides a rapid, detergent-free method to couple virus membranes to NPs and should be readily applicable for many other virus and NP types.


Subject(s)
Cationic Amino Acid Transporter 1/chemistry , Moloney murine leukemia virus/chemistry , Nanoparticles/chemistry , Animals , Cationic Amino Acid Transporter 1/metabolism , Cell Line , Cell Membrane/chemistry , Cell Membrane/metabolism , Cytosol/chemistry , Cytosol/metabolism , Humans , Mice , Moloney murine leukemia virus/metabolism , Particle Size , Surface Properties
17.
Am J Trop Med Hyg ; 75(4): 702-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17038698

ABSTRACT

Virus envelope proteins are the primary targets of neutralizing antibody responses. The epitopes recognized differ sufficiently between virus subtypes and species to distinguish viruses and provide an important basis for disease diagnosis. Venezuelan equine encephalitis virus (VEEV) causes acute febrile illness in humans and has high mortality in equines. The most specific detection methods for serum antibodies use live virus in neutralization assays or in blocking enzyme linked immunosorbent assays. However, work with Venezuelan equine encephalitis virus requires biosafety level 3 containment and select agent security in the United States. We report two new assays for detection of Venezuelan equine encephalitis virus neutralizing antibody responses, based on virus pseudotypes. The first provides detection by marker gene expression after 20 hours and is particularly suited for high-throughput screening; the second uses a new, rapid virus entry assay to give readouts within 1 hour. Both assays are safe, sensitive, and in general recapitulate neutralizing antibody titers obtained by conventional plaque reduction assays. Each is suitable as a rapid primary screen for detection of neutralizing antibodies against Venezuelan equine encephalitis virus.


Subject(s)
Antibodies, Viral/blood , Encephalitis Virus, Venezuelan Equine/immunology , Encephalomyelitis, Venezuelan Equine/diagnosis , Horse Diseases/diagnosis , Analysis of Variance , Animals , Cell Line , Cricetinae , Cross Reactions , Encephalitis Virus, Venezuelan Equine/classification , Encephalitis Virus, Venezuelan Equine/genetics , Encephalomyelitis, Venezuelan Equine/immunology , Epitopes/immunology , Genes, Reporter , Horse Diseases/immunology , Horse Diseases/virology , Horses , Humans , Leukemia Virus, Murine/genetics , Luciferases/analysis , Luciferases/genetics , Luciferases/immunology , Neutralization Tests , Regression Analysis , Vesicular stomatitis Indiana virus/genetics , Viral Envelope Proteins/immunology , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology , Viral Vaccines/immunology
18.
J Virol ; 80(24): 12160-70, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17005642

ABSTRACT

Like many enveloped viruses, human respiratory syncytial virus (RSV) assembles at and buds from lipid rafts. Translocation of the envelope proteins to these membrane subdomains is essential for production of infectious virus, but the targeting mechanism is poorly understood and it is not known if other virus proteins are required. Here we demonstrate that F protein of RSV intrinsically targets to lipid rafts without a requirement for any other virus protein, including the SH and G envelope proteins. Recombinant virus deficient in SH and G but retaining F protein expression was used to demonstrate that F protein still localized in rafts in both A549 and HEp-2 cells. Expression of a recombinant F gene by use of plasmid vectors demonstrated that F contains its own targeting domain and localized to rafts in the absence of other virus proteins. The domain responsible for translocation was then mapped. Unlike most other virus envelope proteins, F is unusual since the target signal is not contained within the cytoplasmic domain nor did it involve fatty acid modified residues. Furthermore, exchange of the transmembrane domain with that of the vesicular stomatitis virus G protein, a nonraft protein, did not alter F protein raft localization. Taken together, these data suggest that domains present in the extracellular portion of the protein are responsible for lipid raft targeting of the RSV F protein.


Subject(s)
Membrane Microdomains/metabolism , Respiratory Syncytial Viruses/genetics , Viral Fusion Proteins/metabolism , Blotting, Western , Cell Line , DNA Primers , Flow Cytometry , Humans , Microscopy, Fluorescence , Mutagenesis , Protein Structure, Tertiary/genetics , Protein Transport/physiology , Viral Fusion Proteins/genetics
19.
J Mol Model ; 12(6): 921-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16607494

ABSTRACT

A new method for predicting interacting residues in protein complexes, InterProSurf, was applied to the E1 envelope protein of Venezuelan equine encephalitis (VEEV). Monomeric and trimeric models of VEEV-E1 were constructed with our MPACK program, using the crystal structure of the E1 protein of Semliki forest virus as a template. An alignment of the E1 sequences from representative alphavirus sequences was used to determine physical chemical property motifs (likely functional areas) with our PCPMer program. Information on residue variability, propensity to be in protein interfaces, and surface exposure on the model was combined to predict surface clusters likely to interact with other viral or cellular proteins. Mutagenesis of these clusters indicated that the predictions accurately detected areas crucial for virus infection. In addition to the fusion peptide area in domain 2, at least two other surface areas play an important role in virus infection. We propose that these may be sites of interaction between the E1-E1 and E1-E2 subdomains of the envelope proteins that are required to assemble the functional unit. The InterProSurf method is, thus, an important new tool for predicting viral protein interactions. These results can aid in the design of new vaccines against alphaviruses and other viruses.


Subject(s)
Encephalitis Virus, Venezuelan Equine/chemistry , Viral Envelope Proteins/chemistry , Amino Acids , Animals , Binding Sites , Dimerization , Encephalitis Virus, Venezuelan Equine/pathogenicity , Horses , Multiprotein Complexes/chemistry
20.
J Virol Methods ; 135(2): 143-50, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16584792

ABSTRACT

Entry is the first and essential step in virus replication and is a target for therapeutic intervention. However, current knowledge on entry mechanism for the majority of viruses is poor, partly due to lack of a simple, sensitive and accurate entry assay that can be applied to diverse viruses. To overcome this obstacle, a novel contents-mixing-based virus entry assay is described that can be broadly applied to many enveloped viruses. By fusing firefly luciferase to the HIV Nef protein, luciferase was directly packaged into HIV particles pseudotyped with envelope proteins of diverse viruses including HIV, rabies and others. Upon cell entry, the luciferase-fusion protein was released into the cell cytoplasm, reacted with its substrates and was detected by light emission. The assay was validated by demonstrating its versatility in measuring virus entry. Entry was detected much more rapidly (in real-time) with higher sensitivity (a multiplicity of infection <0.1 gives a robust signal) and lower background (signal/noise ration >1000) than other comparable assays. In addition to its utility in studying virus entry mechanisms, the assay will aid in screening potential entry/fusion inhibitors and in diagnosis of virus infections.


Subject(s)
HIV/physiology , Rabies virus/physiology , Cells, Cultured , Endosomes/metabolism , HIV Fusion Inhibitors/pharmacology , Humans , Hydrogen-Ion Concentration , Membrane Fusion , Receptors, Virus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...