Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
mBio ; 11(1)2020 02 25.
Article in English | MEDLINE | ID: mdl-32098811

ABSTRACT

Lassa virus (LASV) is endemic in Western Africa and is estimated to infect hundreds of thousands of individuals annually. A considerable number of these infections result in Lassa fever (LF), which is associated with significant morbidity and a case-fatality rate as high as 69% among hospitalized confirmed patients. U.S. Food and Drug Administration-approved LF vaccines are not available. Current antiviral treatment is limited to off-label use of a nucleoside analogue, ribavirin, that is only partially effective and associated with significant side effects. We generated and characterized a recombinant LASV expressing a codon-deoptimized (CD) glycoprotein precursor gene (GPC), rLASV-GPC/CD. Comparison of growth kinetics and peak titers showed that rLASV-GPC/CD is slightly attenuated in cell culture compared to wild-type (WT) recombinant LASV (rLASV-WT). However, rLASV-GPC/CD is highly attenuated in strain 13 and Hartley guinea pigs, as reflected by the absence of detectable clinical signs in animals inoculated with rLASV-GPC/CD. Importantly, a single subcutaneous dose of rLASV-GPC/CD provides complete protection against an otherwise lethal exposure to LASV. Our results demonstrate the feasibility of implementing a CD approach for developing a safe and effective LASV live-attenuated vaccine candidate. Moreover, rLASV-GPC/CD might provide investigators with a tool to safely study LASV outside maximum (biosafety level 4) containment, which could accelerate the elucidation of basic aspects of the molecular and cell biology of LASV and the development of novel LASV medical countermeasures.IMPORTANCE Lassa virus (LASV) infects several hundred thousand people in Western Africa, resulting in many lethal Lassa fever (LF) cases. Licensed LF vaccines are not available, and anti-LF therapy is limited to off-label use of the nucleoside analog ribavirin with uncertain efficacy. We describe the generation of a novel live-attenuated LASV vaccine candidate. This vaccine candidate is based on mutating wild-type (WT) LASV in a key region of the viral genome, the glycoprotein precursor (GPC) gene. These mutations do not change the encoded GPC but interfere with its production in host cells. This mutated LASV (rLASV-GPC/CD) behaves like WT LASV (rLASV-WT) in cell culture, but in contrast to rLASV-WT, does not cause disease in inoculated guinea pigs. Guinea pigs immunized with rLASV-GPC/CD were protected against an otherwise lethal exposure to WT LASV. Our results support the testing of this candidate vaccine in nonhuman primate models ofLF.


Subject(s)
Lassa Fever/prevention & control , Lassa virus/genetics , Lassa virus/immunology , Vaccines, Attenuated/immunology , Viral Vaccines/immunology , A549 Cells , Africa, Western , Amino Acid Sequence , Animals , Arenaviridae , Arenavirus , Bunyaviridae , Chlorocebus aethiops , Codon , Disease Models, Animal , Female , Genes, Viral/genetics , Genome, Viral , Glycoproteins/genetics , Guinea Pigs , Humans , Lassa Fever/immunology , Lassa Fever/virology , Male , Ribavirin , Vaccines, Attenuated/genetics , Vero Cells
2.
Antiviral Res ; 173: 104667, 2020 01.
Article in English | MEDLINE | ID: mdl-31786250

ABSTRACT

The mammarenavirus Lassa (LASV) is highly prevalent in West Africa where it infects several hundred thousand individuals annually resulting in a high number of Lassa fever (LF) cases, a febrile disease associated with high morbidity and significant mortality. Mounting evidence indicates that the worldwide-distributed prototypic mammarenavirus lymphocytic choriomeningitis virus (LCMV) is a neglected human pathogen of clinical significance. There are not Food and Drug Administration (FDA) licensed vaccines and current anti-mammarenavirus therapy is limited to an off-label use of ribavirin that is only partially effective and can cause significant side effects. Therefore, there is an unmet need for novel antiviral drugs to combat LASV. This task would be facilitated by the implementation of high throughput screens (HTS) to identify inhibitors of the activity of the virus ribonucleoprotein (vRNP) responsible for directing virus RNA genome replication and gene transcription. The use of live LASV for this purpose is jeopardized by the requirement of biosafety level 4 (BSL4) containment. We have developed a virus-free cell platform, where expression levels of reporter genes serve as accurate surrogates of vRNP activity, to develop cell-based assays compatible with HTS to identify inhibitors of LASV and LCMV mammarenavirus vRNP activities.


Subject(s)
Antiviral Agents/pharmacology , Drug Evaluation, Preclinical/methods , Lassa virus/drug effects , Ribonucleoproteins/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Animals , Chlorocebus aethiops , Dose-Response Relationship, Drug , Gene Expression , Genetic Engineering , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , RNA Interference , Reproducibility of Results , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Small Molecule Libraries , Vero Cells
3.
Virology ; 501: 35-46, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27855284

ABSTRACT

Several arenaviruses, chiefly Lassa (LASV) in West Africa, cause hemorrhagic fever (HF) disease in humans and pose important public health problems in their endemic regions. To date, there are no FDA-approved arenavirus vaccines and current anti-arenaviral therapy is limited to the use of ribavirin that has very limited efficacy. In this work we document that a recombinant prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) with a codon deoptimized (CD) surface glycoprotein (GP), rLCMV/CD, exhibited wild type (WT)-like growth properties in cultured cells despite barely detectable GP expression levels in rLCMV/CD-infected cells. Importantly, rLCMV/CD was highly attenuated in vivo but able to induce complete protection against a subsequent lethal challenge with rLCMV/WT. Our findings support the feasibility of implementing an arenavirus GP CD-based approach for the development of safe and effective live-attenuated vaccines (LAVs) to combat diseases caused by human pathogenic arenaviruses.


Subject(s)
Arenaviridae Infections/virology , Arenavirus/genetics , Codon/genetics , Glycoproteins/genetics , Vaccines, Attenuated/genetics , Viral Proteins/genetics , Viral Vaccines/genetics , Amino Acid Sequence , Animals , Arenaviridae Infections/immunology , Arenavirus/immunology , Codon/immunology , Glycoproteins/administration & dosage , Glycoproteins/immunology , Humans , Mice , Molecular Sequence Data , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Viral Proteins/administration & dosage , Viral Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
4.
Nat Commun ; 7: 11544, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27161536

ABSTRACT

Lassa fever is a severe multisystem disease that often has haemorrhagic manifestations. The epitopes of the Lassa virus (LASV) surface glycoproteins recognized by naturally infected human hosts have not been identified or characterized. Here we have cloned 113 human monoclonal antibodies (mAbs) specific for LASV glycoproteins from memory B cells of Lassa fever survivors from West Africa. One-half bind the GP2 fusion subunit, one-fourth recognize the GP1 receptor-binding subunit and the remaining fourth are specific for the assembled glycoprotein complex, requiring both GP1 and GP2 subunits for recognition. Notably, of the 16 mAbs that neutralize LASV, 13 require the assembled glycoprotein complex for binding, while the remaining 3 require GP1 only. Compared with non-neutralizing mAbs, neutralizing mAbs have higher binding affinities and greater divergence from germline progenitors. Some mAbs potently neutralize all four LASV lineages. These insights from LASV human mAb characterization will guide strategies for immunotherapeutic development and vaccine design.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Lassa virus/immunology , Antibody Specificity , Antigens, Viral/chemistry , Antigens, Viral/genetics , Antigens, Viral/immunology , Arenavirus/immunology , Cross Reactions , Epitope Mapping , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Humans , Lassa Fever/immunology , Lassa Fever/prevention & control , Lassa virus/genetics , Models, Molecular , Mutagenesis, Site-Directed , Sequence Deletion , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
5.
Methods Mol Biol ; 1403: 313-51, 2016.
Article in English | MEDLINE | ID: mdl-27076139

ABSTRACT

Several arenavirus cause hemorrhagic fever disease in humans and pose a significant public health problem in their endemic regions. To date, no licensed vaccines are available to combat human arenavirus infections, and anti-arenaviral drug therapy is limited to an off-label use of ribavirin that is only partially effective. The development of arenavirus reverse genetics approaches provides investigators with a novel and powerful approach for the investigation of the arenavirus molecular and cell biology. The use of cell-based minigenome systems has allowed examining the cis- and trans-acting factors involved in arenavirus replication and transcription and the identification of novel anti-arenaviral drug targets without requiring the use of live forms of arenaviruses. Likewise, it is now feasible to rescue infectious arenaviruses entirely from cloned cDNAs containing predetermined mutations in their genomes to investigate virus-host interactions and mechanisms of pathogenesis, as well as to facilitate screens to identify anti-arenaviral drugs and development of novel live-attenuated arenavirus vaccines. Recently, reverse genetics have also allowed the generation of tri-segmented arenaviruses expressing foreign genes, facilitating virus detection and opening the possibility of implementing live-attenuated arenavirus-based vaccine vector approaches. Likewise, the development of single-cycle infectious, reporter-expressing, arenaviruses has provided a new experimental method to study some aspects of the biology of highly pathogenic arenaviruses without the requirement of high-security biocontainment required to study HF-causing arenaviruses. In this chapter we summarize the current knowledge on arenavirus reverse genetics and the implementation of plasmid-based reverse genetics techniques for the development of arenavirus vaccines and vaccine vectors.


Subject(s)
Arenaviridae Infections/prevention & control , Arenavirus/immunology , Reverse Genetics/methods , Viral Vaccines/genetics , Viral Vaccines/immunology , Animals , Arenaviridae Infections/immunology , Humans
6.
J Virol ; 89(14): 7373-84, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25972555

ABSTRACT

UNLABELLED: Several members of the Arenaviridae family cause hemorrhagic fever disease in humans and pose serious public health problems in their geographic regions of endemicity as well as a credible biodefense threat. To date, there have been no FDA-approved arenavirus vaccines, and current antiarenaviral therapy is limited to an off-label use of ribavirin that is only partially effective. Arenaviruses are enveloped viruses with a bisegmented negative-stranded RNA genome. Each genome segment uses an ambisense coding strategy to direct the synthesis of two viral polypeptides in opposite orientations, separated by a noncoding intergenic region. Here we have used minigenome-based approaches to evaluate expression levels of reporter genes from the nucleoprotein (NP) and glycoprotein precursor (GPC) loci within the S segment of the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV). We found that reporter genes are expressed to higher levels from the NP than from the GPC locus. Differences in reporter gene expression levels from the NP and GPC loci were confirmed with recombinant trisegmented LCM viruses. We then used reverse genetics to rescue a recombinant LCMV (rLCMV) containing a translocated viral S segment (rLCMV/TransS), where the viral NP and GPC open reading frames replaced one another. The rLCMV/TransS showed slower growth kinetics in cultured cells and was highly attenuated in vivo in a mouse model of lethal LCMV infection, but immunization with rLCMV/TransS conferred complete protection against a lethal challenge with wild-type LCMV. Attenuation of rLCMV/TransS was associated with reduced NP expression levels. These results open a new avenue for the development of arenavirus live attenuated vaccines based on rearrangement of their viral genome. IMPORTANCE: Several arenaviruses cause severe hemorrhagic fever in humans and also pose a credible bioterrorism threat. Currently, no FDA-licensed vaccines are available to combat arenavirus infections and antiarenaviral therapy is limited to the off-label use of ribavirin, which is only partially effective and associated with side effects. Here we describe, for the first time, the generation of a recombinant LCMV where the viral protein products encoded by the S RNA segment (NP and GPC) were swapped to generate rLCMV/TransS. rLCMV/TransS exhibited reduced viral multiplication in cultured cells and was highly attenuated in vivo while conferring protection, upon a single immunization dose, against a lethal challenge with wild-type LCMV. Our studies provide a proof of concept for the rational development of safe and protective live attenuated vaccine candidates based on genome reorganization for the treatment of pathogenic arenavirus infections in humans.


Subject(s)
Gene Rearrangement , Genome, Viral , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Viral Vaccines/immunology , Viral Vaccines/isolation & purification , Animals , Arenaviridae Infections/prevention & control , Disease Models, Animal , Gene Expression Profiling , Genes, Reporter , Lymphocytic Choriomeningitis/prevention & control , Lymphocytic choriomeningitis virus/physiology , Male , Mice, Inbred C57BL , Reverse Genetics , Survival Analysis , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Attenuated/isolation & purification , Viral Vaccines/genetics , Virus Cultivation , Virus Replication
7.
J Virol ; 89(7): 3523-33, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25589652

ABSTRACT

UNLABELLED: Arenaviruses have a significant impact on public health and pose a credible biodefense threat, but the development of safe and effective arenavirus vaccines has remained elusive, and currently, no Food and Drug Administration (FDA)-licensed arenavirus vaccines are available. Here, we explored the use of a codon deoptimization (CD)-based approach as a novel strategy to develop live-attenuated arenavirus vaccines. We recoded the nucleoprotein (NP) of the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) with the least frequently used codons in mammalian cells, which caused lower LCMV NP expression levels in transfected cells that correlated with decreased NP activity in cell-based functional assays. We used reverse-genetics approaches to rescue a battery of recombinant LCMVs (rLCMVs) encoding CD NPs (rLCMV/NP(CD)) that showed attenuated growth kinetics in vitro. Moreover, experiments using the well-characterized mouse model of LCMV infection revealed that rLCMV/NP(CD1) and rLCMV/NP(CD2) were highly attenuated in vivo but, upon a single immunization, conferred complete protection against a subsequent lethal challenge with wild-type (WT) recombinant LCMV (rLCMV/WT). Both rLCMV/NP(CD1) and rLCMV/NP(CD2) were genetically and phenotypically stable during serial passages in FDA vaccine-approved Vero cells. These results provide proof of concept of the safety, efficacy, and stability of a CD-based approach for developing live-attenuated vaccine candidates against human-pathogenic arenaviruses. IMPORTANCE: Several arenaviruses cause severe hemorrhagic fever in humans and pose a credible bioterrorism threat. Currently, no FDA-licensed vaccines are available to combat arenavirus infections, while antiarenaviral therapy is limited to the off-label use of ribavirin, which is only partially effective and is associated with side effects. Here, we describe the generation of recombinant versions of the prototypic arenavirus LCMV encoding codon-deoptimized viral nucleoproteins (rLCMV/NP(CD)). We identified rLCMV/NP(CD1) and rLCMV/NP(CD2) to be highly attenuated in vivo but able to confer protection against a subsequent lethal challenge with wild-type LCMV. These viruses displayed an attenuated phenotype during serial amplification passages in cultured cells. Our findings support the use of this approach for the development of safe, stable, and protective live-attenuated arenavirus vaccines.


Subject(s)
Arenaviridae Infections/prevention & control , Codon , Lymphocytic choriomeningitis virus/growth & development , Lymphocytic choriomeningitis virus/immunology , Viral Vaccines/immunology , Animals , Arenaviridae Infections/immunology , Arenaviridae Infections/pathology , Arenaviridae Infections/virology , Chlorocebus aethiops , Disease Models, Animal , Gene Expression , Genomic Instability , Lymphocytic choriomeningitis virus/genetics , Male , Mice , Survival Analysis , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Vaccines, Attenuated/isolation & purification , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Synthetic/isolation & purification , Vero Cells , Viral Vaccines/administration & dosage , Viral Vaccines/genetics , Viral Vaccines/isolation & purification , Virus Cultivation , Virus Replication
8.
J Vis Exp ; (78)2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23928556

ABSTRACT

The development and implementation of arenavirus reverse genetics represents a significant breakthrough in the arenavirus field. The use of cell-based arenavirus minigenome systems together with the ability to generate recombinant infectious arenaviruses with predetermined mutations in their genomes has facilitated the investigation of the contribution of viral determinants to the different steps of the arenavirus life cycle, as well as virus-host interactions and mechanisms of arenavirus pathogenesis. In addition, the development of trisegmented arenaviruses has permitted the use of the arenavirus genome to express additional foreign genes of interest, thus opening the possibility of arenavirus-based vaccine vector applications. Likewise, the development of single-cycle infectious arenaviruses capable of expressing reporter genes provides a new experimental tool to improve the safety of research involving highly pathogenic human arenaviruses. The generation of recombinant arenaviruses using plasmid-based reverse genetics techniques has so far relied on the use of rodent cell lines, which poses some barriers for the development of Food and Drug Administration (FDA)-licensed vaccine or vaccine vectors. To overcome this obstacle, we describe here the efficient generation of recombinant arenaviruses in FDA-approved Vero cells.


Subject(s)
Arenavirus/genetics , Arenavirus/immunology , Viral Vaccines/genetics , Animals , Chlorocebus aethiops , Humans , Reverse Genetics/methods , Transfection , United States , United States Food and Drug Administration , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vero Cells , Viral Vaccines/immunology , Virus Cultivation/methods
9.
J Gen Virol ; 94(Pt 6): 1175-1188, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23364194

ABSTRACT

Arenaviruses are important human pathogens with no Food and Drug Administration (FDA)-licensed vaccines available and current antiviral therapy being limited to an off-label use of the nucleoside analogue ribavirin of limited prophylactic efficacy. The development of reverse genetics systems represented a major breakthrough in arenavirus research. However, rescue of recombinant arenaviruses using current reverse genetics systems has been restricted to rodent cells. In this study, we describe the rescue of recombinant arenaviruses from human 293T cells and Vero cells, an FDA-approved line for vaccine development. We also describe the generation of novel vectors that mediate synthesis of both negative-sense genome RNA and positive-sense mRNA species of lymphocytic choriomeningitis virus (LCMV) directed by the human RNA polymerases I and II, respectively, within the same plasmid. This approach reduces by half the number of vectors required for arenavirus rescue, which could facilitate virus rescue in cell lines approved for human vaccine production but that cannot be transfected at high efficiencies. We have shown the feasibility of this approach by rescuing both the Old World prototypic arenavirus LCMV and the live-attenuated vaccine Candid#1 strain of the New World arenavirus Junín. Moreover, we show the feasibility of using these novel strategies for efficient rescue of recombinant tri-segmented both LCMV and Candid#1.


Subject(s)
Arenaviridae Infections/virology , Arenavirus/genetics , Reverse Genetics/methods , Viral Vaccines/genetics , Animals , Arenaviridae Infections/prevention & control , Arenavirus/immunology , Chlorocebus aethiops , Cricetinae , Genetic Vectors/genetics , Genetic Vectors/immunology , HEK293 Cells , Humans , Vero Cells , Viral Vaccines/immunology , Virus Cultivation
10.
Viruses ; 4(10): 2137-61, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23202457

ABSTRACT

Arenaviruses merit significant interest because several family members are etiological agents of severe hemorrhagic fevers, representing a major burden to public health. Currently, there are no FDA-licensed vaccines against arenaviruses and the only available antiviral therapy is limited to the use of ribavirin that is partially effective. Arenavirus nucleoprotein (NP) is found associated with the genomic RNA forming the viral ribonucleoproteins (vRNPs) that together with the polymerase (L) direct viral replication and transcription. Virion formation requires the recruitment of vRNPs into budding sites, a process in which the arenavirus matrix-like protein (Z) plays a major role. Therefore, proper NP-NP and NP-Z interactions are required for the generation of infectious progeny. In this work we demonstrate the role of the amino acid residue D471 in the self-association of lymphocytic choriomeningitis virus nucleoprotein (LCMV-NP). Amino acid substitutions at this position abrogate NP oligomerization, affecting its ability to mediate replication and transcription of a minigenome reporter plasmid. However, its ability to interact with the Z protein, counteract the cellular interferon response and bind to dsRNA analogs was retained. Additionally, we also document the dominant negative effect of D471G mutation on viral infection, suggesting that NP self-association is an excellent target for the development of new antivirals against arenaviruses.


Subject(s)
Lymphocytic choriomeningitis virus/metabolism , Mutation , Nucleoproteins/metabolism , RNA, Viral/biosynthesis , Viral Proteins/metabolism , Virus Assembly , Amino Acid Sequence , Amino Acid Substitution , Animals , Aspartic Acid/genetics , Aspartic Acid/metabolism , Blotting, Western , Carrier Proteins/genetics , Carrier Proteins/metabolism , Genes, Reporter , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Immunoprecipitation , Interferon Type I/genetics , Interferon Type I/metabolism , Intracellular Signaling Peptides and Proteins , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/pathogenicity , Lymphocytic choriomeningitis virus/physiology , Molecular Sequence Data , Nucleoproteins/genetics , Plasmids/genetics , Plasmids/metabolism , Protein Binding , Protein Interaction Mapping , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , RNA, Viral/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Species Specificity , Transcription, Genetic , Transfection , Viral Proteins/genetics , Virus Replication
11.
J Virol ; 86(8): 4340-57, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22318145

ABSTRACT

We applied a custom tiled microarray to examine murine gammaherpesvirus 68 (MHV68) polyadenylated transcript expression in a time course of de novo infection of fibroblast cells and following phorbol ester-mediated reactivation from a latently infected B cell line. During de novo infection, all open reading frames (ORFs) were transcribed and clustered into four major temporal groups that were overlapping yet distinct from clusters based on the phorbol ester-stimulated B cell reactivation time course. High-density transcript analysis at 2-h intervals during de novo infection mapped gene boundaries with a 20-nucleotide resolution, including a previously undefined ORF73 transcript and the MHV68 ORF63 homolog of Kaposi's sarcoma-associated herpesvirus vNLRP1. ORF6 transcript initiation was mapped by tiled array and confirmed by 5' rapid amplification of cDNA ends. The ∼1.3-kb region upstream of ORF6 was responsive to lytic infection and MHV68 RTA, identifying a novel RTA-responsive promoter. Transcription in intergenic regions consistent with the previously defined expressed genomic regions was detected during both types of productive infection. We conclude that the MHV68 transcriptome is dynamic and distinct during de novo fibroblast infection and upon phorbol ester-stimulated B cell reactivation, highlighting the need to evaluate further transcript structure and the context-dependent molecular events that govern viral gene expression during chronic infection.


Subject(s)
Gammaherpesvirinae/genetics , Gene Expression Profiling , Transcriptome , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cell Line , Cluster Analysis , Computational Biology , Fibroblasts/metabolism , Fibroblasts/virology , Gene Expression Regulation, Viral , Genome, Viral , Lymphocyte Activation/drug effects , Mice , Oligonucleotide Array Sequence Analysis , Open Reading Frames , Regulatory Elements, Transcriptional , Reproducibility of Results , Tetradecanoylphorbol Acetate/pharmacology
12.
J Virol ; 86(6): 3307-17, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22258244

ABSTRACT

Arenaviruses have a bisegmented, negative-strand RNA genome. Both the large (L) and small (S) genome segments use an ambisense coding strategy to direct the synthesis of two viral proteins. The L segment encodes the virus polymerase (L protein) and the matrix Z protein, whereas the S segment encodes the nucleoprotein (NP) and the glycoprotein precursor (GPC). NPs are the most abundant viral protein in infected cells and virions and encapsidate genomic RNA species to form an NP-RNA complex that, together with the virus L polymerase, forms the virus ribonucleoprotein (RNP) core capable of directing both replication and transcription of the viral genome. RNP formation predicts a self-association property of NPs. Here we document self-association (homotypic interaction) of the NP of the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV), as well as those of the hemorrhagic fever (HF) arenaviruses Lassa virus (LASV) and Machupo virus (MACV). We also show heterotypic interaction between NPs from both closely (LCMV and LASV) and distantly (LCMV and MACV) genetically related arenaviruses. LCMV NP self-association was dependent on the presence of single-stranded RNA and mediated by an N-terminal region of the NP that did not overlap with the previously described C-terminal NP domain involved in either counteracting the host type I interferon response or interacting with LCMV Z.


Subject(s)
Interferon Type I/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/metabolism , Nucleoproteins/metabolism , Viral Proteins/metabolism , Amino Acid Motifs , Animals , Cell Line , Dogs , Humans , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/chemistry , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/immunology , Nucleoproteins/chemistry , Nucleoproteins/genetics , Nucleoproteins/immunology , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/immunology
13.
J Virol ; 85(24): 13038-48, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21976642

ABSTRACT

Several arenaviruses cause hemorrhagic fever (HF) disease in humans that is associated with high morbidity and significant mortality. Arenavirus nucleoprotein (NP), the most abundant viral protein in infected cells and virions, encapsidates the viral genome RNA, and this NP-RNA complex, together with the viral L polymerase, forms the viral ribonucleoprotein (vRNP) that directs viral RNA replication and gene transcription. Formation of infectious arenavirus progeny requires packaging of vRNPs into budding particles, a process in which arenavirus matrix-like protein (Z) plays a central role. In the present study, we have characterized the NP-Z interaction for the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV). The LCMV NP domain that interacted with Z overlapped with a previously documented C-terminal domain that counteracts the host type I interferon (IFN) response. However, we found that single amino acid mutations that affect the anti-IFN function of LCMV NP did not disrupt the NP-Z interaction, suggesting that within the C-terminal region of NP different amino acid residues critically contribute to these two distinct and segregable NP functions. A similar NP-Z interaction was confirmed for the HF arenavirus Lassa virus (LASV). Notably, LCMV NP interacted similarly with both LCMV Z and LASV Z, while LASV NP interacted only with LASV Z. Our results also suggest the presence of a conserved protein domain within NP but with specific amino acid residues playing key roles in determining the specificity of NP-Z interaction that may influence the viability of reassortant arenaviruses. In addition, this NP-Z interaction represents a potential target for the development of antiviral drugs to combat human-pathogenic arenaviruses.


Subject(s)
Carrier Proteins/metabolism , Interferon Type I/antagonists & inhibitors , Lymphocytic choriomeningitis virus/physiology , Nucleoproteins/metabolism , Protein Interaction Domains and Motifs , Protein Interaction Mapping , Virus Assembly , Amino Acid Substitution , Animals , Carrier Proteins/genetics , Cell Line , Intracellular Signaling Peptides and Proteins , Lassa virus/genetics , Lassa virus/physiology , Lymphocytic choriomeningitis virus/immunology , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nucleoproteins/genetics , Point Mutation
14.
PLoS Pathog ; 7(7): e1002150, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21811408

ABSTRACT

During a lytic gammaherpesvirus infection, host gene expression is severely restricted by the global degradation and altered 3' end processing of mRNA. This host shutoff phenotype is orchestrated by the viral SOX protein, yet its functional significance to the viral lifecycle has not been elucidated, in part due to the multifunctional nature of SOX. Using an unbiased mutagenesis screen of the murine gammaherpesvirus 68 (MHV68) SOX homolog, we isolated a single amino acid point mutant that is selectively defective in host shutoff activity. Incorporation of this mutation into MHV68 yielded a virus with significantly reduced capacity for mRNA turnover. Unexpectedly, the MHV68 mutant showed little defect during the acute replication phase in the mouse lung. Instead, the virus exhibited attenuation at later stages of in vivo infections suggestive of defects in both trafficking and latency establishment. Specifically, mice intranasally infected with the host shutoff mutant accumulated to lower levels at 10 days post infection in the lymph nodes, failed to develop splenomegaly, and exhibited reduced viral DNA levels and a lower frequency of latently infected splenocytes. Decreased latency establishment was also observed upon infection via the intraperitoneal route. These results highlight for the first time the importance of global mRNA degradation during a gammaherpesvirus infection and link an exclusively lytic phenomenon with downstream latency establishment.


Subject(s)
Herpesviridae Infections/metabolism , RNA Stability , RNA, Messenger/metabolism , SOX Transcription Factors/metabolism , Viral Proteins/metabolism , Virus Latency/physiology , Animals , COS Cells , Chlorocebus aethiops , Female , HEK293 Cells , Herpesviridae Infections/genetics , Herpesviridae Infections/pathology , Humans , Lung/metabolism , Lung/pathology , Lung/virology , Mice , NIH 3T3 Cells , RNA, Messenger/genetics , Rhadinovirus/genetics , SOX Transcription Factors/genetics , Vero Cells , Viral Proteins/genetics
15.
Arch Phys Med Rehabil ; 92(9): 1484-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21762874

ABSTRACT

OBJECTIVE: To compare the thickness and stiffness of plantar soft tissues between people with diabetic peripheral neuropathy (DPN) and healthy subjects. DESIGN: Cross-sectional study. SETTING: University research laboratory. PARTICIPANTS: Subjects with DPN (n=70 [35 men, 35 women]; mean age ± SD, 65.4 ± 8.6y) and healthy control subjects (n=54 [12 men, 42 women]; mean age ± SD, 57.9 ± 6.1y) were recruited. INTERVENTIONS: Not applicable. MAIN OUTCOME MEASURES: The thickness and stiffness of the plantar soft tissues were measured by the tissue ultrasound palpation system over the pulp of the big toe (BT), first metatarsal head (MTH), second MTH, and the heel. RESULTS: No significant difference in the thickness of the plantar soft tissues was found in any measurement site between the diabetic group and control group. The plantar soft tissues of the DPN group were significantly stiffer than those of the control group at the BT (85.29 kPa vs 50.49 kPa), first MTH (96.29 kPa vs 62.05 kPa), second MTH (84.77 kPa vs 52.93 kPa), and the heel (65.62 kPa vs 44.95 kPa) (all P<.01). CONCLUSIONS: People with DPN tend to have stiffer plantar tissues than do healthy control subjects. The stiffer plantar soft tissues may reduce the cushioning effects of the foot during walking for people with DPN.


Subject(s)
Diabetic Neuropathies/pathology , Foot/pathology , Aged , Cross-Sectional Studies , Female , Humans , Male , Middle Aged
16.
Clin Rehabil ; 24(3): 202-10, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20156983

ABSTRACT

OBJECTIVE: To investigate the immediate effect of transcutaneous electrical nerve stimulation (TENS) on spasticity in patients with spinal cord injury. DESIGN: Randomized controlled trial. SETTING: Extended rehabilitation centre. SUBJECTS AND INTERVENTION: Eighteen subjects with spinal cord injury and symptoms of spasticity over lower limbs were randomly assigned to receive either 60 minutes of active TENS (0.25 ms, 100 Hz, 15 mA) or 60 minutes of placebo non-electrically stimulated TENS over the common peroneal nerve. OUTCOME MEASURES: Composite Spasticity Score was used to assess the spasticity level of ankle plantar flexors immediately before and after TENS application. Composite Spasticity Score consisted of Achilles tendon jerks, resistance to full-range passive ankle dorsiflexion and ankle clonus. Between-group statistical differences of reduction of Composite Spasticity Score, Achilles tendon jerks, resistance to full-range passive ankle dorsiflexion and ankle clonus were calculated using the Mann-Whitney test. Within-group statistical differences of Composite Spasticity Score, Achilles tendon jerks, resistance to full-range passive ankle dorsiflexion and ankle clonus were calculated using the Wilcoxon signed ranks test. RESULTS: Significant reductions were shown in Composite Spasticity Score by 29.5% (p = 0.017), resistance to full-range passive ankle dorsiflexion by 31.0% (p = 0.024) and ankle clonus by 29.6% (p = 0.023) in the TENS group but these reductions were not found in the placebo TENS group. The between-group differences of both Composite Spasticity Score and resistance to full-range passive ankle dorsiflexion were significant (p = 0.027 and p = 0.024, respectively). CONCLUSION: This study showed that a single session of TENS could immediately reduce spasticity.


Subject(s)
Muscle Spasticity/therapy , Spinal Cord Injuries/rehabilitation , Transcutaneous Electric Nerve Stimulation , Adult , Aged , Aged, 80 and over , Ankle/innervation , Female , Humans , Male , Middle Aged , Muscle Spasticity/etiology , Spinal Cord Injuries/complications , Treatment Outcome , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...