Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
J Virol ; 94(24)2020 11 23.
Article in English | MEDLINE | ID: mdl-32999019

ABSTRACT

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus associated with debilitating arthralgia in humans. RNA secondary structure in the viral genome plays an important role in the lifecycle of alphaviruses; however, the specific role of RNA structure in regulating CHIKV replication is poorly understood. Our previous studies found little conservation in RNA secondary structure between alphaviruses, and this structural divergence creates unique functional structures in specific alphavirus genomes. Therefore, to understand the impact of RNA structure on CHIKV biology, we used SHAPE-MaP to inform the modeling of RNA secondary structure throughout the genome of a CHIKV isolate from the 2013 Caribbean outbreak. We then analyzed regions of the genome with high levels of structural specificity to identify potentially functional RNA secondary structures and identified 23 regions within the CHIKV genome with higher than average structural stability, including four previously identified, functionally important CHIKV RNA structures. We also analyzed the RNA flexibility and secondary structures of multiple 3'UTR variants of CHIKV that are known to affect virus replication in mosquito cells. This analysis found several novel RNA structures within these 3'UTR variants. A duplication in the 3'UTR that enhances viral replication in mosquito cells led to an overall increase in the amount of unstructured RNA in the 3'UTR. This analysis demonstrates that the CHIKV genome contains a number of unique, specific RNA secondary structures and provides a strategy for testing these secondary structures for functional importance in CHIKV replication and pathogenesis.IMPORTANCE Chikungunya virus (CHIKV) is a mosquito-borne RNA virus that causes febrile illness and debilitating arthralgia in humans. CHIKV causes explosive outbreaks but there are no approved therapies to treat or prevent CHIKV infection. The CHIKV genome contains functional RNA secondary structures that are essential for proper virus replication. Since RNA secondary structures have only been defined for a small portion of the CHIKV genome, we used a chemical probing method to define the RNA secondary structures of CHIKV genomic RNA. We identified 23 highly specific structured regions of the genome, and confirmed the functional importance of one structure using mutagenesis. Furthermore, we defined the RNA secondary structure of three CHIKV 3'UTR variants that differ in their ability to replicate in mosquito cells. Our study highlights the complexity of the CHIKV genome and describes new systems for designing compensatory mutations to test the functional relevance of viral RNA secondary structures.


Subject(s)
3' Untranslated Regions/genetics , Chikungunya virus/genetics , RNA, Viral/chemistry , RNA, Viral/genetics , Animals , Cell Line , Chikungunya Fever/virology , Chlorocebus aethiops , Culicidae , Cytopathogenic Effect, Viral , Genome, Viral , Mutation , Nucleic Acid Conformation , Sequence Analysis , Vero Cells , Virus Replication/genetics
2.
J Infect Dis ; 221(6): 882-889, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31621854

ABSTRACT

BACKGROUND: Virus infections result in a range of clinical outcomes for the host, from asymptomatic to severe or even lethal disease. Despite global efforts to prevent and treat virus infections to limit morbidity and mortality, the continued emergence and re-emergence of new outbreaks as well as common infections such as influenza persist as a health threat. Challenges to the prevention of severe disease after virus infection include both a paucity of protective vaccines as well as the early identification of individuals with the highest risk that may require supportive treatment. METHODS: We completed a screen of mice from the Collaborative Cross (CC) that we infected with influenza, severe acute respiratory syndrome-coronavirus, and West Nile virus. RESULTS: The CC mice exhibited a range of disease manifestations upon infections, and we used this natural variation to identify strains with mortality after infection and strains exhibiting no mortality. We then used comprehensive preinfection immunophenotyping to identify global baseline immune correlates of protection from mortality to virus infection. CONCLUSIONS: These data suggest that immune phenotypes might be leveraged to identify humans at highest risk of adverse clinical outcomes upon infection, who may most benefit from intensive clinical interventions, in addition to providing insight for rational vaccine design.


Subject(s)
Mortality , RNA Virus Infections/immunology , RNA Virus Infections/mortality , Animals , Collaborative Cross Mice , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Influenza A virus/immunology , Influenza, Human , Male , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , RNA , RNA Virus Infections/virology , Severe acute respiratory syndrome-related coronavirus/immunology , Severe Acute Respiratory Syndrome/immunology , Severe Acute Respiratory Syndrome/mortality , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Viral Vaccines/immunology , West Nile Fever/immunology , West Nile Fever/mortality , West Nile virus/immunology
3.
Cell Rep ; 31(4): 107587, 2020 04 28.
Article in English | MEDLINE | ID: mdl-32348764

ABSTRACT

Host genetic factors play a fundamental role in regulating humoral immunity to viral infection, including influenza A virus (IAV). Here, we utilize the Collaborative Cross (CC), a mouse genetic reference population, to study genetic regulation of variation in antibody response following IAV infection. CC mice show significant heritable variation in the magnitude, kinetics, and composition of IAV-specific antibody response. We map 23 genetic loci associated with this variation. Analysis of a subset of these loci finds that they broadly affect the antibody response to IAV as well as other viruses. Candidate genes are identified based on predicted variant consequences and haplotype-specific expression patterns, and several show overlap with genes identified in human mapping studies. These findings demonstrate that the host antibody response to IAV infection is under complex genetic control and highlight the utility of the CC in modeling and identifying genetic factors with translational relevance to human health and disease.


Subject(s)
Host-Pathogen Interactions/genetics , Influenza, Human/genetics , Virus Replication/genetics , Humans
4.
Water Res ; 42(14): 4001-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18708238

ABSTRACT

Fecal pollution is a serious threat to the estuarine environment along the Georgia coast. Culture-dependant and molecular methodologies were utilized to compare and evaluate the abundance of fecal indicator bacteria in four Georgia estuaries (Darien River, Frederica River, Gulley Hole Creek, and St. Marys River). The functionality of enterococci and bifidobacteria as indicator organisms in marine environments was assessed, as well as Bifidobacterium adolescentis densities. At each study site, enterococci were enumerated as colony forming units (CFU) on mEI agar. For quantitative polymerase chain reaction (qPCR), genus- and species-specific primer sets were used to quantify bifidobacteria and B. adolescentis as 16S rRNA gene copies and enterococci as tuf gene copies. A high correlation (r=0.925) was observed between CFU and qPCR enumeration of enterococci. Enterococci densities in the estuarine rivers ranged from 3-449CFU/100ml on mEI plates and 4.58-5.39Log(10) gene copies/100ml by qPCR. Bifidobacteria densities ranged from 3.62-4.14Log(10) gene copies/100ml and suggested the Frederica River as least affected by fecal bacteria and the Darien River as most affected by fecal pollution. A correlation of 0.46 was observed among qPCR densities of enterococci and bifidobacteria at all sample sites. Quantitative polymerase chain reaction detection of B. adolescentis was a rapid (i.e., less than 2h) indicator of presumptive human fecal pollution and suggested that Gulley Hole Creek, the Darien River, and the St. Marys River were affected by fecal bacteria derived from a human source. Gulley Hole Creek and the Darien River had the highest levels of fecal pollution detected in the studied estuaries. Molecular quantification of bifidobacteria may be a more accurate method of determining immediate health risks associated with fecal pollution in estuarine water than traditional and contemporary assessments of enterococci.


Subject(s)
Bifidobacterium/isolation & purification , Enterococcus/isolation & purification , Rivers/microbiology , Water Microbiology , Georgia , Oceans and Seas , Sewage/microbiology , Water Pollutants
5.
G3 (Bethesda) ; 8(2): 427-445, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29187420

ABSTRACT

Influenza A virus (IAV) is a respiratory pathogen that causes substantial morbidity and mortality during both seasonal and pandemic outbreaks. Infection outcomes in unexposed populations are affected by host genetics, but the host genetic architecture is not well understood. Here, we obtain a broad view of how heritable factors affect a mouse model of response to IAV infection using an 8 × 8 diallel of the eight inbred founder strains of the Collaborative Cross (CC). Expanding on a prior statistical framework for modeling treatment response in diallels, we explore how a range of heritable effects modify acute host response to IAV through 4 d postinfection. Heritable effects in aggregate explained ∼57% of the variance in IAV-induced weight loss. Much of this was attributable to a pattern of additive effects that became more prominent through day 4 postinfection and was consistent with previous reports of antiinfluenza myxovirus resistance 1 (Mx1) polymorphisms segregating between these strains; these additive effects largely recapitulated haplotype effects observed at the Mx1 locus in a previous study of the incipient CC, and are also replicated here in a CC recombinant intercross population. Genetic dominance of protective Mx1 haplotypes was observed to differ by subspecies of origin: relative to the domesticus null Mx1 allele, musculus acts dominantly whereas castaneus acts additively. After controlling for Mx1, heritable effects, though less distinct, accounted for ∼34% of the phenotypic variance. Implications for future mapping studies are discussed.


Subject(s)
Bayes Theorem , Genetic Predisposition to Disease/genetics , Myxovirus Resistance Proteins/genetics , Orthomyxoviridae Infections/genetics , Animals , Disease Models, Animal , Haplotypes , Humans , Influenza A virus/physiology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Inbred Strains , Orthomyxoviridae Infections/virology , Phenotype , Species Specificity
6.
mBio ; 8(6)2017 11 14.
Article in English | MEDLINE | ID: mdl-29138302

ABSTRACT

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus responsible for several significant outbreaks of debilitating acute and chronic arthritis and arthralgia over the past decade. These include a recent outbreak in the Caribbean islands and the Americas that caused more than 1 million cases of viral arthralgia. Despite the major impact of CHIKV on global health, viral determinants that promote CHIKV-induced disease are incompletely understood. Most CHIKV strains contain a conserved opal stop codon at the end of the viral nsP3 gene. However, CHIKV strains that encode an arginine codon in place of the opal stop codon have been described, and deep-sequencing analysis of a CHIKV isolate from the Caribbean identified both arginine and opal variants within this strain. Therefore, we hypothesized that the introduction of the arginine mutation in place of the opal termination codon may influence CHIKV virulence. We tested this by introducing the arginine mutation into a well-characterized infectious clone of a CHIKV strain from Sri Lanka and designated this virus Opal524R. This mutation did not impair viral replication kinetics in vitro or in vivo Despite this, the Opal524R virus induced significantly less swelling, inflammation, and damage within the feet and ankles of infected mice. Further, we observed delayed induction of proinflammatory cytokines and chemokines, as well as reduced CD4+ T cell and NK cell recruitment compared to those in the parental strain. Therefore, the opal termination codon plays an important role in CHIKV pathogenesis, independently of effects on viral replication.IMPORTANCE Chikungunya virus (CHIKV) is a mosquito-borne alphavirus that causes significant outbreaks of viral arthralgia. Studies with CHIKV and other alphaviruses demonstrated that the opal termination codon within nsP3 is highly conserved. However, some strains of CHIKV and other alphaviruses contain mutations in the opal termination codon. These mutations alter the virulence of related alphaviruses in mammalian and mosquito hosts. Here, we report that a clinical isolate of a CHIKV strain from the recent outbreak in the Caribbean islands contains a mixture of viruses encoding either the opal termination codon or an arginine mutation. Mutating the opal stop codon to an arginine residue attenuates CHIKV-induced disease in a mouse model. Compared to infection with the opal-containing parental virus, infection with the arginine mutant causes limited swelling and inflammation, as well as dampened recruitment of immune mediators of pathology, including CD4+ T cells and NK cells. We propose that the opal termination codon plays an essential role in the induction of severe CHIKV disease.


Subject(s)
Arthritis/pathology , Chikungunya Fever/pathology , Chikungunya virus/pathogenicity , Codon, Terminator , Mutation , Viral Nonstructural Proteins/genetics , Virulence Factors/genetics , Animals , Arginine/genetics , Arthritis/virology , Chikungunya Fever/virology , Chikungunya virus/physiology , Disease Models, Animal , Mice , Virus Replication
7.
Microbiol Spectr ; 4(3)2016 06.
Article in English | MEDLINE | ID: mdl-27337473

ABSTRACT

Chikungunya virus (CHIKV) is a mosquito-borne alphavirus in the family Togaviridae that causes outbreaks of debilitating acute and chronic arthralgia in humans. Although historically associated with localized outbreaks in Africa and Asia, recent epidemics in the Indian Ocean region and the Americas have led to the recognition that CHIKV is capable of moving into previously unaffected areas and causing significant levels of human suffering. The severity of CHIKV rheumatic disease, which can severely impact life quality of infected individuals for weeks, months, or even years, combined with the explosive nature of CHIKV outbreaks and its demonstrated ability to quickly spread into new regions, has led to renewed interest in developing strategies for the prevention or treatment of CHIKV-induced disease. Therefore, this chapter briefly discusses the biology of CHIKV and the factors contributing to CHIKV dissemination, while also discussing the pathogenesis of CHIKV-induced disease and summarizing the status of efforts to develop safe and effective therapies and vaccines against CHIKV and related viruses.


Subject(s)
Chikungunya Fever/epidemiology , Chikungunya Fever/pathology , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/pathology , Disease Outbreaks , Animals , Chikungunya Fever/prevention & control , Chikungunya Fever/therapy , Communicable Disease Control/methods , Communicable Diseases, Emerging/prevention & control , Communicable Diseases, Emerging/therapy , Global Health , Humans , Viral Vaccines/immunology , Viral Vaccines/isolation & purification
8.
Virology ; 456-457: 96-107, 2014 May.
Article in English | MEDLINE | ID: mdl-24889229

ABSTRACT

West Nile virus (WNV) is a mosquito-transmitted pathogen, which causes significant disease in humans. The innate immune system is a first-line defense against invading microorganism and many flaviviruses, including WNV, have evolved multifunctional proteins, which actively suppress its activation and antiviral actions. The WNV non-structural protein 1 (NS1) inhibits signal transduction originating from Toll-like receptor 3 (TLR3) and also critically contributes to virus genome replication. In this study we developed a novel FACS-based screen to attempt to separate these two functions. The individual amino acid changes P320S and M333V in NS1 restored TLR3 signaling in virus-infected HeLa cells. However, virus replication was also attenuated, suggesting that the two functions are not easily separated and may be contained within overlapping domains. The residues we identified are completely conserved among several mosquito- and tick-borne flaviviruses, indicating that they are of biological importance to the virus.


Subject(s)
Host-Pathogen Interactions , Immune Evasion , Toll-Like Receptor 3/immunology , Viral Nonstructural Proteins/immunology , Virus Replication , West Nile virus/immunology , West Nile virus/physiology , Amino Acid Substitution , DNA Mutational Analysis , HeLa Cells , Humans , Mutant Proteins/genetics , Mutant Proteins/immunology , Toll-Like Receptor 3/antagonists & inhibitors , Viral Nonstructural Proteins/genetics , West Nile virus/genetics
9.
Virology ; 458-459: 172-82, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24928049

ABSTRACT

West Nile virus (WNV) employs several different strategies to escape the innate immune response. We have previously demonstrated that the WNV NS1 protein interferes with signal transduction from Toll-like receptor 3 (TLR3). NS1 is a glycoprotein that can be found intracellularly or associated with the plasma membrane. In addition, NS1 is secreted to high levels during flavivirus infections. We investigated whether the secreted form of NS1 inhibits innate immune signaling pathways in uninfected cells. Secreted NS1 (sNS1) was purified from supernatants of cells engineered to express the protein. Purified sNS1 associated with and repressed TLR3-induced cytokine production by HeLa cells, and inhibited signaling from TLR3 and other TLRs in bone marrow-derived macrophages and dendritic cells. Footpad administration of sNS1 showed the protein associated predominantly with macrophages and dendritic cells in the draining lymph node. Additionally, sNS1 significantly reduced TLR3 signaling and WNV replicon particle-mediated cytokine transcription in popliteal lymph nodes.


Subject(s)
Immunity, Innate , Signal Transduction/immunology , Viral Nonstructural Proteins/immunology , West Nile virus/immunology , Animals , HeLa Cells , Humans , Luciferases/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/immunology , Myeloid Cells/metabolism , Poly I-C/pharmacology , Specific Pathogen-Free Organisms , Toll-Like Receptor 3/antagonists & inhibitors , Viral Nonstructural Proteins/physiology , West Nile virus/metabolism
10.
J Immunol Methods ; 344(1): 58-63, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19328803

ABSTRACT

We report a streamlined procedure to efficiently carry samples from chromatin to qPCR-compatible DNA in as little as 4 h. We use this streamlined ChIP to quantify histone H3 modifications at active (cad) and repressed (T early alpha) promoters in a Rag1-deficient pro-T cell line after 1-2 h IP. We further show that the protocol readily quantified histone modifications in chromatin from 10(4) Rag-deficient DN thymocytes. Taken together, these data outline a simple, cost-effective procedure for efficient ChIP analysis.


Subject(s)
Chromatin Immunoprecipitation/methods , Chromatin/isolation & purification , Histones/metabolism , Animals , Aspartate Carbamoyltransferase/genetics , Aspartate Carbamoyltransferase/metabolism , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/genetics , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/metabolism , Cell Line , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dihydroorotase/genetics , Dihydroorotase/metabolism , Gene Knockdown Techniques , Histones/genetics , Mice , Mice, Knockout , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , T-Lymphocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL