Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Bull Math Biol ; 86(5): 44, 2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38512541

ABSTRACT

On July 19th, 2023, the National Institute of Allergy and Infectious Diseases co-organized a workshop with the Society of Mathematical Biology, with the authors of this paper as the organizing committee. The workshop, "Bridging multiscale modeling and practical clinical applications in infectious diseases" sought to create an environment for mathematical modelers, statisticians, and infectious disease researchers and clinicians to exchange ideas and perspectives.


Subject(s)
Communicable Diseases , Mathematical Concepts , United States , Humans , National Institute of Allergy and Infectious Diseases (U.S.) , Models, Biological
2.
Am J Epidemiol ; 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37968375

ABSTRACT

The spread of SARS-CoV-2 since late 2019 represented an unprecedented public health emergency, which included a need to fully understand COVID-19 disease across all ages and populations. In response, the US National Institute of Allergy and Infectious Diseases (NIAID) rapidly funded epidemiology studies that monitored COVID-19. However, the diversity and breadth of the populations studied in NIAID-funded COVID-19 observational cohorts were not easy to extrapolate because of siloed approaches to collect and report data within NIAID. Here, we describe the effort to develop a harmonized cohort study reporting tool that includes common epidemiological data elements as well as NIAID priorities. We report its implementation to analyze metadata from 58 COVID-19 cohort studies funded February 2020 to June 2021, visualize key metadata including geographic distribution, study duration, participant demographics, sample types collected, and scientific priorities addressed. A bibliographic analysis highlights the scientific publications and citations across these funded studies and demonstrates their enormous impact on the COVID-19 field. These analyses highlight how common data elements and reporting tools can assist funding agencies to capture the landscape and potential gaps during public health responses and how they can assist in decision making.

3.
PLoS Pathog ; 13(2): e1006164, 2017 02.
Article in English | MEDLINE | ID: mdl-28152048

ABSTRACT

Zika virus (ZIKV) is an emerging mosquito-borne flavivirus that is causally linked to severe neonatal birth defects, including microcephaly, and is associated with Guillain-Barre syndrome in adults. Dendritic cells (DCs) are an important cell type during infection by multiple mosquito-borne flaviviruses, including dengue virus, West Nile virus, Japanese encephalitis virus, and yellow fever virus. Despite this, the interplay between ZIKV and DCs remains poorly defined. Here, we found human DCs supported productive infection by a contemporary Puerto Rican isolate with considerable variability in viral replication, but not viral binding, between DCs from different donors. Historic isolates from Africa and Asia also infected DCs with distinct viral replication kinetics between strains. African lineage viruses displayed more rapid replication kinetics and infection magnitude as compared to Asian lineage viruses, and uniquely induced cell death. Infection of DCs with both contemporary and historic ZIKV isolates led to minimal up-regulation of T cell co-stimulatory and MHC molecules, along with limited secretion of inflammatory cytokines. Inhibition of type I interferon (IFN) protein translation was observed during ZIKV infection, despite strong induction at the RNA transcript level and up-regulation of other host antiviral proteins. Treatment of human DCs with RIG-I agonist potently restricted ZIKV replication, while type I IFN had only modest effects. Mechanistically, we found all strains of ZIKV antagonized type I IFN-mediated phosphorylation of STAT1 and STAT2. Combined, our findings show that ZIKV subverts DC immunogenicity during infection, in part through evasion of type I IFN responses, but that the RLR signaling pathway is still capable of inducing an antiviral state, and therefore may serve as an antiviral therapeutic target.


Subject(s)
DEAD Box Protein 58/immunology , Dendritic Cells/virology , Immune Evasion/immunology , Interferon Type I/immunology , Zika Virus Infection/immunology , Blotting, Western , Dendritic Cells/immunology , Flow Cytometry , Humans , Polymerase Chain Reaction , Receptors, Immunologic , Zika Virus/immunology
4.
J Immunol ; 199(11): 3808-3820, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29070670

ABSTRACT

Type III IFNs are important mediators of antiviral immunity. IFN-λ4 is a unique type III IFN because it is produced only in individuals who carry a dG allele of a genetic variant rs368234815-dG/TT. Counterintuitively, those individuals who can produce IFN-λ4, an antiviral cytokine, are also less likely to clear hepatitis C virus infection. In this study, we searched for unique functional properties of IFN-λ4 that might explain its negative effect on hepatitis C virus clearance. We used fresh primary human hepatocytes (PHHs) treated with recombinant type III IFNs or infected with Sendai virus to model acute viral infection and subsequently validated our findings in HepG2 cell line models. Endogenous IFN-λ4 protein was detectable only in Sendai virus-infected PHHs from individuals with the dG allele, where it was poorly secreted but highly functional, even at concentrations < 50 pg/ml. IFN-λ4 acted faster than other type III IFNs in inducing antiviral genes, as well as negative regulators of the IFN response, such as USP18 and SOCS1 Transient treatment of PHHs with IFN-λ4, but not IFN-λ3, caused a strong and sustained induction of SOCS1 and refractoriness to further stimulation with IFN-λ3. Our results suggest unique functional properties of IFN-λ4 that can be important in viral clearance and other clinical conditions.


Subject(s)
Alleles , Hepatocytes/immunology , Interferons/genetics , Interleukins/genetics , Respirovirus Infections/immunology , Sendai virus/immunology , Adolescent , Adult , Aged , Endopeptidases/genetics , Female , Hep G2 Cells , Hepacivirus/immunology , Hepatitis C/genetics , Hepatitis C/immunology , Hepatocytes/virology , Humans , Immunity , Interferons/metabolism , Interleukins/metabolism , Male , Middle Aged , Polymorphism, Single Nucleotide , Suppressor of Cytokine Signaling 1 Protein/genetics , Ubiquitin Thiolesterase , Up-Regulation , Viral Load , Young Adult
5.
J Virol ; 90(23): 10600-10611, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27654297

ABSTRACT

Since the India and Indian Ocean outbreaks of 2005 and 2006, the global distribution of chikungunya virus (CHIKV) and the locations of epidemics have dramatically shifted. First, the Indian Ocean lineage (IOL) caused sustained epidemics in India and has radiated to many other countries. Second, the Asian lineage has caused frequent outbreaks in the Pacific islands and in 2013 was introduced into the Caribbean, followed by rapid spread to nearly all of the neotropics. Further, CHIKV epidemics, as well as exported cases, have been reported in central Africa after a long period of perceived silence. To understand these changes and to anticipate the future of the virus, the exact distribution, genetic diversity, transmission routes, and future epidemic potential of CHIKV require further assessment. To do so, we conducted the most comprehensive phylogenetic analysis to date, examined CHIKV evolution and transmission, and explored distinct genetic factors associated with the emergence of the East/Central/South African (ECSA) lineage, the IOL, and the Asian lineage. Our results reveal contrasting evolutionary patterns among the lineages, with growing genetic diversities observed in each, and suggest that CHIKV will continue to be a major public health threat with the potential for further emergence and spread. IMPORTANCE: Chikungunya fever is a reemerging infectious disease that is transmitted by Aedes mosquitoes and causes severe health and economic burdens in affected populations. Since the unprecedented Indian Ocean and Indian subcontinent outbreaks of 2005 and 2006, CHIKV has further expanded its geographic range, including to the Americas in 2013. Its evolution and transmission during and following these epidemics, as well as the recent evolution and spread of other lineages, require optimal assessment. Using newly obtained genome sequences, we provide a comprehensive update of the global distribution of CHIKV genetic diversity and analyze factors associated with recent outbreaks. These results provide a solid foundation for future evolutionary studies of CHIKV that can elucidate emergence mechanisms and also may help to predict future epidemics.


Subject(s)
Chikungunya Fever/epidemiology , Chikungunya Fever/virology , Chikungunya virus/classification , Chikungunya virus/genetics , 5' Untranslated Regions , Aedes/virology , Africa/epidemiology , Americas/epidemiology , Animals , Asia/epidemiology , Chikungunya Fever/transmission , Epidemics , Evolution, Molecular , Genetic Variation , Genome, Viral , Humans , India/epidemiology , Insect Vectors/virology , Phylogeny
6.
PLoS Pathog ; 10(10): e1004420, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25275541

ABSTRACT

Bats harbor many viruses, which are periodically transmitted to humans resulting in outbreaks of disease (e.g., Ebola, SARS-CoV). Recently, influenza virus-like sequences were identified in bats; however, the viruses could not be cultured. This discovery aroused great interest in understanding the evolutionary history and pandemic potential of bat-influenza. Using synthetic genomics, we were unable to rescue the wild type bat virus, but could rescue a modified bat-influenza virus that had the HA and NA coding regions replaced with those of A/PR/8/1934 (H1N1). This modified bat-influenza virus replicated efficiently in vitro and in mice, resulting in severe disease. Additional studies using a bat-influenza virus that had the HA and NA of A/swine/Texas/4199-2/1998 (H3N2) showed that the PR8 HA and NA contributed to the pathogenicity in mice. Unlike other influenza viruses, engineering truncations hypothesized to reduce interferon antagonism into the NS1 protein didn't attenuate bat-influenza. In contrast, substitution of a putative virulence mutation from the bat-influenza PB2 significantly attenuated the virus in mice and introduction of a putative virulence mutation increased its pathogenicity. Mini-genome replication studies and virus reassortment experiments demonstrated that bat-influenza has very limited genetic and protein compatibility with Type A or Type B influenza viruses, yet it readily reassorts with another divergent bat-influenza virus, suggesting that the bat-influenza lineage may represent a new Genus/Species within the Orthomyxoviridae family. Collectively, our data indicate that the bat-influenza viruses recently identified are authentic viruses that pose little, if any, pandemic threat to humans; however, they provide new insights into the evolution and basic biology of influenza viruses.


Subject(s)
Chiroptera/virology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Orthomyxoviridae Infections , Virus Replication/genetics , Animals , Cell Line , Humans , Mice , Swine , Viral Proteins/metabolism
7.
PLoS Pathog ; 9(1): e1003147, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23382680

ABSTRACT

Ebolaviruses, highly lethal zoonotic pathogens, possess longer genomes than most other non-segmented negative-strand RNA viruses due in part to long 5' and 3' untranslated regions (UTRs) present in the seven viral transcriptional units. To date, specific functions have not been assigned to these UTRs. With reporter assays, we demonstrated that the Zaire ebolavirus (EBOV) 5'-UTRs lack internal ribosomal entry site function. However, the 5'-UTRs do differentially regulate cap-dependent translation when placed upstream of a GFP reporter gene. Most dramatically, the 5'-UTR derived from the viral polymerase (L) mRNA strongly suppressed translation of GFP compared to a ß-actin 5'-UTR. The L 5'-UTR is one of four viral genes to possess upstream AUGs (uAUGs), and ablation of each uAUG enhanced translation of the primary ORF (pORF), most dramatically in the case of the L 5'-UTR. The L uAUG was sufficient to initiate translation, is surrounded by a "weak" Kozak sequence and suppressed pORF translation in a position-dependent manner. Under conditions where eIF2α was phosphorylated, the presence of the uORF maintained translation of the L pORF, indicating that the uORF modulates L translation in response to cellular stress. To directly address the role of the L uAUG in virus replication, a recombinant EBOV was generated in which the L uAUG was mutated to UCG. Strikingly, mutating two nucleotides outside of previously-defined protein coding and cis-acting regulatory sequences attenuated virus growth to titers 10-100-fold lower than a wild-type virus in Vero and A549 cells. The mutant virus also exhibited decreased viral RNA synthesis as early as 6 hours post-infection and enhanced sensitivity to the stress inducer thapsigargin. Cumulatively, these data identify novel mechanisms by which EBOV regulates its polymerase expression, demonstrate their relevance to virus replication and identify a potential therapeutic target.


Subject(s)
DNA-Directed RNA Polymerases , Ebolavirus/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Viral , Viral Proteins/metabolism , Virus Replication/genetics , 3' Untranslated Regions/genetics , 5' Untranslated Regions/genetics , Animals , Base Sequence , Cell Line , Chlorocebus aethiops , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Ebolavirus/genetics , Enzyme Inhibitors/pharmacology , Humans , Molecular Sequence Data , Protein Biosynthesis , RNA, Viral/biosynthesis , Thapsigargin/pharmacology , Vero Cells
8.
Proc Natl Acad Sci U S A ; 109(50): 20661-6, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23185024

ABSTRACT

Filoviruses, marburgvirus (MARV) and ebolavirus (EBOV), are causative agents of highly lethal hemorrhagic fever in humans. MARV and EBOV share a common genome organization but show important differences in replication complex formation, cell entry, host tropism, transcriptional regulation, and immune evasion. Multifunctional filoviral viral protein (VP) 35 proteins inhibit innate immune responses. Recent studies suggest double-stranded (ds)RNA sequestration is a potential mechanism that allows EBOV VP35 to antagonize retinoic-acid inducible gene-I (RIG-I) like receptors (RLRs) that are activated by viral pathogen-associated molecular patterns (PAMPs), such as double-strandedness and dsRNA blunt ends. Here, we show that MARV VP35 can inhibit IFN production at multiple steps in the signaling pathways downstream of RLRs. The crystal structure of MARV VP35 IID in complex with 18-bp dsRNA reveals that despite the similar protein fold as EBOV VP35 IID, MARV VP35 IID interacts with the dsRNA backbone and not with blunt ends. Functional studies show that MARV VP35 can inhibit dsRNA-dependent RLR activation and interferon (IFN) regulatory factor 3 (IRF3) phosphorylation by IFN kinases TRAF family member-associated NFkb activator (TANK) binding kinase-1 (TBK-1) and IFN kB kinase e (IKKe) in cell-based studies. We also show that MARV VP35 can only inhibit RIG-I and melanoma differentiation associated gene 5 (MDA5) activation by double strandedness of RNA PAMPs (coating backbone) but is unable to inhibit activation of RLRs by dsRNA blunt ends (end capping). In contrast, EBOV VP35 can inhibit activation by both PAMPs. Insights on differential PAMP recognition and inhibition of IFN induction by a similar filoviral VP35 fold, as shown here, reveal the structural and functional plasticity of a highly conserved virulence factor.


Subject(s)
Marburgvirus/immunology , Marburgvirus/pathogenicity , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/immunology , Amino Acid Sequence , Animals , Base Sequence , Crystallography, X-Ray , HEK293 Cells , Host-Pathogen Interactions , Humans , I-kappa B Kinase/antagonists & inhibitors , Immunity, Innate , Interferon Type I/antagonists & inhibitors , Marburg Virus Disease/etiology , Marburg Virus Disease/immunology , Marburg Virus Disease/virology , Marburgvirus/chemistry , Models, Biological , Models, Molecular , Molecular Sequence Data , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Structure, Tertiary , RNA/chemistry , RNA/genetics , RNA/metabolism , Sequence Homology, Amino Acid , Virulence/immunology
9.
J Virol ; 87(7): 3801-14, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23345511

ABSTRACT

Antigen-presenting cells (APCs) are critical targets of Ebola virus (EBOV) infection in vivo. However, the susceptibility of monocytes to infection is controversial. Studies indicate productive monocyte infection, and yet monocytes are also reported to be resistant to EBOV GP-mediated entry. In contrast, monocyte-derived macrophages and dendritic cells are permissive for both EBOV entry and replication. Here, freshly isolated monocytes are demonstrated to indeed be refractory to EBOV entry. However, EBOV binds monocytes, and delayed entry occurs during monocyte differentiation. Cultured monocytes spontaneously downregulate the expression of viral entry restriction factors such as interferon-inducible transmembrane proteins, while upregulating the expression of critical EBOV entry factors cathepsin B and NPC1. Moreover, these processes are accelerated by EBOV infection. Finally, ectopic expression of NPC1 is sufficient to rescue entry into an undifferentiated, normally nonpermissive monocytic cell line. These results define the molecular basis for infection of APCs and suggest means to limit APC infection.


Subject(s)
Cell Differentiation/physiology , Ebolavirus/physiology , Monocytes/virology , Virus Attachment , Virus Internalization , Carrier Proteins/metabolism , Cathepsin B/metabolism , DNA Primers/genetics , Dendritic Cells/virology , Flow Cytometry , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Macrophages/virology , Membrane Glycoproteins/metabolism , Monocytes/physiology , Niemann-Pick C1 Protein , Reverse Transcriptase Polymerase Chain Reaction
10.
PLoS Pathog ; 8(10): e1002934, 2012.
Article in English | MEDLINE | ID: mdl-23055924

ABSTRACT

Dengue virus (DENV) is a pathogen with a high impact on human health. It replicates in a wide range of cells involved in the immune response. To efficiently infect humans, DENV must evade or inhibit fundamental elements of the innate immune system, namely the type I interferon response. DENV circumvents the host immune response by expressing proteins that antagonize the cellular innate immunity. We have recently documented the inhibition of type I IFN production by the proteolytic activity of DENV NS2B3 protease complex in human monocyte derived dendritic cells (MDDCs). In the present report we identify the human adaptor molecule STING as a target of the NS2B3 protease complex. We characterize the mechanism of inhibition of type I IFN production in primary human MDDCs by this viral factor. Using different human and mouse primary cells lacking STING, we show enhanced DENV replication. Conversely, mutated versions of STING that cannot be cleaved by the DENV NS2B3 protease induced higher levels of type I IFN after infection with DENV. Additionally, we show that DENV NS2B3 is not able to degrade the mouse version of STING, a phenomenon that severely restricts the replication of DENV in mouse cells, suggesting that STING plays a key role in the inhibition of DENV infection and spread in mice.


Subject(s)
Dendritic Cells/metabolism , Dengue Virus/immunology , Dengue Virus/pathogenicity , Interferon Type I/biosynthesis , Membrane Proteins/metabolism , Viral Nonstructural Proteins/metabolism , Aedes , Animals , Cells, Cultured , Chlorocebus aethiops , Cricetinae , Dendritic Cells/virology , Dengue Virus/metabolism , HEK293 Cells , Humans , Immune Evasion , Macrophages/metabolism , Macrophages/virology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , RNA Interference , RNA, Small Interfering , Signal Transduction , Vero Cells , Virus Replication
11.
Biochemistry ; 52(47): 8406-19, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24067086

ABSTRACT

Viral protein 35 (VP35), encoded by filoviruses, is a multifunctional dsRNA binding protein that plays important roles in viral replication, innate immune evasion, and pathogenesis. The multifunctional nature of these proteins also presents opportunities to develop countermeasures that target distinct functional regions. However, functional validation and the establishment of therapeutic approaches toward such multifunctional proteins, particularly for nonenzymatic targets, are often challenging. Our previous work on filoviral VP35 proteins defined conserved basic residues located within its C-terminal dsRNA binding interferon (IFN) inhibitory domain (IID) as important for VP35 mediated IFN antagonism and viral polymerase cofactor functions. In the current study, we used a combination of structural and functional data to determine regions of Ebola virus (EBOV) VP35 (eVP35) to target for aptamer selection using SELEX. Select aptamers, representing, two distinct classes, were further characterized based on their interaction properties to eVP35 IID. These results revealed that these aptamers bind to distinct regions of eVP35 IID with high affinity (10-50 nM) and specificity. These aptamers can compete with dsRNA for binding to eVP35 and disrupt the eVP35-nucleoprotein (NP) interaction. Consistent with the ability to antagonize the eVP35-NP interaction, select aptamers can inhibit the function of the EBOV polymerase complex reconstituted by the expression of select viral proteins. Taken together, our results support the identification of two aptamers that bind filoviral VP35 proteins with high affinity and specificity and have the capacity to potentially function as filoviral VP35 protein inhibitors.


Subject(s)
Antiviral Agents/chemistry , Aptamers, Nucleotide/chemistry , RNA/chemistry , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Amino Acid Sequence , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Aptamers, Nucleotide/metabolism , Aptamers, Nucleotide/pharmacology , Binding, Competitive , Conserved Sequence , Ebolavirus/drug effects , Ebolavirus/metabolism , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/metabolism , Kinetics , Molecular Targeted Therapy , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nucleic Acid Conformation , Nucleoproteins/metabolism , Protein Conformation , Protein Interaction Domains and Motifs , RNA/metabolism , RNA/pharmacology , RNA, Double-Stranded/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , SELEX Aptamer Technique , Species Specificity , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism
12.
J Virol ; 86(21): 11779-88, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22915810

ABSTRACT

Inclusion bodies are a characteristic feature of ebolavirus infections in cells. They contain large numbers of preformed nucleocapsids, but their biological significance has been debated, and they have been suggested to be aggregates of viral proteins without any further biological function. However, recent data for other viruses that produce similar structures have suggested that inclusion bodies might be involved in genome replication and transcription. In order to study filovirus inclusion bodies, we fused mCherry to the ebolavirus polymerase L, which is found in inclusion bodies. The resulting L-mCherry fusion protein was functional in minigenome assays and incorporated into virus-like particles. Importantly, L-mCherry fluorescence in transfected cells was readily detectable and distributed in a punctate pattern characteristic for inclusion bodies. A recombinant ebolavirus encoding L-mCherry instead of L was rescued and showed virtually identical growth kinetics and endpoint titers to those for wild-type virus. Using this virus, we showed that the onset of inclusion body formation corresponds to the onset of viral genome replication, but that viral transcription occurs prior to inclusion body formation. Live-cell imaging further showed that inclusion bodies are highly dynamic structures and that they can undergo dramatic reorganization during cell division. Finally, by labeling nascent RNAs using click technology we showed that inclusion bodies are indeed the site of viral RNA synthesis. Based on these data we conclude that, rather than being inert aggregates of nucleocapsids, ebolavirus inclusion bodies are in fact complex and dynamic structures and an important site at which viral RNA replication takes place.


Subject(s)
Ebolavirus/physiology , Inclusion Bodies, Viral/virology , Virus Replication , Animals , Artificial Gene Fusion , Cell Line , Genes, Reporter , Humans , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Microscopy, Fluorescence , RNA-Dependent RNA Polymerase/genetics , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Transfection , Viral Proteins/genetics , Red Fluorescent Protein
13.
Sci Data ; 10(1): 99, 2023 02 23.
Article in English | MEDLINE | ID: mdl-36823157

ABSTRACT

Biomedical datasets are increasing in size, stored in many repositories, and face challenges in FAIRness (findability, accessibility, interoperability, reusability). As a Consortium of infectious disease researchers from 15 Centers, we aim to adopt open science practices to promote transparency, encourage reproducibility, and accelerate research advances through data reuse. To improve FAIRness of our datasets and computational tools, we evaluated metadata standards across established biomedical data repositories. The vast majority do not adhere to a single standard, such as Schema.org, which is widely-adopted by generalist repositories. Consequently, datasets in these repositories are not findable in aggregation projects like Google Dataset Search. We alleviated this gap by creating a reusable metadata schema based on Schema.org and catalogued nearly 400 datasets and computational tools we collected. The approach is easily reusable to create schemas interoperable with community standards, but customized to a particular context. Our approach enabled data discovery, increased the reusability of datasets from a large research consortium, and accelerated research. Lastly, we discuss ongoing challenges with FAIRness beyond discoverability.


Subject(s)
Communicable Diseases , Datasets as Topic , Metadata , Reproducibility of Results , Datasets as Topic/standards , Humans
14.
J Infect Dis ; 204 Suppl 3: S904-10, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21987768

ABSTRACT

The Ebola virus (EBOV) protein VP24 inhibits type I and II interferon (IFN) signaling by binding to NPI-1 subfamily karyopherin α (KPNA) nuclear import proteins, preventing their interaction with tyrosine-phosphorylated STAT1 (phospho-STAT1). This inhibits phospho-STAT1 nuclear import. A biochemical screen now identifies heterogeneous nuclear ribonuclear protein complex C1/C2 (hnRNP C1/C2) nuclear import as an additional target of VP24. Co-immunoprecipitation studies demonstrate that hnRNP C1/C2 interacts with multiple KPNA family members, including KPNA1. Interaction with hnRNP C1/C2 occurs through the same KPNA1 C-terminal region (amino acids 424-457) that binds VP24 and phospho-STAT1. The ability of hnRNP C1/C2 to bind KPNA1 is diminished in the presence of VP24, and cells transiently expressing VP24 redistribute hnRNP C1/C2 from the nucleus to the cytoplasm. These data further define the mechanism of hnRNP C1/C2 nuclear import and demonstrate that the impact of EBOV VP24 on nuclear import extends beyond STAT1.


Subject(s)
Ebolavirus/physiology , Gene Expression Regulation/physiology , Heterogeneous-Nuclear Ribonucleoprotein Group C/metabolism , Viral Proteins/metabolism , alpha Karyopherins/metabolism , Active Transport, Cell Nucleus , Animals , Cell Nucleus/metabolism , Chlorocebus aethiops , Cytoplasm/metabolism , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoprotein Group C/genetics , Humans , Mice , Protein Binding , Protein Structure, Tertiary , Rabbits , Vero Cells , Viral Proteins/genetics , alpha Karyopherins/genetics
15.
J Infect Dis ; 204 Suppl 3: S911-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21987769

ABSTRACT

The Zaire Ebola virus (EBOV) protein VP35 is multifunctional; it inhibits IFN-α/ß production and functions as a cofactor of the viral RNA polymerase. Mass spectrometry identified the double stranded RNA binding protein 76 (DRBP76/NFAR-1/NF90) as a cellular factor that associates with the VP35 C-terminal interferon inhibitory domain (IID). DRBP76 is described to regulate host cell protein synthesis and play an important role in host defense. The VP35-IID-DRBP76 interaction required the addition of exogenous dsRNA, but full-length VP35 associated with DRBP76 in the absence of exogenous dsRNA. Cells infected with a Newcastle disease virus (NDV)-expressing VP35 redistributed DRBP76 from the nucleus to the cytoplasm, the compartment in which EBOV replicates. Overexpression of DRBP76 did not alter the ability of VP35 to inhibit type I IFN production but did impair the function of the EBOV transcription/replication complex. These data suggest that DRBP76, via its association with VP35, exerts an anti-EBOV function.


Subject(s)
DNA-Directed RNA Polymerases/metabolism , Ebolavirus/physiology , Gene Expression Regulation/physiology , Nuclear Factor 90 Proteins/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Animals , Chlorocebus aethiops , DNA-Directed RNA Polymerases/genetics , HEK293 Cells , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Newcastle disease virus/genetics , Nuclear Factor 90 Proteins/genetics , Poly I-C , Promoter Regions, Genetic , Protein Structure, Tertiary , RNA, Double-Stranded , Vero Cells , Viral Regulatory and Accessory Proteins/genetics , Virus Replication
16.
J Virol ; 84(20): 10581-91, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20686031

ABSTRACT

The ebolavirus (EBOV) VP35 protein binds to double-stranded RNA (dsRNA), inhibits host alpha/beta interferon (IFN-α/ß) production, and is an essential component of the viral polymerase complex. Structural studies of the VP35 C-terminal IFN inhibitory domain (IID) identified specific structural features, including a central basic patch and a hydrophobic pocket, that are important for dsRNA binding and IFN inhibition. Several other conserved basic residues bordering the central basic patch and a separate cluster of basic residues, called the first basic patch, were also identified. Functional analysis of alanine substitution mutants indicates that basic residues outside the central basic patch are not required for dsRNA binding or for IFN inhibition. However, minigenome assays, which assess viral RNA polymerase complex function, identified these other basic residues to be critical for viral RNA synthesis. Of these, a subset located within the first basic patch is important for VP35-nucleoprotein (NP) interaction, as evidenced by the inability of alanine substitution mutants to coimmunoprecipitate with NP. Therefore, first basic patch residues are likely critical for replication complex formation through interactions with NP. Coimmunoprecipitation studies further demonstrate that the VP35 IID is sufficient to interact with NP and that dsRNA can modulate VP35 IID interactions with NP. Other basic residue mutations that disrupt the VP35 polymerase cofactor function do not affect interaction with NP or with the amino terminus of the viral polymerase. Collectively, these results highlight the importance of conserved basic residues from the EBOV VP35 C-terminal IID and validate the VP35 IID as a potential therapeutic target.


Subject(s)
Ebolavirus/physiology , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/physiology , Amino Acid Substitution , Animals , Cell Line , Chlorocebus aethiops , Ebolavirus/genetics , Ebolavirus/pathogenicity , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Models, Molecular , Mutagenesis, Site-Directed , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Nucleocapsid Proteins , Nucleoproteins/chemistry , Nucleoproteins/genetics , Nucleoproteins/physiology , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , RNA/genetics , RNA/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Static Electricity , Vero Cells , Viral Core Proteins/chemistry , Viral Core Proteins/genetics , Viral Core Proteins/physiology , Viral Regulatory and Accessory Proteins/genetics , Virulence/genetics , Virulence/physiology
17.
J Virol ; 82(24): 12374-83, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18922878

ABSTRACT

Alphaviruses are mosquito-transmitted viruses that cause significant human disease, and understanding how these pathogens successfully transition from the mosquito vector to the vertebrate host is an important area of research. Previous studies demonstrated that mosquito and mammalian-cell-derived alphaviruses differentially induce type I interferons (alpha/beta interferon [IFN-alpha/beta]) in myeloid dendritic cells (mDCs), where the mosquito cell-derived virus is a poor inducer of IFN-alpha/beta compared to the mammalian-cell-derived virus. Furthermore, the reduced IFN-alpha/beta induction by the mosquito cell-derived virus is attributed to differential N-linked glycosylation. To further evaluate the role of viral envelope glycans in regulating the IFN-alpha/beta response, studies were performed to assess whether the mosquito cell-derived virus actively inhibits IFN-alpha/beta induction or is simply a poor inducer of IFN-alpha/beta. Coinfection studies using mammalian- and mosquito cell-derived Ross River virus (mam-RRV and mos-RRV, respectively) indicated that mos-RRV was unable to suppress IFN-alpha/beta induction by mam-RRV in mDC cultures. Additionally, a panel of mutant viruses lacking either individual or multiple N-linked glycosylation sites was used to demonstrate that N-linked glycans were essential for high-level IFN-alpha/beta induction by the mammalian-cell-derived virus. These results suggest that the failure of the mosquito cell-derived virus to induce IFN-alpha/beta is due to a lack of complex carbohydrates on the virion rather than the active suppression of the DC antiviral response.


Subject(s)
Dendritic Cells/metabolism , Interferon Type I/biosynthesis , Myeloid Cells/metabolism , Polysaccharides/metabolism , Ross River virus/metabolism , Viral Envelope Proteins/metabolism , Aedes , Animals , Cell Line , Cricetinae , Genome, Viral/genetics , Mutation/genetics , Ross River virus/genetics , Viral Envelope Proteins/genetics
18.
Virus Res ; 263: 173-178, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30742853

ABSTRACT

Zika Virus (ZIKV) is a mosquito-borne flavivirus that the World Health Organization (WHO) declared a global concern due to the severity of infection. This study focuses on determining the level of detection of ZIKV RNA in human serum and urine. Known amounts of Zika virus were added to uninfected human serum and urine samples. Different reverse transcriptases were compared to select the optimal enzyme for this application. Zika RNA in these samples was then quantified with qRT-PCR to determine the lower limit of detection in these fluids and to construct a standard curve. Student's t-test of paired samples was used in order to identify statistical differences. The SuperScript III enzyme was able to produce more ZIKV cDNA when compared to PrimeScript. Zika virus RNA was found to be detectable at lower levels (2.5 PFU/mL) in urine than in serum (250 PFU/mL) when using SuperScript III. This study demonstrates how the selection of both the human clinical specimen, and the reverse transcriptase enzyme involved in the molecular detection of ZIKV by quantitative real-time polymerase chain reaction (qRT-PCR), play an important role in enabling improved detection of the virus.


Subject(s)
RNA, Viral/analysis , Real-Time Polymerase Chain Reaction/methods , Reverse Transcriptase Polymerase Chain Reaction/methods , Serum/virology , Urine/virology , Zika Virus Infection/diagnosis , Zika Virus/isolation & purification , Humans , RNA, Viral/genetics , RNA-Directed DNA Polymerase/metabolism , Sensitivity and Specificity , Zika Virus/genetics , Zika Virus Infection/virology
19.
J Virol ; 81(24): 13412-23, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17913817

ABSTRACT

A novel genetic vaccine that is based on a Venezuelan equine encephalitis virus (VEE) replicon launched from plasmid DNA is described. The plasmid encodes a VEE replicon under the transcriptional control of the cytomegalovirus immediate-early promoter (VEE DNA). The VEE DNA consistently expressed 3- to 15-fold more green fluorescent protein in vitro than did a conventional DNA vaccine. Furthermore, transfection with the DNA-launched VEE replicon induced apoptosis and type I interferon production. Inoculation of mice with VEE DNA encoding human immunodeficiency virus type 1 gp160 significantly increased humoral responses by several orders of magnitude compared to an equal dose of a conventional DNA vaccine. These increases were also observed at 10- and 100-fold-lower doses of the VEE DNA. Cellular immune responses measured by gamma interferon and interleukin 2 enzyme-linked immunospot assay were significantly higher in mice immunized with the VEE DNA at decreased doses. The immune responses induced by the VEE DNA-encoded antigen, however, were independent of an intact type I interferon signaling pathway. Moreover, the DNA-launched VEE replicon induced an efficient prime to a VEE replicon particle (VRP) boost, increasing humoral and cellular immunity by at least 1 order of magnitude compared to VEE DNA only. Importantly, immunization with VEE DNA, as opposed to VRP, did not induce any anti-VRP neutralizing antibodies. Increased potency of DNA vaccines and reduced vector immunity may ultimately have an impact on the design of vaccination strategies in humans.


Subject(s)
Antibodies, Viral/blood , Encephalitis Virus, Venezuelan Equine/genetics , Encephalitis Virus, Venezuelan Equine/immunology , Interferon Type I/biosynthesis , Interleukin-2/biosynthesis , Plasmids/genetics , Replicon/immunology , Vaccines, DNA/immunology , Animals , Apoptosis , Cell Line , Chlorocebus aethiops , Genetic Vectors , HIV Envelope Protein gp160/genetics , HIV Envelope Protein gp160/immunology , HIV-1/genetics , Humans , Immunization , Immunoglobulin G/blood , L Cells , Mice , Mice, Inbred BALB C , Mice, Knockout , NIH 3T3 Cells , Promoter Regions, Genetic , Replicon/genetics , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vero Cells
20.
F1000Res ; 7: 98, 2018.
Article in English | MEDLINE | ID: mdl-31231504

ABSTRACT

The human cell lines HepG2, HuH-7, and Jurkat are commonly used for amplification of the RNA viruses present in environmental samples. To assist with assays by RNAseq, we sequenced these cell lines and developed a subtraction database that contains sequences expected in sequence data from uninfected cells. RNAseq data from cell lines infected with Sendai virus were analyzed to test host subtraction. The process of mapping RNAseq reads to our subtraction database vastly reduced the number non-viral reads in the dataset to allow for efficient secondary analyses.


Subject(s)
Databases, Genetic , Cell Line , DNA Viruses , High-Throughput Nucleotide Sequencing , Humans , Viruses
SELECTION OF CITATIONS
SEARCH DETAIL