Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 121(25): e2316376121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38861603

ABSTRACT

Human parainfluenza virus type 3 (HPIV3) is a major pediatric respiratory pathogen lacking available vaccines or antiviral drugs. We generated live-attenuated HPIV3 vaccine candidates by codon-pair deoptimization (CPD). HPIV3 open reading frames (ORFs) encoding the nucleoprotein (N), phosphoprotein (P), matrix (M), fusion (F), hemagglutinin-neuraminidase (HN), and polymerase (L) were modified singly or in combination to generate 12 viruses designated Min-N, Min-P, Min-M, Min-FHN, Min-L, Min-NP, Min-NPM, Min-NPL, Min-PM, Min-PFHN, Min-MFHN, and Min-PMFHN. CPD of N or L severely reduced growth in vitro and was not further evaluated. CPD of P or M was associated with increased and decreased interferon (IFN) response in vitro, respectively, but had little effect on virus replication. In Vero cells, CPD of F and HN delayed virus replication, but final titers were comparable to wild-type (wt) HPIV3. In human lung epithelial A549 cells, CPD F and HN induced a stronger IFN response, viral titers were reduced 100-fold, and the expression of F and HN proteins was significantly reduced without affecting N or P or the relative packaging of proteins into virions. Following intranasal infection in hamsters, replication in the nasal turbinates and lungs tended to be the most reduced for viruses bearing CPD F and HN, with maximum reductions of approximately 10-fold. Despite decreased in vivo replication (and lower expression of CPD F and HN in vitro), all viruses induced titers of serum HPIV3-neutralizing antibodies similar to wt and provided complete protection against HPIV3 challenge. In summary, CPD of HPIV3 yielded promising vaccine candidates suitable for further development.


Subject(s)
Codon , Parainfluenza Virus 3, Human , Vaccines, Attenuated , Virus Replication , Animals , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 3, Human/genetics , Humans , Vaccines, Attenuated/immunology , Vaccines, Attenuated/genetics , Codon/genetics , Cricetinae , Respirovirus Infections/immunology , Respirovirus Infections/prevention & control , Respirovirus Infections/virology , Chlorocebus aethiops , Vero Cells , Open Reading Frames/genetics , Mesocricetus , Antibodies, Viral/immunology , Antibodies, Viral/blood , Viral Vaccines/immunology , Viral Vaccines/genetics , Viral Proteins/immunology , Viral Proteins/genetics , Parainfluenza Vaccines/immunology , Parainfluenza Vaccines/genetics
2.
Viruses ; 13(5)2021 05 12.
Article in English | MEDLINE | ID: mdl-34065980

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is an alphavirus that causes encephalitis. Previous work indicated that VEEV infection induced early growth response 1 (EGR1) expression, leading to cell death via the protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) arm of the unfolded protein response (UPR) pathway. Loss of PERK prevented EGR1 induction and decreased VEEV-induced death. The results presented within show that loss of PERK in human primary astrocytes dramatically reduced VEEV and eastern equine encephalitis virus (EEEV) infectious titers by 4-5 log10. Loss of PERK also suppressed VEEV replication in primary human pericytes and human umbilical vein endothelial cells, but it had no impact on VEEV replication in transformed U87MG and 293T cells. A significant reduction in VEEV RNA levels was observed as early as 3 h post-infection, but viral entry assays indicated that the loss of PERK minimally impacted VEEV entry. In contrast, the loss of PERK resulted in a dramatic reduction in viral nonstructural protein translation and negative-strand viral RNA production. The loss of PERK also reduced the production of Rift Valley fever virus and Zika virus infectious titers. These data indicate that PERK is an essential factor for the translation of alphavirus nonstructural proteins and impacts multiple RNA viruses, making it an exciting target for antiviral development.


Subject(s)
Alphavirus/genetics , Protein Biosynthesis , Viral Nonstructural Proteins/genetics , eIF-2 Kinase/genetics , Alphavirus/classification , Alphavirus/physiology , Astrocytes/metabolism , Astrocytes/virology , Cell Death , Cells, Cultured , Encephalitis Virus, Venezuelan Equine/physiology , Endothelial Cells/metabolism , Endothelial Cells/virology , HEK293 Cells , Humans , Pericytes/metabolism , Pericytes/virology , RNA, Viral/metabolism , Unfolded Protein Response , Viral Nonstructural Proteins/metabolism , eIF-2 Kinase/metabolism
3.
PLoS Pathog ; 16(3): e1008282, 2020 03.
Article in English | MEDLINE | ID: mdl-32150585

ABSTRACT

Protein phosphorylation plays an important role during the life cycle of many viruses. Venezuelan equine encephalitis virus (VEEV) capsid protein has recently been shown to be phosphorylated at four residues. Here those studies are extended to determine the kinase responsible for phosphorylation and the importance of capsid phosphorylation during the viral life cycle. Phosphorylation site prediction software suggests that Protein Kinase C (PKC) is responsible for phosphorylation of VEEV capsid. VEEV capsid co-immunoprecipitated with PKCδ, but not other PKC isoforms and siRNA knockdown of PKCδ caused a decrease in viral replication. Furthermore, knockdown of PKCδ by siRNA decreased capsid phosphorylation. A virus with capsid phosphorylation sites mutated to alanine (VEEV CPD) displayed a lower genomic copy to pfu ratio than the parental virus; suggesting more efficient viral assembly and more infectious particles being released. RNA:capsid binding was significantly increased in the mutant virus, confirming these results. Finally, VEEV CPD is attenuated in a mouse model of infection, with mice showing increased survival and decreased clinical signs as compared to mice infected with the parental virus. Collectively our data support a model in which PKCδ mediated capsid phosphorylation regulates viral RNA binding and assembly, significantly impacting viral pathogenesis.


Subject(s)
Capsid Proteins/metabolism , Encephalitis Virus, Venezuelan Equine/metabolism , Encephalomyelitis, Venezuelan Equine/enzymology , Protein Kinase C-delta/metabolism , RNA, Viral/metabolism , Animals , Capsid/metabolism , Capsid Proteins/genetics , Encephalitis Virus, Venezuelan Equine/genetics , Encephalomyelitis, Venezuelan Equine/genetics , Encephalomyelitis, Venezuelan Equine/virology , Female , Horses , Host-Pathogen Interactions , Mice , Mice, Inbred C3H , Phosphorylation , Protein Binding , Protein Kinase C-delta/genetics , RNA, Viral/genetics
4.
Virology ; 539: 121-128, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31733451

ABSTRACT

Venezuelan equine encephalitis virus (VEEV) is a neurotropic virus that causes significant disease in both humans and equines. Here we characterized the impact of VEEV on signaling pathways regulating cell death in human primary astrocytes. VEEV productively infected primary astrocytes and caused an upregulation of early growth response 1 (EGR1) gene expression at 9 and 18 h post infection. EGR1 induction was dependent on extracellular signal-regulated kinase1/2 (ERK1/2) and protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK), but not on p38 mitogen activated protein kinase (MAPK) or phosphoinositide 3-kinase (PI3K) signaling. Knockdown of EGR1 significantly reduced VEEV-induced apoptosis and impacted viral replication. Knockdown of ERK1/2 or PERK significantly reduced EGR1 gene expression, dramatically reduced viral replication, and increased cell survival as well as rescued cells from VEEV-induced apoptosis. These data indicate that EGR1 activation and subsequent cell death are regulated through ERK and PERK pathways in VEEV infected primary astrocytes.


Subject(s)
Cell Death , Early Growth Response Protein 1/genetics , Encephalitis Virus, Venezuelan Equine/physiology , Encephalomyelitis, Venezuelan Equine/virology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , eIF-2 Kinase/metabolism , Apoptosis , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/virology , Cell Survival , Cells, Cultured , Early Growth Response Protein 1/metabolism , Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalomyelitis, Venezuelan Equine/metabolism , Encephalomyelitis, Venezuelan Equine/pathology , Gene Expression , Gene Knockdown Techniques , Humans , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Signal Transduction , Virus Replication , eIF-2 Kinase/genetics
5.
Int J Antimicrob Agents ; 54(1): 80-84, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30930299

ABSTRACT

Zika virus (ZIKV) is a re-emerging Flavivirus that has been linked to microcephaly and other neurological pathologies. In this study, phloretin, a glucose transporter inhibitor naturally derived from plants, was used to investigate the glucose dependence of ZIKV replication in host cells. The results showed that phloretin significantly decreased infectious titres of two ZIKV strains, namely MR766 (African genotype) and PRVABC59 (Puerto Rico genotype). The 50% effective concentration (EC50) of phloretin against MR766 and PRVABC59 was 22.85 µM and 9.31 µM, respectively. Further analyses demonstrated that decreased viral production was due to host-targeted inhibition, including decreased apoptotic caspase-3 and -7 activities and reduced phosphorylation of Akt/mTOR pathways. In addition, upon disruption of cellular glucose availability within host cells using 2-deoxy-d-glucose, ZIKV propagation was inhibited. Collectively, we demonstrate phloretin inhibition of ZIKV propagation and provide evidence of glucose utilization pathways as being important for ZIKV propagation. The activity of phloretin and its role in inhibiting glucose uptake could provide a useful foundation for the development of ZIKV antivirals.


Subject(s)
Antiviral Agents/pharmacology , Carbohydrate Metabolism/drug effects , Enzyme Inhibitors/pharmacology , Phloretin/pharmacology , Virus Replication/drug effects , Zika Virus/drug effects , Animals , Chlorocebus aethiops , Vero Cells , Viral Load , Zika Virus/growth & development
6.
Antiviral Res ; 157: 57-67, 2018 09.
Article in English | MEDLINE | ID: mdl-29981794

ABSTRACT

The New World alphaviruses -Venezuelan, eastern, and western equine encephalitis viruses (VEEV, EEEV, and WEEV respectively) - cause a febrile disease that is often lethal in equines and children and leads to long-term neurological sequelae in survivors. Endemic to the Americas, epizootic outbreaks of the three viruses occur sporadically in the continental United States. All three viruses aerosolize readily, replicate to high titers in cell culture, and have low infectious doses. Additionally, there are no FDA-approved vaccines or therapeutics for human use. To address the therapeutic gap, a high throughput assay utilizing a luciferase reporter virus, TC83-luc, was performed to screen a library of commercially available, FDA-approved drugs for antiviral activity. From a group of twenty compounds found to significantly decrease luminescence, the carcinoma therapeutic sorafenib inhibited replication of VEEV-TC83 and TrD in vitro. Additionally, sorafenib inhibited replication of EEEV and two Old World alphaviruses, Sindbis virus and chikungunya virus, at 8 and 16 h post-infection. Sorafenib caused no toxicity in Vero cells, and coupled with a low EC50 value, yielded a selectivity index of >19. Mechanism of actions studies suggest that sorafenib inhibited viral translation through dephosphorylation of several key proteins, including eIF4E and p70S6K, leading to a reduction in viral protein production and overall viral replication.


Subject(s)
Alphavirus/drug effects , Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , Drug Repositioning , Sorafenib/pharmacology , Virus Replication/drug effects , Alphavirus/growth & development , Animals , Cell Line , Drug Evaluation, Preclinical/methods , Genes, Reporter , High-Throughput Screening Assays , Luciferases/analysis , Luciferases/genetics , Luminescent Measurements , Reverse Genetics
7.
Viruses ; 10(4)2018 04 13.
Article in English | MEDLINE | ID: mdl-29652799

ABSTRACT

Viruses must parasitize host cell translational machinery in order to make proteins for viral progeny. In this study, we sought to use this signal transduction conduit against them by inhibiting multiple kinases that influence translation. Previous work indicated that several kinases involved in translation, including p70 S6K, p90RSK, ERK, and p38 MAPK, are phosphorylated following Rift Valley fever virus (RVFV) infection. Furthermore, inhibiting p70 S6K through treatment with the FDA approved drug rapamycin prevents RVFV pathogenesis in a mouse model of infection. We hypothesized that inhibiting either p70 S6K, p90RSK, or p90RSK’s upstream kinases, ERK and p38 MAPK, would decrease translation and subsequent viral replication. Treatment with the p70 S6K inhibitor PF-4708671 resulted in decreased phosphorylation of translational proteins and reduced RVFV titers. In contrast, treatment with the p90RSK inhibitor BI-D1870, p38MAPK inhibitor SB203580, or the ERK inhibitor PD0325901 alone had minimal influence on RVFV titers. The combination of PF-4708671 and BI-D1870 treatment resulted in robust inhibition of RVFV replication. Likewise, a synergistic inhibition of RVFV replication was observed with p38MAPK inhibitor SB203580 or the ERK inhibitor PD0325901 combined with rapamycin treatment. These findings serve as a proof of concept regarding combination kinase inhibitor treatment for RVFV infection.


Subject(s)
Antiviral Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Rift Valley fever virus/drug effects , Rift Valley fever virus/physiology , Virus Replication/drug effects , Animals , Cell Line , Mice , Phosphorylation , Protein Biosynthesis/drug effects , Protein Processing, Post-Translational , Ribosomal Proteins/metabolism
8.
Sci Rep ; 7(1): 14385, 2017 10 30.
Article in English | MEDLINE | ID: mdl-29085037

ABSTRACT

Rift Valley fever virus (RVFV) causes major outbreaks among livestock, characterized by "abortion storms" in which spontaneous abortion occurs in almost 100% of pregnant ruminants. Humans can also become infected with mild symptoms that can progress to more severe symptoms, such as hepatitis, encephalitis, and hemorrhagic fever. The goal of this study was to use RNA-sequencing (RNA-seq) to analyze the host transcriptome in response to RVFV infection. G2/M DNA damage checkpoint, ATM signaling, mitochondrial dysfunction, regulation of the antiviral response, and integrin-linked kinase (ILK) signaling were among the top altered canonical pathways with both the attenuated MP12 strain and the fully virulent ZH548 strain. Although several mRNA transcripts were highly upregulated, an increase at the protein level was not observed for the selected genes, which was at least partially due to the NSs dependent block in mRNA export. Inhibition of ILK signaling, which is involved in cell motility and cytoskeletal reorganization, resulted in reduced RVFV replication, indicating that this pathway is important for viral replication. Overall, this is the first global transcriptomic analysis of the human host response following RVFV infection, which could give insight into novel host responses that have not yet been explored.


Subject(s)
Rift Valley Fever/genetics , Cell Culture Techniques , Cell Cycle Checkpoints , Epithelial Cells , Humans , Protein Serine-Threonine Kinases , RNA, Messenger/genetics , Rift Valley Fever/metabolism , Rift Valley fever virus/genetics , Rift Valley fever virus/pathogenicity , Sequence Analysis, RNA , Signal Transduction , Transcriptome/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/physiology
9.
Microbiol Res ; 195: 31-39, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28024524

ABSTRACT

Biological Nitrogen Fixation is critical for ecosystem productivity. Select members of Bacteria and Archaea express a nitrogenase enzyme complex that reduces atmospheric nitrogen to ammonia. Several nitrogen fixing bacteria form symbiotic associations with plants, but free-living diazotrophs also contribute a substantial amount of nitrogen to ecosystems. The aim of this study was to isolate and characterize free-living diazotrophs in arid lands of South Dakota Badlands. Samples were obtained from sod tables and the surrounding base in spring and fall. Diazotrophs were isolated on solid nitrogen free medium (NFM) under hypoxic conditions, and their16S rRNA and nifH genes sequenced. nifH was also amplified directly from soil DNA extracts. The 16S rRNA gene data indicated a diversity of putative free-living diazotrophs across 4 phyla (Actinomycetes, Proteobacteria, Bacteroidetes, and Firmicutes), but ∼50% of these clustered with Streptomyces. These Streptomyces isolates grew in liquid NFM in an ammonia-depleted environment. Only 5 of these yielded a nifH gene product using the PolF/PolR primer set. Four of these aligned with nifH of the cyanobacteria Scytonema and Nostoc, and the other one aligned with nifH of Bradyrhizobium. Six selected Streptomyces isolates, three of which were nifH positive by PCR, all indicated 15N2 incorporation, providing strong support of nitrogen fixation. All nifH amplicons from soil DNA extract resembled Cyanobacteria. This is the first known report of diazotrophic Streptomyces, other than the thermophilic, autotrophic S. thermoautotrophicus. nifH genes of these Streptomyces were related to those from Cyanobacteria. It is possible that the cyanobacteria-like nifH amplicons obtained from soil DNA were associated with Streptomyces.


Subject(s)
Biodiversity , Nitrogen Fixation , Soil Microbiology , Streptomyces/classification , Streptomyces/isolation & purification , Cluster Analysis , Culture Media/chemistry , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , DNA, Ribosomal/chemistry , DNA, Ribosomal/genetics , Oxidoreductases/genetics , Phylogeny , Polymerase Chain Reaction , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , South Dakota , Streptomyces/genetics , Streptomyces/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...