Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 186
Filter
Add more filters

Publication year range
1.
J Infect Dis ; 229(Supplement_2): S265-S274, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-37995376

ABSTRACT

Variola virus (VARV), the etiological agent of smallpox, had enormous impacts on global health prior to its eradication. In the absence of global vaccination programs, mpox virus (MPXV) has become a growing public health threat that includes endemic and nonendemic regions across the globe. While human mpox resembles smallpox in clinical presentation, there are considerable knowledge gaps regarding conserved molecular pathogenesis between these 2 orthopoxviruses. Thus, we sought to compare MPXV and VARV infections in human monocytes through kinome analysis. We performed a longitudinal analysis of host cellular responses to VARV infection in human monocytes as well as a comparative analysis to clade I MPXV-mediated responses. While both viruses elicited strong activation of cell responses early during infection as compared to later time points, several key differences in cell signaling events were identified and validated. These observations will help in the design and development of panorthopoxvirus therapeutics.


Subject(s)
Orthopoxvirus , Smallpox , Variola virus , Humans , Monkeypox virus , Monocytes
2.
Emerg Infect Dis ; 30(4): 721-731, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38526136

ABSTRACT

Genetically diverse simian arteriviruses (simarteriviruses) naturally infect geographically and phylogenetically diverse monkeys, and cross-species transmission and emergence are of considerable concern. Characterization of most simarteriviruses beyond sequence analysis has not been possible because the viruses fail to propagate in the laboratory. We attempted to isolate 4 simarteriviruses, Kibale red colobus virus 1, Pebjah virus, simian hemorrhagic fever virus, and Southwest baboon virus 1, by inoculating an immortalized grivet cell line (known to replicate simian hemorrhagic fever virus), primary macaque cells, macrophages derived from macaque induced pluripotent stem cells, and mice engrafted with macaque CD34+-enriched hematopoietic stem cells. The combined effort resulted in successful virus isolation; however, no single approach was successful for all 4 simarteriviruses. We describe several approaches that might be used to isolate additional simarteriviruses for phenotypic characterization. Our results will expedite laboratory studies of simarteriviruses to elucidate virus-host interactions, assess zoonotic risk, and develop medical countermeasures.


Subject(s)
Arterivirus , Animals , Mice , Arterivirus/genetics , Macaca , Macrophages , Cell Line
3.
Am J Pathol ; 192(1): 121-129, 2022 01.
Article in English | MEDLINE | ID: mdl-34626576

ABSTRACT

The pathogenesis of Ebola virus disease (EVD) is still incomplete, in spite of the availability of a nonhuman primate modelfor more than 4 decades. To further investigate EVD pathogenesis, a natural history study was conducted using 27 Chinese-origin rhesus macaques. Of these, 24 macaques were exposed intramuscularly to Kikwit Ebola virus and euthanized at predetermined time points or when end-stage clinical disease criteria were met, and 3 sham-exposed macaques were euthanized on study day 0. This study showed for the first time that Ebola virus causes uterine cervicitis, vaginitis, posthitis, and medullary adrenalitis. Not only was Ebola virus detected in the interstitial stromal cells of the genital tract, but it was also present in the epididymal and seminal vesicular tubular epithelial cells, ectocervical and vaginal squamous epithelial cells, and seminal fluid. Furthermore, as early as day 3 after exposure, Ebola virus replicative intermediate RNA was detected in Kupffer cells and hepatocytes. These findings in the nonhuman model provide additional insight into potential sexual transmission, possible disruption of sympathetic hormone production, and early virus replication sites in human EVD patients.


Subject(s)
Ebolavirus/physiology , Hormones/metabolism , Liver/virology , Tropism/physiology , Virus Replication/physiology , Animals , Chromaffin Cells/pathology , Chromaffin Cells/virology , Disease Models, Animal , Epididymis/pathology , Epididymis/virology , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Hepatocytes/pathology , Hepatocytes/virology , Kupffer Cells/pathology , Kupffer Cells/virology , Macaca mulatta , Male , Uterine Cervicitis/pathology , Uterine Cervicitis/virology , Vaginitis/pathology , Vaginitis/virology
4.
Proc Natl Acad Sci U S A ; 117(48): 30687-30698, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33184176

ABSTRACT

The SARS-CoV-2 pandemic has made it clear that we have a desperate need for antivirals. We present work that the mammalian SKI complex is a broad-spectrum, host-directed, antiviral drug target. Yeast suppressor screening was utilized to find a functional genetic interaction between proteins from influenza A virus (IAV) and Middle East respiratory syndrome coronavirus (MERS-CoV) with eukaryotic proteins that may be potential host factors involved in replication. This screening identified the SKI complex as a potential host factor for both viruses. In mammalian systems siRNA-mediated knockdown of SKI genes inhibited replication of IAV and MERS-CoV. In silico modeling and database screening identified a binding pocket on the SKI complex and compounds predicted to bind. Experimental assays of those compounds identified three chemical structures that were antiviral against IAV and MERS-CoV along with the filoviruses Ebola and Marburg and two further coronaviruses, SARS-CoV and SARS-CoV-2. The mechanism of antiviral activity is through inhibition of viral RNA production. This work defines the mammalian SKI complex as a broad-spectrum antiviral drug target and identifies lead compounds for further development.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus/drug effects , Filoviridae/drug effects , Host-Pathogen Interactions/drug effects , Multiprotein Complexes/metabolism , Orthomyxoviridae/drug effects , Cell Line , Genes, Suppressor , Models, Molecular , Molecular Targeted Therapy , Protein Binding , RNA, Small Interfering/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Saccharomyces cerevisiae/genetics , Viral Proteins/metabolism , Virus Replication/drug effects
5.
Am J Pathol ; 190(9): 1867-1880, 2020 09.
Article in English | MEDLINE | ID: mdl-32479821

ABSTRACT

The most commonly reported symptom of post-Ebola virus disease syndrome in survivors is arthralgia, yet involvement of the joints in acute or convalescent Ebola virus infection is not well characterized in human patients or animal models. Through immunohistochemistry, we found that the lining synovial intima of the stifle (knee) is a target for acute infection by Ebola virus/Kikwit, Ebola virus/Makona-C05, and Marburg virus/Angola in the rhesus macaque model. Furthermore, histologic analysis, immunohistochemistry, RNAscope in situ hybridization, and transmission electron microscopy showed that synoviocytes of the stifle, shoulder, and hip are a target for mouse-adapted Ebola virus/Yambuku-Mayinga infection during acute disease in rhesus macaques. A time course of infection study with Ebola virus/Kikwit found that the large joint synovium became immunopositive beginning on postinfection day 6. In total, the synovium of 28 of 30 rhesus macaques with terminal filovirus disease had evidence of infection (64 of 96 joints examined). On the basis of immunofluorescence, infected cell types included CD68+ type A (macrophage-like) synoviocytes and CD44+ type B (fibroblast-like) synoviocytes. Cultured primary human fibroblast-like synoviocytes were permissive to infection with Ebola and Marburg viruses in vitro. Because synovial joints include immune privileged sites, these findings are significant for future investigations of filovirus pathogenesis and persistence as well as arthralgias in acute and convalescent filovirus disease.


Subject(s)
Filoviridae Infections/virology , Synoviocytes/virology , Animals , Cells, Cultured , Filoviridae , Humans , Macaca mulatta
6.
J Infect Dis ; 219(11): 1818-1822, 2019 05 05.
Article in English | MEDLINE | ID: mdl-30517671

ABSTRACT

Lassa fever (LF) survivors develop various clinical manifestations including polyserositis, myalgia, epididymitis, and hearing loss weeks to months after recovery from acute infection. We demonstrate a systemic lymphoplasmacytic and histiocytic arteritis and periarteritis in guinea pigs more than 2 months after recovery from acute Lassa virus (LASV) infection. LASV was detected in the arterial tunica media smooth muscle cells by immunohistochemistry, in situ hybridization, and transmission electron microscopy. Our results suggest that the sequelae of LASV infection may be due to virus persistence resulting in systemic vascular damage. These findings shed light on the pathogenesis of LASV sequelae in convalescent human survivors.


Subject(s)
Lassa Fever/virology , Lassa virus/immunology , Animals , Convalescence , Disease Models, Animal , Disease Progression , Female , Guinea Pigs , Humans , Immunohistochemistry , Inflammation , Lassa Fever/pathology , Male
7.
Am J Pathol ; 188(3): 550-558, 2018 03.
Article in English | MEDLINE | ID: mdl-29429544

ABSTRACT

Sexual transmission of Ebola virus (EBOV) has been demonstrated more than a year after recovery from the acute phase of Ebola virus disease (EVD). The mechanisms underlying EBOV persistence and sexual transmission are not currently understood. Using the acute macaque model of EVD, we hypothesized EBOV would infect the reproductive tissues and sought to localize the infection in these tissues using immunohistochemistry and transmission electron microscopy. In four female and eight male macaques that succumbed to EVD between 6 and 9 days after EBOV challenge, we demonstrate widespread EBOV infection of the interstitial tissues and endothelium in the ovary, uterus, testis, seminal vesicle, epididymis, and prostate gland, with minimal associated tissue immune response or organ pathology. Given the widespread involvement of EBOV in the reproductive tracts of both male and female macaques, it is reasonable to surmise that our understanding of the mechanisms underlying sexual transmission of EVD and persistence of EBOV in immune-privileged sites would be facilitated by the development of a nonhuman primate model in which the macaques survived past the acute stage into convalescence.


Subject(s)
Ebolavirus/isolation & purification , Hemorrhagic Fever, Ebola/virology , Ovary/virology , Prostate/virology , Testis/virology , Uterus/virology , Animals , Female , Hemorrhagic Fever, Ebola/pathology , Macaca , Male , Ovary/pathology , Prostate/pathology , Testis/pathology , Uterus/pathology
8.
J Infect Dis ; 218(suppl_5): S423-S433, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30053050

ABSTRACT

Previously, several studies have been performed to delineate the development and progression of Marburg virus infection in nonhuman primates (NHPs), primarily to clarify the mechanisms of severe (fatal) disease. After the 2013-2016 Ebola virus disease (EVD) epidemic in Western Africa, there has been a reassessment of the available filovirus animal models and the utility of these to faithfully recapitulate human disease. The high lethality of the NHP models has raised doubts as to their ability to provide meaningful data for the full spectrum of disease observed in humans. Of particular interest are the etiologic and pathophysiologic mechanisms underlying postconvalescent sequelae observed in human survivors of EVD and Marburg virus disease (MVD). In the current study, we evaluated the lesions of MVD in NHPs; however, in contrast to previous studies, we focused on the potential for development of sequelae similar to those reported in human survivors of MVD and EVD. We found that during acute MVD in the macaque model, there is frequent inflammation of peripheral nerves, autonomic ganglia, and the iris of the eye. Furthermore, we demonstrate viral infection of the ocular ciliary body and retina, testis, epididymis, ovary, oviduct, uterine endometrium, prostate, and mammary gland. These findings are relevant for both development of postconvalescent sequelae and the natural transmission of virus.


Subject(s)
Marburg Virus Disease/pathology , Animals , Disease Models, Animal , Eye/pathology , Female , Ganglia/pathology , Humans , Macaca mulatta , Male , Mammary Glands, Human/pathology , Peripheral Nerves/pathology , Urogenital System/pathology
9.
J Infect Dis ; 218(suppl_5): S588-S591, 2018 11 22.
Article in English | MEDLINE | ID: mdl-29982632

ABSTRACT

No therapeutics are approved for the treatment of filovirus infections. Bepridil, a calcium channel blocker developed for treating angina, was identified as a potent inhibitor of filoviruses in vitro, including Ebola and Marburg viruses, and Ebola virus in vivo. We evaluated the efficacy of bepridil in a lethal mouse model of Marburg virus disease. A dose of 12 mg/kg bepridil once or twice daily resulted in 80% or 90% survival, respectively. These data confirm bepridil's broad-spectrum anti-filovirus activity warranting further investigation of bepridil, or improved compounds with a similar mechanism, as a pan-filovirus therapeutic agent.


Subject(s)
Bepridil/therapeutic use , Calcium Channel Blockers/therapeutic use , Marburg Virus Disease/drug therapy , Animals , Chlorocebus aethiops , Disease Models, Animal , Female , Marburg Virus Disease/mortality , Mice , Mice, Inbred BALB C , Vero Cells
10.
J Infect Dis ; 218(suppl_5): S592-S596, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30016444

ABSTRACT

At the onset of the 2013-2016 epidemic of Ebola virus disease (EVD), no vaccine or antiviral medication was approved for treatment. Therefore, considerable efforts were directed towards the concept of drug repurposing or repositioning. Amiodarone, an approved multi-ion channel blocker for the treatment of cardiac arrhythmia, was reported to inhibit filovirus entry in vitro. Compassionate use of amiodarone in EVD patients indicated a possible survival benefit. In support of further clinical testing, we confirmed anti-Ebola virus activity of amiodarone in different cell types. Despite promising in vitro results, amiodarone failed to protect guinea pigs from a lethal dose of Ebola virus.


Subject(s)
Amiodarone/pharmacology , Ebolavirus/drug effects , Amiodarone/pharmacokinetics , Amiodarone/therapeutic use , Animals , Chlorocebus aethiops , Female , Guinea Pigs , Hemorrhagic Fever, Ebola/drug therapy , Male , Vero Cells
11.
J Infect Dis ; 218(suppl_5): S636-S648, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30010950

ABSTRACT

Transchromosomic bovines (Tc-bovines) adaptively produce fully human polyclonal immunoglobulin (Ig)G antibodies after exposure to immunogenic antigen(s). The National Interagency Confederation for Biological Research and collaborators rapidly produced and then evaluated anti-Ebola virus IgG immunoglobulins (collectively termed SAB-139) purified from Tc-bovine plasma after sequential hyperimmunization with an Ebola virus Makona isolate glycoprotein nanoparticle vaccine. SAB-139 was characterized by several in vitro production, research, and clinical level assays using wild-type Makona-C05 or recombinant virus/antigens from different Ebola virus variants. SAB-139 potently activates natural killer cells, monocytes, and peripheral blood mononuclear cells and has high-binding avidity demonstrated by surface plasmon resonance. SAB-139 has similar concentrations of galactose-α-1,3-galactose carbohydrates compared with human-derived intravenous Ig, and the IgG1 subclass antibody is predominant. All rhesus macaques infected with Ebola virus/H.sapiens-tc/GIN/2014/Makona-C05 and treated with sufficient SAB-139 at 1 day (n = 6) or 3 days (n = 6) postinfection survived versus 0% of controls. This study demonstrates that Tc-bovines can produce pathogen-specific human Ig to prevent and/or treat patients when an emerging infectious disease either threatens to or becomes an epidemic.


Subject(s)
Antibodies, Viral/therapeutic use , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/drug therapy , Immunoglobulin G/therapeutic use , Animals , Cattle , Chlorocebus aethiops , Female , Humans , Macaca mulatta , Male , Vero Cells
12.
J Infect Dis ; 218(suppl_5): S672-S678, 2018 11 22.
Article in English | MEDLINE | ID: mdl-29939303

ABSTRACT

Background: A need to develop therapeutics to treat Ebola virus disease patients in remote and resource-challenged settings remains in the wake of the 2013-2016 epidemic in West Africa. Toward this goal, we screened drugs under consideration as treatment options and other drugs of interest, most being small molecules approved by the Food and Drug Administration. Drugs demonstrating in vitro antiviral activity were advanced for evaluation in combinations because of advantages often provided by drug cocktails. Methods: Drugs were screened for blockade of Ebola virus infection in cultured cells. Twelve drugs were tested in all (78 pair-wise) combinations, and 3 were tested in a subset of combinations. Results: Multiple synergistic drug pairs emerged, with the majority comprising 2 entry inhibitors. For the pairs of entry inhibitors studied, synergy was demonstrated at the level of virus entry into host cells. Highly synergistic pairs included aripiprazole/piperacetazine, sertraline/toremifene, sertraline/bepridil, and amodiaquine/clomiphene. Conclusions: Our study shows the feasibility of identifying pairs of approved drugs that synergistically block Ebola virus infection in cell cultures. We discuss our findings in terms of the theoretic ability of these or alternate combinations to reach therapeutic levels. Future research will assess selected combinations in small-animal models of Ebola virus disease.


Subject(s)
Antiviral Agents/administration & dosage , Ebolavirus/drug effects , Animals , Antiviral Agents/therapeutic use , Chlorocebus aethiops , Drug Approval , Drug Synergism , Drug Therapy, Combination , Vero Cells , Virion/drug effects , Virus Internalization/drug effects
13.
J Virol ; 91(2)2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27807241

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) emerged in 2012 and is a highly pathogenic respiratory virus. There are no treatment options against MERS-CoV for humans or animals, and there are no large-scale clinical trials for therapies against MERS-CoV. To address this need, we developed an inactivated rabies virus (RABV) that contains the MERS-CoV spike (S) protein expressed on its surface. Our initial recombinant vaccine, BNSP333-S, expresses a full-length wild-type MERS-CoV S protein; however, it showed significantly reduced viral titers compared to those of the parental RABV strain and only low-level incorporation of full-length MERS-CoV S into RABV particles. Therefore, we developed a RABV-MERS vector that contained the MERS-CoV S1 domain of the MERS-CoV S protein fused to the RABV G protein C terminus (BNSP333-S1). BNSP333-S1 grew to titers similar to those of the parental vaccine vector BNSP333, and the RABV G-MERS-CoV S1 fusion protein was efficiently expressed and incorporated into RABV particles. When we vaccinated mice, chemically inactivated BNSP333-S1 induced high-titer neutralizing antibodies. Next, we challenged both vaccinated mice and control mice with MERS-CoV after adenovirus transduction of the human dipeptidyl peptidase 4 (hDPP4) receptor and then analyzed the ability of mice to control MERS-CoV infection. Our results demonstrated that vaccinated mice were fully protected from the MERS-CoV challenge, as indicated by the significantly lower MERS-CoV titers and MERS-CoV and mRNA levels in challenged mice than those in unvaccinated controls. These data establish that an inactivated RABV-MERS S-based vaccine may be effective for use in animals and humans in areas where MERS-CoV is endemic. IMPORTANCE: Rabies virus-based vectors have been proven to be efficient dual vaccines against rabies and emergent infectious diseases such as Ebola virus. Here we show that inactivated rabies virus particles containing the MERS-CoV S1 protein induce potent immune responses against MERS-CoV and RABV. This novel vaccine is easy to produce and may be useful to protect target animals, such as camels, as well as humans from deadly MERS-CoV and RABV infections. Our results indicate that this vaccine approach can prevent disease, and the RABV-based vaccine platform may be a valuable tool for timely vaccine development against emerging infectious diseases.


Subject(s)
Coronavirus Infections/immunology , Cross Protection/immunology , Middle East Respiratory Syndrome Coronavirus/immunology , Rabies virus/immunology , Rabies/immunology , Viral Vaccines/immunology , Animals , Coronavirus Infections/prevention & control , Coronavirus Infections/virology , Disease Models, Animal , Gene Expression Regulation, Viral , Humans , Immunization , Mice , Microbial Interactions , Middle East Respiratory Syndrome Coronavirus/genetics , Rabies/prevention & control , Rabies/virology , Rabies virus/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Attenuated , Vaccines, Synthetic , Viral Proteins/genetics , Viral Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/adverse effects , Viral Vaccines/genetics , Virus Assembly
14.
J Virol ; 91(21)2017 11 01.
Article in English | MEDLINE | ID: mdl-28814515

ABSTRACT

Real-time bioimaging of infectious disease processes may aid countermeasure development and lead to an improved understanding of pathogenesis. However, few studies have identified biomarkers for monitoring infections using in vivo imaging. Previously, we demonstrated that positron emission tomography/computed tomography (PET/CT) imaging with [18F]-fluorodeoxyglucose (FDG) can monitor monkeypox disease progression in vivo in nonhuman primates (NHPs). In this study, we investigated [18F]-FDG-PET/CT imaging of immune processes in lymphoid tissues to identify patterns of inflammation in the monkepox NHP model and to determine the value of [18F]-FDG-PET/CT as a biomarker for disease and treatment outcomes. Quantitative analysis of [18F]-FDG-PET/CT images revealed differences between moribund and surviving animals at two sites vital to the immune response to viral infections, bone marrow and lymph nodes (LNs). Moribund NHPs demonstrated increased [18F]-FDG uptake in bone marrow 4 days postinfection compared to surviving NHPs. In surviving, treated NHPs, increase in LN volume correlated with [18F]-FDG uptake and peaked 10 days postinfection, while minimal lymphadenopathy and higher glycolytic activity were observed in moribund NHPs early in infection. Imaging data were supported by standard virology, pathology, and immunology findings. Even with the limited number of subjects, imaging was able to differentiate the difference between disease outcomes, warranting additional studies to demonstrate whether [18F]-FDG-PET/CT can identify other, subtler effects. Visualizing altered metabolic activity at sites involved in the immune response by [18F]-FDG-PET/CT imaging is a powerful tool for identifying key disease-specific time points and locations that are most relevant for pathogenesis and treatment.IMPORTANCE Positron emission tomography and computed tomography (PET/CT) imaging is a universal tool in oncology and neuroscience. The application of this technology to infectious diseases is far less developed. We used PET/CT imaging with [18F]-labeled fluorodeoxyglucose ([18F]-FDG) in monkeys after monkeypox virus exposure to monitor the immune response in lymphoid tissues. In lymph nodes of surviving monkeys, changes in [18F]-FDG uptake positively correlated with enlargement of the lymph nodes and peaked on day 10 postinfection. In contrast, the bone marrow and lymph nodes of nonsurvivors showed increased [18F]-FDG uptake by day 4 postinfection with minimal lymph node enlargement, indicating that elevated cell metabolic activity early after infection is predictive of disease outcome. [18F]-FDG-PET/CT imaging can provide real-time snapshots of metabolic activity changes in response to viral infections and identify key time points and locations most relevant for monitoring the development of pathogenesis and for potential treatment to be effective.


Subject(s)
Cytosine/analogs & derivatives , Fluorodeoxyglucose F18/metabolism , Lymphadenopathy/pathology , Lymphoid Tissue/pathology , Monkeypox virus/pathogenicity , Mpox (monkeypox)/pathology , Organophosphonates/pharmacology , Positron Emission Tomography Computed Tomography/methods , Animals , Antiviral Agents/pharmacology , Bone Marrow/diagnostic imaging , Bone Marrow/drug effects , Bone Marrow/pathology , Cidofovir , Cytosine/pharmacology , Lymphadenopathy/diagnostic imaging , Lymphoid Tissue/diagnostic imaging , Lymphoid Tissue/drug effects , Macaca mulatta/virology , Male , Mpox (monkeypox)/diagnostic imaging , Mpox (monkeypox)/drug therapy , Mpox (monkeypox)/virology , Prognosis , Radiopharmaceuticals/metabolism , Survival Rate
15.
J Virol ; 91(4)2017 02 15.
Article in English | MEDLINE | ID: mdl-27974564

ABSTRACT

Simian arteriviruses are a diverse clade of viruses infecting captive and wild nonhuman primates. We recently reported that Kibale red colobus virus 1 (KRCV-1) causes a mild and self-limiting disease in experimentally infected crab-eating macaques, while simian hemorrhagic fever virus (SHFV) causes lethal viral hemorrhagic fever. Here we characterize how these viruses evolved during replication in cell culture and in experimentally infected macaques. During passage in cell culture, 68 substitutions that were localized in open reading frames (ORFs) likely associated with host cell entry and exit became fixed in the KRCV-1 genome. However, we did not detect any strong signatures of selection during replication in macaques. We uncovered patterns of evolution that were distinct from those observed in surveys of wild red colobus monkeys, suggesting that these species may exert different adaptive challenges for KRCV-1. During SHFV infection, we detected signatures of selection on ORF 5a and on a small subset of sites in the genome. Overall, our data suggest that patterns of evolution differ markedly among simian arteriviruses and among host species. IMPORTANCE: Certain RNA viruses can cross species barriers and cause disease in new hosts. Simian arteriviruses are a diverse group of related viruses that infect captive and wild nonhuman primates, with associated disease severity ranging from apparently asymptomatic infections to severe, viral hemorrhagic fevers. We infected nonhuman primate cell cultures and then crab-eating macaques with either simian hemorrhagic fever virus (SHFV) or Kibale red colobus virus 1 (KRCV-1) and assessed within-host viral evolution. We found that KRCV-1 quickly acquired a large number of substitutions in its genome during replication in cell culture but that evolution in macaques was limited. In contrast, we detected selection focused on SHFV ORFs 5a and 5, which encode putative membrane proteins. These patterns suggest that in addition to diverse pathogenic phenotypes, these viruses may also exhibit distinct patterns of within-host evolution both in vitro and in vivo.


Subject(s)
Arterivirus Infections/veterinary , Arterivirus/physiology , Biological Evolution , Host-Pathogen Interactions , Monkey Diseases/virology , Animals , Host-Pathogen Interactions/genetics , Macaca fascicularis , Monkey Diseases/genetics , Open Reading Frames , Polymorphism, Single Nucleotide , RNA, Viral , Selection, Genetic , Virus Internalization , Virus Replication
16.
Curr Top Microbiol Immunol ; 411: 171-193, 2017.
Article in English | MEDLINE | ID: mdl-28643203

ABSTRACT

Ebola virus disease (EVD) in humans is associated with four ebolaviruses: Ebola virus (EBOV), Sudan virus (SUDV), Bundibugyo virus (BDBV), and Taï Forest virus. To date, no documented cases of human disease have been associated with Reston virus. Here, we describe the nonhuman primate (NHP) models that currently serve as gold standards for testing ebolavirus vaccines and therapeutic agents and elucidating underlying mechanisms of pathogenesis. Although multiple models have been explored over the past 50 years, the predominance of published work has been performed in macaque models. This chapter will focus on the most commonly used models.


Subject(s)
Disease Models, Animal , Ebolavirus , Hemorrhagic Fever, Ebola , Macaca/virology , Animals , Ebolavirus/classification , Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/therapy , Hemorrhagic Fever, Ebola/virology , Humans
17.
Virol J ; 15(1): 135, 2018 08 31.
Article in English | MEDLINE | ID: mdl-30165875

ABSTRACT

BACKGROUND: Currently, no FDA-approved vaccines or treatments are available for Ebola virus disease (EVD), and therapy remains largely supportive. Ebola virus (EBOV) has broad tissue tropism and can infect a variety of cells including epithelial cells. Epithelial cells differ from most other cell types by their polarized phenotype and barrier function. In polarized cells, the apical and basolateral membrane domains are demarcated by tight junctions, and specialized sorting machinery, which results in a difference in composition between the two membrane domains. These specialized sorting functions can have important consequences for viral infections. Differential localization of a viral receptor can restrict virus entry to a particular membrane while polarized sorting can lead to a vectorial virus release. The present study investigated the impact of cell polarity on EBOV infection. METHODS: Characteristics of EBOV infection in polarized cells were evaluated in the polarized Caco-2 model grown on semipermeable transwells. Transepithelial resistance (TEER), which is a function of tight junctions, was used to assess epithelial cell polarization. EBOV infection was assessed with immunofluorescence microscopy and qPCR. Statistical significance was calculated using one-way ANOVA and significance was set at p < 0.05. RESULTS: Our data indicate that EBOV preferentially infects cells from the basolateral route, and this preference may be influenced by the resistance across the Caco-2 monolayer. Infection occurs without changes in cellular permeability. Further, our data show that basolateral infection bias may be dependent on polarized distribution of heparan sulfate, a known viral attachment factor. Treatment with iota-carrageenan, or heparin lyase, which interrupts viral interaction with cellular heparan sulfate, significantly reduced cell susceptibility to basolateral infection, likely by inhibiting virus attachment. CONCLUSIONS: Our results show cell polarity has an impact on EBOV infection. EBOV preferentially infects polarized cells through the basolateral route. Access to heparan sulfate is an important factor during basolateral infection and blocking interaction of cellular heparan sulfate with virus leads to significant inhibition of basolateral infection in the polarized Caco-2 cell model.


Subject(s)
Ebolavirus/physiology , Epithelial Cells/virology , Heparitin Sulfate/metabolism , Virus Attachment , Caco-2 Cells , Humans , Microscopy, Fluorescence , Real-Time Polymerase Chain Reaction
18.
J Infect Dis ; 215(9): 1416-1420, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28368541

ABSTRACT

Previous studies have demonstrated little efficacy of interferons (IFNs) in animal models of Ebola virus disease. However, these studies were limited to a small number of type I IFNs and, during the most recent outbreak of Ebola virus, questions regarding the suitability of the animal models to evaluate IFNs were raised. To address the potential that anti-Ebola virus activity was overlooked, type I and type II IFNs (α-2a, α-2b, -ß, -γ, and -universal) were tested in a variety of cell types (Vero E6, Huh 7 cells, and human macrophages). IFNs are weak inhibitors of Ebola virus Makona in these cell lines.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Animals , Cell Line, Tumor , Chlorocebus aethiops , Drug Synergism , Humans , Vero Cells
19.
Mol Cell Proteomics ; 14(3): 646-57, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25573744

ABSTRACT

The Syrian golden hamster has been increasingly used to study viral hemorrhagic fever (VHF) pathogenesis and countermeasure efficacy. As VHFs are a global health concern, well-characterized animal models are essential for both the development of therapeutics and vaccines as well as for increasing our understanding of the molecular events that underlie viral pathogenesis. However, the paucity of reagents or platforms that are available for studying hamsters at a molecular level limits the ability to extract biological information from this important animal model. As such, there is a need to develop platforms/technologies for characterizing host responses of hamsters at a molecular level. To this end, we developed hamster-specific kinome peptide arrays to characterize the molecular host response of the Syrian golden hamster. After validating the functionality of the arrays using immune agonists of defined signaling mechanisms (lipopolysaccharide (LPS) and tumor necrosis factor (TNF)-α), we characterized the host response in a hamster model of VHF based on Pichinde virus (PICV(1)) infection by performing temporal kinome analysis of lung tissue. Our analysis revealed key roles for vascular endothelial growth factor (VEGF), interleukin (IL) responses, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, and Toll-like receptor (TLR) signaling in the response to PICV infection. These findings were validated through phosphorylation-specific Western blot analysis. Overall, we have demonstrated that hamster-specific kinome arrays are a robust tool for characterizing the species-specific molecular host response in a VHF model. Further, our results provide key insights into the hamster host response to PICV infection and will inform future studies with high-consequence VHF pathogens.


Subject(s)
Hemorrhagic Fever, American/virology , Lung/enzymology , Pichinde virus/physiology , Protein Kinases/isolation & purification , Proteome/analysis , Animals , Disease Models, Animal , Female , Hemorrhagic Fever, American/enzymology , Interleukins/isolation & purification , Lung/virology , Mesocricetus , NF-kappa B/isolation & purification , Phosphorylation , Signal Transduction , Species Specificity , Toll-Like Receptors/isolation & purification , Vascular Endothelial Growth Factor A/isolation & purification
20.
J Infect Dis ; 213(8): 1237-9, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26374912

ABSTRACT

Sexual transmission of Ebola virus in Liberia has now been documented and associated with new clusters in regions previously declared Ebola free. Assays that have Emergency Use Authorization (EUA) and are routinely used to detect Ebola virus RNA in whole blood and plasma specimens at the Liberian Institute for Biomedical Research were tested for their suitability in detecting the presence of Ebola virus RNA in semen. Qiagen AVL extraction protocols, as well as the Ebola Zaire Target 1 and major groove binder quantitative reverse-transcription polymerase chain reaction assays, were demonstrably suitable for this purpose and should facilitate epidemiologic investigations, including those involving long-term survivors of Ebola.


Subject(s)
Ebolavirus/genetics , Hemorrhagic Fever, Ebola/diagnosis , Reverse Transcriptase Polymerase Chain Reaction/methods , Semen/virology , Virology/methods , Hemorrhagic Fever, Ebola/virology , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL