Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 186
Filtrar
1.
Emerg Infect Dis ; 30(4): 721-731, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38526136

RESUMEN

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.


Asunto(s)
Arterivirus , Animales , Ratones , Arterivirus/genética , Macaca , Macrófagos , Línea Celular
2.
J Infect Dis ; 229(Supplement_2): S265-S274, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-37995376

RESUMEN

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.


Asunto(s)
Orthopoxvirus , Viruela , Virus de la Viruela , Humanos , Monkeypox virus , Monocitos
3.
Microbiol Spectr ; 11(3): e0349422, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37036346

RESUMEN

Marburg virus (MARV) is a highly virulent zoonotic filovirid that causes Marburg virus disease (MVD) in humans. The pathogenesis of MVD remains poorly understood, partially due to the low number of cases that can be studied, the absence of state-of-the-art medical equipment in areas where cases are reported, and limitations on the number of animals that can be safely used in experimental studies under maximum containment animal biosafety level 4 conditions. Medical imaging modalities, such as whole-body computed tomography (CT), may help to describe disease progression in vivo, potentially replacing ethically contentious and logistically challenging serial euthanasia studies. Towards this vision, we performed a pilot study, during which we acquired whole-body CT images of 6 rhesus monkeys before and 7 to 9 days after intramuscular MARV exposure. We identified imaging abnormalities in the liver, spleen, and axillary lymph nodes that corresponded to clinical, virological, and gross pathological hallmarks of MVD in this animal model. Quantitative image analysis indicated hepatomegaly with a significant reduction in organ density (indicating fatty infiltration of the liver), splenomegaly, and edema that corresponded with gross pathological and histopathological findings. Our results indicated that CT imaging could be used to verify and quantify typical MVD pathogenesis versus altered, diminished, or absent disease severity or progression in the presence of candidate medical countermeasures, thus possibly reducing the number of animals needed and eliminating serial euthanasia. IMPORTANCE Marburg virus (MARV) is a highly virulent zoonotic filovirid that causes Marburg virus disease (MVD) in humans. Much is unknown about disease progression and, thus, prevention and treatment options are limited. Medical imaging modalities, such as whole-body computed tomography (CT), have the potential to improve understanding of MVD pathogenesis. Our study used CT to identify abnormalities in the liver, spleen, and axillary lymph nodes that corresponded to known clinical signs of MVD in this animal model. Our results indicated that CT imaging and analyses could be used to elucidate pathogenesis and possibly assess the efficacy of candidate treatments.


Asunto(s)
Enfermedad del Virus de Marburg , Marburgvirus , Humanos , Animales , Enfermedad del Virus de Marburg/diagnóstico por imagen , Enfermedad del Virus de Marburg/patología , Proyectos Piloto , Tomografía Computarizada por Rayos X , Progresión de la Enfermedad , Primates
4.
Am J Pathol ; 192(1): 121-129, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34626576

RESUMEN

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.


Asunto(s)
Ebolavirus/fisiología , Hormonas/metabolismo , Hígado/virología , Tropismo/fisiología , Replicación Viral/fisiología , Animales , Células Cromafines/patología , Células Cromafines/virología , Modelos Animales de Enfermedad , Epidídimo/patología , Epidídimo/virología , Células Epiteliales/patología , Células Epiteliales/virología , Femenino , Hepatocitos/patología , Hepatocitos/virología , Macrófagos del Hígado/patología , Macrófagos del Hígado/virología , Macaca mulatta , Masculino , Cervicitis Uterina/patología , Cervicitis Uterina/virología , Vaginitis/patología , Vaginitis/virología
5.
Viruses ; 13(8)2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34452435

RESUMEN

Hemorrhagic smallpox, caused by variola virus (VARV), was a rare but nearly 100% lethal human disease manifestation. Hemorrhagic smallpox is frequently characterized by secondary bacterial infection, coagulopathy, and myocardial and subendocardial hemorrhages. Previous experiments have demonstrated that intravenous (IV) cowpox virus (CPXV) exposure of macaques mimics human hemorrhagic smallpox. The goal of this experiment was to further understand the onset, nature, and severity of cardiac pathology and how it may contribute to disease. The findings support an acute late-stage myocarditis with lymphohistiocytic infiltrates in the CPXV model of hemorrhagic smallpox.


Asunto(s)
Virus de la Viruela Vacuna/patogenicidad , Hemorragia/virología , Miocarditis/virología , Viruela/fisiopatología , Viruela/virología , Enfermedad Aguda , Animales , Modelos Animales de Enfermedad , Femenino , Macaca fascicularis/virología , Masculino , Miocarditis/veterinaria , Viruela/complicaciones
6.
Microorganisms ; 9(3)2021 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-33801811

RESUMEN

Outbreaks of Ebola ebolavirus (EBOV) have been associated with high morbidity and mortality. Milestones have been reached recently in the management of EBOV disease (EVD) with licensure of an EBOV vaccine and two monoclonal antibody therapies. However, neither vaccines nor therapies are available for other disease-causing filoviruses. In preparation for such outbreaks, and for more facile and cost-effective management of EVD, we seek a cocktail containing orally available and room temperature stable drugs with strong activity against multiple filoviruses. We previously showed that (bepridil + sertraline) and (sertraline + toremifene) synergistically suppress EBOV in cell cultures. Here, we describe steps towards testing these combinations in a mouse model of EVD. We identified a vehicle suitable for oral delivery of the component drugs and determined that, thus formulated the drugs are equally active against EBOV as preparations in DMSO, and they maintain activity upon storage in solution for up to seven days. Pharmacokinetic (PK) studies indicated that the drugs in the oral delivery vehicle are well tolerated in mice at the highest doses tested. Collectively the data support advancement of these combinations to tests for synergy in a mouse model of EVD. Moreover, mathematical modeling based on human oral PK projects that the combinations would be more active in humans than their component single drugs.

7.
Viruses ; 13(4)2021 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-33917085

RESUMEN

Simian hemorrhagic fever virus (SHFV) causes acute, lethal disease in macaques. We developed a single-plasmid cDNA-launch infectious clone of SHFV (rSHFV) and modified the clone to rescue an enhanced green fluorescent protein-expressing rSHFV-eGFP that can be used for rapid and quantitative detection of infection. SHFV has a narrow cell tropism in vitro, with only the grivet MA-104 cell line and a few other grivet cell lines being susceptible to virion entry and permissive to infection. Using rSHFV-eGFP, we demonstrate that one cricetid rodent cell line and three ape cell lines also fully support SHFV replication, whereas 55 human cell lines, 11 bat cell lines, and three rodent cells do not. Interestingly, some human and other mammalian cell lines apparently resistant to SHFV infection are permissive after transfection with the rSHFV-eGFP cDNA-launch plasmid. To further demonstrate the investigative potential of the infectious clone system, we introduced stop codons into eight viral open reading frames (ORFs). This approach suggested that at least one ORF, ORF 2b', is dispensable for SHFV in vitro replication. Our proof-of-principle experiments indicated that rSHFV-eGFP is a useful tool for illuminating the understudied molecular biology of SHFV.


Asunto(s)
Arterivirus/genética , ADN Complementario/genética , Proteínas Fluorescentes Verdes/genética , Sistemas de Lectura Abierta , ARN Viral/genética , Recombinación Genética , Replicación Viral/genética , Animales , Arterivirus/fisiología , Línea Celular , Quirópteros , Hominidae , Humanos , Plásmidos/genética , Prueba de Estudio Conceptual , Roedores
8.
Viruses ; 12(12)2020 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-33327396

RESUMEN

We report the discovery and sequence-based molecular characterization of a novel virus, lanama virus (LNMV), in blood samples obtained from two wild vervet monkeys (Chlorocebus pygerythrus), sampled near Lake Nabugabo, Masaka District, Uganda. Sequencing of the complete viral genomes and subsequent phylogenetic analysis identified LNMV as a distinct member of species Kunsagivirus C, in the undercharacterized picornavirid genus Kunsagivirus.


Asunto(s)
Chlorocebus aethiops/virología , Enfermedades de los Monos/virología , Infecciones por Picornaviridae/veterinaria , Picornaviridae/clasificación , Animales , Genoma Viral , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Filogenia
9.
Proc Natl Acad Sci U S A ; 117(48): 30687-30698, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33184176

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Coronavirus/efectos de los fármacos , Filoviridae/efectos de los fármacos , Interacciones Huésped-Patógeno/efectos de los fármacos , Complejos Multiproteicos/metabolismo , Orthomyxoviridae/efectos de los fármacos , Línea Celular , Genes Supresores , Modelos Moleculares , Terapia Molecular Dirigida , Unión Proteica , ARN Interferente Pequeño/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Saccharomyces cerevisiae/genética , Proteínas Virales/metabolismo , Replicación Viral/efectos de los fármacos
10.
Viruses ; 12(8)2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32717786

RESUMEN

Over the last 15 years, advances in immunofluorescence-imaging based cycling methods, antibody conjugation methods, and automated image processing have facilitated the development of a high-resolution, multiplexed tissue immunofluorescence (MxIF) method with single cell-level quantitation termed Cell DIVETM. Originally developed for fixed oncology samples, here it was evaluated in highly fixed (up to 30 days), archived monkeypox virus-induced inflammatory skin lesions from a retrospective study in 11 rhesus monkeys to determine whether MxIF was comparable to manual H-scoring of chromogenic stains. Six protein markers related to immune and cellular response (CD68, CD3, Hsp70, Hsp90, ERK1/2, ERK1/2 pT202_pY204) were manually quantified (H-scores) by a pathologist from chromogenic IHC double stains on serial sections and compared to MxIF automated single cell quantification of the same markers that were multiplexed on a single tissue section. Overall, there was directional consistency between the H-score and the MxIF results for all markers except phosphorylated ERK1/2 (ERK1/2 pT202_pY204), which showed a decrease in the lesion compared to the adjacent non-lesioned skin by MxIF vs an increase via H-score. Improvements to automated segmentation using machine learning and adding additional cell markers for cell viability are future options for improvement. This method could be useful in infectious disease research as it conserves tissue, provides marker colocalization data on thousands of cells, allowing further cell level data mining as well as a reduction in user bias.


Asunto(s)
Técnica del Anticuerpo Fluorescente/métodos , Inmunohistoquímica/métodos , Mpox/patología , Piel/virología , Animales , Biomarcadores/análisis , Compuestos Cromogénicos , Femenino , Procesamiento de Imagen Asistido por Computador , Macaca mulatta , Masculino , Monkeypox virus/patogenicidad , Estudios Retrospectivos , Análisis de la Célula Individual , Piel/patología , Coloración y Etiquetado
11.
Viruses ; 12(7)2020 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-32674252

RESUMEN

Ongoing Ebola virus disease outbreaks in the Democratic Republic of the Congo follow the largest recorded outbreak in Western Africa (2013-2016). To combat outbreaks, testing of medical countermeasures (therapeutics or vaccines) requires a well-defined, reproducible, animal model. Here we present Ebola virus disease kinetics in 24 Chinese-origin rhesus monkeys exposed intramuscularly to a highly characterized, commercially available Kikwit Ebola virus Filovirus Animal Non-Clinical Group (FANG) stock. Until reaching predetermined clinical disease endpoint criteria, six animals underwent anesthesia for repeated clinical sampling and were compared to six that did not. Groups of three animals were euthanized and necropsied on days 3, 4, 5, and 6 post-exposure, respectively. In addition, three uninfected animals served as controls. Here, we present detailed characterization of clinical and laboratory disease kinetics and complete blood counts, serum chemistries, Ebola virus titers, and disease kinetics for future medical countermeasure (MCM) study design and control data. We measured no statistical difference in hematology, chemistry values, or time to clinical endpoint in animals that were anesthetized for clinical sampling during the acute disease compared to those that were not.


Asunto(s)
Modelos Animales de Enfermedad , Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/fisiopatología , Macaca mulatta , Animales , Progresión de la Enfermedad , Ebolavirus/clasificación , Femenino , Masculino , Carga Viral , Viremia
12.
Am J Pathol ; 190(9): 1867-1880, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32479821

RESUMEN

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.


Asunto(s)
Infecciones por Filoviridae/virología , Sinoviocitos/virología , Animales , Células Cultivadas , Filoviridae , Humanos , Macaca mulatta
13.
Expert Rev Vaccines ; 19(1): 85-103, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31937163

RESUMEN

Introduction: The development of therapeutics and vaccines to combat Risk Group 4 pathogens, which are associated with high case-fatality rates, is a high priority. Postexposure prophylactic vaccines have the potential to bridge classical therapeutic and vaccine applications, but little progress has been reported to date.Areas covered: This review provides an overview of postexposure prophylactic vaccine candidates against Risk Group 4 pathogens.Expert opinion: A few candidate postexposure prophylactic vaccines protect experimental animals infected with a few Risk Group 4 pathogens, such as filoviruses or hantaviruses, but the efficacy of candidate vaccines has not been similarly reported for most other high-consequence pathogens. A major drawback for the further development of existing candidates is the lack of understanding of their mechanisms of action, knowledge of which could help to identify focused paths forward in vaccine development and licensure. These drawbacks to further development ultimately slow progress toward postexposure prophylactic vaccine licensure.


Asunto(s)
Profilaxis Posexposición/métodos , Vacunas Virales/administración & dosificación , Virosis/prevención & control , Animales , Humanos , Vacunación , Vacunas Virales/inmunología , Virosis/inmunología , Virosis/virología
14.
Sci Rep ; 9(1): 20199, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31882748

RESUMEN

During the Ebola virus disease (EVD) epidemic in Western Africa (2013‒2016), antimalarial treatment was administered to EVD patients due to the high coexisting malaria burden in accordance with World Health Organization guidelines. In an Ebola treatment center in Liberia, EVD patients receiving the combination antimalarial artesunate-amodiaquine had a lower risk of death compared to those treated with artemether-lumefantrine. As artemether and artesunate are derivatives of artemisinin, the beneficial anti-Ebola virus (EBOV) effect observed could possibly be attributed to the change from lumefantrine to amodiaquine. Amodiaquine is a widely used antimalarial in the countries that experience outbreaks of EVD and, therefore, holds promise as an approved drug that could be repurposed for treating EBOV infections. We investigated the potential anti-EBOV effect of amodiaquine in a well-characterized nonhuman primate model of EVD. Using a similar 3-day antimalarial dosing strategy as for human patients, plasma concentrations of amodiaquine in healthy animals were similar to those found in humans. However, the treatment regimen did not result in a survival benefit or decrease of disease signs in EBOV-infected animals. While amodiaquine on its own failed to demonstrate efficacy, we cannot exclude potential therapeutic value of amodiaquine when used in combination with artesunate or another antiviral.


Asunto(s)
Amodiaquina/uso terapéutico , Antivirales/uso terapéutico , Artemisininas/uso terapéutico , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Combinación de Medicamentos , Femenino , Macaca mulatta , Masculino
16.
PLoS One ; 14(8): e0221407, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31454374

RESUMEN

Antibody titers against a viral pathogen are typically measured using an antigen binding assay, such as an enzyme-linked immunosorbent assay (ELISA), which only measures the ability of antibodies to identify a viral antigen of interest. Neutralization assays measure the presence of virus-neutralizing antibodies in a sample. Traditional neutralization assays, such as the plaque reduction neutralization test (PRNT), are often difficult to use on a large scale due to being both labor and resource intensive. Here we describe an Ebola virus fluorescence reduction neutralization assay (FRNA), which tests for neutralizing antibodies, that requires only a small volume of sample in a 96-well format and is easy to automate. The readout of the FRNA is the percentage of Ebola virus-infected cells measured with an optical reader or overall chemiluminescence that can be generated by multiple reading platforms. Using blinded human clinical samples (EVD survivors or contacts) obtained in Liberia during the 2013-2016 Ebola virus disease outbreak, we demonstrate there was a high degree of agreement between the FRNA-measured antibody titers and the Filovirus Animal Non-clinical Group (FANG) ELISA titers with the FRNA providing information on the neutralizing capabilities of the antibodies.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Animales , Anticuerpos Bloqueadores/inmunología , Chlorocebus aethiops , Brotes de Enfermedades , Ebolavirus/patogenicidad , Ensayo de Inmunoadsorción Enzimática , Fiebre Hemorrágica Ebola/prevención & control , Fiebre Hemorrágica Ebola/virología , Humanos , Liberia , Pruebas de Neutralización/métodos , Células Vero
17.
PLoS Negl Trop Dis ; 13(7): e0007595, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31356611

RESUMEN

Ebolaviruses cause an often rapidly fatal syndrome known as Ebola virus disease (EVD), with average case fatality rates of ~50%. There is no licensed vaccine or treatment for EVD, underscoring the urgent need to develop new anti-ebolavirus agents, especially in the face of an ongoing outbreak in the Democratic Republic of the Congo and the largest ever outbreak in Western Africa in 2013-2016. Lectins have been investigated as potential antiviral agents as they bind glycans present on viral surface glycoproteins, but clinical use of them has been slowed by concerns regarding their mitogenicity, i.e. ability to cause immune cell proliferation. We previously engineered a banana lectin (BanLec), a carbohydrate-binding protein, such that it retained antiviral activity but lost mitogenicity by mutating a single amino acid, yielding H84T BanLec (H84T). H84T shows activity against viruses containing high-mannose N-glycans, including influenza A and B, HIV-1 and -2, and hepatitis C virus. Since ebolavirus surface glycoproteins also contain many high-mannose N-glycans, we assessed whether H84T could inhibit ebolavirus replication. H84T inhibited Ebola virus (EBOV) replication in cell cultures. In cells, H84T inhibited both virus-like particle (VLP) entry and transcription/replication of the EBOV mini-genome at high micromolar concentrations, while inhibiting infection by transcription- and replication-competent VLPs, which measures the full viral life cycle, in the low micromolar range. H84T did not inhibit assembly, budding, or release of VLPs. These findings suggest that H84T may exert its anti-ebolavirus effect(s) by blocking both entry and transcription/replication. In a mouse model, H84T partially (maximally, ~50-80%) protected mice from an otherwise lethal mouse-adapted EBOV infection. Interestingly, a single dose of H84T pre-exposure to EBOV protected ~80% of mice. Thus, H84T shows promise as a new anti-ebolavirus agent with potential to be used in combination with vaccination or other agents in a prophylactic or therapeutic regimen.


Asunto(s)
Antivirales/farmacología , Ebolavirus/efectos de los fármacos , Musa/química , Lectinas de Plantas/farmacología , Animales , Antivirales/síntesis química , Línea Celular Tumoral , Ebolavirus/genética , Ebolavirus/inmunología , Escherichia coli , Femenino , Ingeniería Genética , Células HEK293 , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Fiebre Hemorrágica Ebola/prevención & control , Humanos , Región Variable de Inmunoglobulina/genética , Ratones , Ratones Endogámicos C57BL , Lectinas de Plantas/síntesis química , Replicación Viral/efectos de los fármacos
18.
PLoS Negl Trop Dis ; 13(6): e0007454, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31166946

RESUMEN

The ability to appropriately mimic human disease is critical for using animal models as a tool for understanding virus pathogenesis. In the case of Nipah virus (NiV), infection of humans appears to occur either through inhalation, contact with or consumption of infected material. In two of these circumstances, respiratory or sinusoidal exposure represents a likely route of infection. In this study, intermediate-size aerosol particles (~7 µm) of NiV-Malaysia were used to mimic potential routes of exposure by focusing viral deposition in the upper respiratory tract. Our previous report showed this route of exposure extended the disease course and a single animal survived the infection. Here, analysis of the peripheral immune response found minimal evidence of systemic inflammation and depletion of B cells during acute disease. However, the animal that survived infection developed an early IgM response with rapid development of neutralizing antibodies that likely afforded protection. The increase in NiV-specific antibodies correlated with an expansion of the B cell population in the survivor. Cell-mediated immunity was not clearly apparent in animals that succumbed during the acute phase of disease. However, CD4+ and CD8+ effector memory cells increased in the survivor with correlating increases in cytokines and chemokines associated with cell-mediated immunity. Interestingly, kinetic changes of the CD4+ and CD8bright T cell populations over the course of acute disease were opposite from animals that succumbed to infection. In addition, increases in NK cells and basophils during convalescence of the surviving animal were also evident, with viral antigen found in NK cells. These data suggest that a systemic inflammatory response and "cytokine storm" are not major contributors to NiV-Malaysia pathogenesis in the AGM model using this exposure route. Further, these data demonstrate that regulation of cell-mediated immunity, in addition to rapid production of NiV specific antibodies, may be critical for surviving NiV infection.


Asunto(s)
Aerosoles , Infecciones por Henipavirus/inmunología , Inmunidad Humoral , Exposición por Inhalación , Virus Nipah/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Chlorocebus aethiops , Citocinas/análisis , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad Celular , Inmunoglobulina M/sangre , Células Asesinas Naturales/inmunología , Masculino
19.
Viruses ; 11(2)2019 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-30781518

RESUMEN

Following the largest Ebola virus disease outbreak from 2013 to 2016, viral RNA has been detected in survivors from semen and breast milk long after disease recovery. However, as there have been few cases of sexual transmission, it is unclear whether every RNA positive fluid sample contains infectious virus. Virus isolation, typically using cell culture or animal models, can serve as a tool to determine the infectivity of patient samples. However, the sensitivity of these methods has not been assessed for the Ebola virus isolate, Makona. Described here is an efficiency comparison of Ebola virus Makona isolation using Vero E6, Huh-7, monocyte-derived macrophage cells, and suckling laboratory mice. Isolation sensitivity was similar in all methods tested. Laboratory mice and Huh-7 cells were less affected by toxicity from breast milk than Vero E6 and MDM cells. However, the advantages associated with isolation in Huh-7 cells over laboratory mice, including cost effectiveness, sample volume preservation, and a reduction in animal use, make Huh-7 cells the preferred substrate tested for Ebola virus Makona isolation.


Asunto(s)
Ebolavirus/aislamiento & purificación , Virología/métodos , Animales , Animales Lactantes , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Fiebre Hemorrágica Ebola/virología , Humanos , Macrófagos/virología , Ratones , Ratones Endogámicos BALB C , Leche Humana/virología , Semen/virología , Sensibilidad y Especificidad , Células Vero
20.
Viruses ; 11(1)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30650570

RESUMEN

Simian hemorrhagic fever virus (SHFV) causes a fulminant and typically lethal viral hemorrhagic fever (VHF) in macaques (Cercopithecinae: Macaca spp.) but causes subclinical infections in patas monkeys (Cercopithecinae: Erythrocebus patas). This difference in disease course offers a unique opportunity to compare host responses to infection by a VHF-causing virus in biologically similar susceptible and refractory animals. Patas and rhesus monkeys were inoculated side-by-side with SHFV. Unlike the severe disease observed in rhesus monkeys, patas monkeys developed a limited clinical disease characterized by changes in complete blood counts, serum chemistries, and development of lymphadenopathy. Viral RNA was measurable in circulating blood 2 days after exposure, and its duration varied by species. Infectious virus was detected in terminal tissues of both patas and rhesus monkeys. Varying degrees of overlap in changes in serum concentrations of interferon (IFN)-γ, monocyte chemoattractant protein (MCP)-1, and interleukin (IL)-6 were observed between patas and rhesus monkeys, suggesting the presence of common and species-specific cytokine responses to infection. Similarly, quantitative immunohistochemistry of livers from terminal monkeys and whole blood flow cytometry revealed varying degrees of overlap in changes in macrophages, natural killer cells, and T-cells. The unexpected degree of overlap in host response suggests that relatively small subsets of a host's response to infection may be responsible for driving hemorrhagic fever pathogenesis. Furthermore, comparative SHFV infection in patas and rhesus monkeys offers an experimental model to characterize host⁻response mechanisms associated with viral hemorrhagic fever and evaluate pan-viral hemorrhagic fever countermeasures.


Asunto(s)
Infecciones por Arterivirus/veterinaria , Arterivirus/patogenicidad , Fiebres Hemorrágicas Virales/veterinaria , Interacciones Huésped-Patógeno , Enfermedades de los Monos/inmunología , Animales , Anticuerpos Antivirales/sangre , Arterivirus/inmunología , Infecciones por Arterivirus/inmunología , Citocinas/sangre , Erythrocebus , Femenino , Fiebres Hemorrágicas Virales/inmunología , Macaca , Macrófagos/virología , Masculino , Enfermedades de los Monos/virología , ARN Viral , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA