Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
1.
Clin Infect Dis ; 78(4): 870-879, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-37967326

RESUMEN

BACKGROUND: The rVSVΔG-ZEBOV-GP vaccine (ERVEBO®) is a single-dose, live-attenuated, recombinant vesicular stomatitis virus vaccine indicated for the prevention of Ebola virus disease (EVD) caused by Zaire ebolavirus in individuals 12 months of age and older. METHODS: The Partnership for Research on Ebola VACcination (PREVAC) is a multicenter, phase 2, randomized, double-blind, placebo-controlled trial of 3 vaccine strategies in healthy children (ages 1-17) and adults, with projected 5 years of follow-up (NCT02876328). Using validated assays (GP-ELISA and PRNT), we measured antibody responses after 1-dose rVSVΔG-ZEBOV-GP, 2-dose rVSVΔG-ZEBOV-GP (given on Day 0 and Day 56), or placebo. Furthermore, we quantified vaccine virus shedding in a subset of children's saliva using RT-PCR. RESULTS: In total, 819 children and 783 adults were randomized to receive rVSVΔG-ZEBOV-GP (1 or 2 doses) or placebo. A single dose of rVSVΔG-ZEBOV-GP increased antibody responses by Day 28 that were sustained through Month 12. A second dose of rVSVΔG-ZEBOV-GP given on Day 56 transiently boosted antibody concentrations. In vaccinated children, GP-ELISA titers were superior to placebo and non-inferior to vaccinated adults. Vaccine virus shedding was observed in 31.7% of children, peaking by Day 7, with no shedding observed after Day 28 post-dose 1 or any time post-dose 2. CONCLUSIONS: A single dose of rVSVΔG-ZEBOV-GP induced robust antibody responses in children that was non-inferior to the responses induced in vaccinated adults. Vaccine virus shedding in children was time-limited and only observed after the first dose. Overall, these data support the use of rVSVΔG-ZEBOV-GP for the prevention of EVD in at-risk children. Clinical Trials Registration. The study is registered at ClinicalTrials.gov (NCT02876328), the Pan African Clinical Trials Registry (PACTR201712002760250), and the European Clinical Trials Register (EudraCT number: 2017-001798-18).


Asunto(s)
Vacunas contra el Virus del Ébola , Ebolavirus , Fiebre Hemorrágica Ebola , Adulto , Niño , Humanos , Anticuerpos Antivirales , Proteínas del Envoltorio Viral , Vacunas Sintéticas , Vacunación/métodos , Vacunas Atenuadas , Inmunogenicidad Vacunal
2.
Am J Pathol ; 193(12): 2031-2046, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37689386

RESUMEN

The pathophysiology of long-recognized hematologic abnormalities in Ebolavirus (EBOV) disease (EVD) is unknown. From limited human sampling (of peripheral blood), it has been postulated that emergency hematopoiesis plays a role in severe EVD, but the systematic characterization of the bone marrow (BM) has not occurred in human disease or in nonhuman primate models. In a lethal rhesus macaque model of EVD, 18 sternal BM samples exposed to the Kikwit strain of EBOV were compared to those from uninfected controls (n = 3). Immunohistochemistry, RNAscope in situ hybridization, transmission electron microscopy, and confocal microscopy showed that EBOV infects BM monocytes/macrophages and megakaryocytes. EBOV exposure was associated with severe BM hypocellularity, including depletion of myeloid, erythroid, and megakaryocyte hematopoietic cells. These depletions were negatively correlated with cell proliferation (Ki67 expression) and were not associated with BM apoptosis during disease progression. In EBOV-infected rhesus macaques with terminal disease, BM showed marked hemophagocytosis, megakaryocyte emperipolesis, and the release of immature hematopoietic cells into the sinusoids. Collectively, these data demonstrate not only direct EBOV infection of BM monocytes/macrophages and megakaryocytes but also that disease progression is associated with hematopoietic failure, notably in peripheral cytopenia. These findings inform current pathophysiologic unknowns and suggest a crucial role for BM dysfunction and/or failure, including emergency hematopoiesis, as part of the natural history of severe human disease.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Animales , Humanos , Ebolavirus/fisiología , Macaca mulatta , Médula Ósea , Progresión de la Enfermedad
3.
J Infect Dis ; 228(4): 371-382, 2023 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-37279544

RESUMEN

BACKGROUND: Ebola virus (EBOV) disease (EVD) is one of the most severe and fatal viral hemorrhagic fevers and appears to mimic many clinical and laboratory manifestations of hemophagocytic lymphohistiocytosis syndrome (HLS), also known as macrophage activation syndrome. However, a clear association is yet to be firmly established for effective host-targeted, immunomodulatory therapeutic approaches to improve outcomes in patients with severe EVD. METHODS: Twenty-four rhesus monkeys were exposed intramuscularly to the EBOV Kikwit isolate and euthanized at prescheduled time points or when they reached the end-stage disease criteria. Three additional monkeys were mock-exposed and used as uninfected controls. RESULTS: EBOV-exposed monkeys presented with clinicopathologic features of HLS, including fever, multiple organomegaly, pancytopenia, hemophagocytosis, hyperfibrinogenemia with disseminated intravascular coagulation, hypertriglyceridemia, hypercytokinemia, increased concentrations of soluble CD163 and CD25 in serum, and the loss of activated natural killer cells. CONCLUSIONS: Our data suggest that EVD in the rhesus macaque model mimics pathophysiologic features of HLS/macrophage activation syndrome. Hence, regulating inflammation and immune function might provide an effective treatment for controlling the pathogenesis of acute EVD.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Linfohistiocitosis Hemofagocítica , Síndrome de Activación Macrofágica , Animales , Síndrome de Activación Macrofágica/terapia , Macaca mulatta
4.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-35110410

RESUMEN

Despite more than 300,000 rVSVΔG-ZEBOV-glycoprotein (GP) vaccine doses having been administered during Ebola virus disease (EVD) outbreaks in the Democratic Republic of the Congo (DRC) between 2018 and 2020, seroepidemiologic studies of vaccinated Congolese populations are lacking. This study examines the antibody response at 21 d and 6 mo postvaccination after single-dose rVSVΔG-ZEBOV-GP vaccination among EVD-exposed and potentially exposed populations in the DRC. We conducted a longitudinal cohort study of 608 rVSVΔG-ZEBOV-GP-vaccinated individuals during an EVD outbreak in North Kivu Province, DRC. Participants provided questionnaires and blood samples at three study visits (day 0, visit 1; day 21, visit 2; and month 6, visit 3). Anti-GP immunoglobulin G (IgG) antibody titers were measured in serum by the Filovirus Animal Nonclinical Group anti-Ebola virus GP IgG enzyme-linked immunosorbent assay. Antibody response was defined as an antibody titer that had increased fourfold from visit 1 to visit 2 and was above four times the lower limit of quantification at visit 2; antibody persistence was defined as a similar increase from visit 1 to visit 3. We then examined demographics for associations with follow-up antibody titers using generalized linear mixed models. A majority of the sample, 87.2%, had an antibody response at visit 2, and 95.6% demonstrated antibody persistence at visit 3. Being female and of young age was predictive of a higher antibody titer postvaccination. Antibody response and persistence after Ebola vaccination was robust in this cohort, confirming findings from outside of the DRC.


Asunto(s)
Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Inmunogenicidad Vacunal/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Antivirales/inmunología , Niño , República Democrática del Congo , Brotes de Enfermedades/prevención & control , Femenino , Glicoproteínas/inmunología , Humanos , Masculino , Persona de Mediana Edad , Estudios Seroepidemiológicos , Vacunación/métodos , Proteínas del Envoltorio Viral/inmunología , Adulto Joven
5.
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
6.
Acad Radiol ; 28 Suppl 1: S37-S44, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-32943333

RESUMEN

With the advent of deep learning, convolutional neural networks (CNNs) have evolved as an effective method for the automated segmentation of different tissues in medical image analysis. In certain infectious diseases, the liver is one of the more highly affected organs, where an accurate liver segmentation method may play a significant role to improve the diagnosis, quantification, and follow-up. Although several segmentation algorithms have been proposed for liver or liver-tumor segmentation in computed tomography (CT) of human subjects, none of them have been investigated for nonhuman primates (NHPs), where the livers have a wide range in size and morphology. In addition, the unique characteristics of different infections or the heterogeneous immune responses of different NHPs to the infections appear with a diverse radiodensity distribution in the CT imaging. In this study, we investigated three state-of-the-art algorithms; VNet, UNet, and feature pyramid network (FPN) for automated liver segmentation in whole-body CT images of NHPs. The efficacy of the CNNs were evaluated on 82 scans of 37 animals, including pre and post-exposure to different viruses such as Ebola, Marburg, and Lassa. Using a 10-fold cross-validation, the best performance for the segmented liver was provided by the FPN; an average 94.77% Dice score, and 3.6% relative absolute volume difference. Our study demonstrated the efficacy of multiple CNNs, wherein the FPN outperforms VNet and UNet for liver segmentation in infectious disease imaging research.


Asunto(s)
Enfermedades Transmisibles , Aprendizaje Profundo , Animales , Procesamiento de Imagen Asistido por Computador , Hígado/diagnóstico por imagen , Primates
7.
Cell ; 183(5): 1383-1401.e19, 2020 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-33159858

RESUMEN

Ebola virus (EBOV) causes epidemics with high mortality yet remains understudied due to the challenge of experimentation in high-containment and outbreak settings. Here, we used single-cell transcriptomics and CyTOF-based single-cell protein quantification to characterize peripheral immune cells during EBOV infection in rhesus monkeys. We obtained 100,000 transcriptomes and 15,000,000 protein profiles, finding that immature, proliferative monocyte-lineage cells with reduced antigen-presentation capacity replace conventional monocyte subsets, while lymphocytes upregulate apoptosis genes and decline in abundance. By quantifying intracellular viral RNA, we identify molecular determinants of tropism among circulating immune cells and examine temporal dynamics in viral and host gene expression. Within infected cells, EBOV downregulates STAT1 mRNA and interferon signaling, and it upregulates putative pro-viral genes (e.g., DYNLL1 and HSPA5), nominating pathways the virus manipulates for its replication. This study sheds light on EBOV tropism, replication dynamics, and elicited immune response and provides a framework for characterizing host-virus interactions under maximum containment.


Asunto(s)
Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/genética , Fiebre Hemorrágica Ebola/virología , Interacciones Huésped-Patógeno/genética , Análisis de la Célula Individual , Animales , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Efecto Espectador , Diferenciación Celular , Proliferación Celular , Citocinas/metabolismo , Ebolavirus/genética , Chaperón BiP del Retículo Endoplásmico , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Regulación Viral de la Expresión Génica , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/patología , Antígenos de Histocompatibilidad Clase II/metabolismo , Interferones/genética , Interferones/metabolismo , Macaca mulatta , Macrófagos/metabolismo , Monocitos/metabolismo , Mielopoyesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo , Transcriptoma/genética
8.
J Infect Dis ; 222(10): 1745-1755, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-32498080

RESUMEN

Neurological signs and symptoms are the most common complications of Ebola virus disease. However, the mechanisms underlying the neurologic manifestations in Ebola patients are not known. In this study, peripheral ganglia were collected from 12 rhesus macaques that succumbed to Ebola virus (EBOV) disease from 5 to 8 days post exposure. Ganglionitis, characterized by neuronal degeneration, necrosis, and mononuclear leukocyte infiltrates, was observed in the dorsal root, autonomic, and enteric ganglia. By immunohistochemistry, RNAscope in situ hybridization, transmission electron microscopy, and confocal microscopy, we confirmed that CD68+ macrophages are the target cells for EBOV in affected ganglia. Further, we demonstrated that EBOV can induce satellite cell and neuronal apoptosis and microglial activation in infected ganglia. Our results demonstrate that EBOV can infect peripheral ganglia and results in ganglionopathy in rhesus macaques, which may contribute to the neurological signs and symptoms observed in acute and convalescent Ebola virus disease in human patients.


Asunto(s)
Fiebre Hemorrágica Ebola/complicaciones , Fiebre Hemorrágica Ebola/patología , Degeneración Nerviosa/complicaciones , Degeneración Nerviosa/patología , Enfermedades del Sistema Nervioso Periférico/complicaciones , Enfermedades del Sistema Nervioso Periférico/patología , Animales , Antígenos CD , Antígenos de Diferenciación Mielomonocítica , Modelos Animales de Enfermedad , Ebolavirus , Femenino , Ganglios , Ganglios Espinales/patología , Ganglios Espinales/virología , Ganglión/patología , Fiebre Hemorrágica Ebola/virología , Humanos , Inmunohistoquímica , Leucocitos Mononucleares , Macaca mulatta , Macrófagos/patología , Masculino , Microglía/patología , Microglía/virología , Necrosis , Sistema Nervioso Parasimpático/patología , Enfermedades del Sistema Nervioso Periférico/virología , Células Receptoras Sensoriales/patología , Células Receptoras Sensoriales/virología , Sistema Nervioso Simpático/patología
9.
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
10.
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
11.
PLoS One ; 13(3): e0194868, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29566060

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) presents an emerging threat to public health worldwide by causing severe respiratory disease in humans with high virulence and case fatality rate (about 35%) since 2012. Little is known about the pathogenesis and innate antiviral response in primary human monocyte-derived macrophages (MDMs) and dendritic cells (MDDCs) upon MERS-CoV infection. In this study, we assessed MERS-CoV replication as well as induction of inflammatory cytokines and chemokines in MDMs and immature and mature MDDCs. Immature MDDCs and MDMs were permissive for MERS-CoV infection, while mature MDDCs were not, with stimulation of proinflammatory cytokine and chemokine upregulation in MDMs, but not in MDDCs. To further evaluate the antiviral activity of well-defined drugs in primary antigen presenting cells (APCs), three compounds (chloroquine, chlorpromazine and toremifine), each with broad-spectrum antiviral activity in immortalized cell lines, were evaluated in MDMs and MDDCs to determine their antiviral effect on MERS-CoV infection. While chloroquine was not active in these primary cells, chlorpromazine showed strong anti-MERS-CoV activity, but it was associated with high cytotoxicity narrowing the potential window for drug utilization. Unlike in established cells, toremifene had marginal activity when tested in antigen presenting cells, with high apparent cytotoxicity, also limiting its potential as a therapeutic option. These results demonstrate the value of testing drugs in primary cells, in addition to established cell lines, before initiating preclinical or clinical studies for MERS treatment and the importance of carefully assessing cytotoxicity in drug screen assays. Furthermore, these studies also highlight the role of APCs in stimulating a robust protective immune response to MERS-CoV infection.


Asunto(s)
Células Presentadoras de Antígenos/efectos de los fármacos , Antivirales/uso terapéutico , Infecciones por Coronavirus/tratamiento farmacológico , Macrófagos/efectos de los fármacos , Coronavirus del Síndrome Respiratorio de Oriente Medio , Animales , Células Presentadoras de Antígenos/fisiología , Células Cultivadas , Chlorocebus aethiops , Infecciones por Coronavirus/inmunología , Aprobación de Drogas , Evaluación Preclínica de Medicamentos , Humanos , Macrófagos/fisiología , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Monocitos/fisiología , Resultado del Tratamiento , Células Vero
12.
Drugs ; 77(18): 1935-1966, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29143192

RESUMEN

No specific antivirals are currently available for two emerging infectious diseases, Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS). A literature search was performed covering pathogenesis, clinical features and therapeutics, clinically developed drugs for repurposing and novel drug targets. This review presents current knowledge on the epidemiology, pathogenesis and clinical features of the SARS and MERS coronaviruses. The rationale for and outcomes with treatments used for SARS and MERS is discussed. The main focus of the review is on drug development and the potential that drugs approved for other indications provide for repurposing. The drugs we discuss belong to a wide range of different drug classes, such as cancer therapeutics, antipsychotics, and antimalarials. In addition to their activity against MERS and SARS coronaviruses, many of these approved drugs have broad-spectrum potential and have already been in clinical use for treating other viral infections. A wealth of knowledge is available for these drugs. However, the information in this review is not meant to guide clinical decisions, and any therapeutic described here should only be used in context of a clinical trial. Potential targets for novel antivirals and antibodies are discussed as well as lessons learned from treatment development for other RNA viruses. The article concludes with a discussion of the gaps in our knowledge and areas for future research on emerging coronaviruses.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Coronavirus/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Antivirales/química , Infecciones por Coronavirus/virología , Aprobación de Drogas , Humanos , Coronavirus del Síndrome Respiratorio de Oriente Medio , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo , Síndrome Respiratorio Agudo Grave/virología
13.
PLoS Negl Trop Dis ; 11(4): e0005540, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28403145

RESUMEN

Phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) is a lipid kinase involved in endosome maturation that emerged from a haploid genetic screen as being required for Ebola virus (EBOV) infection. Here we analyzed the effects of apilimod, a PIKfyve inhibitor that was reported to be well tolerated in humans in phase 2 clinical trials, for its effects on entry and infection of EBOV and Marburg virus (MARV). We first found that apilimod blocks infections by EBOV and MARV in Huh 7, Vero E6 and primary human macrophage cells, with notable potency in the macrophages (IC50, 10 nM). We next observed that similar doses of apilimod block EBOV-glycoprotein-virus like particle (VLP) entry and transcription-replication competent VLP infection, suggesting that the primary mode of action of apilimod is as an entry inhibitor, preventing release of the viral genome into the cytoplasm to initiate replication. After providing evidence that the anti-EBOV action of apilimod is via PIKfyve, we showed that it blocks trafficking of EBOV VLPs to endolysosomes containing Niemann-Pick C1 (NPC1), the intracellular receptor for EBOV. Concurrently apilimod caused VLPs to accumulate in early endosome antigen 1-positive endosomes. We did not detect any effects of apilimod on bulk endosome acidification, on the activity of cathepsins B and L, or on cholesterol export from endolysosomes. Hence by antagonizing PIKfyve, apilimod appears to block EBOV trafficking to its site of fusion and entry into the cytoplasm. Given the drug's observed anti-filoviral activity, relatively unexplored mechanism of entry inhibition, and reported tolerability in humans, we propose that apilimod be further explored as part of a therapeutic regimen to treat filoviral infections.


Asunto(s)
Antivirales/farmacología , Ebolavirus/efectos de los fármacos , Marburgvirus/efectos de los fármacos , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Triazinas/farmacología , Internalización del Virus/efectos de los fármacos , Animales , Transporte Biológico , Línea Celular , Chlorocebus aethiops , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Humanos , Hidrazonas , Lisosomas/metabolismo , Macrófagos/virología , Marburgvirus/fisiología , Nocodazol/farmacología , Pirimidinas , Toremifeno/farmacología , Células Vero
14.
J Infect Dis ; 215(9): 1416-1420, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28368541

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Ebolavirus/efectos de los fármacos , Interferón beta/farmacología , Interferón gamma/farmacología , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Sinergismo Farmacológico , Humanos , Células Vero
15.
J Virol ; 91(2)2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27807241

RESUMEN

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.


Asunto(s)
Infecciones por Coronavirus/inmunología , Protección Cruzada/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Virus de la Rabia/inmunología , Rabia/inmunología , Vacunas Virales/inmunología , Animales , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/virología , Modelos Animales de Enfermedad , Regulación Viral de la Expresión Génica , Humanos , Inmunización , Ratones , Interacciones Microbianas , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Rabia/prevención & control , Rabia/virología , Virus de la Rabia/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/inmunología , Vacunas Atenuadas , Vacunas Sintéticas , Proteínas Virales/genética , Proteínas Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Virales/efectos adversos , Vacunas Virales/genética , Ensamble de Virus
16.
PLoS One ; 11(11): e0166318, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27902714

RESUMEN

In the fall of 2014, an international news agency reported that patients suffering from Ebola virus disease (EVD) in Liberia were treated successfully with lamivudine, an antiviral drug used to treat human immunodeficiency virus-1 and hepatitis B virus infections. According to the report, 13 out of 15 patients treated with lamivudine survived and were declared free from Ebola virus disease. In this study, the anti-Ebola virus (EBOV) activity of lamivudine and another antiretroviral, zidovudine, were evaluated in a diverse set of cell lines against two variants of wild-type EBOV. Variable assay parameters were assessed to include different multiplicities of infection, lengths of inoculation times, and durations of dosing. At a multiplicity of infection of 1, lamivudine and zidovudine had no effect on EBOV propagation in Vero E6, Hep G2, or HeLa cells, or in primary human monocyte-derived macrophages. At a multiplicity of infection of 0.1, zidovudine demonstrated limited anti-EBOV activity in Huh 7 cells. Under certain conditions, lamivudine had low anti-EBOV activity at the maximum concentration tested (320 µM). However, lamivudine never achieved greater than 30% viral inhibition, and the activity was not consistently reproducible. Combination of lamivudine and zidovudine showed no synergistic antiviral activity. Independently, a set of in vitro experiments testing lamivudine and zidovudine for antiviral activity against an Ebola-enhanced green fluorescent protein reporter virus was performed at the Centers for Disease Control and Prevention. No antiviral activity was observed for either compound. A study evaluating the efficacy of lamivudine in a guinea pig model of EVD found no survival benefit. This lack of benefit was observed despite plasma lamivudine concentrations in guinea pig of about 4 µg/ml obtained in a separately conducted pharmacokinetics study. These studies found no evidence to support the therapeutic use of lamivudine for the treatment of EVD.


Asunto(s)
Fármacos Anti-VIH/farmacología , Ebolavirus/efectos de los fármacos , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Lamivudine/farmacología , Zidovudina/farmacología , Animales , Chlorocebus aethiops , Ebolavirus/aislamiento & purificación , Cobayas , Células HeLa , Fiebre Hemorrágica Ebola/virología , Humanos , Macrófagos , Proyectos Piloto , Células Vero , Replicación Viral/efectos de los fármacos
17.
Sci Transl Med ; 8(326): 326ra21, 2016 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-26888429

RESUMEN

As of 13 November 2015, 1618 laboratory-confirmed human cases of Middle East respiratory syndrome coronavirus (MERS-CoV) infection, including 579 deaths, had been reported to the World Health Organization. No specific preventive or therapeutic agent of proven value against MERS-CoV is currently available. Public Health England and the International Severe Acute Respiratory and Emerging Infection Consortium identified passive immunotherapy with neutralizing antibodies as a treatment approach that warrants priority study. Two experimental MERS-CoV vaccines were used to vaccinate two groups of transchromosomic (Tc) bovines that were genetically modified to produce large quantities of fully human polyclonal immunoglobulin G (IgG) antibodies. Vaccination with a clade A γ-irradiated whole killed virion vaccine (Jordan strain) or a clade B spike protein nanoparticle vaccine (Al-Hasa strain) resulted in Tc bovine sera with high enzyme-linked immunosorbent assay (ELISA) and neutralizing antibody titers in vitro. Two purified Tc bovine human IgG immunoglobulins (Tc hIgG), SAB-300 (produced after Jordan strain vaccination) and SAB-301 (produced after Al-Hasa strain vaccination), also had high ELISA and neutralizing antibody titers without antibody-dependent enhancement in vitro. SAB-301 was selected for in vivo and preclinical studies. Administration of single doses of SAB-301 12 hours before or 24 and 48 hours after MERS-CoV infection (Erasmus Medical Center 2012 strain) of Ad5-hDPP4 receptor-transduced mice rapidly resulted in viral lung titers near or below the limit of detection. Tc bovines, combined with the ability to quickly produce Tc hIgG and develop in vitro assays and animal model(s), potentially offer a platform to rapidly produce a therapeutic to prevent and/or treat MERS-CoV infection and/or other emerging infectious diseases.


Asunto(s)
Cromosomas de los Mamíferos/genética , Inmunoglobulina G/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Acrecentamiento Dependiente de Anticuerpo , Bovinos , Dipeptidil Peptidasa 4/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Ratones Endogámicos BALB C , Coronavirus del Síndrome Respiratorio de Oriente Medio/genética , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Pruebas de Neutralización , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción Genética , Vacunación , Replicación Viral
18.
Antiviral Res ; 125: 71-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26526586

RESUMEN

Brincidofovir (BCV) is the 3-hexadecyloxy-1-propanol (HDP) lipid conjugate of the acyclic nucleoside phosphonate cidofovir (CDV). BCV has established broad-spectrum activity against double-stranded DNA (dsDNA) viruses; however, its activity against RNA viruses has been less thoroughly evaluated. Here, we report that BCV inhibited infection of Ebola virus in multiple human cell lines. Unlike the mechanism of action for BCV against cytomegalovirus and other dsDNA viruses, phosphorylation of CDV to the diphosphate form appeared unnecessary. Instead, antiviral activity required the lipid moiety and in vitro activity against EBOV was observed for several HDP-nucleotide conjugates.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Citosina/análogos & derivados , Ebolavirus/efectos de los fármacos , Fiebre Hemorrágica Ebola/tratamiento farmacológico , Organofosfonatos/química , Organofosfonatos/farmacología , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Cidofovir , Citosina/química , Citosina/farmacología , Evaluación Preclínica de Medicamentos/métodos , Células HeLa , Fiebre Hemorrágica Ebola/prevención & control , Fiebre Hemorrágica Ebola/virología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lípidos/química , Lípidos/farmacología , Masculino , Relación Estructura-Actividad , Células Vero , Replicación Viral/efectos de los fármacos
19.
Virology ; 485: 422-30, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26342468

RESUMEN

Middle East Respiratory Syndrome Coronavirus (MERS-CoV) continues to be a threat to human health in the Middle East. Development of countermeasures is ongoing; however, an animal model that faithfully recapitulates human disease has yet to be defined. A recent study indicated that inoculation of common marmosets resulted in inconsistent lethality. Based on these data we sought to compare two isolates of MERS-CoV. We followed disease progression in common marmosets after intratracheal exposure with: MERS-CoV-EMC/2012, MERS-CoV-Jordan-n3/2012, media, or inactivated virus. Our data suggest that common marmosets developed a mild to moderate non-lethal respiratory disease, which was quantifiable by computed tomography (CT), with limited other clinical signs. Based on CT data, clinical data, and virological data, MERS-CoV inoculation of common marmosets results in mild to moderate clinical signs of disease that are likely due to manipulations of the marmoset rather than as a result of robust viral replication.


Asunto(s)
Infecciones por Coronavirus/veterinaria , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Enfermedades de los Monos/mortalidad , Enfermedades de los Monos/virología , Animales , Anticuerpos Antivirales/inmunología , Biopsia , Callithrix , Chlorocebus aethiops , Modelos Animales de Enfermedad , Riñón/patología , Riñón/virología , Pulmón/patología , Pulmón/virología , Enfermedades de los Monos/diagnóstico , Enfermedades de los Monos/inmunología , ARN Viral/genética , Índice de Severidad de la Enfermedad , Tomografía Computarizada por Rayos X , Células Vero
20.
Sci Transl Med ; 7(290): 290ra89, 2015 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-26041706

RESUMEN

Currently, no approved therapeutics exist to treat or prevent infections induced by Ebola viruses, and recent events have demonstrated an urgent need for rapid discovery of new treatments. Repurposing approved drugs for emerging infections remains a critical resource for potential antiviral therapies. We tested ~2600 approved drugs and molecular probes in an in vitro infection assay using the type species, Zaire ebolavirus. Selective antiviral activity was found for 80 U.S. Food and Drug Administration-approved drugs spanning multiple mechanistic classes, including selective estrogen receptor modulators, antihistamines, calcium channel blockers, and antidepressants. Results using an in vivo murine Ebola virus infection model confirmed the protective ability of several drugs, such as bepridil and sertraline. Viral entry assays indicated that most of these antiviral drugs block a late stage of viral entry. By nature of their approved status, these drugs have the potential to be rapidly advanced to clinical settings and used as therapeutic countermeasures for Ebola virus infections.


Asunto(s)
Antivirales/uso terapéutico , Aprobación de Drogas , Fiebre Hemorrágica Ebola/terapia , Sondas Moleculares , Animales , Bepridil/farmacología , Ebolavirus/efectos de los fármacos , Humanos , Ratones , Sertralina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA