Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Infect Dis ; 218(suppl_5): S612-S626, 2018 11 22.
Article in English | MEDLINE | ID: mdl-29860496

ABSTRACT

Background: For most classes of drugs, rapid development of therapeutics to treat emerging infections is challenged by the timelines needed to identify compounds with the desired efficacy, safety, and pharmacokinetic profiles. Fully human monoclonal antibodies (mAbs) provide an attractive method to overcome many of these hurdles to rapidly produce therapeutics for emerging diseases. Methods: In this study, we deployed a platform to generate, test, and develop fully human antibodies to Zaire ebolavirus. We obtained specific anti-Ebola virus (EBOV) antibodies by immunizing VelocImmune mice that use human immunoglobulin variable regions in their humoral responses. Results: Of the antibody clones isolated, 3 were selected as best at neutralizing EBOV and triggering FcγRIIIa. Binding studies and negative-stain electron microscopy revealed that the 3 selected antibodies bind to non-overlapping epitopes, including a potentially new protective epitope not targeted by other antibody-based treatments. When combined, a single dose of a cocktail of the 3 antibodies protected nonhuman primates (NHPs) from EBOV disease even after disease symptoms were apparent. Conclusions: This antibody cocktail provides complementary mechanisms of actions, incorporates novel specificities, and demonstrates high-level postexposure protection from lethal EBOV disease in NHPs. It is now undergoing testing in normal healthy volunteers in preparation for potential use in future Ebola epidemics.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Hemorrhagic Fever, Ebola/drug therapy , Animals , Antibodies, Monoclonal/isolation & purification , Glycoproteins/immunology , Guinea Pigs , HEK293 Cells , Humans , Macaca mulatta , Male , Mice
2.
bioRxiv ; 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38410448

ABSTRACT

Infection with Sudan virus (SUDV) is characterized by an aggressive disease course with case fatality rates between 40-100% and no approved vaccines or therapeutics. SUDV causes sporadic outbreaks in sub-Saharan Africa, including a recent outbreak in Uganda which has resulted in over 100 confirmed cases in one month. Prior vaccine and therapeutic efforts have historically prioritized Ebola virus (EBOV), leading to a significant gap in available treatments. Two vaccines, Erbevo ® and Zabdeno ® /Mvabea ® , are licensed for use against EBOV but are ineffective against SUDV. Recombinant adenovirus vector vaccines have been shown to be safe and effective against filoviruses, but efficacy depends on having low seroprevalence to the vector in the target human population. For this reason, and because of an excellent safety and immunogenicity profile, ChAd3 was selected as a superior vaccine vector. Here, a ChAd3 vaccine expressing the SUDV glycoprotein (GP) was evaluated for immunogenicity and efficacy in nonhuman primates. We demonstrate that a single dose of ChAd3-SUDV confers acute and durable protection against lethal SUDV challenge with a strong correlation between the SUDV GP-specific antibody titers and survival outcome. Additionally, we show that a bivalent ChAd3 vaccine encoding the GP from both EBOV and SUDV protects against both parenteral and aerosol lethal SUDV challenge. Our data indicate that the ChAd3-SUDV vaccine is a suitable candidate for a prophylactic vaccination strategy in regions at high risk of filovirus outbreaks. One Sentence Summary: A single-dose of ChAd3 vaccine protected macaques from lethal challenge with Sudan virus (SUDV) by parenteral and aerosol routes of exposure.

3.
PLoS Negl Trop Dis ; 16(5): e0010081, 2022 05.
Article in English | MEDLINE | ID: mdl-35533188

ABSTRACT

Eastern equine encephalitis virus (EEEV) is mosquito-borne virus that produces fatal encephalitis in humans. We recently conducted a first of its kind study to investigate EEEV clinical disease course following aerosol challenge in a cynomolgus macaque model utilizing the state-of-the-art telemetry to measure critical physiological parameters. Here, we report the results of a comprehensive pathology study of NHP tissues collected at euthanasia to gain insights into EEEV pathogenesis. Viral RNA and proteins as well as microscopic lesions were absent in the visceral organs. In contrast, viral RNA and proteins were readily detected throughout the brain including autonomic nervous system (ANS) control centers and spinal cord. However, despite presence of viral RNA and proteins, majority of the brain and spinal cord tissues exhibited minimal or no microscopic lesions. The virus tropism was restricted primarily to neurons, and virus particles (~61-68 nm) were present within axons of neurons and throughout the extracellular spaces. However, active virus replication was absent or minimal in majority of the brain and was limited to regions proximal to the olfactory tract. These data suggest that EEEV initially replicates in/near the olfactory bulb following aerosol challenge and is rapidly transported to distal regions of the brain by exploiting the neuronal axonal transport system to facilitate neuron-to-neuron spread. Once within the brain, the virus gains access to the ANS control centers likely leading to disruption and/or dysregulation of critical physiological parameters to produce severe disease. Moreover, the absence of microscopic lesions strongly suggests that the underlying mechanism of EEEV pathogenesis is due to neuronal dysfunction rather than neuronal death. This study is the first comprehensive investigation into EEEV pathology in a NHP model and will provide significant insights into the evaluation of countermeasure.


Subject(s)
Encephalitis Virus, Eastern Equine , Encephalomyelitis, Equine , Aerosols , Animals , Brain , Disease Models, Animal , Encephalomyelitis, Equine/pathology , Horses , Macaca fascicularis , RNA, Viral , Spinal Cord/pathology
4.
Sci Transl Med ; 14(631): eabi5229, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35138912

ABSTRACT

Effective therapeutics have been developed against acute Ebola virus disease (EVD) in both humans and experimentally infected nonhuman primates. However, the risk of viral persistence and associated disease recrudescence in survivors receiving these therapeutics remains unclear. In contrast to rhesus macaques that survived Ebola virus (EBOV) exposure in the absence of treatment, we discovered that EBOV, despite being cleared from all other organs, persisted in the brain ventricular system of rhesus macaque survivors that had received monoclonal antibody (mAb) treatment. In mAb-treated macaque survivors, EBOV persisted in macrophages infiltrating the brain ventricular system, including the choroid plexuses. This macrophage infiltration was accompanied by severe tissue damage, including ventriculitis, choroid plexitis, and meningoencephalitis. Specifically, choroid plexus endothelium-derived EBOV infection led to viral persistence in the macaque brain ventricular system. This resulted in apoptosis of ependymal cells, which constitute the blood-cerebrospinal fluid barrier of the choroid plexuses. Fatal brain-confined recrudescence of EBOV infection manifested as severe inflammation, local pathology, and widespread infection of the ventricular system and adjacent neuropil in some of the mAb-treated macaque survivors. This study highlights organ-specific EBOV persistence and fatal recrudescent disease in rhesus macaque survivors after therapeutic treatment and has implications for the long-term follow-up of human survivors of EVD.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Animals , Antibodies, Monoclonal , Brain , Humans , Macaca mulatta , Recurrence , Survivors
5.
PLoS Negl Trop Dis ; 15(6): e0009424, 2021 06.
Article in English | MEDLINE | ID: mdl-34138849

ABSTRACT

Most alphaviruses are mosquito-borne and can cause severe disease in humans and domesticated animals. In North America, eastern equine encephalitis virus (EEEV) is an important human pathogen with case fatality rates of 30-90%. Currently, there are no therapeutics or vaccines to treat and/or prevent human infection. One critical impediment in countermeasure development is the lack of insight into clinically relevant parameters in a susceptible animal model. This study examined the disease course of EEEV in a cynomolgus macaque model utilizing advanced telemetry technology to continuously and simultaneously measure temperature, respiration, activity, heart rate, blood pressure, electrocardiogram (ECG), and electroencephalography (EEG) following an aerosol challenge at 7.0 log10 PFU. Following challenge, all parameters were rapidly and substantially altered with peak alterations from baseline ranged as follows: temperature (+3.0-4.2°C), respiration rate (+56-128%), activity (-15-76% daytime and +5-22% nighttime), heart rate (+67-190%), systolic (+44-67%) and diastolic blood pressure (+45-80%). Cardiac abnormalities comprised of alterations in QRS and PR duration, QTc Bazett, T wave morphology, amplitude of the QRS complex, and sinoatrial arrest. An unexpected finding of the study was the first documented evidence of a critical cardiac event as an immediate cause of euthanasia in one NHP. All brain waves were rapidly (~12-24 hpi) and profoundly altered with increases of up to 6,800% and severe diffuse slowing of all waves with decreases of ~99%. Lastly, all NHPs exhibited disruption of the circadian rhythm, sleep, and food/fluid intake. Accordingly, all NHPs met the euthanasia criteria by ~106-140 hpi. This is the first of its kind study utilizing state of the art telemetry to investigate multiple clinical parameters relevant to human EEEV infection in a susceptible cynomolgus macaque model. The study provides critical insights into EEEV pathogenesis and the parameters identified will improve animal model development to facilitate rapid evaluation of vaccines and therapeutics.


Subject(s)
Alphavirus Infections/virology , Disease Models, Animal , Electroencephalography , Encephalitis Virus, Eastern Equine , Monitoring, Physiologic/instrumentation , Telemetry/instrumentation , Aerosols , Alphavirus Infections/pathology , Animals , Blood Pressure , Body Temperature , Chlorocebus aethiops , Female , Heart Rate , Humans , Macaca fascicularis , Male , Monitoring, Physiologic/methods , Motor Activity , Respiratory Physiological Phenomena , Telemetry/methods , Vero Cells
6.
J Am Chem Soc ; 132(32): 10970-2, 2010 Aug 18.
Article in English | MEDLINE | ID: mdl-20698645

ABSTRACT

A proposed tangential flow ultrafiltration method was compared to the widely used ultracentrifugation method for efficiency and efficacy in concentrating, size selecting, and minimizing the aggregation state of a silver nanoparticle (AgNP) colloid while probing the AgNPs' SERS-based sensing capabilities. The ultrafiltration method proved to be more efficient and more effective and was found to tremendously boost the SERS-based sensing capabilities of these AgNPs through the increased number of homogeneous SERS hot spots available for a biotarget molecule within a minimal focal volume. Future research studies and applications addressing the physiochemical properties or biological impact of AgNPs would greatly benefit from ultrafiltration for its ability to generate monodisperse colloidal nanoparticles, to eliminate excess toxic chemicals from nanoparticle synthesis, and to obtain minimum levels of aggregation during nanoparticle concentration.


Subject(s)
Biosensing Techniques/methods , Metal Nanoparticles/chemistry , Silver/chemistry , Spectrum Analysis, Raman , Ultrafiltration/methods , Particle Size
7.
Viruses ; 12(6)2020 05 29.
Article in English | MEDLINE | ID: mdl-32485952

ABSTRACT

Lassa virus (LASV), an arenavirus causing Lassa fever, is endemic to West Africa with up to 300,000 cases and between 5000 and 10,000 deaths per year. Rarely seen in the United States, Lassa virus is a CDC category A biological agent inasmuch deliberate aerosol exposure can have high mortality rates compared to naturally acquired infection. With the need for an animal model, specific countermeasures remain elusive as there is no FDA-approved vaccine. This natural history of aerosolized Lassa virus exposure in Macaca fascicularis was studied under continuous telemetric surveillance. The macaque response to challenge was largely analogous to severe human disease with fever, tachycardia, hypotension, and tachypnea. During initial observations, an increase trend of activated monocytes positive for viral glycoprotein was accompanied by lymphocytopenia. Disease uniformly progressed to high viremia followed by low anion gap, alkalosis, anemia, and thrombocytopenia. Hypoproteinemia occurred late in infection followed by increased levels of white blood cells, cytokines, chemokines, and biochemical markers of liver injury. Viral nucleic acids were detected in tissues of three non­survivors at endpoint, but not in the lone survivor. This study provides useful details to benchmark a pivotal model of Lassa fever in support of medical countermeasure development for both endemic disease and traditional biodefense purposes.


Subject(s)
Aerosols/adverse effects , Lassa Fever/etiology , Animals , Flow Cytometry , Inhalation Exposure , Lassa Fever/diagnosis , Lassa Fever/virology , Lassa virus/pathogenicity , Macaca fascicularis , Male , Real-Time Polymerase Chain Reaction , Telemetry , Viral Plaque Assay , Viremia/diagnosis
8.
Viruses ; 10(10)2018 09 20.
Article in English | MEDLINE | ID: mdl-30241284

ABSTRACT

In the 2014⁻2016 West Africa Ebola Virus (EBOV) outbreak, there was a significant concern raised about the potential for secondary bacterial infection originating from the gastrointestinal tract, which led to the empiric treatment of many patients with antibiotics. This retrospective pathology case series summarizes the gastrointestinal pathology observed in control animals in the rhesus EBOV-Kikwit intramuscular 1000 plaque forming unit infection model. All 31 Non-human primates (NHPs) exhibited lymphoid depletion of gut-associated lymphoid tissue (GALT) but the severity and the specific location of the depletion varied. Mesenteric lymphoid depletion and necrosis were present in 87% (27/31) of NHPs. There was mucosal barrier disruption of the intestinal tract with mucosal necrosis and/or ulceration most notably in the duodenum (16%), cecum (16%), and colon (29%). In the intestinal tract, hemorrhage was noted most frequently in the duodenum (52%) and colon (45%). There were focal areas of bacterial submucosal invasion in the gastrointestinal (GI) tract in 9/31 (29%) of NHPs. Only 2/31 (6%) had evidence of pancreatic necrosis. One NHP (3%) experienced jejunal intussusception which may have been directly related to EBOV. Immunofluorescence assays demonstrated EBOV antigen in CD68+ macrophage/monocytes and endothelial cells in areas of GI vascular injury or necrosis.


Subject(s)
Ebolavirus/immunology , Gastrointestinal Tract/pathology , Hemorrhagic Fever, Ebola/pathology , Animals , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/immunology , Antigens, Viral/immunology , Cohort Studies , Disease Models, Animal , Female , Gastrointestinal Hemorrhage/pathology , Gastrointestinal Hemorrhage/virology , Gastrointestinal Tract/virology , Humans , Lymphoid Tissue/pathology , Lymphoid Tissue/virology , Macaca mulatta , Male , Necrosis/pathology , Necrosis/virology , Retrospective Studies
9.
Cell Rep ; 24(4): 1050-1059.e5, 2018 07 24.
Article in English | MEDLINE | ID: mdl-30044972

ABSTRACT

Development of an effective vaccine became a worldwide priority after the devastating 2013-2016 Ebola disease outbreak. To qualitatively profile the humoral response against advanced filovirus vaccine candidates, we developed Domain Programmable Arrays (DPA), a systems serology platform to identify epitopes targeted after vaccination or filovirus infection. We optimized the assay using a panel of well-characterized monoclonal antibodies. After optimization, we utilized the system to longitudinally characterize the immunoglobulin (Ig) isotype-specific responses in non-human primates vaccinated with rVSV-ΔG-EBOV-glycoprotein (GP). Strikingly, we observed that, although the IgM response was directed against epitopes over the whole GP, the IgG and IgA responses were almost exclusively directed against the mucin-like domain (MLD) of the glycan cap. Further research will be needed to characterize this possible biased IgG and IgA response toward the MLD, but the results corroborate that DPA is a valuable tool to qualitatively measure the humoral response after vaccination.


Subject(s)
Ebola Vaccines/immunology , Ebolavirus/immunology , Immunity, Humoral/genetics , Animals , Ebola Vaccines/blood , Humans , Macaca fascicularis , Mice
10.
Parasit Vectors ; 10(1): 218, 2017 May 03.
Article in English | MEDLINE | ID: mdl-28468673

ABSTRACT

BACKGROUND: Ebola virus (EBOV) infection results in high morbidity and mortality and is primarily transmitted in communities by contact with infectious bodily fluids. While clinical and experimental evidence indicates that EBOV is transmitted via mucosal exposure, the ability of non-biting muscid flies to mechanically transmit EBOV following exposure to the face had not been assessed. RESULTS: To investigate this transmission route, house flies (Musca domestica Linnaeus) were used to deliver an EBOV/blood mixture to the ocular/nasal/oral facial mucosa of four cynomolgus macaques (Macaca fascicularis Raffles). Following exposure, macaques were monitored for evidence of infection through the conclusion of the study, days 57 and 58. We found no evidence of systemic infection in any of the exposed macaques. CONCLUSIONS: The results of this study indicate that there is a low potential for the mechanical transmission of EBOV via house flies - the conditions in this study were not sufficient to initiate infection.


Subject(s)
Ebolavirus/isolation & purification , Hemorrhagic Fever, Ebola/transmission , Houseflies/virology , Insect Vectors/virology , Animals , Eye/virology , Face/virology , Feces/virology , Hemorrhagic Fever, Ebola/blood , Hemorrhagic Fever, Ebola/virology , Macaca fascicularis , Mouth Mucosa/virology , Mucous Membrane/virology , Nose/virology
11.
Science ; 351(6279): 1339-42, 2016 Mar 18.
Article in English | MEDLINE | ID: mdl-26917593

ABSTRACT

Ebola virus disease in humans is highly lethal, with case fatality rates ranging from 25 to 90%. There is no licensed treatment or vaccine against the virus, underscoring the need for efficacious countermeasures. We ascertained that a human survivor of the 1995 Kikwit Ebola virus disease outbreak maintained circulating antibodies against the Ebola virus surface glycoprotein for more than a decade after infection. From this survivor we isolated monoclonal antibodies (mAbs) that neutralize recent and previous outbreak variants of Ebola virus and mediate antibody-dependent cell-mediated cytotoxicity in vitro. Strikingly, monotherapy with mAb114 protected macaques when given as late as 5 days after challenge. Treatment with a single human mAb suggests that a simplified therapeutic strategy for human Ebola infection may be possible.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antibodies, Viral/administration & dosage , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Adult , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/isolation & purification , Antibodies, Viral/immunology , Antibodies, Viral/isolation & purification , Clinical Trials as Topic , Disease Outbreaks , Female , Hemorrhagic Fever, Ebola/epidemiology , Humans , Macaca , Male , Molecular Sequence Data , Survivors
12.
Viruses ; 7(12): 6739-54, 2015 Dec 19.
Article in English | MEDLINE | ID: mdl-26703716

ABSTRACT

Recent experimentation with the variants of the Ebola virus that differ in the glycoprotein's poly-uridine site, which dictates the form of glycoprotein produced through a transcriptional stutter, has resulted in questions regarding the pathogenicity and lethality of the stocks used to develop products currently undergoing human clinical trials to combat the disease. In order to address these concerns and prevent the delay of these critical research programs, we designed an experiment that permitted us to intramuscularly challenge statistically significant numbers of naïve and vaccinated cynomolgus macaques with either a 7U or 8U variant of the Ebola virus, Kikwit isolate. In naïve animals, no difference in survivorship was observed; however, there was a significant delay in the disease course between the two groups. Significant differences were also observed in time-of-fever, serum chemistry, and hematology. In vaccinated animals, there was no statistical difference in survivorship between either challenge groups, with two succumbing in the 7U group compared to 1 in the 8U challenge group. In summary, survivorship was not affected, but the Ebola virus disease course in nonhuman primates is temporally influenced by glycoprotein poly-U editing site populations.


Subject(s)
Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/virology , Poly U/analysis , Viral Envelope Proteins/chemistry , Virulence Factors/chemistry , Animals , Disease Models, Animal , Injections, Intramuscular , Macaca fascicularis , Survival Analysis , Viral Envelope Proteins/metabolism , Virulence Factors/metabolism
13.
Viruses ; 6(11): 4666-82, 2014 Nov 24.
Article in English | MEDLINE | ID: mdl-25421892

ABSTRACT

Multiple products are being developed for use against filoviral infections. Efficacy for these products will likely be demonstrated in nonhuman primate models of filoviral disease to satisfy licensure requirements under the Animal Rule, or to supplement human data. Typically, the endpoint for efficacy assessment will be survival following challenge; however, there exists no standardized approach for assessing the health or euthanasia criteria for filovirus-exposed nonhuman primates. Consideration of objective criteria is important to (a) ensure test subjects are euthanized without unnecessary distress; (b) enhance the likelihood that animals exhibiting mild or moderate signs of disease are not prematurely euthanized; (c) minimize the occurrence of spontaneous deaths and loss of end-stage samples; (d) enhance the reproducibility of experiments between different researchers; and (e) provide a defensible rationale for euthanasia decisions that withstands regulatory scrutiny. Historic records were compiled for 58 surviving and non-surviving monkeys exposed to Ebola virus at the US Army Medical Research Institute of Infectious Diseases. Clinical pathology parameters were statistically analyzed and those exhibiting predicative value for survival are reported. These findings may be useful for standardization of objective euthanasia assessments in rhesus monkeys exposed to Ebola virus and may serve as a useful approach for other standardization efforts.


Subject(s)
Euthanasia, Animal , Haplorhini , Hemorrhagic Fever, Ebola/pathology , Primate Diseases/pathology , Animals , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Hemorrhagic Fever, Ebola/therapy , Primate Diseases/therapy , Survival Analysis
14.
Nat Med ; 20(10): 1126-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25194571

ABSTRACT

Ebolavirus disease causes high mortality, and the current outbreak has spread unabated through West Africa. Human adenovirus type 5 vectors (rAd5) encoding ebolavirus glycoprotein (GP) generate protective immunity against acute lethal Zaire ebolavirus (EBOV) challenge in macaques, but fail to protect animals immune to Ad5, suggesting natural Ad5 exposure may limit vaccine efficacy in humans. Here we show that a chimpanzee-derived replication-defective adenovirus (ChAd) vaccine also rapidly induced uniform protection against acute lethal EBOV challenge in macaques. Because protection waned over several months, we boosted ChAd3 with modified vaccinia Ankara (MVA) and generated, for the first time, durable protection against lethal EBOV challenge.


Subject(s)
Ebola Vaccines/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/prevention & control , Adenovirus Vaccines/administration & dosage , Adenovirus Vaccines/genetics , Adenovirus Vaccines/immunology , Adenoviruses, Human/genetics , Adenoviruses, Human/immunology , Adenoviruses, Simian/genetics , Adenoviruses, Simian/immunology , Animals , Defective Viruses/genetics , Defective Viruses/immunology , Ebola Vaccines/administration & dosage , Ebola Vaccines/genetics , Ebolavirus/genetics , Female , Genetic Vectors , Hemorrhagic Fever, Ebola/virology , Humans , Immunization, Secondary , Macaca fascicularis , Pan troglodytes , RNA, Viral/blood , RNA, Viral/genetics , Time Factors , Vaccinia virus/genetics , Vaccinia virus/immunology
15.
J Biomed Nanotechnol ; 9(9): 1624-35, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23980510

ABSTRACT

Silver nanoparticles have been shown to inhibit viruses. However, very little is known about the mechanism of antiviral activity. This study tested the hypothesis that 25-nm silver nanoparticles inhibited Vaccinia virus replication by preventing viral entry. Plaque reduction, confocal microscopy, and beta-galactosidase reporter gene assays were used to examine viral attachment and entry in the presence and absence of silver nanoparticles. To explore the mechanism of inhibition, viral entry experiments were conducted with silver nanoparticles and small interfering RNAs designed to silence the gene coding for p21-activated kinase 1, a key mediator of macropinocytosis. The silver nanoparticles caused a 4- to 5-log reduction in viral titer at concentrations that were not toxic to cells. Virus was capable of adsorbing to cells but could not enter cells in the presence of silver nanoparticles. Virus particles that had adsorbed to cells in the presence of silver nanoparticles were found to be infectious upon removal from the cells, indicating lack of direct virucidal effect. The half maximal inhibitory concentration for viral entry in the presence of silver nanoparticles was 27.4+/-3.3 microg/ml. When macropinocytosis was blocked, this inhibition was significantly reduced. Thus, macropinocytosis was required for the full antiviral effect. For the first time, this study points to the novel result that a cellular process involved in viral entry is responsible for the antiviral effects of silver nanoparticles.


Subject(s)
Kidney/physiology , Kidney/virology , Metal Nanoparticles/administration & dosage , Pinocytosis/physiology , Silver/administration & dosage , Vaccinia virus/physiology , Virus Internalization/drug effects , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Cell Line , Haplorhini , HeLa Cells , Humans , Materials Testing , Metal Nanoparticles/chemistry , Pinocytosis/drug effects , Silver/chemistry , Vaccinia/drug therapy , Vaccinia/virology , Vaccinia virus/drug effects
16.
PLoS Negl Trop Dis ; 7(4): e2171, 2013.
Article in English | MEDLINE | ID: mdl-23638192

ABSTRACT

Lassa virus (LASV) is a significant human pathogen that is endemic to several countries in West Africa. Infection with LASV leads to the development of hemorrhagic fever in a significant number of cases, and it is estimated that thousands die each year from the disease. Little is known about the complex immune mechanisms governing the response to LASV or the genetic determinants of susceptibility and resistance to infection. In the study presented here, we have used a whole-genome, microarray-based approach to determine the temporal host response in the peripheral blood mononuclear cells (PBMCs) of non-human primates (NHP) following aerosol exposure to LASV. Sequential sampling over the entire disease course showed that there are strong transcriptional changes of the immune response to LASV exposure, including the early induction of interferon-responsive genes and Toll-like receptor signaling pathways. However, this increase in early innate responses was coupled with a lack of pro-inflammatory cytokine response in LASV exposed NHPs. There was a distinct lack of cytokines such as IL1ß and IL23α, while immunosuppressive cytokines such as IL27 and IL6 were upregulated. Comparison of IRF/STAT1-stimulated gene expression with the viral load in LASV exposed NHPs suggests that mRNA expression significantly precedes viremia, and thus might be used for early diagnostics of the disease. Our results provide a transcriptomic survey of the circulating immune response to hemorrhagic LASV exposure and provide a foundation for biomarker identification to allow clinical diagnosis of LASV infection through analysis of the host response.


Subject(s)
Gene Expression Profiling/methods , Lassa Fever/genetics , Lassa virus/immunology , Leukocytes, Mononuclear/metabolism , Macaca/immunology , Macaca/virology , Animals , Female , Immunity, Innate/immunology , Lassa Fever/virology , Lassa virus/pathogenicity , Male , Toll-Like Receptors/metabolism
17.
J Virol Methods ; 183(1): 19-24, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22465243

ABSTRACT

This study centers on the development of a new screening tool for simultaneously evaluating the antiviral and cytotoxic properties of antiviral agents against an HIV-1-based, pseudotyped virus particle engineered to encode antibiotic resistance. The traditional colony-forming-unit assay for quantifying this type of virus was impractical as a screening tool due to the cumbersome nature of the setup and high costs in labor and supplies. Therefore, a smaller-scale and higher-throughput means of scoring antiviral activity was successfully developed and used to evaluate a specific batch of 25-nm silver nanoparticles (AgNPs). The new assay employed a unique application of the traditional cell proliferation/cytotoxicity test that is based on the chemical 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, which produces a colorimetric readout. The AgNPs showed a half maximal inhibitory concentration against the virus of 11.2±0.6 µg/ml (p<0.0001) with no significant toxicity against the cells. Because the virus was engineered to undergo only the first half of its replication cycle, the observed AgNP inhibition must have occurred at one of the early stages of infection. Overall, the new assay was very efficient and will be useful for testing different viral pseudotypes, screening different types of nanomaterials, and investigating other antiviral agents.


Subject(s)
Antiviral Agents/pharmacology , Antiviral Agents/toxicity , Colorimetry/methods , Nanoparticles , Silver/pharmacology , Silver/toxicity , Animals , Cell Line , Cell Survival/drug effects , HIV-1/drug effects , Humans , Microbial Sensitivity Tests/methods , Staining and Labeling/methods , Tetrazolium Salts/metabolism , Thiazoles/metabolism
18.
Viruses ; 4(9): 1619-50, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23170176

ABSTRACT

Viruses of the family Filoviridae represent significant health risks as emerging infectious diseases as well as potentially engineered biothreats. While many research efforts have been published offering possibilities toward the mitigation of filoviral infection, there remain no sanctioned therapeutic or vaccine strategies. Current progress in the development of filovirus therapeutics and vaccines is outlined herein with respect to their current level of testing, evaluation, and proximity toward human implementation, specifically with regard to human clinical trials, nonhuman primate studies, small animal studies, and in vitro development. Contemporary methods of supportive care and previous treatment approaches for human patients are also discussed.


Subject(s)
Filoviridae Infections/therapy , Filoviridae/immunology , Post-Exposure Prophylaxis/methods , Viral Vaccines/immunology , Animals , Clinical Trials as Topic , Humans , Immunotherapy/methods , Primates
19.
J Vis Exp ; (68)2012 Oct 04.
Article in English | MEDLINE | ID: mdl-23070148

ABSTRACT

Nowadays, AgNPs are extensively used in the manufacture of consumer products,(1) water disinfectants,(2) therapeutics,(1, 3) and biomedical devices(4) due to their powerful antimicrobial properties.(3-6) These nanoparticle applications are strongly influenced by the AgNP size and aggregation state. Many challenges exist in the controlled fabrication(7) and size-based isolation(4,8) of unfunctionalized, homogenous AgNPs that are free from chemically aggressive capping/stabilizing agents or organic solvents.(7-13) Limitations emerge from the toxicity of reagents, high costs or reduced efficiency of the AgNP synthesis or isolation methods (e.g., centrifugation, size-dependent solubility, size-exclusion chromatography, etc.).(10,14-18) To overcome this, we recently showed that TFU permits greater control over the size, concentration and aggregation state of Creighton AgNPs (300 ml of 15.3 µg ml(-1) down to 10 ml of 198.7 µg ml(-1)) than conventional methods of isolation such as ultracentrifugation.(19) TFU is a recirculation method commonly used for the weight-based isolation of proteins, viruses and cells.(20,21) Briefly, the liquid sample is passed through a series of hollow fiber membranes with pore size ranging from 1,000 kD to 10 kD. Smaller suspended or dissolved constituents in the sample will pass through the porous barrier together with the solvent (filtrate), while the larger constituents are retained (retentate). TFU may be considered a "green" method as it neither damages the sample nor requires additional solvent to eliminate toxic excess reagents and byproducts. Furthermore, TFU may be applied to a large variety of nanoparticles as both hydrophobic and hydrophilic filters are available. The two main objectives of this study were: 1) to illustrate the experimental aspects of the TFU approach through an invited video experience and 2) to demonstrate the feasibility of the TFU method for larger volumes of colloidal nanoparticles and smaller volumes of retentate. First, unfuctionalized AgNPs (4 L, 15.2 µg ml(-1)) were synthesized using the well-established Creighton method(22,23) by the reduction of AgNO3 with NaBH4. AgNP polydispersity was then minimized via a 3-step TFU using a 50-nm filter (460 cm(2)) to remove AgNPs and AgNP-aggregates larger than 50 nm, followed by two 100-kD (200 cm(2) and 20 cm(2)) filters to concentrate the AgNPs. Representative samples were characterized using transmission electron microscopy, UV-Vis absorption spectrophotometry, Raman spectroscopy, and inductively coupled plasma optical emission spectroscopy. The final retentate consisted of highly concentrated (4 ml, 8,539.9 µg ml(-1)) yet lowly aggregated and homogeneous AgNPs of 1-20 nm in diameter. This corresponds to a silver concentration yield of about 62%.


Subject(s)
Colloids/chemistry , Metal Nanoparticles/chemistry , Silver/chemistry , Ultrafiltration/methods , Colloids/isolation & purification , Silver/isolation & purification , Spectrophotometry, Ultraviolet , Spectrum Analysis, Raman , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL