Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 184(19): 4969-4980.e15, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34332650

ABSTRACT

Memory B cell reserves can generate protective antibodies against repeated SARS-CoV-2 infections, but with unknown reach from original infection to antigenically drifted variants. We charted memory B cell receptor-encoded antibodies from 19 COVID-19 convalescent subjects against SARS-CoV-2 spike (S) and found seven major antibody competition groups against epitopes recurrently targeted across individuals. Inclusion of published and newly determined structures of antibody-S complexes identified corresponding epitopic regions. Group assignment correlated with cross-CoV-reactivity breadth, neutralization potency, and convergent antibody signatures. Although emerging SARS-CoV-2 variants of concern escaped binding by many members of the groups associated with the most potent neutralizing activity, some antibodies in each of those groups retained affinity-suggesting that otherwise redundant components of a primary immune response are important for durable protection from evolving pathogens. Our results furnish a global atlas of S-specific memory B cell repertoires and illustrate properties driving viral escape and conferring robustness against emerging variants.

2.
PLoS Pathog ; 20(4): e1012134, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38603762

ABSTRACT

Monoclonal antibodies (mAbs) are an important class of antiviral therapeutics. MAbs are highly selective, well tolerated, and have long in vivo half-life as well as the capacity to induce immune-mediated virus clearance. Their activities can be further enhanced by integration of their variable fragments (Fvs) into bispecific antibodies (bsAbs), affording simultaneous targeting of multiple epitopes to improve potency and breadth and/or to mitigate against viral escape by a single mutation. Here, we explore a bsAb strategy for generation of pan-ebolavirus and pan-filovirus immunotherapeutics. Filoviruses, including Ebola virus (EBOV), Sudan virus (SUDV), and Marburg virus (MARV), cause severe hemorrhagic fever. Although there are two FDA-approved mAb therapies for EBOV infection, these do not extend to other filoviruses. Here, we combine Fvs from broad ebolavirus mAbs to generate novel pan-ebolavirus bsAbs that are potently neutralizing, confer protection in mice, and are resistant to viral escape. Moreover, we combine Fvs from pan-ebolavirus mAbs with those of protective MARV mAbs to generate pan-filovirus protective bsAbs. These results provide guidelines for broad antiviral bsAb design and generate new immunotherapeutic candidates.


Subject(s)
Antibodies, Bispecific , Antibodies, Viral , Ebolavirus , Hemorrhagic Fever, Ebola , Animals , Mice , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Bispecific/therapeutic use , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/virology , Antibodies, Viral/immunology , Humans , Filoviridae/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Antibodies, Monoclonal/immunology , Female , Mice, Inbred BALB C , Filoviridae Infections/immunology , Filoviridae Infections/therapy , Filoviridae Infections/prevention & control
4.
Clin Infect Dis ; 74(6): 1081-1084, 2022 03 23.
Article in English | MEDLINE | ID: mdl-34245255

ABSTRACT

The clinical significance of severe acute respiratory syndrome coronavirus 2 (SARS CoV-2) RNA in stool remains uncertain. We found that extrapulmonary dissemination of infection to the gastrointestinal tract, assessed by the presence of SARS-CoV-2 RNA in stool, is associated with decreased coronavirus disease 2019 (COVID-19) survival. Measurement of SARS-CoV-2 RNA in stool may have utility for clinical risk assessment.


Subject(s)
COVID-19 , SARS-CoV-2 , Feces , Gastrointestinal Tract , Humans , RNA, Viral , SARS-CoV-2/genetics
5.
J Am Chem Soc ; 143(42): 17615-17621, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34647745

ABSTRACT

Cellular binding and entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are mediated by its spike glycoprotein (S protein), which binds with not only the human angiotensin-converting enzyme 2 (ACE2) receptor but also glycosaminoglycans such as heparin. Cell membrane-coated nanoparticles ("cellular nanosponges") mimic the host cells to attract and neutralize SARS-CoV-2 through natural cellular receptors, leading to a broad-spectrum antiviral strategy. Herein, we show that increasing surface heparin density on the cellular nanosponges can promote their inhibition against SARS-CoV-2. Specifically, cellular nanosponges are made with azido-expressing host cell membranes followed by conjugating heparin to the nanosponge surfaces. Cellular nanosponges with a higher heparin density have a larger binding capacity with viral S proteins and a significantly higher inhibition efficacy against SARS-CoV-2 infectivity. Overall, surface glycan engineering of host-mimicking cellular nanosponges is a facile method to enhance SARS-CoV-2 inhibition. This approach can be readily generalized to promote the inhibition of other glycan-dependent viruses.


Subject(s)
COVID-19 Drug Treatment , Heparin/administration & dosage , Nanostructures/therapeutic use , Polysaccharides/administration & dosage , SARS-CoV-2/metabolism , COVID-19/virology , Heparin/metabolism , Humans , Polysaccharides/metabolism
6.
Nano Lett ; 20(7): 5570-5574, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32551679

ABSTRACT

We report cellular nanosponges as an effective medical countermeasure to the SARS-CoV-2 virus. Two types of cellular nanosponges are made of the plasma membranes derived from human lung epithelial type II cells or human macrophages. These nanosponges display the same protein receptors, both identified and unidentified, required by SARS-CoV-2 for cellular entry. It is shown that, following incubation with the nanosponges, SARS-CoV-2 is neutralized and unable to infect cells. Crucially, the nanosponge platform is agnostic to viral mutations and potentially viral species, as well. As long as the target of the virus remains the identified host cell, the nanosponges will be able to neutralize the virus.


Subject(s)
Betacoronavirus , Coronavirus Infections/prevention & control , Nanostructures , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , Betacoronavirus/pathogenicity , Betacoronavirus/physiology , COVID-19 , Cell Membrane/virology , Coronavirus Infections/virology , Epithelial Cells/virology , Host Microbial Interactions , Humans , Lung/cytology , Lung/virology , Macrophages/virology , Nanostructures/ultrastructure , Nanotechnology , Pneumonia, Viral/virology , Receptors, Virus/physiology , SARS-CoV-2 , Virus Internalization
7.
PLoS Pathog ; 14(9): e1007322, 2018 09.
Article in English | MEDLINE | ID: mdl-30265711

ABSTRACT

Lassa fever virus (LASV) is endemic in West Africa and causes severe hemorrhagic fever and sensorineural hearing loss. We identified a small molecule inhibitor of LASV and used it to analyze the mechanism of entry. Using a photo-reactive analog that retains antiviral activity as a probe, we identified the inhibitor target as lysosome-associated membrane protein 1 (LAMP1), a host factor that binds to the LASV glycoprotein (GP) during infection. We found that LAMP1 binding to LASV GP is cholesterol-dependent, and that the inhibitor blocks infection by competing with cholesterol in LAMP1. Mutational analysis of a docking-based model identified a putative inhibitor binding site in the cholesterol-binding pocket within the LAMP1 domain that binds GP. These findings identify a critical role for cholesterol in LASV entry and a potential target for therapeutic intervention.


Subject(s)
Cholesterol/metabolism , Lassa virus/physiology , Lassa virus/pathogenicity , Lysosomal Membrane Proteins/physiology , Receptors, Virus/physiology , Adamantane/analogs & derivatives , Adamantane/chemistry , Adamantane/pharmacology , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Cell Line , Chlorocebus aethiops , HEK293 Cells , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Lassa Fever/etiology , Lassa virus/drug effects , Lysosomal Membrane Proteins/antagonists & inhibitors , Lysosomal Membrane Proteins/genetics , Models, Molecular , Mutation , Protein Stability , Protein Structure, Tertiary , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/genetics , Vero Cells , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/physiology , Virus Internalization/drug effects
8.
Proc Natl Acad Sci U S A ; 113(16): 4458-63, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27044104

ABSTRACT

Countermeasures against potential biothreat agents remain important to US Homeland Security, and many of these pharmaceuticals could have dual use in the improvement of global public health. Junin virus, the causative agent of Argentine hemorrhagic fever (AHF), is an arenavirus identified as a category A high-priority agent. There are no Food and Drug Administration (FDA) approved drugs available for preventing or treating AHF, and the current treatment option is limited to administration of immune plasma. Whereas immune plasma demonstrates the feasibility of passive immunotherapy, it is limited in quantity, variable in quality, and poses safety risks such as transmission of transfusion-borne diseases. In an effort to develop a monoclonal antibody (mAb)-based alternative to plasma, three previously described neutralizing murine mAbs were expressed as mouse-human chimeric antibodies and evaluated in the guinea pig model of AHF. These mAbs provided 100% protection against lethal challenge when administered 2 d after infection (dpi), and one of them (J199) was capable of providing 100% protection when treatment was initiated 6 dpi and 92% protection when initiated 7 dpi. The efficacy of J199 is superior to that previously described for all other evaluated drugs, and its high potency suggests that mAbs like J199 offer an economical alternative to immune plasma and an effective dual use (bioterrorism/public health) therapeutic.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Viral/pharmacology , Hemorrhagic Fever, American/drug therapy , Hemorrhagic Fever, American/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Disease Models, Animal , Drug Evaluation, Preclinical , Guinea Pigs , Humans , Junin virus , Mice , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology
9.
J Infect Dis ; 218(suppl_5): S486-S495, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30476250

ABSTRACT

The domestic ferret is a uniformly lethal model of infection for 3 species of Ebolavirus known to be pathogenic in humans. Reagents to systematically analyze the ferret host response to infection are lacking; however, the recent publication of a draft ferret genome has opened the potential for transcriptional analysis of ferret models of disease. In this work, we present comparative analysis of longitudinally sampled blood taken from ferrets and nonhuman primates infected with lethal doses of the Makona variant of Zaire ebolavirus. Strong induction of proinflammatory and prothrombotic signaling programs were present in both ferrets and nonhuman primates, and both transcriptomes were similar to previously published datasets of fatal cases of human Ebola virus infection.


Subject(s)
Ferrets/genetics , Hemorrhagic Fever, Ebola/genetics , Macaca mulatta/genetics , Transcriptome , Animals , Cytokines/genetics , Disease Models, Animal , Female , Humans
10.
J Infect Dis ; 218(suppl_5): S592-S596, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30016444

ABSTRACT

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


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

ABSTRACT

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


Subject(s)
Antibodies, Viral/therapeutic use , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/drug therapy , Immunoglobulin G/therapeutic use , Animals , Cattle , Chlorocebus aethiops , Female , Humans , Macaca mulatta , Male , Vero Cells
12.
J Infect Dis ; 215(9): 1416-1420, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28368541

ABSTRACT

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


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Interferon-beta/pharmacology , Interferon-gamma/pharmacology , Animals , Cell Line, Tumor , Chlorocebus aethiops , Drug Synergism , Humans , Vero Cells
13.
J Infect Dis ; 213(1): 57-60, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26153408

ABSTRACT

A 3.5-year-old adult female rhesus macaque (Macaca mulatta) manifested swelling of the left upper eyelid and conjunctiva and a decline in clinical condition 18 days following intramuscular challenge with Ebola virus (EBOV; Kikwit-1995), after apparent clinical recovery. Histologic lesions with strong EBOV antigen staining were noted in the left eye (scleritis, conjunctivitis, and peri-optic neuritis), brain (choriomeningoencephalitis), stomach, proximal duodenum, and pancreas. Spleen, liver, and adrenal glands, common targets for acute infection, appeared histologically normal with no evidence of EBOV immunoreactivity. These findings may provide important insight for understanding sequelae seen in West African survivors of Ebola virus disease.


Subject(s)
Brain/pathology , Central Nervous System Viral Diseases/pathology , Conjunctivitis/pathology , Ebolavirus/isolation & purification , Hemorrhagic Fever, Ebola/pathology , Scleritis/pathology , Animals , Brain/virology , Central Nervous System Viral Diseases/etiology , Central Nervous System Viral Diseases/virology , Conjunctivitis/etiology , Conjunctivitis/virology , Disease Models, Animal , Female , Hemorrhagic Fever, Ebola/complications , Hemorrhagic Fever, Ebola/physiopathology , Macaca mulatta , Necrosis , Scleritis/etiology , Scleritis/virology
14.
BMC Genomics ; 17: 707, 2016 09 05.
Article in English | MEDLINE | ID: mdl-27595844

ABSTRACT

BACKGROUND: Ebola virus is the causative agent of a severe syndrome in humans with a fatality rate that can approach 90 %. During infection, the host immune response is thought to become dysregulated, but the mechanisms through which this happens are not entirely understood. In this study, we analyze RNA sequencing data to determine the host response to Ebola virus infection in circulating immune cells. RESULTS: Approximately half of the 100 genes with the strongest early increases in expression were interferon-stimulated genes, such as ISG15, OAS1, IFIT2, HERC5, MX1 and DHX58. Other highly upregulated genes included cytokines CXCL11, CCL7, IL2RA, IL2R1, IL15RA, and CSF2RB, which have not been previously reported to change during Ebola virus infection. Comparing this response in two different models of exposure (intramuscular and aerosol) revealed a similar signature of infection. The strong innate response in the aerosol model was seen not only in circulating cells, but also in primary and secondary target tissues. Conversely, the innate immune response of vaccinated macaques was almost non-existent. This suggests that the innate response is a major aspect of the cellular response to Ebola virus infection in multiple tissues. CONCLUSIONS: Ebola virus causes a severe infection in humans that is associated with high mortality. The host immune response to virus infection is thought to be an important aspect leading to severe pathology, but the components of this overactive response are not well characterized. Here, we analyzed how circulating immune cells respond to the virus and found that there is a strong innate response dependent on active virus replication. This finding is in stark contrast to in vitro evidence showing a suppression of innate immune signaling, and it suggests that the strong innate response we observe in infected animals may be an important contributor to pathogenesis.


Subject(s)
Ebolavirus/physiology , Hemorrhagic Fever, Ebola/immunology , Immunity, Innate , Leukocytes, Mononuclear/immunology , Animals , Ebolavirus/pathogenicity , Gene Expression Profiling/methods , Gene Expression Regulation , Gene Regulatory Networks , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/virology , Leukocytes, Mononuclear/metabolism , Macaca/virology , Mice , Sequence Analysis, RNA/methods , Virus Replication
15.
Emerg Infect Dis ; 22(8): 1448-51, 2016 08.
Article in English | MEDLINE | ID: mdl-27191483

ABSTRACT

Nanopore sequencing, a novel genomics technology, has potential applications for routine biosurveillance, clinical diagnosis, and outbreak investigation of virus infections. Using rapid sequencing of unamplified RNA/cDNA hybrids, we identified Venezuelan equine encephalitis virus and Ebola virus in 3 hours from sample receipt to data acquisition, demonstrating a fieldable technique for RNA virus characterization.


Subject(s)
DNA, Complementary/genetics , Ebolavirus/isolation & purification , Encephalitis Virus, Venezuelan Equine/isolation & purification , Nucleic Acid Amplification Techniques/methods , RNA Viruses/isolation & purification , RNA/genetics , Nanopores , Time Factors
16.
J Virol ; 89(19): 9875-85, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26202230

ABSTRACT

UNLABELLED: Marburg virus (MARV) infection is a lethal hemorrhagic fever for which no licensed vaccines or therapeutics are available. Development of appropriate medical countermeasures requires a thorough understanding of the interaction between the host and the pathogen and the resulting disease course. In this study, 15 rhesus macaques were sequentially sacrificed following aerosol exposure to the MARV variant Angola, with longitudinal changes in physiology, immunology, and histopathology used to assess disease progression. Immunohistochemical evidence of infection and resulting histopathological changes were identified as early as day 3 postexposure (p.e.). The appearance of fever in infected animals coincided with the detection of serum viremia and plasma viral genomes on day 4 p.e. High (>10(7) PFU/ml) viral loads were detected in all major organs (lung, liver, spleen, kidney, brain, etc.) beginning day 6 p.e. Clinical pathology findings included coagulopathy, leukocytosis, and profound liver destruction as indicated by elevated liver transaminases, azotemia, and hypoalbuminemia. Altered cytokine expression in response to infection included early increases in Th2 cytokines such as interleukin 10 (IL-10) and IL-5 and late-stage increases in Th1 cytokines such as IL-2, IL-15, and granulocyte-macrophage colony-stimulating factor (GM-CSF). This study provides a longitudinal examination of clinical disease of aerosol MARV Angola infection in the rhesus macaque model. IMPORTANCE: In this study, we carefully analyzed the timeline of Marburg virus infection in nonhuman primates in order to provide a well-characterized model of disease progression following aerosol exposure.


Subject(s)
Cytokines/blood , Host-Pathogen Interactions , Marburg Virus Disease/physiopathology , Marburgvirus/pathogenicity , Aerosols , Animals , Disease Progression , Immunohistochemistry , Longitudinal Studies , Macaca mulatta , Marburg Virus Disease/blood , Time Factors , Viral Load
17.
Inhal Toxicol ; 28(14): 670-676, 2016 12.
Article in English | MEDLINE | ID: mdl-27919178

ABSTRACT

For inhalational studies and aerosol exposures to viruses, head-out plethysmography acquisition has been traditionally used for the determination of estimated inhaled dose in anesthetized nonhuman primates prior to or during an aerosol exposure. A pressure drop across a pneumotachograph is measured within a sealed chamber during inspiration/exhalation of the nonhuman primate, generating respiratory values and breathing frequencies. Due to the fluctuation of depth of anesthesia, pre-exposure respiratory values can be variable, leading to less precise and accurate dosing calculations downstream. Although an anesthesia infusion pump may help stabilize the depth of sedation, pumps are difficult to use within a sealed head-out plethysmography chamber. Real-time, head-out plethysmography acquisition could increase precision and accuracy of the measurements, but the bulky equipment needed for head-out plethysmography precludes real-time use inside a Class III biological safety cabinet, where most aerosol exposures occur. However, the respiratory inductive plethysmography (RIP) acquisition method measures the same respiratory parameters by detecting movement of the chest and abdomen during breathing using two elastic bands within the Class III biological safety cabinet. As respiratory values are relayed to a computer for software integration and analysis real-time, adjustment of aerosol exposure duration is based on the depth of sedation of the animal. The objective of this study was to compare values obtained using two methodologies (pre-exposure head-out plethysmography and real-time RIP). Transitioning to RIP technology with real-time acquisition provides more consistent, precise, and accurate aerosol dosing by reducing reported errors in respiratory values from anesthesia variability when using pre-exposure head-out plethysmography acquisition.


Subject(s)
Plethysmography/methods , Respiration , Toxicity Tests/methods , Administration, Inhalation , Aerosols/administration & dosage , Anesthesia , Animals , Containment of Biohazards , Female , Macaca mulatta , Male , Tidal Volume
18.
BMC Genomics ; 16: 95, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25765146

ABSTRACT

BACKGROUND: MicroRNAs (miRNAs) represent new and potentially informative diagnostic targets for disease detection and prognosis. However, little work exists documenting the effect of TRIzol, a common viral inactivation and nucleic acid extraction reagent, on miRNA purification. Here, we developed an optimized protocol for miRNA extraction from plasma samples by evaluating five different RNA extraction kits, TRIzol phase separation, purification additives, and initial plasma sample volume. This method was then used for downstream profiling of plasma miRNAs found in archived samples from one nonhuman primate (NHP) experimentally challenged with Ebola virus by the aerosol route. RESULTS: Comparison of real-time RT-PCR results for spiked-in and endogenous miRNA sequences determined extraction efficiencies from five different RNA purification kits. These experiments showed that 50 µL plasma processed using the QIAGEN miRNeasy Mini Kit with 5 µg of glycogen as a co-precipitant yielded the highest recovery of endogenous miRNAs. Using this optimized protocol, miRNAs from archived plasma samples of one rhesus macaque challenged with aerosolized Ebola virus was profiled using a targeted real-time PCR array. A total of 519 of the 752 unique miRNAs assayed were present in the plasma samples at day 0 and day 7 (time of death) post-exposure. Statistical analyses revealed 25 sequences significantly up- or down-regulated between day 0 and day 7 post infection, validating the utility of the extraction method for plasma miRNA profiling. CONCLUSIONS: This study contributes to the knowledgebase of circulating miRNA extraction methods and expands on the potential applications of cell-free miRNA profiling for diagnostics and pathogenesis studies. Specifically, we optimized an extraction protocol for miRNAs from TRIzol-inactivated plasma samples that can be used for highly pathogenic viruses.


Subject(s)
Hemorrhagic Fever, Ebola/genetics , MicroRNAs/genetics , MicroRNAs/isolation & purification , Animals , Ebolavirus/genetics , Guanidines/pharmacology , Hemorrhagic Fever, Ebola/blood , Hemorrhagic Fever, Ebola/virology , Macaca mulatta/blood , Macaca mulatta/genetics , Macaca mulatta/virology , MicroRNAs/blood , Phenols/pharmacology
19.
BMC Genomics ; 15: 960, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25377889

ABSTRACT

BACKGROUND: Lassa virus and Marburg virus are two causative agents of viral hemorrhagic fever. Their diagnosis is difficult because patients infected with either pathogen present similar nonspecific symptoms early after infection. Current diagnostic tests are based on detecting viral proteins or nucleic acids in the blood, but these cannot be found during the early stages of disease, before the virus starts replicating in the blood. Using the transcriptional response of the host during infection can lead to earlier diagnoses compared to those of traditional methods. RESULTS: In this study, we use RNA sequencing to obtain a high-resolution view of the in vivo transcriptional dynamics of peripheral blood mononuclear cells (PBMCs) throughout both types of infection. We report a subset of host mRNAs, including heat-shock proteins like HSPA1B, immunoglobulins like IGJ, and cell adhesion molecules like SIGLEC1, whose differences in expression are strong enough to distinguish Lassa infection from Marburg infection in non-human primates. We have validated these infection-specific expression differences by using microarrays on a larger set of samples, and by quantifying the expression of individual genes using RT-PCR. CONCLUSIONS: These results suggest that host transcriptional signatures are correlated with specific viral infections, and that they can be used to identify highly pathogenic viruses during the early stages of disease, before standard detection methods become effective.


Subject(s)
Hemorrhagic Fevers, Viral/veterinary , Host-Pathogen Interactions/genetics , Lassa virus , Marburgvirus , Monkey Diseases/genetics , Transcription, Genetic , Animals , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Interferon Type I/pharmacology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Macaca fascicularis , Male , Monkey Diseases/diagnosis , Monkey Diseases/virology , Reproducibility of Results , Sequence Analysis, RNA
20.
J Virol ; 87(7): 3801-14, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23345511

ABSTRACT

Antigen-presenting cells (APCs) are critical targets of Ebola virus (EBOV) infection in vivo. However, the susceptibility of monocytes to infection is controversial. Studies indicate productive monocyte infection, and yet monocytes are also reported to be resistant to EBOV GP-mediated entry. In contrast, monocyte-derived macrophages and dendritic cells are permissive for both EBOV entry and replication. Here, freshly isolated monocytes are demonstrated to indeed be refractory to EBOV entry. However, EBOV binds monocytes, and delayed entry occurs during monocyte differentiation. Cultured monocytes spontaneously downregulate the expression of viral entry restriction factors such as interferon-inducible transmembrane proteins, while upregulating the expression of critical EBOV entry factors cathepsin B and NPC1. Moreover, these processes are accelerated by EBOV infection. Finally, ectopic expression of NPC1 is sufficient to rescue entry into an undifferentiated, normally nonpermissive monocytic cell line. These results define the molecular basis for infection of APCs and suggest means to limit APC infection.


Subject(s)
Cell Differentiation/physiology , Ebolavirus/physiology , Monocytes/virology , Virus Attachment , Virus Internalization , Carrier Proteins/metabolism , Cathepsin B/metabolism , DNA Primers/genetics , Dendritic Cells/virology , Flow Cytometry , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins , Macrophages/virology , Membrane Glycoproteins/metabolism , Monocytes/physiology , Niemann-Pick C1 Protein , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL