Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
1.
Viruses ; 7(2): 559-89, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25674766

ABSTRACT

Sin Nombre Hantavirus (SNV, Bunyaviridae Hantavirus) is a Category A pathogen that causes Hantavirus Cardiopulmonary Syndrome (HCPS) with case fatality ratios generally ranging from 30% to 50%. HCPS is characterized by vascular leakage due to dysregulation of the endothelial barrier function. The loss of vascular integrity results in non-cardiogenic pulmonary edema, shock, multi-organ failure and death. Using Electric Cell-substrate Impedance Sensing (ECIS) measurements, we found that plasma samples drawn from University of New Mexico Hospital patients with serologically-confirmed HCPS, induce loss of cell-cell adhesion in confluent epithelial and endothelial cell monolayers grown in ECIS cultureware. We show that the loss of cell-cell adhesion is sensitive to both thrombin and plasmin inhibitors in mild cases, and to thrombin only inhibition in severe cases, suggesting an increasing prothrombotic state with disease severity. A proteomic profile (2D gel electrophoresis and mass spectrometry) of HCPS plasma samples in our cohort revealed robust antifibrinolytic activity among terminal case patients. The prothrombotic activity is highlighted by acute ≥30 to >100 fold increases in active plasminogen activator inhibitor (PAI-1) which, preceded death of the subjects within 48 h. Taken together, this suggests that PAI-1 might be a response to the severe pathology as it is expected to reduce plasmin activity and possibly thrombin activity in the terminal patients.


Subject(s)
Cytokines/blood , Hantavirus Pulmonary Syndrome/blood , Hantavirus Pulmonary Syndrome/virology , Plasminogen Activator Inhibitor 1/blood , Sin Nombre virus/physiology , Thrombin/metabolism , Animals , Blood Proteins/metabolism , Chlorocebus aethiops , Cytopathogenic Effect, Viral , Endothelial Cells/metabolism , Endothelial Cells/virology , Hantavirus Pulmonary Syndrome/diagnosis , Hantavirus Pulmonary Syndrome/immunology , Humans , Models, Biological , Proteome , Proteomics/methods , Retrospective Studies , Severity of Illness Index , Vero Cells
2.
Antivir Ther ; 20(4): 377-86, 2015.
Article in English | MEDLINE | ID: mdl-25316807

ABSTRACT

BACKGROUND: In Chile, Andes virus (ANDV) is the sole aetiological agent of hantavirus cardiopulmonary syndrome (HCPS) with mean annual incidence of 55 cases, 32% case fatality rate (CFR) and no specific treatment. Neutralizing antibody (NAb) titres at hospital admission correlate inversely with HCPS severity. We designed an open trial to explore safety and efficacy and evaluate pharmacokinetics of immune plasma as a treatment strategy for this disease. METHODS: We performed plasmapheresis on donors at least 6 months after HCPS and measured NAb titres through a focus-reduction neutralization test. Subjects admitted to 10 study sites with suspected/confirmed HCPS were eligible for treatment with immune plasma by intravenous infusion at an ANDV NAb dose of 5,000 U/kg. HCPS was confirmed through immunoglobulin M serology or reverse transcriptase-PCR. The main outcome was mortality within 30 days. RESULTS: From 2008-2012, we enrolled and treated 32 cases and confirmed HCPS in 29. CFR of hantavirus plasma-treated cases was 4/29 (14%); CFR of non-treated cases in the same period in Chile was 63/199 (32%; P=0.049, OR=0.35, CI=0.12, 0.99); CFR of non-treated cases at the same study sites between 2005-2012 was 18/66 (27%; (P=0.15, OR=0.43, CI=0.14, 1.34) and CFR in a previous methylprednisolone treatment study was 20/60 (33%; P=0.052, OR=0.32, CI=0.10, 1.00). We detected no serious adverse events associated to plasma infusion. Plasma NAb titres reached in recipients were variable and viral load remained stable. CONCLUSIONS: Human ANDV immune plasma infusion appears safe for HCPS. We observed a decrease in CFR in treated cases with borderline significance that will require further studies for confirmation.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Hantavirus Infections/therapy , Immune Sera/pharmacology , RNA, Viral/antagonists & inhibitors , Adult , Female , Glucocorticoids/therapeutic use , Orthohantavirus/drug effects , Orthohantavirus/growth & development , Orthohantavirus/immunology , Hantavirus Infections/immunology , Hantavirus Infections/mortality , Hantavirus Infections/virology , Heart/drug effects , Heart/physiopathology , Heart/virology , Humans , Lung/drug effects , Lung/immunology , Lung/pathology , Lung/virology , Male , Methylprednisolone/therapeutic use , Middle Aged , Neutralization Tests , Plasmapheresis , RNA, Viral/blood , RNA, Viral/immunology , Severity of Illness Index , Survival Analysis , Syndrome , Viral Load/drug effects
3.
Proc Natl Acad Sci U S A ; 111(19): 7114-9, 2014 May 13.
Article in English | MEDLINE | ID: mdl-24778254

ABSTRACT

The pathophysiology of hantavirus pulmonary syndrome (HPS) remains unclear because of a lack of surrogate disease models with which to perform pathogenesis studies. Nonhuman primates (NHP) are considered the gold standard model for studying the underlying immune activation/suppression associated with immunopathogenic viruses such as hantaviruses; however, to date an NHP model for HPS has not been described. Here we show that rhesus macaques infected with Sin Nombre virus (SNV), the primary etiological agent of HPS in North America, propagated in deer mice develop HPS, which is characterized by thrombocytopenia, leukocytosis, and rapid onset of respiratory distress caused by severe interstitial pneumonia. Despite establishing a systemic infection, SNV differentially activated host responses exclusively in the pulmonary endothelium, potentially the mechanism leading to acute severe respiratory distress. This study presents a unique chronological characterization of SNV infection and provides mechanistic data into the pathophysiology of HPS in a closely related surrogate animal model. We anticipate this model will advance our understanding of HPS pathogenesis and will greatly facilitate research toward the development of effective therapeutics and vaccines against hantaviral diseases.


Subject(s)
Disease Models, Animal , Hantavirus Pulmonary Syndrome/physiopathology , Macaca mulatta/virology , Monkey Diseases/virology , Peromyscus/virology , Sin Nombre virus/genetics , Animals , Chlorocebus aethiops , Hantavirus Pulmonary Syndrome/diagnostic imaging , Hantavirus Pulmonary Syndrome/transmission , Lung/diagnostic imaging , Lung/virology , Molecular Sequence Data , Monkey Diseases/physiopathology , Monkey Diseases/transmission , North America , RNA, Viral/genetics , Radiography , Vero Cells , Viremia/physiopathology
4.
Viruses ; 6(3): 1091-111, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24618810

ABSTRACT

Decay accelerating factor (DAF/CD55) is targeted by many pathogens for cell entry. It has been implicated as a co-receptor for hantaviruses. To examine the binding of hantaviruses to DAF, we describe the use of Protein G beads for binding human IgG Fc domain-functionalized DAF ((DAF)2-Fc). When mixed with Protein G beads the resulting DAF beads can be used as a generalizable platform for measuring kinetic and equilibrium binding constants of DAF binding targets. The hantavirus interaction has high affinity (24-30 nM; k(on) ~ 105 M⁻¹ s⁻¹, k(off) ~ 0.0045 s⁻¹). The bivalent (DAF)2-Fc/SNV data agree with hantavirus binding to DAF expressed on Tanoue B cells (K(d) = 14.0 nM). Monovalent affinity interaction between SNV and recombinant DAF of 58.0 nM is determined from competition binding. This study serves a dual purpose of presenting a convenient and quantitative approach of measuring binding affinities between DAF and the many cognate viral and bacterial ligands and providing new data on the binding constant of DAF and Sin Nombre hantavirus. Knowledge of the equilibrium binding constant allows for the determination of the relative fractions of bound and free virus particles in cell entry assays. This is important for drug discovery assays for cell entry inhibitors.


Subject(s)
CD55 Antigens/metabolism , Receptors, Virus/metabolism , Sin Nombre virus/physiology , Virus Attachment , Humans , Microspheres
5.
Am J Trop Med Hyg ; 89(3): 489-94, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23836565

ABSTRACT

In Panama, hantavirus pulmonary syndrome (HPS) is caused by Choclo virus, a species phylogenetically related to Andes and Maporal viruses. Up to 60% of the population has been positive for specific serum antibody in community-based surveys, but mortality is very uncommon. In four western Panama clinics, we tested individuals presenting with a severe febrile prodrome for acute hantavirus (HV) infection by immunoglobulin M enzyme-linked immunosorbent assay and reverse transcription polymerase chain reaction as well as clinically similar infections, such as dengue and leptospirosis. From 2006 to 2009, at least 21% of 117 patients diagnosed with HV infection had HV Fever (HF) with no evidence of pulmonary edema (no respiratory distress or radiographic lung infiltrates), and 44% of patients had very mild HPS (radiographic pulmonary edema but no respiratory insufficiency). HV infection caused by Choclo virus in Panama presents often as HF, which contrasts with HV in the Americas but is consistent with the high seroprevalence in endemic regions.


Subject(s)
Hantavirus Pulmonary Syndrome/diagnosis , Hantavirus Pulmonary Syndrome/epidemiology , Hemorrhagic Fever with Renal Syndrome/diagnosis , Hemorrhagic Fever with Renal Syndrome/epidemiology , Orthohantavirus/isolation & purification , Adolescent , Adult , Aged , Antibodies, Viral/blood , Child , Child, Preschool , Female , Orthohantavirus/genetics , Humans , Immunoglobulin M/blood , Male , Middle Aged , Panama/epidemiology , RNA, Viral/isolation & purification , Seroepidemiologic Studies , Young Adult
6.
mBio ; 4(2): e00216-13, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23572554

ABSTRACT

UNLABELLED: Hepatitis C virus (HCV) and human pegivirus (HPgV or GB virus C) are globally distributed and infect 2 to 5% of the human population. The lack of tractable-animal models for these viruses, in particular for HCV, has hampered the study of infection, transmission, virulence, immunity, and pathogenesis. To address this challenge, we searched for homologous viruses in small mammals, including wild rodents. Here we report the discovery of several new hepaciviruses (HCV-like viruses) and pegiviruses (GB virus-like viruses) that infect wild rodents. Complete genome sequences were acquired for a rodent hepacivirus (RHV) found in Peromyscus maniculatus and a rodent pegivirus (RPgV) found in Neotoma albigula. Unique genomic features and phylogenetic analyses confirmed that these RHV and RPgV variants represent several novel virus species in the Hepacivirus and Pegivirus genera within the family Flaviviridae. The genetic diversity of the rodent hepaciviruses exceeded that observed for hepaciviruses infecting either humans or non-primates, leading to new insights into the origin, evolution, and host range of hepaciviruses. The presence of genes, encoded proteins, and translation elements homologous to those found in human hepaciviruses and pegiviruses suggests the potential for the development of new animal systems with which to model HCV pathogenesis, vaccine design, and treatment. IMPORTANCE: The genetic and biological characterization of animal homologs of human viruses provides insights into the origins of human infections and enhances our ability to study their pathogenesis and explore preventive and therapeutic interventions. Horses are the only reported host of nonprimate homologs of hepatitis C virus (HCV). Here, we report the discovery of HCV-like viruses in wild rodents. The majority of HCV-like viruses were found in deer mice (Peromyscus maniculatus), a small rodent used in laboratories to study viruses, including hantaviruses. We also identified pegiviruses in rodents that are distinct from the pegiviruses found in primates, bats, and horses. These novel viruses may enable the development of small-animal models for HCV, the most common infectious cause of liver failure and hepatocellular carcinoma after hepatitis B virus, and help to explore the health relevance of the highly prevalent human pegiviruses.


Subject(s)
Flaviviridae/classification , Flaviviridae/isolation & purification , Genome, Viral , Peromyscus/virology , Sigmodontinae/virology , Animals , Cluster Analysis , Flaviviridae/genetics , Genetic Variation , Molecular Sequence Data , Phylogeny , RNA, Viral/genetics , Sequence Analysis, DNA
7.
J Biol Chem ; 288(12): 8531-8543, 2013 Mar 22.
Article in English | MEDLINE | ID: mdl-23382385

ABSTRACT

Cdc42 plays important roles in cytoskeleton organization, cell cycle progression, signal transduction, and vesicle trafficking. Overactive Cdc42 has been implicated in the pathology of cancers, immune diseases, and neuronal disorders. Therefore, Cdc42 inhibitors would be useful in probing molecular pathways and could have therapeutic potential. Previous inhibitors have lacked selectivity and trended toward toxicity. We report here the characterization of a Cdc42-selective guanine nucleotide binding lead inhibitor that was identified by high throughput screening. A second active analog was identified via structure-activity relationship studies. The compounds demonstrated excellent selectivity with no inhibition toward Rho and Rac in the same GTPase family. Biochemical characterization showed that the compounds act as noncompetitive allosteric inhibitors. When tested in cellular assays, the lead compound inhibited Cdc42-related filopodia formation and cell migration. The lead compound was also used to clarify the involvement of Cdc42 in the Sin Nombre virus internalization and the signaling pathway of integrin VLA-4. Together, these data present the characterization of a novel Cdc42-selective allosteric inhibitor and a related analog, the use of which will facilitate drug development targeting Cdc42-related diseases and molecular pathway studies that involve GTPases.


Subject(s)
Enzyme Inhibitors/pharmacology , Molecular Probes/pharmacology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , cdc42 GTP-Binding Protein/antagonists & inhibitors , 3T3 Cells , Allosteric Regulation , Animals , Antiviral Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Humans , Integrin alpha4beta1/antagonists & inhibitors , Integrin alpha4beta1/physiology , Mice , Oligopeptides/metabolism , Phenylurea Compounds/metabolism , Protein Binding , Pseudopodia/drug effects , Sin Nombre virus/physiology , Structure-Activity Relationship , Virus Internalization/drug effects , cdc42 GTP-Binding Protein/chemistry , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rhoA GTP-Binding Protein/metabolism
8.
PLoS One ; 8(1): e55310, 2013.
Article in English | MEDLINE | ID: mdl-23383148

ABSTRACT

New World hantaviruses can cause hantavirus cardiopulmonary syndrome with high mortality in humans. Distinct virus species are hosted by specific rodent reservoirs, which also serve as the vectors. Although regional spillover has been documented, it is unknown whether rodent reservoirs are competent for infection by hantaviruses that are geographically separated, and known to have related, but distinct rodent reservoir hosts. We show that Andes virus (ANDV) of South America, carried by the long tailed pygmy rice rat (Oligoryzomys longicaudatus), infects and replicates in vitro and in vivo in the deer mouse (Peromyscus maniculatus), the reservoir host of Sin Nombre virus (SNV), found in North America. In experimentally infected deer mice, viral RNA was detected in the blood, lung, heart and spleen, but virus was cleared by 56 days post inoculation (dpi). All of the inoculated deer mice mounted a humoral immune response by 14 dpi, and produced measurable amounts of neutralizing antibodies by 21 dpi. An up-regulation of Ccl3, Ccl4, Ccl5, and Tgfb, a strong CD4⁺ T-cell response, and down-regulation of Il17, Il21 and Il23 occurred during infection. Infection was transient with an absence of clinical signs or histopathological changes. This is the first evidence that ANDV asymptomatically infects, and is immunogenic in deer mice, a non-natural host species of ANDV. Comparing the immune response in this model to that of the immune response in the natural hosts upon infection with their co-adapted hantaviruses may help clarify the mechanisms governing persistent infection in the natural hosts of hantaviruses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Disease Reservoirs/virology , Disease Vectors , Gene Expression Regulation/immunology , Hantavirus Infections/immunology , Orthohantavirus , Virus Replication/physiology , Animals , Antibodies, Viral/blood , Arvicolinae , Chemokines/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/genetics , Hantavirus Infections/physiopathology , Immunohistochemistry , Microscopy, Confocal , Microscopy, Fluorescence , Neutralization Tests , Peromyscus , Rats , Real-Time Polymerase Chain Reaction , South America
9.
J Virol ; 86(18): 10015-27, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22787210

ABSTRACT

Deer mice are the principal reservoir hosts of Sin Nombre virus, the etiologic agent of most hantavirus cardiopulmonary syndrome cases in North America. Infection of deer mice results in persistence without conspicuous pathology, and most, if not all, infected mice remain infected for life, with periods of viral shedding. The kinetics of viral load, histopathology, virus distribution, and immune gene expression in deer mice were examined. Viral antigen was detected as early as 5 days postinfection and peaked on day 15 in the lungs, hearts, kidneys, and livers. Viral RNA levels varied substantially but peaked on day 15 in the lungs and heart, and antinucleocapsid IgG antibodies appeared in some animals on day 10, but a strong neutralizing antibody response failed to develop during the 20-day experiment. No clinical signs of disease were observed in any of the infected deer mice. Most genes were repressed on day 2, suggesting a typical early downregulation of gene expression often observed in viral infections. Several chemokine and cytokine genes were elevated, and markers of a T cell response occurred but then declined days later. Splenic transforming growth factor beta (TGF-ß) expression was elevated early in infection, declined, and then was elevated again late in infection. Together, these data suggest that a subtle immune response that fails to clear the virus occurs in deer mice.


Subject(s)
Peromyscus/immunology , Peromyscus/virology , Sin Nombre virus/immunology , Sin Nombre virus/pathogenicity , Animals , Antibodies, Viral/blood , Base Sequence , Cytokines/genetics , DNA Primers/genetics , Disease Reservoirs/virology , Female , Gene Expression , Hantavirus Pulmonary Syndrome/genetics , Hantavirus Pulmonary Syndrome/immunology , Hantavirus Pulmonary Syndrome/pathology , Hantavirus Pulmonary Syndrome/virology , Humans , Immunoglobulin G/blood , Kinetics , Male , RNA, Viral/genetics , RNA, Viral/metabolism , Sin Nombre virus/genetics , Viral Load , Virus Shedding
11.
Emerg Infect Dis ; 17(10): 1936-9, 2011 Oct.
Article in English | MEDLINE | ID: mdl-22000376

ABSTRACT

During 2001-2007, to determine incidence of all hantavirus infections, including those without pulmonary syndrome, in western Panama, we conducted 11 communitywide surveys. Among 1,129 persons, antibody prevalence was 16.5%-60.4%. Repeat surveys of 476 found that patients who seroconverted outnumbered patients with hantavirus pulmonary syndrome by 14 to 1.


Subject(s)
Hantavirus Infections/epidemiology , Orthohantavirus/immunology , Adult , Antibodies, Viral/blood , Antibodies, Viral/immunology , Hantavirus Infections/virology , Hantavirus Pulmonary Syndrome/epidemiology , Hantavirus Pulmonary Syndrome/virology , Humans , Incidence , Middle Aged , Panama/epidemiology , Young Adult
12.
Mol Ecol ; 20(19): 4109-22, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21880089

ABSTRACT

Co-divergence between host and parasites suggests that evolutionary processes act across similar spatial and temporal scales. Although there has been considerable work on the extent and correlates of co-divergence of RNA viruses and their mammalian hosts, relatively little is known about the extent to which virus evolution is determined by the phylogeographic history of host species. To test hypotheses related to co-divergence across a variety of spatial and temporal scales, we explored phylogenetic signatures in Andes virus (ANDV) sampled from Chile and its host rodent, Oligoryzomys longicaudatus. ANDV showed strong spatial subdivision, a phylogeographic pattern also recovered in the host using both spatial and genealogical approaches, and despite incomplete lineage sorting. Lineage structure in the virus seemed to be a response to current population dynamics in the host at the spatial scale of ecoregions. However, finer scale analyses revealed contrasting patterns of genetic structure across a latitudinal gradient. As predicted by their higher substitution rates, ANDV showed greater genealogical resolution than the rodent, with topological congruence influenced by the degree of lineage sorting within the host. However, despite these major differences in evolutionary dynamics, the geographic structure of host and virus converged across large spatial scales.


Subject(s)
Arvicolinae/virology , Orthohantavirus/genetics , Phylogeny , Animals , Arvicolinae/genetics , Population Dynamics , Selection, Genetic
13.
Drug Discov Today Ther Strateg ; 8(3-4): 61-69, 2011.
Article in English | MEDLINE | ID: mdl-22368688

ABSTRACT

Academia and small business research units are poised to play an increasing role in drug discovery, with drug repurposing as one of the major areas of activity. Here we summarize project status for a number of drugs or classes of drugs: raltegravir, cyclobenzaprine, benzbromarone, mometasone furoate, astemizole, R-naproxen, ketorolac, tolfenamic acid, phenothiazines, methylergonovine maleate and beta-adrenergic receptor drugs, respectively. Based on this multi-year, multi-project experience we discuss strengths and weaknesses of academic-based drug repurposing research. Translational, target and disease foci are strategic advantages fostered by close proximity and frequent interactions between basic and clinical scientists, which often result in discovering new modes of action for approved drugs. On the other hand, lack of integration with pharmaceutical sciences and toxicology, lack of appropriate intellectual coverage and issues related to dosing and safety may lead to significant drawbacks. The development of a more streamlined regulatory process world-wide, and the development of pre-competitive knowledge transfer systems such as a global healthcare database focused on regulatory and scientific information for drugs world-wide, are among the ideas proposed to improve the process of academic drug discovery and repurposing, and to overcome the "valley of death" by bridging basic to clinical sciences.

14.
Arch Virol ; 156(3): 443-56, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21161552

ABSTRACT

Monoclonal antibodies are important tools for various applications in hantavirus diagnostics. Recently, we generated Puumala virus (PUUV)-reactive monoclonal antibodies (mAbs) by immunisation of mice with chimeric polyomavirus-derived virus-like particles (VLPs) harbouring the 120-amino-acid-long amino-terminal region of the PUUV nucleocapsid (N) protein. Here, we describe the generation of two mAbs by co-immunisation of mice with hexahistidine-tagged full-length N proteins of Sin Nombre virus (SNV) and Andes virus (ANDV), their characterization by different immunoassays and comparison with the previously generated mAbs raised against a segment of PUUV N protein inserted into VLPs. All of the mAbs reacted strongly in ELISA and western blot tests with the antigens used for immunization and cross-reacted to varying extents with N proteins of other hantaviruses. All mAbs raised against a segment of the PUUV N protein presented on chimeric VLPs and both mAbs raised against the full-length AND/SNV N protein reacted with Vero cells infected with different hantaviruses. The reactivity of mAbs with native viral nucleocapsids was also confirmed by their reactivity in immunohistochemistry assays with kidney tissue specimens from experimentally SNV-infected rodents and human heart tissue specimens from hantavirus cardiopulmonary syndrome patients. Therefore, the described mAbs represent useful tools for the immunodetection of hantavirus infection.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Hantavirus Infections/diagnosis , Nucleocapsid Proteins/immunology , Orthohantavirus/immunology , Sin Nombre virus/immunology , Virology/methods , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Viral/isolation & purification , Blotting, Western , Chlorocebus aethiops , Cross Reactions , Enzyme-Linked Immunosorbent Assay , Hantavirus Infections/virology , Immunohistochemistry/methods , Mice , Puumala virus/immunology , Vero Cells
15.
PLoS Negl Trop Dis ; 4(10): e848, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-21049014

ABSTRACT

The exogenous RNA interference (RNAi) pathway is an important antiviral defense against arboviruses in mosquitoes, and virus-specific small interfering (si)RNAs are key components of this pathway. Understanding the biogenesis of siRNAs in mosquitoes could have important ramifications in using RNAi to control arbovirus transmission. Using deep sequencing technology, we characterized dengue virus type 2 (DENV2)-specific small RNAs produced during infection of Aedes aegypti mosquitoes and A. aegypti Aag2 cell cultures and compared them to those produced in the C6/36 Aedes albopictus cell line. We show that the size and mixed polarity of virus-specific small RNAs from DENV-infected A. aegypti cells indicate that they are products of Dicer-2 (Dcr2) cleavage of long dsRNA, whereas C6/36 cells generate DENV2-specific small RNAs that are longer and predominantly positive polarity, suggesting that they originate from a different small RNA pathway. Examination of virus-specific small RNAs after infection of the two mosquito cell lines with the insect-only flavivirus cell fusing agent virus (CFAV) corroborated these findings. An in vitro assay also showed that Aag2 A. aegypti cells are capable of siRNA production, while C6/36 A. albopictus cells exhibit inefficient Dcr2 cleavage of long dsRNA. Defective expression or function of Dcr2, the key initiator of the RNAi pathway, might explain the comparatively robust growth of arthropod-borne viruses in the C6/36 cell line, which has been used frequently as a surrogate for studying molecular interactions between arboviruses and cells of their mosquito hosts.


Subject(s)
Aedes/virology , Dengue Virus/immunology , RNA Interference , RNA, Small Interfering/genetics , Aedes/immunology , Animals , Cells, Cultured , Dengue Virus/genetics , RNA, Small Interfering/immunology , Ribonuclease III/metabolism
16.
Bioorg Med Chem Lett ; 20(23): 7085-91, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20951038

ABSTRACT

Hantaviruses use α(v)ß(3) integrins on the surface of human host cells as a gateway to invasion, hence compounds that target this receptor may be used as antiviral agents. To accomplish this aim, new peptidomimetic compounds were selected based on similarity to a cyclic peptide known to bind the α(v)ß(3) receptor. This first round of biological screening identified peptidomimetic molecules which were effective hantavirus inhibitors in the low micromolar range, two thousand times more potent than the original cyclic peptide. Pharmacophore models were built to broaden the structural diversity of the second set of compounds screened. Structure-activity relationships (SAR) were drawn from the entire dataset. Further characterization by dose-response studies revealed that three compounds had potency in the nanomolar range. Selectivity assays with a panel of hantaviruses supported the mechanism of inhibition by targeting the α(v)ß(3) receptor, through the ß(3) integrin.


Subject(s)
Hantavirus Infections/prevention & control , Integrin alphaVbeta3/antagonists & inhibitors , Peptides, Cyclic/chemistry , Dose-Response Relationship, Drug , Orthohantavirus/drug effects , Humans , Integrin beta3 , Peptides, Cyclic/pharmacology , Structure-Activity Relationship
17.
J Gen Virol ; 91(Pt 11): 2698-705, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20631087

ABSTRACT

Powassan virus (POW) is a tick-borne flavivirus distributed in Canada, the northern USA and the Primorsky region of Russia. POW is the only tick-borne flavivirus endemic to the western hemisphere, where it is transmitted mainly between Ixodes cookei and groundhogs (Marmota monax). Deer tick virus (DTV), a genotype of POW that has been frequently isolated from deer ticks (Ixodes scapularis), appears to be maintained in an enzootic cycle between these ticks and white-footed mice (Peromyscus leucopus). DTV has been isolated from ticks in several regions of North America, including the upper Midwest and the eastern seaboard. The incidence of human disease due to POW is apparently increasing. Previous analysis of tick-borne flaviviruses endemic to North America have been limited to relatively short genome fragments. We therefore assessed the evolutionary dynamics of POW using newly generated complete and partial genome sequences. Maximum-likelihood and Bayesian phylogenetic inferences showed two well-supported, reciprocally monophyletic lineages corresponding to POW and DTV. Bayesian skyline plots based on year-of-sampling data indicated no significant population size change for either virus lineage. Statistical model-based selection analyses showed evidence of purifying selection in both lineages. Positive selection was detected in NS-5 sequences for both lineages and envelope sequences for POW. Our findings confirm that POW and DTV sequences are relatively stable over time, which suggests strong evolutionary constraint, and support field observations that suggest that tick-borne flavivirus populations are extremely stable in enzootic foci.


Subject(s)
Encephalitis Viruses, Tick-Borne/classification , Encephalitis Viruses, Tick-Borne/genetics , Animals , Cluster Analysis , Encephalitis Viruses, Tick-Borne/isolation & purification , Evolution, Molecular , Genome, Viral , Molecular Epidemiology , Molecular Sequence Data , North America/epidemiology , Phylogeny , RNA, Viral/genetics , Selection, Genetic , Sequence Analysis, DNA , Sequence Homology , Ticks/virology
18.
PLoS One ; 5(6): e11159, 2010 Jun 17.
Article in English | MEDLINE | ID: mdl-20567522

ABSTRACT

BACKGROUND: Hantaviruses indigenous to the New World are the etiologic agents of hantavirus cardiopulmonary syndrome (HCPS). These viruses induce a strong interferon-stimulated gene (ISG) response in human endothelial cells. African green monkey-derived Vero E6 cells are used to propagate hantaviruses as well as many other viruses. The utility of the Vero E6 cell line for virus production is thought to owe to their lack of genes encoding type I interferons (IFN), rendering them unable to mount an efficient innate immune response to virus infection. Interferon lambda, a more recently characterized type III IFN, is transcriptionally controlled much like the type I IFNs, and activates the innate immune system in a similar manner. METHODOLOGY/PRINCIPAL FINDINGS: We show that Vero E6 cells respond to hantavirus infection by secreting abundant IFNlambda. Three New World hantaviruses were similarly able to induce IFNlambda expression in this cell line. The IFNlambda contained within virus preparations generated with Vero E6 cells independently activates ISGs when used to infect several non-endothelial cell lines, whereas innate immune responses by endothelial cells are specifically due to viral infection. We show further that Sin Nombre virus replicates to high titer in human hepatoma cells (Huh7) without inducing ISGs. CONCLUSIONS/SIGNIFICANCE: Herein we report that Vero E6 cells respond to viral infection with a highly active antiviral response, including secretion of abundant IFNlambda. This cytokine is biologically active, and when contained within viral preparations and presented to human epithelioid cell lines, results in the robust activation of innate immune responses. We also show that both Huh7 and A549 cell lines do not respond to hantavirus infection, confirming that the cytoplasmic RNA helicase pathways possessed by these cells are not involved in hantavirus recognition. We demonstrate that Vero E6 actively respond to virus infection and inhibiting IFNlambda production in these cells might increase their utility for virus propagation.


Subject(s)
Interferon Type I/genetics , Interferons/biosynthesis , Orthohantavirus/physiology , Animals , Cell Line, Tumor , Cells, Cultured , Chlorocebus aethiops , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Interferons/metabolism , Vero Cells , Virus Replication
19.
Anal Biochem ; 402(2): 151-60, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20363206

ABSTRACT

Hantaviruses cause two severe diseases in humans: hemorrhagic fever with renal syndrome (HFRS) and hantavirus cardiopulmonary syndrome (HCPS). The lack of vaccines or specific drugs to prevent or treat HFRS and HCPS and the requirement for conducting experiments in a biosafety level 3 laboratory (BSL-3) limit the ability to probe the mechanism of infection and disease pathogenesis. In this study, we developed a generalizable spectroscopic assay to quantify saturable fluorophore sites solubilized in envelope membranes of Sin Nombre virus (SNV) particles. We then used flow cytometry and live cell confocal fluorescence microscopy imaging to show that ultraviolet (UV)-killed SNV particles bind to the cognate receptors of live virions, namely, decay accelerating factor (DAF/CD55) expressed on Tanoue B cells and alpha(v)beta(3) integrins expressed on Vero E6 cells. SNV binding to DAF is multivalent and of high affinity (K(d) approximately 26pM). Self-exchange competition binding assays between fluorescently labeled SNV and unlabeled SNV are used to evaluate an infectious unit-to-particle ratio of approximately 1:14,000. We configured the assay for measuring the binding of fluorescently labeled SNV to Tanoue B suspension cells using a high-throughput flow cytometer. In this way, we established a proof-of-principle high-throughput screening (HTS) assay for binding inhibition. This is a first step toward developing HTS format assays for small molecule inhibitors of viral-cell interactions as well as dissecting the mechanism of infection in a BSL-2 environment.


Subject(s)
CD55 Antigens/metabolism , Flow Cytometry/methods , High-Throughput Screening Assays/methods , Integrin alphaVbeta3/metabolism , Sin Nombre virus/metabolism , Virion/metabolism , Animals , Calibration , Cell Line , Chlorocebus aethiops , Hantavirus Pulmonary Syndrome/metabolism , Humans , Protein Binding , Sin Nombre virus/chemistry , Sin Nombre virus/ultrastructure , Ultraviolet Rays , Vero Cells , Virion/chemistry , Virion/ultrastructure
20.
PLoS Pathog ; 6(2): e1000779, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20174562

ABSTRACT

In man, infection with South American Andes virus (ANDV) causes hantavirus cardiopulmonary syndrome (HCPS). HCPS due to ANDV is endemic in Southern Chile and much of Argentina and increasing numbers of cases are reported all over South America. A case-fatality rate of about 36% together with the absence of successful antiviral therapies urge the development of a vaccine. Although T-cell responses were shown to be critically involved in immunity to hantaviruses in mouse models, no data are available on the magnitude, specificity and longevity of ANDV-specific memory T-cell responses in patients. Using sets of overlapping peptides in IFN-gamma ELISPOT assays, we herein show in 78 Chilean convalescent patients that Gn-derived epitopes were immunodominant as compared to those from the N- and Gc-proteins. Furthermore, while the relative contribution of the N-specific response significantly declined over time, Gn-specific responses remained readily detectable ex vivo up to 13 years after the acute infection. Tetramer analysis further showed that up to 16.8% of all circulating CD3(+)CD8(+) T cells were specific for the single HLA-B*3501-restricted epitope Gn(465-473) years after the acute infection. Remarkably, Gn(465-473)-specific cells readily secreted IFN-gamma, granzyme B and TNF-alpha but not IL-2 upon stimulation and showed a 'revertant' CD45RA(+)CD27(-)CD28(-)CCR7(-)CD127(-) effector memory phenotype, thereby resembling a phenotype seen in other latent virus infections. Most intriguingly, titers of neutralizing antibodies increased over time in 10/17 individuals months to years after the acute infection and independently of whether they were residents of endemic areas or not. Thus, our data suggest intrinsic, latent antigenic stimulation of Gn-specific T-cells. However, it remains a major task for future studies to proof this hypothesis by determination of viral antigen in convalescent patients. Furthermore, it remains to be seen whether Gn-specific T cells are critical for viral control and protective immunity. If so, Gn-derived immunodominant epitopes could be of high value for future ANDV vaccines.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Hantavirus Infections/immunology , Immunologic Memory/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Cell Separation , Chile , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Orthohantavirus/immunology , Humans , Immunodominant Epitopes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...