Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 55
1.
Am J Clin Pathol ; 157(3): 470-475, 2022 03 03.
Article En | MEDLINE | ID: mdl-34643226

OBJECTIVES: Navajo Nation is disproportionately affected by hantavirus cardiopulmonary syndrome (HCPS), a severe respiratory disease that can quickly progress to respiratory failure and cardiogenic shock. The initial signs and symptoms of HCPS are indistinguishable from coronavirus disease 2019 (COVID-19). However, this distinction is critical, as the disease course differs greatly, with most patients with COVID-19 experiencing mild to moderate illness. We set out to determine if the evaluation of peripheral blood smears for five hematopathologic criteria previously identified as hallmarks of hantavirus infection, or "the hantavirus 5-point screen," could distinguish between COVID-19 and HCPS. METHODS: The hantavirus 5-point screen was performed on peripheral blood smears from 139 patients positive for COVID-19 seeking treatment from Tséhootsooí Medical Center and two Emory University hospitals. RESULTS: Of these 139 individuals, 136 (98%) received a score of 3/5 or below, indicating low suspicion for HCPS. While thrombocytopenia, one of the key signs of HCPS, was seen in the patients with COVID-19, it was generally mild and remained stable on repeat specimens collected 12 to 24 hours later. CONCLUSIONS: Given these findings, the 5-point screen remains a useful rapid screening tool for potential HCPS cases and may be useful to distinguish early HCPS from COVID-19 in HCPS endemic regions.


COVID-19 , Hantavirus Infections , Hantavirus Pulmonary Syndrome , Orthohantavirus , Hantavirus Infections/diagnosis , Hantavirus Pulmonary Syndrome/diagnosis , Hantavirus Pulmonary Syndrome/epidemiology , Hantavirus Pulmonary Syndrome/pathology , Humans , SARS-CoV-2
2.
PLoS Negl Trop Dis ; 15(8): e0009592, 2021 08.
Article En | MEDLINE | ID: mdl-34339406

BACKGROUND: Syrian hamsters infected with Andes virus (ANDV) develop a disease that recapitulates many of the salient features of human hantavirus pulmonary syndrome (HPS), including lethality. Infection of hamsters with Hantaan virus (HTNV) results in an asymptomatic, disseminated infection. In order to explore this dichotomy, we examined the transcriptome of ANDV- and HTNV-infected hamsters. RESULTS: Using NanoString technology, we examined kinetic transcriptional responses in whole blood collected from ANDV- and HTNV-infected hamsters. Of the 770 genes analyzed, key differences were noted in the kinetics of type I interferon sensing and signaling responses, complement activation, and apoptosis pathways between ANDV- and HTNV-infected hamsters. CONCLUSIONS: Delayed activation of type I interferon responses in ANDV-infected hamsters represents a potential mechanism that ANDV uses to subvert host immune responses and enhance disease. This is the first genome-wide analysis of hantavirus-infected hamsters and provides insight into potential avenues for therapeutics to hantavirus disease.


Hantavirus Infections/pathology , Hantavirus Pulmonary Syndrome/pathology , High-Throughput Nucleotide Sequencing/methods , Orthohantavirus/genetics , Orthohantavirus/pathogenicity , Animals , Chlorocebus aethiops , Cricetinae , Female , Orthohantavirus/isolation & purification , Mesocricetus , Vero Cells
3.
Article En | MEDLINE | ID: mdl-31618375

The hantavirus cardiopulmonary syndrome is considered an emerging disease in the Americas. Since 1993, thousands of cases have been reported from different countries, but mainly from Brazil. This study aims to describe some epidemiological, clinical and anatomopathological aspects of patients with hantavirus who presented poor outcome and were autopsied in a teaching hospital in Brazil, from 2000 to 2014. Of the 10 patients included, nine were male (mean age 43.5 years) and seven reported previous contact with rodents. Fever was present in eight of ten patients, dyspnea in nine of ten and myalgia in seven of ten patients; hemoconcentration, leukocytosis, thrombocytopenia and renal involvement were evidenced in all the 10 cases. At autopsy, the main alterations were seen in the lungs: pleural effusion (8/10 cases), increased weight 2.5 to 3 times, congestion/edema (10/10), interstitial mononuclear inflammation (10/10), alveolar hemorrhage (7/10), pulmonary collapse (7/10), hyaline membranes (7/10) and alveolar neutrophilic infiltrate (2/10). Pericardial effusion (2/10), mild myocardium inflammation (4/10), right ventricle dilation (1/10), polyploidy nuclei (3/10) and pericardial diffuse petechial (1/10) were also observed. The other organs exhibited discrete and non-specific alterations. Currently, this syndrome continues to be associated with high mortality directly linked to a late diagnosis and/or a misdiagnosis in the medical centers where these patients were seen for the first time. The anatomopathological findings at autopsy revealed the final phase of the process with pulmonary alterations, allowing a direct correlation with the severity of respiratory distress observed in these patients at admission.


Hantavirus Pulmonary Syndrome/pathology , Adolescent , Adult , Brazil/epidemiology , Female , Hantavirus Pulmonary Syndrome/mortality , Humans , Male , Middle Aged , Retrospective Studies , Severity of Illness Index
4.
Sci Rep ; 9(1): 834, 2019 01 29.
Article En | MEDLINE | ID: mdl-30696898

Orthohantaviruses, previously known as hantaviruses, are zoonotic viruses that can cause hantavirus pulmonary syndrome (HPS) and hemorrhagic fever with renal syndrome (HFRS) in humans. The HPS-causing Andes virus (ANDV) and the HFRS-causing Hantaan virus (HTNV) have anti-apoptotic effects. To investigate if this represents a general feature of orthohantaviruses, we analysed the capacity of six different orthohantaviruses - belonging to three distinct phylogroups and representing both pathogenic and non-pathogenic viruses - to inhibit apoptosis in infected cells. Primary human endothelial cells were infected with ANDV, HTNV, the HFRS-causing Puumala virus (PUUV) and Seoul virus, as well as the putative non-pathogenic Prospect Hill virus and Tula virus. Infected cells were then exposed to the apoptosis-inducing chemical staurosporine or to activated human NK cells exhibiting a high cytotoxic potential. Strikingly, all orthohantaviruses inhibited apoptosis in both settings. Moreover, we show that the nucleocapsid (N) protein from all examined orthohantaviruses are potential targets for caspase-3 and granzyme B. Recombinant N protein from ANDV, PUUV and the HFRS-causing Dobrava virus strongly inhibited granzyme B activity and also, to certain extent, caspase-3 activity. Taken together, this study demonstrates that six different orthohantaviruses inhibit apoptosis, suggesting this to be a general feature of orthohantaviruses likely serving as a mechanism of viral immune evasion.


Apoptosis/immunology , Hantaan virus/immunology , Immune Evasion/immunology , Orthohepadnavirus/immunology , Puumala virus/immunology , Seoul virus/immunology , A549 Cells , Caspase 3/immunology , Cell Line, Tumor , Endothelial Cells/virology , Granzymes/immunology , Orthohantavirus/immunology , Hantavirus Pulmonary Syndrome/pathology , Hantavirus Pulmonary Syndrome/virology , Hemorrhagic Fever with Renal Syndrome/pathology , Hemorrhagic Fever with Renal Syndrome/virology , Human Umbilical Vein Endothelial Cells , Humans , K562 Cells , Killer Cells, Natural/virology , Nucleocapsid/genetics , Nucleocapsid/immunology , Orthohepadnavirus/classification , Orthohepadnavirus/genetics
5.
Clin Microbiol Infect ; 21S: e6-e16, 2019 Apr.
Article En | MEDLINE | ID: mdl-24750436

Over the past few decades understanding and recognition of hantavirus infection has greatly improved worldwide, but both the amplitude and the magnitude of hantavirus outbreaks have been increasing. Several novel hantaviruses with unknown pathogenic potential have been identified in a variety of insectivore hosts. With the new hosts, new geographical distributions of hantaviruses have also been discovered and several new species were found in Africa. Hantavirus infection in humans can result in two clinical syndromes: haemorrhagic fever with renal syndrome (HFRS) and hantavirus cardiopulmonary syndrome (HCPS) caused by Old World and New World hantaviruses, respectively. The clinical presentation of HFRS varies from subclinical, mild, and moderate to severe, depending in part on the causative agent of the disease. In general, HFRS caused by Hantaan virus, Amur virus and Dobrava virus are more severe with mortality rates from 5 to 15%, whereas Seoul virus causes moderate and Puumala virus and Saaremaa virus cause mild forms of disease with mortality rates <1%. The central phenomena behind the pathogenesis of both HFRS and HCPS are increased vascular permeability and acute thrombocytopenia. The pathogenesis is likely to be a complex multifactorial process that includes contributions from immune responses, platelet dysfunction and the deregulation of endothelial cell barrier functions. Also a genetic predisposition, related to HLA type, seems to be important for the severity of the disease. As there is no effective treatment or vaccine approved for use in the USA and Europe, public awareness and precautionary measures are the only ways to minimize the risk of hantavirus disease.


Hantavirus Infections/diagnosis , Hantavirus Infections/epidemiology , Orthohantavirus/physiology , Animals , Disease Outbreaks/prevention & control , Disease Reservoirs , Orthohantavirus/classification , Orthohantavirus/isolation & purification , Hantavirus Infections/pathology , Hantavirus Infections/virology , Hantavirus Pulmonary Syndrome/diagnosis , Hantavirus Pulmonary Syndrome/epidemiology , Hantavirus Pulmonary Syndrome/pathology , Hantavirus Pulmonary Syndrome/virology , Hemorrhagic Fever with Renal Syndrome/diagnosis , Hemorrhagic Fever with Renal Syndrome/epidemiology , Hemorrhagic Fever with Renal Syndrome/pathology , Hemorrhagic Fever with Renal Syndrome/virology , Humans
6.
Article En | MEDLINE | ID: mdl-29930915

Sin Nombre virus (SNV) causes hantavirus cardiopulmonary pulmonary syndrome (HCPS) with the loss of pulmonary vascular endothelial integrity, and pulmonary edema without causing cytopathic effects on the vascular endothelium. HCPS is associated primarily with a dysregulated immune response. We previously found occult signs of hemostatic imbalance in the form of a sharp >30-100 fold increase in the expression of plasminogen activator inhibitor type 1 (PAI-1), in serial blood plasma draws of terminal stage-patients. However, the mechanism of the increase in PAI-1 remains unclear. PAI-1 is a primary inhibitor of fibrinolysis caused by tissue plasminogen activator (tPA) and urokinase plasminogen activator plasma (uPA). Here, we investigate factors that contribute to PAI-1 upregulation during HCPS. Using zymography, we found evidence of PAI-1-refractory uPA activity and no tPA activity in plasma samples drawn from HCPS patients. The sole prevalence of uPA activity suggested that severe inflammation drove PAI-1 activity. We have recently reported that the P2Y2 receptor (P2Y2R) mediates SNV infectivity by interacting in cis with ß3 integrins, which activates the latter during infection. P2Y2R is a known effector for several biological processes relevant to HCPS pathogenesis, such as upregulation of tissue factor (TF), a primary initiator of the coagulation cascade, stimulating vascular permeability and leukocyte homing to sites of infection. As P2Y2R is prone to upregulation under conditions of inflammation, we compared the expression level of P2Y2R in formalin fixed tissues of HCPS decedents using a TaqMan assay and immunohistochemistry. Our TaqMan results show that the expression of P2Y2R is upregulated significantly in HCPS cases compared to non- HCPS controls (P < 0.001). Immunohistochemistry showed that lung macrophages were the primary reservoir of high and coincident localization of P2Y2R, uPA, PAI-1, and TF antigens. We also observed increased staining for SNV antigens in the same tissue segments where P2Y2R expression was upregulated. Conversely, sections of low P2Y2R expression showed weak manifestations of macrophages, SNV, PAI-1, and TF. Coincident localization of P2Y2R and PAI-1 on macrophage deposits suggests an inflammation-dependent mechanism of increasing pro-coagulant activity in HCPS in the absence of tissue injury.


Hantavirus Infections , Orthohantavirus/pathogenicity , Plasminogen Activator Inhibitor 1/metabolism , Receptors, Purinergic P2Y2/metabolism , Up-Regulation , Urokinase-Type Plasminogen Activator/metabolism , Adult , Aged , Capillary Permeability , Female , Fibrinolysis , Hantavirus Infections/diagnostic imaging , Hantavirus Infections/immunology , Hantavirus Infections/pathology , Hantavirus Pulmonary Syndrome/diagnostic imaging , Hantavirus Pulmonary Syndrome/immunology , Hantavirus Pulmonary Syndrome/pathology , Humans , Immunohistochemistry , Inflammation , Leukocytes , Lung/diagnostic imaging , Lung/pathology , Male , Middle Aged , New Mexico , Signal Transduction , Tissue Plasminogen Activator , Urokinase-Type Plasminogen Activator/blood
7.
J Med Virol ; 90(6): 1003-1009, 2018 06.
Article En | MEDLINE | ID: mdl-29446472

Hantavirus cardiopulmonary syndrome is characterized by pulmonary capillary leakage and alveolar flooding, resulting in 50% mortality due to fulminant hypoxic respiratory failure. In addition, depression of cardiac function ensues, which complicates the picture with cardiogenic shock. Early diagnosis and appropriate use of extracorporeal membrane oxygenation (ECMO) are amongst the lifesaving interventions in this fatal illness. However, a recent case report demonstrates that implementation of high volume continuous hemofilteration along with protective ventilation reverses the cardiogenic shock within few hours in hantavirus infected patients. This review article is focused on the recent advances in clinical features, diagnosis, management, epidemiology, and pathogenesis of hantavirus induced cardiopulmonary syndrome. It provides information for clinicians to help in correct diagnosis during the early stages of viral infection that could improve the prognosis of this viral illness.


Hantavirus Infections/complications , Hantavirus Pulmonary Syndrome/diagnosis , Hantavirus Pulmonary Syndrome/pathology , Shock, Cardiogenic/diagnosis , Shock, Cardiogenic/pathology , Disease Management , Early Diagnosis , Extracorporeal Membrane Oxygenation , Hantavirus Pulmonary Syndrome/mortality , Hantavirus Pulmonary Syndrome/therapy , Hemofiltration , Humans , Respiration, Artificial , Shock, Cardiogenic/mortality , Shock, Cardiogenic/therapy , Treatment Outcome
8.
9.
Eur J Clin Microbiol Infect Dis ; 35(4): 713-21, 2016 Apr.
Article En | MEDLINE | ID: mdl-26873376

Hantavirus infections may cause severe and sometime life-threatening lung failure. The pathogenesis is not fully known and there is an urgent need for effective treatment. We aimed to investigate the association between pulmonary viral load and immune responses, and their relation to disease severity. Bronchoscopy with sampling of bronchoalveolar lavage (BAL) fluid was performed in 17 patients with acute Puumala hantavirus infection and 16 healthy volunteers acting as controls. Lymphocyte subsets, granzyme concentrations, and viral load were determined by flow cytometry, enzyme-linked immunosorbent assay (ELISA), and quantitative reverse transcription polymerase chain reaction (RT-PCR), respectively. Analyses of BAL fluid revealed significantly higher numbers of activated CD8(+) T cells and natural killer (NK) cells, as well as higher concentrations of the cytotoxins granzymes A and B in hantavirus-infected patients, compared to controls. In patients, Puumala hantavirus RNA was detected in 88 % of BAL cell samples and correlated inversely to the T cell response. The magnitude of the pulmonary cytotoxic lymphocyte response correlated to the severity of disease and systemic organ dysfunction, in terms of need for supplemental oxygen treatment, hypotension, and laboratory data indicating renal failure, cardiac dysfunction, vascular leakage, and cell damage. Regulatory T cell numbers were significantly lower in patients compared to controls, and may reflect inadequate immune regulation during hantavirus infection. Hantavirus infection elicits a pronounced cytotoxic lymphocyte response in the lungs. The magnitude of the immune response was associated with disease severity. These results give insights into the pathogenesis and possibilities for new treatments.


Cytotoxicity, Immunologic , Hantavirus Pulmonary Syndrome/pathology , Lung/pathology , Puumala virus/isolation & purification , Adult , Aged , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Bronchoscopy , Granzymes/analysis , Hantavirus Pulmonary Syndrome/immunology , Humans , Lung/virology , Lymphocyte Subsets/cytology , Male , Middle Aged , Severity of Illness Index , Viral Load
10.
Vet Pathol ; 53(1): 182-9, 2016 Jan.
Article En | MEDLINE | ID: mdl-25722219

Laguna Negra virus (LNV) is a New World hantavirus associated with severe and often fatal cardiopulmonary disease in humans, known as hantavirus pulmonary syndrome (HPS). Five hamster species were evaluated for clinical and serologic responses following inoculation with 4 hantaviruses. Of the 5 hamster species, only Turkish hamsters infected with LNV demonstrated signs consistent with HPS and a fatality rate of 43%. Clinical manifestations in infected animals that succumbed to disease included severe and rapid onset of dyspnea, weight loss, leukopenia, and reduced thrombocyte numbers as compared to uninfected controls. Histopathologic examination revealed lung lesions that resemble the hallmarks of HPS in humans, including interstitial pneumonia and pulmonary edema, as well as generalized infection of endothelial cells and macrophages in major organ tissues. Histologic lesions corresponded to the presence of viral antigen in affected tissues. To date, there have been no small animal models available to study LNV infection and pathogenesis. The Turkish hamster model of LNV infection may be important in the study of LNV-induced HPS pathogenesis and development of disease treatment and prevention strategies.


Antigens, Viral/immunology , Disease Models, Animal , Hantavirus Pulmonary Syndrome/pathology , Lung Diseases, Interstitial/pathology , Mesocricetus , Orthohantavirus/immunology , Pulmonary Edema/pathology , Animals , Cricetinae , Female , Humans , Lung/pathology , Male
11.
J Med Virol ; 87(11): 1823-30, 2015 Nov.
Article En | MEDLINE | ID: mdl-26087934

Hantavirus causes severe illness including pneumonia, which leads to hospitalization and often death. At present, there is no specific treatment available. The hantavirus pathogenesis is not well understood, but most likely both virus-mediated and host-mediated mechanisms, are involved. The aim of this study was to correlate viral load in samples of hantavirus pulmonary syndrome cases and hantavirus infected individuals, with clinical epidemiological parameters and disease outcome. The variables that could potentially be related with viral load were analyzed. The retrospective study included 73 cases or household contacts, with different clinical evolution. Viral load was measured by reverse-transcription and real time polymerase chain reaction. There was no statistically significant association between blood viral RNA levels and severity of disease. However, viral load was inversely correlated with IgG response in a statistically significant manner. The level of viral RNA was significantly higher in patients infected with Andes virus South lineage, and was markedly low in persons infected with Laguna Negra virus. These results suggest that the infecting viral genotype is associated with disease severity, and that high viral load is associated with a low specific IgG response. Sex, age and disease severity were not related with viral load. Further investigations increasing strikingly the number of cases and also limiting the variables to be studied are necessary.


Blood/virology , Hantavirus Pulmonary Syndrome/pathology , Hantavirus Pulmonary Syndrome/virology , Orthohantavirus/isolation & purification , Viral Load , Adolescent , Adult , Antibodies, Viral/blood , Argentina , Child , Child, Preschool , Female , Humans , Immunoglobulin G/blood , Infant , Male , RNA, Viral/blood , Real-Time Polymerase Chain Reaction , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Severity of Illness Index , Young Adult
12.
PLoS Pathog ; 10(11): e1004521, 2014 Nov.
Article En | MEDLINE | ID: mdl-25412359

Clinical infection with hantaviruses cause two severe acute diseases, hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). These diseases are characterized by strong immune activation, increased vascular permeability, and up to 50% case-fatality rates. One prominent feature observed in clinical hantavirus infection is rapid expansion of natural killer (NK) cells in peripheral blood of affected individuals. We here describe an unusually high state of activation of such expanding NK cells in the acute phase of clinical Puumala hantavirus infection. Expanding NK cells expressed markedly increased levels of activating NK cell receptors and cytotoxic effector molecules. In search for possible mechanisms behind this NK cell activation, we observed virus-induced IL-15 and IL-15Rα on infected endothelial and epithelial cells. Hantavirus-infected cells were shown to strongly activate NK cells in a cell-cell contact-dependent way, and this response was blocked with anti-IL-15 antibodies. Surprisingly, the strength of the IL-15-dependent NK cell response was such that it led to killing of uninfected endothelial cells despite expression of normal levels of HLA class I. In contrast, hantavirus-infected cells were resistant to NK cell lysis, due to a combination of virus-induced increase in HLA class I expression levels and hantavirus-mediated inhibition of apoptosis induction. In summary, we here describe a possible mechanism explaining the massive NK cell activation and proliferation observed in HFRS patients caused by Puumala hantavirus infection. The results add further insights into mechanisms behind the immunopathogenesis of hantavirus infections in humans and identify new possible targets for intervention.


Gene Expression Regulation/immunology , Hantavirus Pulmonary Syndrome/immunology , Hemorrhagic Fever with Renal Syndrome/immunology , Interleukin-15/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation , Puumala virus/immunology , Receptors, Interleukin-15/immunology , Endothelial Cells/immunology , Endothelial Cells/pathology , Endothelial Cells/virology , Hantavirus Pulmonary Syndrome/pathology , Hemorrhagic Fever with Renal Syndrome/pathology , Humans , K562 Cells , Killer Cells, Natural/pathology , Killer Cells, Natural/virology
13.
PLoS One ; 9(6): e99764, 2014.
Article En | MEDLINE | ID: mdl-24924189

Andes virus (ANDV) is the most common causative agent of hantavirus pulmonary syndrome (HPS) in the Americas, and is the only hantavirus associated with human-to-human transmission. Case fatality rates of ANDV-induced HPS are approximately 40%. There are currently no effective vaccines or antivirals against ANDV. Since HPS severity correlates with viral load, we tested small interfering RNA (siRNA) directed against ANDV genes as a potential antiviral strategy. We designed pools of 4 siRNAs targeting each of the ANDV genome segments (S, M, and L), and tested their efficacy in reducing viral replication in vitro. The siRNA pool targeting the S segment reduced viral transcription and replication in Vero-E6 cells more efficiently than those targeting the M and L segments. In contrast, siRNAs targeting the S, M, or L segment were similar in their ability to reduce viral replication in human lung microvascular endothelial cells. Importantly, these siRNAs inhibit ANDV replication even if given after infection. Taken together, our findings indicate that siRNAs targeting the ANDV genome efficiently inhibit ANDV replication, and show promise as a strategy for developing therapeutics against ANDV infection.


Orthohantavirus/drug effects , Orthohantavirus/physiology , RNA, Small Interfering/pharmacology , Virus Replication/drug effects , Animals , Cells, Cultured , Chlorocebus aethiops , Endothelial Cells/drug effects , Endothelial Cells/virology , Orthohantavirus/genetics , Hantavirus Pulmonary Syndrome/pathology , Hantavirus Pulmonary Syndrome/virology , Humans , Lung/cytology , Lung/drug effects , Lung/pathology , Lung/virology , Primary Cell Culture , RNA, Viral/antagonists & inhibitors , RNA, Viral/genetics , Vero Cells , Virus Replication/genetics
14.
Virus Res ; 187: 2-5, 2014 Jul 17.
Article En | MEDLINE | ID: mdl-24412711

We three authors, the two past presidents (HWL and AV) and the current president (CSS) of the International Society for Hantaviruses (ISH) have attended most of the nine International Conferences on HFRS, HPS and Hantaviruses (Table 1). These conferences have provided a forum for a synergistic group of clinicians, basic researchers, mammalogists, epidemiologists and ecologists to share their expertise and interests in all aspects of hantavirus research. Much of what is now hantavirus dogma was only conjecture when HWL organized the first conference in Seoul, Korea in 1989. Herein, we provide our reflections on key events in hantavirus research. As we come from distinct areas of the world and have had individual historical experiences, we certainly have our own geocentric opinions about the key events. Nevertheless, we agree that the discovery of hantaviruses has taken an interesting and unpredictable track to where we are today.


Hantavirus Infections/veterinary , Hantavirus Pulmonary Syndrome/history , Hemorrhagic Fever with Renal Syndrome/history , Orthohantavirus/physiology , Rodent Diseases/epidemiology , Virology/history , Animals , Congresses as Topic , Orthohantavirus/pathogenicity , Hantavirus Infections/epidemiology , Hantavirus Infections/transmission , Hantavirus Infections/virology , Hantavirus Pulmonary Syndrome/epidemiology , Hantavirus Pulmonary Syndrome/pathology , Hantavirus Pulmonary Syndrome/transmission , Hemorrhagic Fever with Renal Syndrome/epidemiology , Hemorrhagic Fever with Renal Syndrome/pathology , Hemorrhagic Fever with Renal Syndrome/transmission , History, 20th Century , History, 21st Century , Humans , Republic of Korea/epidemiology , Rodent Diseases/transmission , Rodent Diseases/virology , Rodentia , United States/epidemiology
15.
J Virol ; 88(2): 811-9, 2014 Jan.
Article En | MEDLINE | ID: mdl-24198421

Sin Nombre virus (SNV) is a rodent-borne hantavirus that causes hantavirus pulmonary syndrome (HPS) predominantly in North America. SNV infection of immunocompetent hamsters results in an asymptomatic infection; the only lethal disease model for a pathogenic hantavirus is Andes virus (ANDV) infection of Syrian hamsters. Efforts to create a lethal SNV disease model in hamsters by repeatedly passaging virus through the hamster have demonstrated increased dissemination of the virus but no signs of disease. In this study, we demonstrate that immunosuppression of hamsters through the administration of a combination of dexamethasone and cyclophosphamide, followed by infection with SNV, results in a vascular leak syndrome that accurately mimics both HPS disease in humans and ANDV infection of hamsters. Immunosuppressed hamsters infected with SNV have a mean number of days to death of 13 and display clinical signs associated with HPS, including pulmonary edema. Viral antigen was widely detectable throughout the pulmonary endothelium. Histologic analysis of lung sections showed marked inflammation and edema within the alveolar septa of SNV-infected hamsters, results which are similar to what is exhibited by hamsters infected with ANDV. Importantly, SNV-specific neutralizing polyclonal antibody administered 5 days after SNV infection conferred significant protection against disease. This experiment not only demonstrated that the disease was caused by SNV, it also demonstrated the utility of this animal model for testing candidate medical countermeasures. This is the first report of lethal disease caused by SNV in an adult small-animal model.


Disease Models, Animal , Hantavirus Pulmonary Syndrome/immunology , Hantavirus Pulmonary Syndrome/virology , Mesocricetus , Sin Nombre virus/physiology , Animals , Antibodies, Viral/therapeutic use , Cricetinae , Cyclophosphamide/administration & dosage , Dexamethasone/administration & dosage , Female , Hantavirus Pulmonary Syndrome/drug therapy , Hantavirus Pulmonary Syndrome/mortality , Hantavirus Pulmonary Syndrome/pathology , Humans , Immunosuppression Therapy , Immunosuppressive Agents/administration & dosage
16.
J Virol ; 86(18): 10015-27, 2012 Sep.
Article En | MEDLINE | ID: mdl-22787210

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.


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
17.
Antiviral Res ; 95(3): 282-92, 2012 Sep.
Article En | MEDLINE | ID: mdl-22705798

Hantavirus pulmonary syndrome (HPS) is a relatively rare, but frequently fatal disease associated with New World hantaviruses, most commonly Sin Nombre and Andes viruses in North and South America, respectively. It is characterized by fever and the sudden, rapid onset of severe respiratory distress and cardiogenic shock, which can be fatal in up to 50% of cases. Currently there are no approved antiviral therapies or vaccines for the treatment or prevention of HPS. A major obstacle in the development of effective medical countermeasures against highly pathogenic agents like the hantaviruses is recapitulating the human disease as closely as possible in an appropriate and reliable animal model. To date, the only animal model that resembles HPS in humans is the Syrian hamster model. Following infection with Andes virus, hamsters develop HPS-like disease which faithfully mimics the human condition with respect to incubation period and pathophysiology of disease. Perhaps most importantly, the sudden and rapid onset of severe respiratory distress observed in humans also occurs in hamsters. The last several years has seen an increase in studies utilizing the Andes virus hamster model which have provided unique insight into HPS pathogenesis as well as potential therapeutic and vaccine strategies to treat and prevent HPS. The purpose of this article is to review the current understanding of HPS disease progression in Syrian hamsters and discuss the suitability of utilizing this model to evaluate potential medical countermeasures against HPS.


Disease Models, Animal , Hantavirus Pulmonary Syndrome/pathology , Orthohantavirus/pathogenicity , Animals , Cricetinae , Humans , Mesocricetus
18.
Vector Borne Zoonotic Dis ; 12(5): 393-9, 2012 May.
Article En | MEDLINE | ID: mdl-22214273

Clinical cases of hantavirus pulmonary syndrome (HPS) can be challenging to differentiate from other acute respiratory diseases, which can lead to delays in diagnosis, treatment, and disease reporting. Rapid onset of severe disease occurs, at times before diagnostic test results are available. This study's objective was to examine the clinical characteristics of patients that would indicate HPS to aid in detection and reporting. Test results of blood samples from U.S. patients suspected of having HPS submitted to the Centers for Disease Control and Prevention from 1998-2010 were reviewed. Patient information collected by case report forms was compared between HPS-confirmed and test-negative patients. Diagnostic sensitivity, specificity, predictive values, and likelihood ratios were calculated for individual clinical findings and combinations of variables. Of 567 patients included, 36% were HPS-confirmed. Thrombocytopenia, chest x-rays with suggestive signs, and receiving supplemental oxygenation were highly sensitive (>95%), while elevated hematocrit was highly specific (83%) in detecting HPS. Combinations that maximized sensitivity required the presence of thrombocytopenia. Using a national sample of suspect patients, we found that thrombocytopenia was a highly sensitive indicator of HPS and should be included in surveillance definitions for suspected HPS. Using a sensitive suspect case definition to identify potential HPS patients that are confirmed by highly specific diagnostic testing will ensure accurate reporting of this disease.


Hantavirus Pulmonary Syndrome/diagnosis , Fever/pathology , Hantavirus Pulmonary Syndrome/diagnostic imaging , Hantavirus Pulmonary Syndrome/epidemiology , Hantavirus Pulmonary Syndrome/pathology , Hematocrit , Humans , Leukocyte Count , Population Surveillance , Radiography , Risk Factors , Thrombocytopenia/pathology , United States/epidemiology
20.
Clin Infect Dis ; 54(1): 91-4, 2012 Jan 01.
Article En | MEDLINE | ID: mdl-22042876

Hantavirus is known to cause 2 distinct clinical syndromes: hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome. Seoul virus is an Old World hantavirus known to cause HFRS. We report a case attributed to domestically acquired Seoul hantavirus with prominent pulmonary involvement and a fatal outcome.


Community-Acquired Infections/diagnosis , Community-Acquired Infections/pathology , Hantavirus Pulmonary Syndrome/diagnosis , Hantavirus Pulmonary Syndrome/pathology , Seoul virus/isolation & purification , Adult , Bronchoalveolar Lavage Fluid/cytology , Community-Acquired Infections/virology , Hantavirus Pulmonary Syndrome/virology , Humans , Male , Microscopy , Seoul virus/pathogenicity , United States
...