Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 89
Filter
1.
Sci Rep ; 11(1): 17440, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34465819

ABSTRACT

The use of antibody-based therapies for the treatment of high consequence viral pathogens has gained interest over the last fifteen years. Here, we sought to evaluate the use of unique camelid-based IgG antibodies to prevent lethal hantavirus pulmonary syndrome (HPS) in Syrian hamsters. Using purified, polyclonal IgG antibodies generated in DNA-immunized alpacas, we demonstrate that post-exposure treatments reduced viral burdens and organ-specific pathology associated with lethal HPS. Antibody treated animals did not exhibit signs of disease and were completely protected. The unique structures and properties, particularly the reduced size, distinct paratope formation and increased solubility of camelid antibodies, in combination with this study support further pre-clinical evaluation of heavy-chain only antibodies for treatment of severe respiratory diseases, including HPS.


Subject(s)
Antibodies, Viral/administration & dosage , Disease Models, Animal , Glycoproteins/immunology , Hantavirus Infections/prevention & control , Hantavirus Pulmonary Syndrome/prevention & control , Immunoglobulin G/administration & dosage , Orthohantavirus/immunology , Animals , Antibodies, Viral/immunology , Camelids, New World , Female , Hantavirus Infections/immunology , Hantavirus Infections/virology , Hantavirus Pulmonary Syndrome/immunology , Hantavirus Pulmonary Syndrome/virology , Immunoglobulin G/immunology , Male , Mesocricetus
2.
Viruses ; 13(8)2021 08 12.
Article in English | MEDLINE | ID: mdl-34452463

ABSTRACT

Pathogenic New World orthohantaviruses cause hantavirus cardiopulmonary syndrome (HCPS), a severe immunopathogenic disease in humans manifested by pulmonary edema and respiratory distress, with case fatality rates approaching 40%. High levels of inflammatory mediators are present in the lungs and systemic circulation of HCPS patients. Previous studies have provided insights into the pathophysiology of HCPS. However, the longitudinal correlations of innate and adaptive immune responses and disease outcomes remain unresolved. This study analyzed serial immune responses in 13 HCPS cases due to Sin Nombre orthohantavirus (SNV), with 11 severe cases requiring extracorporeal membrane oxygenation (ECMO) treatment and two mild cases. We measured viral load, levels of various cytokines, urokinase plasminogen activator (uPA), and plasminogen activator inhibitor-1 (PAI-1). We found significantly elevated levels of proinflammatory cytokines and PAI-1 in five end-stage cases. There was no difference between the expression of active uPA in survivors' and decedents' cases. However, total uPA in decedents' cases was significantly higher compared to survivors'. In some end-stage cases, uPA was refractory to PAI-1 inhibition as measured by zymography, where uPA and PAI-1 were strongly correlated to lymphocyte counts and IFN-γ. We also found bacterial co-infection influencing the etiology and outcome of immune response in two cases. Unsupervised Principal Component Analysis and hierarchical cluster analyses resolved separate waves of correlated immune mediators expressed in one case patient due to a sequential co-infection of bacteria and SNV. Overall, a robust proinflammatory immune response, characterized by an imbalance in T helper 17 (Th17) and regulatory T-cells (Treg) subsets, was correlated with dysregulated inflammation and mortality. Our sample size is small; however, the core differences correlated to survivors and end-stage HCPS are instructive.


Subject(s)
Cytokines/genetics , Cytokines/immunology , Hantavirus Infections/complications , Hantavirus Infections/immunology , Hantavirus Pulmonary Syndrome/immunology , Plasminogen/genetics , Sin Nombre virus/pathogenicity , Adolescent , Adult , Coinfection/complications , Coinfection/microbiology , Coinfection/virology , Cytokines/classification , Female , Hantavirus Infections/physiopathology , Hantavirus Pulmonary Syndrome/physiopathology , Humans , Inflammation/immunology , Inflammation/virology , Longitudinal Studies , Lung/immunology , Lung/pathology , Lung/virology , Male , Middle Aged , Patient Acuity , Plasminogen/analysis , Plasminogen/immunology , Retrospective Studies , Sin Nombre virus/immunology , Young Adult
3.
Viruses ; 11(9)2019 09 13.
Article in English | MEDLINE | ID: mdl-31540199

ABSTRACT

Hantaviruses are widespread zoonotic pathogens found around the globe. Depending on their geographical location, hantaviruses can cause two human syndromes, haemorrhagic fever with renal syndrome (HFRS) or hantavirus pulmonary syndrome (HPS). HPS and HFRS have many commonalities amongst which excessive activation of immune cells is a prominent feature. Hantaviruses replicate in endothelial cells (ECs), the major battlefield of hantavirus-induced pathogenesis, without causing cytopathic effects. This indicates that a misdirected response of human immune cells to hantaviruses is causing damage. As dendritic cells (DCs) orchestrate antiviral immune responses, they are in the focus of research analysing hantavirus-induced immunopathogenesis. In this review, we discuss the interplay between hantaviruses and DCs and the immunological consequences thereof.


Subject(s)
Dendritic Cells/microbiology , Dendritic Cells/virology , Hantavirus Infections/immunology , Hantavirus Infections/physiopathology , Endothelial Cells/immunology , Endothelial Cells/virology , Orthohantavirus , Hantavirus Pulmonary Syndrome/immunology , Hantavirus Pulmonary Syndrome/physiopathology , Hemorrhagic Fever with Renal Syndrome/immunology , Hemorrhagic Fever with Renal Syndrome/physiopathology , Humans
4.
J Intern Med ; 285(5): 510-523, 2019 05.
Article in English | MEDLINE | ID: mdl-30663801

ABSTRACT

Two related hyperinflammatory syndromes are distinguished following infection of humans with hantaviruses: haemorrhagic fever with renal syndrome (HFRS) seen in Eurasia and hantavirus pulmonary syndrome (HPS) seen in the Americas. Fatality rates are high, up to 10% for HFRS and around 35%-40% for HPS. Puumala virus (PUUV) is the most common HFRS-causing hantavirus in Europe. Here, we describe recent insights into the generation of innate and adaptive cell-mediated immune responses following clinical infection with PUUV. First described are studies demonstrating a marked redistribution of peripheral blood mononuclear phagocytes (MNP) to the airways, a process that may underlie local immune activation at the site of primary infection. We then describe observations of an excessive natural killer (NK) cell activation and the persistence of highly elevated numbers of NK cells in peripheral blood following PUUV infection. A similar vigorous CD8 Tcell response is also described, though Tcell responses decline with viraemia. Like MNPs, many NK cells and CD8 T cells also localize to the lung upon acute PUUV infection. Following this, findings demonstrating the ability of hantaviruses, including PUUV, to cause apoptosis resistance in infected target cells, are described. These observations, and associated inflammatory cytokine responses, may provide new insights into HFRS and HPS disease pathogenesis. Based on similarities between inflammatory responses in severe hantavirus infections and other hyperinflammatory disease syndromes, we speculate whether some therapeutic interventions that have been successful in the latter conditions may also be applicable in severe hantavirus infections.


Subject(s)
Adaptive Immunity , Hantavirus Pulmonary Syndrome/immunology , Hemorrhagic Fever with Renal Syndrome/immunology , Immunity, Innate , Puumala virus , Hantavirus Pulmonary Syndrome/therapy , Hemorrhagic Fever with Renal Syndrome/therapy , Humans , Severity of Illness Index
5.
Article in English | MEDLINE | ID: mdl-29930915

ABSTRACT

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.


Subject(s)
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
6.
PLoS Negl Trop Dis ; 11(10): e0006042, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29077702

ABSTRACT

BACKGROUND: Hantavirus, the hemorrhagic causative agent of two clinical diseases, is found worldwide with variation in severity, incidence and mortality. The most lethal hantaviruses are found on the American continent where the most prevalent viruses like Andes virus and Sin Nombre virus are known to cause hantavirus pulmonary syndrome. New World hantavirus infection of immunocompetent hamsters results in an asymptomatic infection except for Andes virus and Maporal virus; the only hantaviruses causing a lethal disease in immunocompetent Syrian hamsters mimicking hantavirus pulmonary syndrome in humans. METHODOLOGY/PRINCIPAL FINDINGS: Hamsters, immunosuppressed with dexamethasone and cyclophosphamide, were infected intramuscularly with different New World hantavirus strains (Bayou virus, Black Creek Canal virus, Caño Delgadito virus, Choclo virus, Laguna Negra virus, and Maporal virus). In the present study, we show that immunosuppression of hamsters followed by infection with a New World hantavirus results in an acute disease that precisely mimics both hantavirus disease in humans and Andes virus infection of hamsters. CONCLUSIONS/ SIGNIFICANCE: Infected hamsters showed specific clinical signs of disease and moreover, histological analysis of lung tissue showed signs of pulmonary edema and inflammation within alveolar septa. In this study, we were able to infect immunosuppressed hamsters with different New World hantaviruses reaching a lethal outcome with signs of disease mimicking human disease.


Subject(s)
Disease Models, Animal , Hantavirus Infections , Hantavirus Pulmonary Syndrome , Immunosuppression Therapy , Mesocricetus , Orthohantavirus/pathogenicity , Animals , Anti-Inflammatory Agents/administration & dosage , Cricetinae , Cyclophosphamide/administration & dosage , Dexamethasone/administration & dosage , Female , Orthohantavirus/isolation & purification , Hantavirus Infections/immunology , Hantavirus Infections/virology , Hantavirus Pulmonary Syndrome/immunology , Hantavirus Pulmonary Syndrome/virology , Humans , Immunocompromised Host , Immunosuppressive Agents/administration & dosage , Lung/immunology , Lung/pathology , Lung/virology , Survival Analysis
7.
PLoS Pathog ; 13(6): e1006462, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28640917

ABSTRACT

Hantaviruses infect humans via inhalation of virus-contaminated rodent excreta. Infection can cause severe disease with up to 40% mortality depending on the viral strain. The virus primarily targets the vascular endothelium without direct cytopathic effects. Instead, exaggerated immune responses may inadvertently contribute to disease development. Mononuclear phagocytes (MNPs), including monocytes and dendritic cells (DCs), orchestrate the adaptive immune responses. Since hantaviruses are transmitted via inhalation, studying immunological events in the airways is of importance to understand the processes leading to immunopathogenesis. Here, we studied 17 patients infected with Puumala virus that causes a mild form of hemorrhagic fever with renal syndrome (HFRS). Bronchial biopsies as well as longitudinal blood draws were obtained from the patients. During the acute stage of disease, a significant influx of MNPs expressing HLA-DR, CD11c or CD123 was detected in the patients' bronchial tissue. In parallel, absolute numbers of MNPs were dramatically reduced in peripheral blood, coinciding with viremia. Expression of CCR7 on the remaining MNPs in blood suggested migration to peripheral and/or lymphoid tissues. Numbers of MNPs in blood subsequently normalized during the convalescent phase of the disease when viral RNA was no longer detectable in plasma. Finally, we exposed blood MNPs in vitro to Puumala virus, and demonstrated an induction of CCR7 expression on MNPs. In conclusion, the present study shows a marked redistribution of blood MNPs to the airways during acute hantavirus disease, a process that may underlie the local immune activation and contribute to immunopathogenesis in hantavirus-infected patients.


Subject(s)
Endothelium, Vascular/virology , Hantavirus Infections/immunology , Hemorrhagic Fever with Renal Syndrome/virology , Phagocytes/virology , Hantavirus Pulmonary Syndrome/immunology , Hantavirus Pulmonary Syndrome/virology , Hemorrhagic Fever with Renal Syndrome/immunology , Humans , Immunity, Humoral/immunology , Phagocytes/immunology , RNA, Viral/genetics
8.
Immunology ; 151(1): 122-135, 2017 05.
Article in English | MEDLINE | ID: mdl-28106253

ABSTRACT

Beside its key diagnostic value, the humoral immune response is thought to play a protective role in hantavirus pulmonary syndrome. However, little is known about the cell source of these antibodies during ongoing human infection. Herein we characterized B-cell subsets circulating in Andes-virus-infected patients. A notable potent plasmablast (PB) response that increased 100-fold over the baseline levels was observed around 1 week after the onset of symptoms. These PB present a CD3neg CD19low CD20neg CD38hi CD27hi CD138+/- IgA+/- surface phenotype together with the presence of cytoplasmic functional immunoglobulins. They are large lymphocytes (lymphoblasts) morphologically coincident with the 'immunoblast-like' cells that have been previously described during blood cytology examinations of hantavirus-infected patients. Immunoreactivity analysis of white blood cell lysates suggests that some circulating PB are virus-specific but we also observed a significant increase of reactivity against virus-unrelated antigens, which suggests a possible bystander effect by polyclonal B-cell activation. The presence of this large and transient PB response raises the question as to whether these cells might have a protective or pathological role during the ongoing hantavirus pulmonary syndrome and suggest their practical application as a diagnostic/prognostic biomarker.


Subject(s)
B-Lymphocyte Subsets/immunology , Hantavirus Pulmonary Syndrome/immunology , Orthohantavirus/immunology , Plasma Cells/immunology , Precursor Cells, B-Lymphoid/immunology , Acute Disease , Adult , Antibodies, Viral/blood , Antigens, CD/metabolism , Autoantigens/immunology , B-Lymphocyte Subsets/virology , Biomarkers/metabolism , Cell Proliferation , Female , Hantavirus Pulmonary Syndrome/diagnosis , Humans , Immunoglobulin A/metabolism , Lymphocyte Activation , Male , Middle Aged , Plasma Cells/virology , Precursor Cells, B-Lymphoid/virology , Young Adult
9.
J Cell Biochem ; 118(8): 2320-2324, 2017 08.
Article in English | MEDLINE | ID: mdl-28106282

ABSTRACT

Hantaviruses are emerging viral pathogens that causes hantavirus cardiopulmonary syndrome (HCPS) in the Americas, a severe, sometimes fatal, respiratory disease in humans with a case fatality rate of ≥50%. IgM and IgG-based serological detection methods are the most common approaches used for laboratory diagnosis of hantaviruses. Such emerging viral pathogens emphasizes the need for improved rapid diagnostic devices and vaccines incorporating pan-specific epitopes of genotypes. We predicted linear B-cell epitopes for hantaviruses that are specific to genotypes causing HCPS in humans using in silico prediction servers. We modeled the Andes and Sin Nombre hantavirus nucleocapsid protein to locate the identified epitopes. Based on the mean percent prediction probability score, epitope IMASKSVGS/TAEEKLKKKSAF was identified as the best candidate B-cell epitope specific for hantaviruses causing HCPS. Promiscuous epitopes were identified in the C-terminal of the protein. Our study for the first time has reported pan-specific B-cell epitopes for developing immunoassays in the detection of antibodies to hantaviruses causing HCPS. Identification of epitopes with pan-specific recognition of all genotypes causing HCPS could be valuable for the development of immunodiagnositic tools toward pan-detection of hantavirus antibodies in ELISA. J. Cell. Biochem. 118: 2320-2324, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/immunology , Hantavirus Pulmonary Syndrome/immunology , Hantavirus Pulmonary Syndrome/metabolism , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/immunology , Orthohantavirus/immunology , Genotype , Orthohantavirus/pathogenicity , Hantavirus Infections/immunology , Hantavirus Infections/metabolism , Humans , Immunoassay , Protein Structure, Secondary
10.
Chest ; 150(2): e49-52, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27502993

ABSTRACT

A 44-year-old man from Connecticut with no significant past medical history presented to the ED with a 2-week history of sore throat and fatigue, subsequently developing cough, dyspnea, fevers, and chills. The patient reported buying an old camper van and noticed a large infestation of rodent droppings, which he had cleaned thoroughly from the cabin. He used the camper van on several camping trips in Vermont, and symptoms started on his return.


Subject(s)
Fatigue/etiology , Hantavirus Pulmonary Syndrome/complications , Pharyngitis/etiology , Adult , Antibodies, Viral/immunology , Cough/etiology , Dyspnea/etiology , Fever/etiology , Hantavirus Pulmonary Syndrome/diagnostic imaging , Hantavirus Pulmonary Syndrome/immunology , Humans , Lung/diagnostic imaging , Male , Radiography, Thoracic , Tomography, X-Ray Computed
11.
Eur J Clin Microbiol Infect Dis ; 35(4): 713-21, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26873376

ABSTRACT

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.


Subject(s)
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
12.
PLoS One ; 10(4): e0122935, 2015.
Article in English | MEDLINE | ID: mdl-25856432

ABSTRACT

Long-tailed pygmy rice rats (Oligoryzomys longicaudatus) are principal reservoir hosts of Andes virus (ANDV) (Bunyaviridae), which causes most hantavirus cardiopulmonary syndrome cases in the Americas. To develop tools for the study of the ANDV-host interactions, we used RNA-Seq to generate a de novo transcriptome assembly. Splenic RNA from five rice rats captured in Chile, three of which were ANDV-infected, was used to generate an assembly of 66,173 annotated transcripts, including noncoding RNAs. Phylogenetic analysis of selected predicted proteins showed similarities to those of the North American deer mouse (Peromyscus maniculatus), the principal reservoir of Sin Nombre virus (SNV). One of the infected rice rats had about 50-fold more viral burden than the others, suggesting acute infection, whereas the remaining two had levels consistent with persistence. Differential expression analysis revealed distinct signatures among the infected rodents. The differences could be due to 1) variations in viral load, 2) dimorphic or reproductive differences in splenic homing of immune cells, or 3) factors of unknown etiology. In the two persistently infected rice rats, suppression of the JAK-STAT pathway at Stat5b and Ccnot1, elevation of Casp1, RIG-I pathway factors Ppp1cc and Mff, and increased FC receptor-like transcripts occurred. Caspase-1 and Stat5b activation pathways have been shown to stimulate T helper follicular cell (TFH) development in other species. These data are also consistent with reports suggestive of TFH stimulation in deer mice experimentally infected with hantaviruses. In the remaining acutely infected rice rat, the apoptotic pathway marker Cox6a1 was elevated, and putative anti-viral factors Abcb1a, Fam46c, Spp1, Rxra, Rxrb, Trmp2 and Trim58 were modulated. Transcripts for preproenkephalin (Prenk) were reduced, which may be predictive of an increased T cell activation threshold. Taken together, this transcriptome dataset will permit rigorous examination of rice rat-ANDV interactions and may lead to better understanding of virus ecology.


Subject(s)
Hantavirus Infections/veterinary , Hantavirus Pulmonary Syndrome/veterinary , Orthohantavirus/genetics , Sigmodontinae/genetics , Sin Nombre virus/genetics , Transcriptome , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/immunology , Animals , Caspase 1/genetics , Caspase 1/immunology , Gene Expression Regulation , Genetic Markers , Orthohantavirus/pathogenicity , Hantavirus Infections/immunology , Hantavirus Infections/virology , Hantavirus Pulmonary Syndrome/immunology , Hantavirus Pulmonary Syndrome/virology , Host-Pathogen Interactions , Male , Mitochondrial Proteins/genetics , Mitochondrial Proteins/immunology , Peromyscus/classification , Peromyscus/genetics , Peromyscus/immunology , Peromyscus/virology , Phylogeny , RNA/genetics , RNA/immunology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Sigmodontinae/classification , Sigmodontinae/immunology , Sigmodontinae/virology , Signal Transduction , Sin Nombre virus/pathogenicity , Spleen/immunology , Spleen/virology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/virology , Viral Load/genetics
13.
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
14.
PLoS Pathog ; 10(11): e1004521, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25412359

ABSTRACT

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.


Subject(s)
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
15.
Sci Transl Med ; 6(264): 264ra162, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25429055

ABSTRACT

Polyclonal immunoglobulin-based medical products have been used successfully to treat diseases caused by viruses for more than a century. We demonstrate the use of DNA vaccine technology and transchromosomal bovines (TcBs) to produce fully human polyclonal immunoglobulins (IgG) with potent antiviral neutralizing activity. Specifically, two hantavirus DNA vaccines [Andes virus (ANDV) DNA vaccine and Sin Nombre virus (SNV) DNA vaccine] were used to produce a candidate immunoglobulin product for the prevention and treatment of hantavirus pulmonary syndrome (HPS). A needle-free jet injection device was used to vaccinate TcB, and high-titer neutralizing antibodies (titers >1000) against both viruses were produced within 1 month. Plasma collected at day 10 after the fourth vaccination was used to produce purified α-HPS TcB human IgG. Treatment with 20,000 neutralizing antibody units (NAU)/kg starting 5 days after challenge with ANDV protected seven of eight animals, whereas zero of eight animals treated with the same dose of normal TcB human IgG survived. Likewise, treatment with 20,000 NAU/kg starting 5 days after challenge with SNV protected immunocompromised hamsters from lethal HPS, protecting five of eight animals. Our findings that the α-HPS TcB human IgG is capable of protecting in animal models of lethal HPS when administered after exposure provides proof of concept that this approach can be used to develop candidate next-generation polyclonal immunoglobulin-based medical products without the need for human donors, despeciation protocols, or inactivated/attenuated vaccine antigen.


Subject(s)
Hantavirus Pulmonary Syndrome/immunology , Hantavirus Pulmonary Syndrome/prevention & control , Immunoglobulin G/biosynthesis , Vaccines, DNA/immunology , Animals , Animals, Genetically Modified , Antibodies, Neutralizing/immunology , Antibody Formation/immunology , Biological Availability , Cattle , Cricetinae , Disease Models, Animal , Female , Orthohantavirus/immunology , Hantavirus Pulmonary Syndrome/virology , Humans , Sin Nombre virus/immunology , Vaccination
16.
PLoS One ; 9(9): e108067, 2014.
Article in English | MEDLINE | ID: mdl-25243411

ABSTRACT

In addition to conventional antibodies, camelids produce immunoglobulins G composed exclusively of heavy chains in which the antigen binding site is formed only by single domains called VHH. Their particular characteristics make VHHs interesting tools for drug-delivery, passive immunotherapy and high-throughput diagnosis. Hantaviruses are rodent-borne viruses of the Bunyaviridae family. Two clinical forms of the infection are known. Hemorrhagic Fever with Renal Syndrome (HFRS) is present in the Old World, while Hantavirus Pulmonary Syndrome (HPS) is found on the American continent. There is no specific treatment for HPS and its diagnosis is carried out by molecular or serological techniques, using mainly monoclonal antibodies or hantavirus nucleoprotein (N) to detect IgM and IgG in patient serum. This study proposes the use of camelid VHHs to develop alternative methods for diagnosing and confirming HPS. Phage display technology was employed to obtain VHHs. After immunizing one Lama glama against the recombinant N protein (prNΔ85) of a Brazilian hantavirus strain, VHH regions were isolated to construct an immune library. VHHs were displayed fused to the M13KO7 phage coat protein III and the selection steps were performed on immobilized prNΔ85. After selection, eighty clones recognized specifically the N protein. These were sequenced, grouped based mainly on the CDRs, and five clones were analyzed by western blot (WB), surface plasmon resonance (SPR) device, and ELISA. Besides the ability to recognize prNΔ85 by WB, all selected clones showed affinity constants in the nanomolar range. Additionaly, the clone KC329705 is able to detect prNΔ85 in solution, as well as the native viral antigen. Findings support the hypothesis that selected VHHs could be a powerful tool in the development of rapid and accurate HPS diagnostic assays, which are essential to provide supportive care to patients and reduce the high mortality rate associated with hantavirus infections.


Subject(s)
Camelus/immunology , Hantavirus Pulmonary Syndrome/diagnosis , Immunoglobulin Fragments/immunology , Nucleoproteins/immunology , Orthohantavirus/immunology , Amino Acid Sequence , Animals , Antibodies, Viral/biosynthesis , Early Diagnosis , Hantavirus Pulmonary Syndrome/immunology , Humans , Immunoglobulin Fragments/chemistry , Male , Molecular Sequence Data , Sequence Homology, Amino Acid , Surface Plasmon Resonance
17.
Curr Gene Ther ; 14(3): 200-10, 2014.
Article in English | MEDLINE | ID: mdl-24867065

ABSTRACT

Sin Nombre virus (SNV) and Andes virus (ANDV) cause most of the hantavirus pulmonary syndrome (HPS) cases in North and South America, respectively. The chances of a patient surviving HPS are only two in three. Previously, we demonstrated that SNV and ANDV DNA vaccines encoding the virus envelope glycoproteins elicit high-titer neutralizing antibodies in laboratory animals, and (for ANDV) in nonhuman primates (NHPs). In those studies, the vaccines were delivered by gene gun or muscle electroporation. Here, we tested whether a combined SNV/ANDV DNA vaccine (HPS DNA vaccine) could be delivered effectively using a disposable syringe jet injection (DSJI) system (PharmaJet, Inc). PharmaJet intramuscular (IM) and intradermal (ID) needle-free devices are FDA 510(k)-cleared, simple to use, and do not require electricity or pressurized gas. First, we tested the SNV DNA vaccine delivered by PharmaJet IM or ID devices in rabbits and NHPs. Both IM and ID devices produced high-titer anti-SNV neutralizing antibody responses in rabbits and NHPs. However, the ID device required at least two vaccinations in NHP to detect neutralizing antibodies in most animals, whereas all animals vaccinated once with the IM device seroconverted. Because the IM device was more effective in NHP, the Stratis(®) (PharmaJet IM device) was selected for follow-up studies. We evaluated the HPS DNA vaccine delivered using Stratis(®) and found that it produced high-titer anti-SNV and anti-ANDV neutralizing antibodies in rabbits (n=8/group) as measured by a classic plaque reduction neutralization test and a new pseudovirion neutralization assay. We were interested in determining if the differences between DSJI delivery (e.g., high-velocity liquid penetration through tissue) and other methods of vaccine injection, such as needle/syringe, might result in a more immunogenic DNA vaccine. To accomplish this, we compared the HPS DNA vaccine delivered by DSJI versus needle/syringe in NHPs (n=8/group). We found that both the anti-SNV and anti-ANDV neutralizing antibody titers were significantly higher (p-value 0.0115) in the DSJI-vaccinated groups than the needle/syringe group. For example, the anti-SNV and anti-ANDV PRNT50 geometric mean titers (GMTs) were 1,974 and 349 in the DSJI-vaccinated group versus 87 and 42 in the needle/syringe group. These data demonstrate, for the first time, that a spring-powered DSJI device is capable of effectively delivering a DNA vaccine to NHPs. Whether this HPS DNA vaccine, or any DNA vaccine, delivered by spring-powered DSJI will elicit a strong immune response in humans, requires clinical trials.


Subject(s)
Hantavirus Pulmonary Syndrome/prevention & control , Vaccination/instrumentation , Vaccines, DNA/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Electroporation , Female , Orthohantavirus , Hantavirus Infections/immunology , Hantavirus Infections/prevention & control , Hantavirus Pulmonary Syndrome/immunology , Injections, Intramuscular , Neutralization Tests , Primates , Rabbits , Sin Nombre virus
18.
Swiss Med Wkly ; 144: w13937, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24652684

ABSTRACT

In humans, hantaviruses can cause haemorrhagic fever with renal syndrome (HFRS) or hantavirus cardiopulmonary syndrome (HCPS). Currently it is estimated that 150,000 to 200,000 cases of hantavirus disease occur each year, the majority being reported in Asia. However, human hantavirus infections are increasingly reported in the Americas and Europe. Although many of the underlying pathogenic mechanisms still remain unclear, recent evidence rather argues against a purely immune-mediated pathophysiology of human disease. Despite the high morbidity and case-fatality rates of HFRS and HCPS, respectively, no vaccine or drug is currently proven to be preventive or therapeutic. This review summarises clinical features and current epidemiological findings, as well as concepts regarding the immunology, pathogenesis and intervention strategies of human hantaviral diseases.


Subject(s)
Hantavirus Pulmonary Syndrome , Hemorrhagic Fever with Renal Syndrome , Animals , Hantavirus Pulmonary Syndrome/diagnosis , Hantavirus Pulmonary Syndrome/drug therapy , Hantavirus Pulmonary Syndrome/epidemiology , Hantavirus Pulmonary Syndrome/immunology , Hemorrhagic Fever with Renal Syndrome/diagnosis , Hemorrhagic Fever with Renal Syndrome/drug therapy , Hemorrhagic Fever with Renal Syndrome/epidemiology , Hemorrhagic Fever with Renal Syndrome/immunology , Humans
19.
J Virol ; 88(2): 811-9, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24198421

ABSTRACT

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.


Subject(s)
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
20.
Viruses ; 5(9): 2320-8, 2013 Sep 24.
Article in English | MEDLINE | ID: mdl-24064796

ABSTRACT

Hantaviruses are widespread emergent zoonotic agents that cause unapparent or limited disease in their rodent hosts, yet cause acute, often fatal pulmonary or renal infections in humans. Previous laboratory experiments with rodent reservoir hosts indicate that hantaviruses can be cleared from host blood early in the infection cycle, while sequestered long term in various host organs. Field studies of North American deer mice (Peromyscus maniculatus), the natural reservoir of Sin Nombre hantavirus, have shown that viral RNA can be transiently detected well past the early acute infection stage, but only in the minority of infected mice. Here, using a non-degenerate RT-PCR assay optimized for SNV strains known to circulate in Montana, USA, we show that viral RNA can be repeatedly detected on a monthly basis in up to 75% of antibody positive deer mice for periods up to 3-6 months. More importantly, our data show that antibody positive male deer mice are more than twice as likely to have detectable SNV RNA in their blood as antibody positive females, suggesting that SNV-infected male deer mice are more likely to shed virus and for longer periods of time.


Subject(s)
Antibodies, Viral/immunology , Hantavirus Pulmonary Syndrome/veterinary , Rodent Diseases/virology , Sin Nombre virus/isolation & purification , Viremia/veterinary , Animals , Female , Hantavirus Pulmonary Syndrome/immunology , Hantavirus Pulmonary Syndrome/virology , Male , Mice , Montana , Peromyscus , Rodent Diseases/immunology , Sin Nombre virus/genetics , Sin Nombre virus/immunology , Species Specificity , Viremia/immunology , Viremia/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...