Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Front Immunol ; 12: 603228, 2021.
Article in English | MEDLINE | ID: mdl-33815363

ABSTRACT

Background: New World Hantaviruses (NWHs) are the etiological agent underlying hantavirus cardiopulmonary syndrome (HCPS), a severe respiratory disease with high mortality rates in humans. In Panama, infections with Choclo Orthohantavirus (CHOV) cause a much milder illness characterized by higher seroprevalence and lower mortality rates. To date, the cytokine profiles and antibody responses associated with this milder form of HCPS have not been defined. Therefore, in this study, we examined immune serological profiles associated with CHOV infections. Methods: For this retrospective study, sera from fifteen individuals with acute CHOV-induced HCPS, were analyzed alongside sera from fifteen convalescent phase individuals and thirty-three asymptomatic, CHOV-seropositive individuals. Cytokine profiles were analyzed by multiplex immunoassay. Antibody subclasses, binding, and neutralization against CHOV-glycoprotein (CHOV-GP) were evaluated by ELISA, and flow cytometry. Results: High titers of IFNγ, IL-4, IL-8, and IL-10 serum cytokines were found in the acute individuals. Elevated IL-4 serum levels were found in convalescent and asymptomatic seropositive individuals. High titers of IgG1 subclass were observed across the three cohorts analyzed. Neutralizing antibody response against CHOV-GP was detectable in few acute individuals but was strong in both convalescent and asymptomatic seropositive individuals. Conclusion: A Th1/Th2 cytokine signature is characteristic during acute mild HCPS caused by CHOV infection. High expression of Th2 and IL-8 cytokines are correlated with clinical parameters in acute mild HCPS. In addition, a strong IL-4 signature is associated with different cohorts, including asymptomatic individuals. Furthermore, asymptomatic individuals presented high titers of neutralizing antibodies.


Subject(s)
Antibodies, Viral , Cytokines , Hantavirus Infections , Immunoglobulin G , Orthohantavirus , Adult , Antibodies, Viral/blood , Antibodies, Viral/immunology , Cytokines/blood , Cytokines/immunology , Female , Orthohantavirus/immunology , Orthohantavirus/metabolism , Hantavirus Infections/blood , Hantavirus Infections/immunology , Humans , Immunoglobulin G/blood , Immunoglobulin G/immunology , Male , Middle Aged , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
2.
Anal Chim Acta ; 1147: 30-37, 2021 Feb 22.
Article in English | MEDLINE | ID: mdl-33485583

ABSTRACT

Simple, low-cost, and sensitive new platforms for electrochemical immunosensors for virus detection have been attracted attention due to the recent pandemic caused by a new type of coronavirus (SARS-CoV-2). In the present work, we report for the first time the construction of an immunosensor using a commercial 3D conductive filament of carbon black and polylactic acid (PLA) to detect Hantavirus Araucaria nucleoprotein (Np) as a proof-of-concept. The recognition biomolecule was anchored directly at the filament surface by using N-(3-Dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride and N-Hydroxysuccinimide (EDC/NHS). Conductive and non-conductive composites of PLA were characterized using thermal gravimetric analysis (TGA), revealing around 30% w/w of carbon in the filament. Morphological features of composites were obtained from SEM and TEM measurements. FTIR measurement revealed that crosslinking agents were covalently bonded at the filament surface. Electrochemical techniques such as cyclic voltammetry (CV) and electrochemical impedance spectroscopy (EIS) were used for the evaluation of each step involved in the construction of the proposed immunosensor. The results showed the potentiality of the device for the quantitative detection of Hantavirus Araucaria nucleoprotein (Np) from 30 µg mL-1 to 240 µg mL-1 with a limit of detection of 22 µg mL-1. Also, the proposed immunosensor was applied with success for virus detection in 100x diluted human serum samples. Therefore, the PLA conductive filament with carbon black is a simple and excellent platform for immunosensing, which offers naturally carboxylic groups able to anchor covalently biomolecules.


Subject(s)
Antibodies, Viral/immunology , Immunoassay/methods , Nucleocapsid Proteins/immunology , Printing, Three-Dimensional , Antibodies, Immobilized/chemistry , Antibodies, Immobilized/immunology , COVID-19/diagnosis , COVID-19/virology , Dielectric Spectroscopy , Electrodes , Orthohantavirus/isolation & purification , Orthohantavirus/metabolism , Hantavirus Infections/diagnosis , Hantavirus Infections/virology , Humans , Immunoassay/instrumentation , Limit of Detection , Nucleocapsid Proteins/blood , SARS-CoV-2/isolation & purification , Soot/chemistry
3.
Cell ; 183(2): 442-456.e16, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32937107

ABSTRACT

Hantaviruses are rodent-borne viruses causing serious zoonotic outbreaks worldwide for which no treatment is available. Hantavirus particles are pleomorphic and display a characteristic square surface lattice. The envelope glycoproteins Gn and Gc form heterodimers that further assemble into tetrameric spikes, the lattice building blocks. The glycoproteins, which are the sole targets of neutralizing antibodies, drive virus entry via receptor-mediated endocytosis and endosomal membrane fusion. Here we describe the high-resolution X-ray structures of the heterodimer of Gc and the Gn head and of the homotetrameric Gn base. Docking them into an 11.4-Å-resolution cryoelectron tomography map of the hantavirus surface accounted for the complete extramembrane portion of the viral glycoprotein shell and allowed a detailed description of the surface organization of these pleomorphic virions. Our results, which further revealed a built-in mechanism controlling Gc membrane insertion for fusion, pave the way for immunogen design to protect against pathogenic hantaviruses.


Subject(s)
Membrane Glycoproteins/metabolism , Membrane Glycoproteins/ultrastructure , Orthohantavirus/chemistry , Glycoproteins/chemistry , Glycoproteins/ultrastructure , Orthohantavirus/metabolism , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/physiology , Protein Conformation , RNA Viruses , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/ultrastructure , Virion , Virus Internalization
4.
PLoS Pathog ; 16(4): e1008483, 2020 04.
Article in English | MEDLINE | ID: mdl-32330200

ABSTRACT

Pathogenic hantaviruses, genus Orthohantaviridae, are maintained in rodent reservoirs with zoonotic transmission to humans occurring through inhalation of rodent excreta. Hantavirus disease in humans is characterized by localized vascular leakage and elevated levels of circulating proinflammatory cytokines. Despite the constant potential for deadly zoonotic transmission to humans, specific virus-host interactions of hantaviruses that lead to innate immune activation, and how these processes impart disease, remain unclear. In this study, we examined the mechanisms of viral recognition and innate immune activation of Hantaan orthohantavirus (HTNV) infection. We identified the RIG-I-like receptor (RLR) pathway as essential for innate immune activation, interferon (IFN) production, and interferon stimulated gene (ISG) expression in response to HTNV infection in human endothelial cells, and in murine cells representative of a non-reservoir host. Our results demonstrate that innate immune activation and signaling through the RLR pathway depends on viral replication wherein the host response can significantly restrict replication in target cells in a manner dependent on the type 1 interferon receptor (IFNAR). Importantly, following HTNV infection of a non-reservoir host murine model, IFNAR-deficient mice had higher viral loads, increased persistence, and greater viral dissemination to lung, spleen, and kidney compared to wild-type animals. Surprisingly, this response was MAVS independent in vivo. Innate immune profiling in these tissues demonstrates that HTNV infection triggers expression of IFN-regulated cytokines early during infection. We conclude that the RLR pathway is essential for recognition of HTNV infection to direct innate immune activation and control of viral replication in vitro, and that additional virus sensing and innate immune response pathways of IFN and cytokine regulation contribute to control of HTNV in vivo. These results reveal a critical role for innate immune regulation in driving divergent outcomes of HTNV infection, and serve to inform studies to identify therapeutic targets to alleviate human hantavirus disease.


Subject(s)
DEAD Box Protein 58/immunology , Hantavirus Infections/immunology , Interferon Type I/immunology , Orthohantavirus/physiology , Virus Replication/physiology , Animals , Chlorocebus aethiops , Cytokines/immunology , Cytokines/metabolism , DEAD Box Protein 58/metabolism , DEAD-box RNA Helicases/metabolism , Endothelial Cells/metabolism , Orthohantavirus/immunology , Orthohantavirus/metabolism , Orthohantavirus/pathogenicity , Hantavirus Infections/metabolism , Hantavirus Infections/virology , Human Umbilical Vein Endothelial Cells , Humans , Immunity, Innate , Interferon Type I/metabolism , Interferon-beta/metabolism , Mice , Receptor, Interferon alpha-beta/metabolism , Receptors, Immunologic , Signal Transduction/immunology , Vero Cells
5.
Virology ; 543: 54-62, 2020 04.
Article in English | MEDLINE | ID: mdl-32056847

ABSTRACT

Hantaviruses are rodent-borne hemorrhagic fever viruses leading to serious diseases. Viral attachment and entry represent the first steps in virus transmission and are promising targets for antiviral therapeutic intervention. Here we investigated receptor use in human airway epithelium of the Old and New World hantaviruses Hantaan virus (HTNV) and Andes virus (ANDV). Using a biocontained recombinant vesicular stomatitis virus pseudotype platform, we provide first evidence for a role of the cellular phosphatidylserine (PS) receptors of the T-cell immunoglobulin and mucin (TIM) protein family in HTNV and ANDV infection. In line with previous studies, HTNV, but not ANDV, was able to use glycosaminoglycan heparan sulfate and αvß3 integrin as co-receptors. In sum, our studies demonstrate for the first time that hantaviruses make use of apoptotic mimicry for infection of human airway epithelium, which may explain why these viruses can easily break the species barrier.


Subject(s)
Hantaan virus/metabolism , Membrane Glycoproteins/metabolism , Orthohantavirus/metabolism , Receptors, Cell Surface/metabolism , Receptors, Virus/metabolism , Respiratory Mucosa/virology , Viral Envelope Proteins/metabolism , Animals , Bacteriocins/pharmacology , Cell Line, Tumor , Epithelial Cells/metabolism , Epithelial Cells/virology , Hantaan virus/drug effects , Hantaan virus/pathogenicity , Hantaan virus/physiology , Orthohantavirus/physiology , Haplorhini , Heparitin Sulfate/pharmacology , Hepatitis A Virus Cellular Receptor 1/metabolism , Humans , Integrins/metabolism , Membrane Proteins/metabolism , Molecular Mimicry , Peptides/pharmacology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Respiratory Mucosa/metabolism , Vesiculovirus/metabolism , Vesiculovirus/physiology , Axl Receptor Tyrosine Kinase
6.
J Gen Virol ; 100(8): 1208-1221, 2019 08.
Article in English | MEDLINE | ID: mdl-31268416

ABSTRACT

The family Hantaviridae mostly comprises rodent-borne segmented negative-sense RNA viruses, many of which are capable of causing devastating disease in humans. In contrast, hantavirus infection of rodent hosts results in a persistent and inapparent infection through their ability to evade immune detection and inhibit apoptosis. In this study, we used Tula hantavirus (TULV) to investigate the interplay between viral and host apoptotic responses during early, peak and persistent phases of virus infection in cell culture. Examination of early-phase TULV infection revealed that infected cells were refractory to apoptosis, as evidenced by the complete lack of cleaved caspase-3 (casp-3C) staining, whereas in non-infected bystander cells casp-3C was highly abundant. Interestingly, at later time points, casp-3C was abundant in infected cells, but the cells remained viable and able to continue shedding infectious virus, and together these observations were suggestive of a TULV-associated apoptotic block. To investigate this block, we viewed TULV-infected cells using laser scanning confocal and wide-field deconvolution microscopy, which revealed that TULV nucleocapsid protein (NP) colocalized with, and sequestered, casp-3C within cytoplasmic ultrastructures. Consistent with casp-3C colocalization, we showed for the first time that TULV NP was cleaved in cells and that TULV NP and casp-3C could be co-immunoprecipitated, suggesting that this interaction was stable and thus unlikely to be solely confined to NP binding as a substrate to the casp-3C active site. To account for these findings, we propose a novel mechanism by which TULV NP inhibits apoptosis by spatially sequestering casp-3C from its downstream apoptotic targets within the cytosol.


Subject(s)
Apoptosis , Caspase 3/metabolism , Hantavirus Infections/enzymology , Nucleocapsid Proteins/metabolism , Orthohantavirus/metabolism , Animals , Caspase 3/genetics , Cytosol/enzymology , Cytosol/virology , Orthohantavirus/genetics , Hantavirus Infections/genetics , Hantavirus Infections/physiopathology , Hantavirus Infections/virology , Host-Pathogen Interactions , Humans , Nucleocapsid Proteins/genetics , Protein Binding
7.
Elife ; 82019 06 10.
Article in English | MEDLINE | ID: mdl-31180319

ABSTRACT

The hantavirus envelope glycoproteins Gn and Gc mediate virion assembly and cell entry, with Gc driving fusion of viral and endosomal membranes. Although the X-ray structures and overall arrangement of Gn and Gc on the hantavirus spikes are known, their detailed interactions are not. Here we show that the lateral contacts between spikes are mediated by the same 2-fold contacts observed in Gc crystals at neutral pH, allowing the engineering of disulfide bonds to cross-link spikes. Disrupting the observed dimer interface affects particle assembly and overall spike stability. We further show that the spikes display a temperature-dependent dynamic behavior at neutral pH, alternating between 'open' and 'closed' forms. We show that the open form exposes the Gc fusion loops but is off-pathway for productive Gc-induced membrane fusion and cell entry. These data also provide crucial new insights for the design of optimized Gn/Gc immunogens to elicit protective immune responses.


Subject(s)
Glycoproteins/metabolism , Orthohantavirus/metabolism , Viral Envelope Proteins/metabolism , Virus Assembly , Virus Internalization , Amino Acid Sequence , Crystallography, X-Ray , Glycoproteins/chemistry , Glycoproteins/genetics , Orthohantavirus/genetics , Orthohantavirus/physiology , Hydrogen-Ion Concentration , Membrane Fusion , Models, Molecular , Protein Conformation , Protein Multimerization , Protein Stability , Sequence Homology, Amino Acid , Temperature , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
8.
J Virol ; 93(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-30867297

ABSTRACT

Andes virus (ANDV) causes hantavirus pulmonary syndrome (HPS) and is the only hantavirus shown to spread person to person and cause a highly lethal HPS-like disease in Syrian hamsters. The unique ability of ANDV N protein to inhibit beta interferon (IFNß) induction may contribute to its virulence and spread. Here we analyzed IFNß regulation by ANDV N protein substituted with divergent residues from the nearly identical Maporal virus (MAPV) N protein. We found that MAPV N fails to inhibit IFNß signaling and that replacing ANDV residues 252 to 296 with a hypervariable domain (HVD) from MAPV N prevents IFNß regulation. In addition, changing ANDV residue S386 to the histidine present in MAPV N or the alanine present in other hantaviruses prevented ANDV N from regulating IFNß induction. In contrast, replacing serine with phosphoserine-mimetic aspartic acid (S386D) in ANDV N robustly inhibited interferon regulatory factor 3 (IRF3) phosphorylation and IFNß induction. Additionally, the MAPV N protein gained the ability to inhibit IRF3 phosphorylation and IFNß induction when ANDV HVD and H386D replaced MAPV residues. Mass spectroscopy analysis of N protein from ANDV-infected cells revealed that S386 is phosphorylated, newly classifying ANDV N as a phosphoprotein and phosphorylated S386 as a unique determinant of IFN regulation. In this context, the finding that the ANDV HVD is required for IFN regulation by S386 but dispensable for IFN regulation by D386 suggests a role for HVD in kinase recruitment and S386 phosphorylation. These findings delineate elements within the ANDV N protein that can be targeted to attenuate ANDV and suggest targeting cellular kinases as potential ANDV therapeutics.IMPORTANCE ANDV contains virulence determinants that uniquely permit it to spread person to person and cause highly lethal HPS in immunocompetent hamsters. We discovered that ANDV S386 and an ANDV-specific hypervariable domain permit ANDV N to inhibit IFN induction and that IFN regulation is directed by phosphomimetic S386D substitutions in ANDV N. In addition, MAPV N proteins containing D386 and ANDV HVD gained the ability to inhibit IFN induction. Validating these findings, mass spectroscopy analysis revealed that S386 of ANDV N protein is uniquely phosphorylated during ANDV infection. Collectively, these findings reveal new paradigms for ANDV N protein as a phosphoprotein and IFN pathway regulator and suggest new mechanisms for hantavirus regulation of cellular kinases and signaling pathways. Our findings define novel IFN-regulating virulence determinants of ANDV, identify residues that can be modified to attenuate ANDV for vaccine development, and suggest the potential for kinase inhibitors to therapeutically restrict ANDV replication.


Subject(s)
Interferon-beta/metabolism , Nucleocapsid Proteins/metabolism , Orthohantavirus/metabolism , Animals , Chlorocebus aethiops , Endothelial Cells/virology , HEK293 Cells , Orthohantavirus/pathogenicity , Hantavirus Infections/virology , Host-Pathogen Interactions , Humans , Interferon-beta/physiology , Interferons/metabolism , Interferons/physiology , Nucleocapsid Proteins/physiology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Serine/metabolism , Signal Transduction , Vero Cells , Virulence , Virulence Factors/metabolism , Virus Replication
9.
Vopr Virusol ; 64(1): 36-41, 2019.
Article in Russian | MEDLINE | ID: mdl-30893528

ABSTRACT

BACKGROUND: Hemorrhagic fever with renal syndrome (HFRS) occupies a leading place among natural focal human diseases in the Russian Federation. Sporadic incidence of HFRS-Sochi has been annually recorded in the Krasnodar Territory since 2000. The group outbreak of the HFRS-Sochi was first registered in Gelendzhik in the fall of 2013. METHODS: Serological methods were used: indirect immunofluorescence, enzyme immunoassay, FRNT in Vero cells, and methods for the viral RNA detection: PCR and RT-PCR. RESULTS: Data of clinical, epidemiological, immunological and molecular studies of 3 out of 4 cases in HFRS-Sochi outbreak are presented. Severity of the disease correlated with early gastrointestinal disorders appearance. Patient MA gastrointestinal disorders were joined on day 3 of a fever. Clinical and laboratory studies revealed signs of kidneys, liver, pancreas damage, bilateral hydrothorax, bilateral polysegmental pneumonia and polyneuropathy. As a result of long-term treatment, the patient recovered. Patient AA had gastrointestinal disturbances the next day after fever onset. The patient was not saved, despite early hospitalization. Hantavirus antigen and RNA were detected in the lung tissues 2 out of 10 Black-Sea field mice captured in the affected area, as well as in the organs of deceased patient. The most severe clinical course of the disease in close relatives, son and father, with a fatal outcome in the latter case may be the result of genetic features. The severity and outcome of the disease was not depend on day of hospitalization and correlated with the early manifestations of gastrointestinal disorders. CONCLUSIONS: Presented data confirm high virulence and pantropism of the Sochi virus, as well as the epidemiological role of Black-Sea field mouse (Apodemus ponticus) as the host of the Sochi virus and the source of human infection.


Subject(s)
Disease Outbreaks , Hemorrhagic Fever with Renal Syndrome , Orthohantavirus , RNA, Viral/blood , Adult , Animals , Chlorocebus aethiops , Female , Orthohantavirus/isolation & purification , Orthohantavirus/metabolism , Hemorrhagic Fever with Renal Syndrome/blood , Hemorrhagic Fever with Renal Syndrome/epidemiology , Humans , Incidence , Male , Middle Aged , Murinae , Russia , Vero Cells
10.
Adv Virus Res ; 98: 83-118, 2017.
Article in English | MEDLINE | ID: mdl-28433053

ABSTRACT

The Bunyavirales Order encompasses nine families of enveloped viruses containing a single-stranded negative-sense RNA genome divided into three segments. The small (S) and large (L) segments encode proteins participating in genome replication in the infected cell cytoplasm. The middle (M) segment encodes the viral glycoproteins Gn and Gc, which are derived from a precursor polyprotein by host cell proteases. Entry studies are available only for a few viruses in the Order, and in each case they were shown to enter cells via receptor-mediated endocytosis. The acidic endosomal pH triggers the fusion of the viral envelope with the membrane of an endosome. Structural studies on two members of this Order, the phleboviruses and the hantaviruses, have shown that the membrane fusion protein Gc displays a class II fusion protein fold and is homologous to its counterparts in flaviviruses and alphaviruses, which are positive-sense, single-stranded RNA viruses. We analyze here recent data on the structure and function of the structure of the phlebovirus Gc and hantavirus Gn and Gc glycoproteins, and extrapolate common features identified in the amino acid sequences to understand also the structure and function of their counterparts in other families of the Bunyavirales Order. Our analysis also identified clear structural homology between the hantavirus Gn and alphavirus E2 glycoproteins, which make a heterodimer with the corresponding fusion proteins Gc and E1, respectively, revealing that not only the fusion protein has been conserved across viral families.


Subject(s)
Genome, Viral , Orthohantavirus/genetics , Phlebovirus/genetics , RNA, Viral/genetics , Viral Fusion Proteins/chemistry , Alphavirus/classification , Alphavirus/genetics , Alphavirus/metabolism , Alphavirus/ultrastructure , Animals , Biological Evolution , Bunyaviridae Infections/virology , Endosomes/metabolism , Endosomes/virology , Orthohantavirus/classification , Orthohantavirus/metabolism , Orthohantavirus/ultrastructure , Humans , Models, Molecular , Phlebovirus/classification , Phlebovirus/metabolism , Phlebovirus/ultrastructure , Phylogeny , Protein Folding , RNA, Viral/metabolism , Structural Homology, Protein , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Virus Internalization
11.
Sci Rep ; 6: 36081, 2016 10 27.
Article in English | MEDLINE | ID: mdl-27786303

ABSTRACT

Hemorrhagic fever with renal syndrome (HFRS) was considered to be transmitted by Apodemus agrarius and Rattus norvegicus, the principal animal hosts of Hantaan virus and Seoul virus, respectively. The aim of this study is to determine the correlation of HFRS incidence with capture rate and hantavirus infection rate of rodent species in Qingdao City, China. We collected HFRS patients' information and captured field and residential rodents in Qingdao City, China from 2010 to 2014. The correlations of HFRS incidence to rodent capture rate and hantavirus infection rate of rodents were analyzed statistically. The main findings of this study are that the high HFRS incidence (19.3/100,000) is correlated to the capture rate of field Mus musculus (p = 0.011, r = 0.037); but surprisingly it did not correlated to the capture rate of the principal rodent hosts Apodemus agrarius and Rattus norvegicus and the hantavirus infection rate of these rodent species in the field or residential area. These novel findings suggest that Mus musculus, a nontraditional animal host of hantavirus may play an important role in hantavirus transmission in Qingdao City.


Subject(s)
Hemorrhagic Fever with Renal Syndrome/epidemiology , Animals , Antigens, Viral/analysis , China/epidemiology , Disease Reservoirs/virology , Fluorescent Antibody Technique, Indirect , Orthohantavirus/isolation & purification , Orthohantavirus/metabolism , Hantavirus Infections/epidemiology , Hantavirus Infections/veterinary , Hantavirus Infections/virology , Humans , Lung/metabolism , Mice , Prevalence , Rats
12.
J Biol Chem ; 291(47): 24702-24714, 2016 Nov 18.
Article in English | MEDLINE | ID: mdl-27733686

ABSTRACT

An evolutionarily conserved sequence at the 5' terminus of hantaviral genomic RNA plays an important role in viral transcription initiation and packaging of the viral genome into viral nucleocapsids. Interaction of viral nucleocapsid protein (N) with this conserved sequence facilitates mRNA translation by a unique N-mediated translation strategy. Whereas this evolutionarily conserved sequence facilitates virus replication with the assistance of N in eukaryotic hosts having multifaceted antiviral defense, we demonstrate its interaction with N presents a novel target for therapeutic intervention of hantavirus disease. Using a high throughput screening approach, we identified three lead inhibitors that bind and induce structural perturbations in N. The inhibitors interrupt N-RNA interaction and abrogate both viral genomic RNA synthesis and N-mediated translation strategy without affecting the canonical translation machinery of the host cell. The inhibitors are well tolerated by cells and inhibit hantavirus replication with the same potency as ribavarin, a commercially available antiviral. We report the identification of a unique chemical scaffold that disrupts a critical RNA-protein interaction in hantaviruses and holds promise for the development of the first anti-hantaviral therapeutic with broad spectrum antiviral activity.


Subject(s)
Antiviral Agents/pharmacology , Hantavirus Infections/drug therapy , Hantavirus Infections/metabolism , Nucleocapsid Proteins/metabolism , Orthohantavirus/metabolism , RNA, Viral/biosynthesis , Antiviral Agents/chemistry , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans
13.
Curr Opin Virol ; 7: 134-40, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25063986

ABSTRACT

The endothelial lining of the vasculature performs a vital role in maintaining fluid barrier functions despite balancing nutrient and fluid content of tissues, repairing localized damage, coordinating responses of a plethora of factors, immune cells and platelets through a multitude of endothelial cell surface receptors. Viruses that nonlytically cause lethal hemorrhagic or edematous diseases engage receptors on vascular and lymphatic endothelial cells, altering normal cellular responses that control capillary leakage and fluid clearance functions with lethal consequences. Recent studies indicate that receptors directing dengue virus and hantavirus infection of the endothelium contribute to the dysregulation of normal endothelial cell signaling responses that control capillary permeability and immune responses that contribute to pathogenesis. Here we present recent studies of virally altered endothelial functions that provide new insight into targeting barrier functions of the endothelium as a potential therapeutic approach.


Subject(s)
Dengue Virus/metabolism , Dengue/metabolism , Endothelial Cells/virology , Hantavirus Infections/metabolism , Orthohantavirus/metabolism , Receptors, Virus/metabolism , Animals , Dengue/virology , Dengue Virus/genetics , Dengue Virus/pathogenicity , Orthohantavirus/genetics , Orthohantavirus/pathogenicity , Hantavirus Infections/virology , Humans
14.
Virus Res ; 187: 72-6, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-24412713

ABSTRACT

Hantaviruses have tri-segmented negative sense RNA genome. The viral M-segment RNA encodes a glycoprotein precursor (GPC), which is cleaved into two glycoprotein molecules Gn and Gc that form spikes on the viral envelope. We previously reported that Gn is degraded shortly after synthesis by the host autophagy machinery. However, Gn being an important integral component of the virion, must escape degradation during the packaging and assembly stage of virus replication cycle. The mechanism regulating the intrinsic steady-state levels of Gn during the course of virus replication cycle is not clear. We transfected cells with plasmids expressing viral S-segment RNA, nucleocapsid protein and glycoproteins Gn and Gc and monitored their expression levels over time. These studies revealed that accumulation of nucleocapsid protein, glycoprotein Gc and viral S-segment RNA helped to stabilize Gn. These observations suggest that initiation of virus assembly may help Gn to escape autophagic degradation by yet unknown mechanism.


Subject(s)
Capsid Proteins/genetics , Gene Expression Regulation, Viral , Genome, Viral , Orthohantavirus/genetics , RNA, Viral/genetics , Viral Core Proteins/genetics , Viral Envelope Proteins/genetics , Animals , Autophagy/genetics , Capsid Proteins/metabolism , Orthohantavirus/metabolism , HeLa Cells , Host-Pathogen Interactions , Humans , Plasmids/chemistry , Protein Stability , Proteolysis , RNA, Viral/metabolism , Transfection , Viral Core Proteins/metabolism , Viral Envelope Proteins/metabolism , Virus Assembly
15.
J Virol ; 88(4): 2344-8, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24335294

ABSTRACT

How hantaviruses assemble and exit infected cells remains largely unknown. Here, we show that the expression of Andes (ANDV) and Puumala (PUUV) hantavirus Gn and Gc envelope glycoproteins lead to their self-assembly into virus-like particles (VLPs) which were released to cell supernatants. The viral nucleoprotein was not required for particle formation. Further, a Gc endodomain deletion mutant did not abrogate VLP formation. The VLPs were pleomorphic, exposed protrusions and reacted with patient sera.


Subject(s)
Orthohantavirus/metabolism , Puumala virus/metabolism , Viral Envelope Proteins/metabolism , Virion/metabolism , Blotting, Western , Cross Reactions/immunology , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Orthohantavirus/genetics , Humans , Puumala virus/genetics , Virion/genetics
16.
Ann Agric Environ Med ; 20(2): 283-6, 2013.
Article in English | MEDLINE | ID: mdl-23772576

ABSTRACT

Samples of 30 dead small mammals each were collected on area 'A' located in eastern Poland which is exposed to flooding by the Vistula river, and on the area 'B', also located in eastern Poland but not exposed to flooding. Kidneys and livers of the mammals were examined by the PCR and nested PCR methods for the presence of hantavirus RNA. Out of 7 species of small mammals examined, the presence of hantaviruses was detected in 4 of them. Hantavirus prevalence was low in Apodemus agrarius (2.6%), the most numerous mammal species, whereas in the remaining 3 positive species (Microtus agrestis, Myodes glareolus, Sorex araneus) this was 12.5-100%. The presence of hantaviruses was detected only in the animals found on area 'A' exposed to flooding, and their prevalence was statistically greater compared to area 'B' not exposed to flooding (16.7% vs. 0%, p=0.0345). The overall positivity of the examined small mammals population from the areas 'A' and 'B' was 8.3%. The sequence analysis of the samples positive for hantavirus proved that the amplified products showed 77-86% homology with the L segment sequence of hantavirus Fusong-Mf-731 isolated from Microtus fortis in China. The presented study is the first to demonstrate the occurrence of hantavirus infection in small mammals from eastern Poland, and the first to demonstrate the significant relationship between flooding and the prevalence of hantaviruses in small mammals.


Subject(s)
Hantavirus Infections/veterinary , Muridae/virology , Orthohantavirus/isolation & purification , Rodent Diseases/epidemiology , Shrews/virology , Animals , Disease Reservoirs/virology , Orthohantavirus/genetics , Orthohantavirus/metabolism , Hantavirus Infections/epidemiology , Hantavirus Infections/virology , Poland/epidemiology , Polymerase Chain Reaction/veterinary , RNA, Viral/genetics , RNA, Viral/metabolism , Rodent Diseases/virology , Sequence Analysis, DNA/veterinary , Species Specificity
17.
Front Biosci (Elite Ed) ; 5(1): 154-66, 2013 01 01.
Article in English | MEDLINE | ID: mdl-23276978

ABSTRACT

Hantaviruses are zoonotic category-A pathogens that cause highly fatal diseases in humans. The hantaviral genome encodes three viral proteins: RNA-dependent RNA polymerase (RdRp or L protein), nucleocapsid protein (N), and a glycoprotein precursor (GPC), which is post-translationally cleaved into two surface glycoproteins Gn and Gc. The cytoplasmic tail of Gn interferes with interferon signaling pathways. N is a multifunctional molecule that was shown to be involved in the transcription and translation of viral proteins. N binds to the host mRNA caps and protects the degradation of mRNA 5' termini, which are later snatched and used as primers by the viral RdRp during transcription initiation. N also seems to lure the host translation machinery for the preferential translation of viral transcripts. Moreover, N was shown to delay the induction of cellular apoptosis and facilitate the transport and localization of viral ribonucleoproteins (RNPs) by exploiting the cellular cytoskeleton and SUMOlyation machinery. Therefore, with their limited protein coding capacity, hantaviruses have evolved several strategies to modulate cellular pathways for their efficient replication.


Subject(s)
Gene Expression Regulation, Viral/physiology , Immunity, Innate/immunology , Membrane Glycoproteins/metabolism , Orthohantavirus/immunology , Orthohantavirus/physiology , Signal Transduction/physiology , Virus Replication/physiology , Apoptosis/physiology , Orthohantavirus/metabolism , Humans , Models, Biological , Nucleocapsid Proteins/metabolism , RNA Caps/metabolism , Sumoylation
18.
Scand J Infect Dis ; 44(9): 663-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22831170

ABSTRACT

BACKGROUND: Matrix metalloproteinases (MMPs) are a family of proteolytic enzymes with numerous roles in the normal immune response to infection. However, excess MMP activity following infection may lead to immunopathological processes that cause tissue damage. Their activity in normal tissues is subject to tight control, which is regulated by its specific endogenous tissue inhibitors (TIMPs). It is known that MMPs bind to cell surface proteins (e.g. integrins) and that such interactions can have modulatory effects on MMP functionality. The objective of this study was to determine whether there are differences in MMP and TIMP production during the acute phase of infection with different pathogens that use ß-integrins as their receptors for cell entry. METHODS: We measured the total amounts of soluble MMP-2, MMP-9, TIMP-1, and TIMP-2 in the sera from patients infected with Dobrava virus (DOBV), Coxiella burnetii, or uropathogenic Escherichia coli. Statistical analyses were used to correlate MMP/TIMP serum levels with different clinical laboratory parameters. RESULTS: The results showed that both of the bacterial infections generally manifested the stronger effect on MMP production, while in contrast, viral infection introduced stronger changes to metalloproteinase inhibitors. MMPs and TIMPs were significantly correlated with some of the clinical laboratory parameters in both bacterial infections, but no correlations were found for DOBV infection. CONCLUSIONS: These findings suggest diverse mechanisms by which MMP activity could be implicated in the pathology of these 2 bacterial infections versus the viral DOBV infection, despite the type of their cellular entry receptors.


Subject(s)
Collagenases/blood , Gram-Negative Bacterial Infections/blood , Hantavirus Infections/blood , Integrins/metabolism , Tissue Inhibitor of Metalloproteinases/blood , Analysis of Variance , Collagenases/immunology , Coxiella burnetii/metabolism , Escherichia coli/metabolism , Gram-Negative Bacterial Infections/enzymology , Gram-Negative Bacterial Infections/immunology , Orthohantavirus/metabolism , Hantavirus Infections/enzymology , Hantavirus Infections/immunology , Humans , Matrix Metalloproteinase 2/blood , Matrix Metalloproteinase 2/immunology , Matrix Metalloproteinase 9/blood , Matrix Metalloproteinase 9/immunology , Statistics, Nonparametric , Tissue Inhibitor of Metalloproteinase-1/blood , Tissue Inhibitor of Metalloproteinase-1/immunology , Tissue Inhibitor of Metalloproteinase-2/blood , Tissue Inhibitor of Metalloproteinase-2/immunology , Tissue Inhibitor of Metalloproteinases/immunology
19.
J Virol ; 86(18): 10173-85, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22787213

ABSTRACT

Hantaviruses, similarly to other negative-strand segmented RNA viruses, initiate the synthesis of translation-competent capped mRNAs by a unique cap-snatching mechanism. Hantavirus nucleocapsid protein (N) binds to host mRNA caps and requires four nucleotides adjacent to the 5' cap for high-affinity binding. N protects the 5' caps of cellular transcripts from degradation by the cellular decapping machinery. The rescued 5' capped mRNA fragments are stored in cellular P bodies by N, which are later efficiently used as primers by the hantaviral RNA-dependent RNA polymerase (RdRp) for transcription initiation. We showed that N also protects the host mRNA caps in P-body-deficient cells. However, the rescued caps were not effectively used by the hantavirus RdRp during transcription initiation, suggesting that caps stored in cellular P bodies by N are preferred for cap snatching. We examined the characteristics of the 5' terminus of a capped test mRNA to delineate the minimum requirements for a capped transcript to serve as an efficient cap donor during hantavirus cap snatching. We showed that hantavirus RdRp preferentially snatches caps from the nonsense mRNAs compared to mRNAs engaged in translation. Hantavirus RdRp preferentially cleaves the cap donor mRNA at a G residue located 14 nucleotides downstream of the 5' cap. The sequence complementarity between the 3' terminus of viral genomic RNA and the nucleotides located in the vicinity of the cleavage site of the cap donor mRNA favors cap snatching. Our results show that hantavirus RdRp snatches caps from viral mRNAs. However, the negligible cap-donating efficiency of wild-type mRNAs in comparison to nonsense mRNAs suggests that viral mRNAs will not be efficiently used for cap snatching during viral infection due to their continuous engagement in protein synthesis. Our results suggest that efficiency of an mRNA to donate caps for viral mRNA synthesis is primarily regulated at the translational level.


Subject(s)
Orthohantavirus/genetics , Orthohantavirus/metabolism , RNA Caps/genetics , RNA Caps/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Animals , Base Sequence , Binding Sites/genetics , Chlorocebus aethiops , Codon, Nonsense , Gene Knockdown Techniques , Genome, Viral , Orthohantavirus/pathogenicity , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Models, Biological , Nucleocapsid Proteins/genetics , Nucleocapsid Proteins/metabolism , Protein Biosynthesis , RNA, Small Interfering/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Vero Cells
20.
PLoS One ; 7(4): e34307, 2012.
Article in English | MEDLINE | ID: mdl-22506017

ABSTRACT

Hantaviruses (Bunyaviridae) are negative-strand RNA viruses with a tripartite genome. The small (S) segment encodes the nucleocapsid protein and, in some hantaviruses, also the nonstructural protein (NSs). The aim of this study was to find potential cellular partners for the hantaviral NSs protein. Toward this aim, yeast two-hybrid (Y2H) screening of mouse cDNA library was performed followed by a search for potential NSs protein counterparts via analyzing a cellular interactome. The resulting interaction network was shown to form logical, clustered structures. Furthermore, several potential binding partners for the NSs protein, for instance ACBD3, were identified and, to prove the principle, interaction between NSs and ACBD3 proteins was demonstrated biochemically.


Subject(s)
Orthohantavirus/genetics , Orthohantavirus/metabolism , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Gene Library , Host-Pathogen Interactions , Membrane Proteins/metabolism , Mice , Protein Interaction Domains and Motifs , Protein Interaction Mapping/methods , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL