Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
2.
J Virol ; 91(10)2017 05 15.
Article En | MEDLINE | ID: mdl-28250124

Crimean-Congo hemorrhagic fever virus (CCHFV) is a bunyavirus causing severe hemorrhagic fever disease in humans, with high mortality rates. The requirement of a high-containment laboratory and the lack of an animal model hampered the study of the immune response and protection of vaccine candidates. Using the recently developed interferon alpha receptor knockout (IFNAR-/-) mouse model, which replicates human disease, we investigated the immunogenicity and protection of two novel CCHFV vaccine candidates: a DNA vaccine encoding a ubiquitin-linked version of CCHFV Gc, Gn, and N and one using transcriptionally competent virus-like particles (tc-VLPs). In contrast to most studies that focus on neutralizing antibodies, we measured both humoral and cellular immune responses. We demonstrated a clear and 100% efficient preventive immunity against lethal CCHFV challenge with the DNA vaccine. Interestingly, there was no correlation with the neutralizing antibody titers alone, which were higher in the tc-VLP-vaccinated mice. However, the animals with a lower neutralizing titer, but a dominant cell-mediated Th1 response and a balanced Th2 response, resisted the CCHFV challenge. Moreover, we found that in challenged mice with a Th1 response (immunized by DNA/DNA and boosted by tc-VLPs), the immune response changed to Th2 at day 9 postchallenge. In addition, we were able to identify new linear B-cell epitope regions that are highly conserved between CCHFV strains. Altogether, our results suggest that a predominantly Th1-type immune response provides the most efficient protective immunity against CCHFV challenge. However, we cannot exclude the importance of the neutralizing antibodies as the surviving immunized mice exhibited substantial amounts of them.IMPORTANCE Crimean-Congo hemorrhagic fever virus (CCHFV) is responsible for hemorrhagic diseases in humans, with a high mortality rate. There is no FDA-approved vaccine, and there are still gaps in our knowledge of the immune responses to infection. The recently developed mouse models mimic human CCHF disease and are useful to study the immunogenicity and the protection by vaccine candidates. Our study shows that mice vaccinated with a specific DNA vaccine were fully protected. Importantly, we show that neutralizing antibodies are not sufficient for protection against CCHFV challenge but that an extra Th1-specific cellular response is required. Moreover, we describe the identification of five conserved B-cell epitopes, of which only one was previously known, that could be of great importance for the development of diagnostics tools and the improvement of vaccine candidates.


Capsid Proteins/immunology , Hemorrhagic Fever, Crimean/immunology , Hemorrhagic Fever, Crimean/prevention & control , Plasmids/genetics , Vaccines, DNA/immunology , Vaccines, Virus-Like Particle/immunology , Viral Envelope Proteins/immunology , Animals , Antibodies, Neutralizing/blood , Capsid Proteins/genetics , Disease Models, Animal , Epitopes, B-Lymphocyte/immunology , Hemorrhagic Fever Virus, Crimean-Congo/chemistry , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever Virus, Crimean-Congo/immunology , Hemorrhagic Fever, Crimean/virology , Humans , Immunity, Cellular , Immunization , Immunogenicity, Vaccine , Interferon-alpha/deficiency , Interferon-alpha/genetics , Mice , Mice, Knockout , Plasmids/administration & dosage , Th1 Cells , Th2 Cells , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Vaccines, Virus-Like Particle/administration & dosage , Viral Envelope Proteins/genetics
3.
J Virol Methods ; 242: 35-45, 2017 04.
Article En | MEDLINE | ID: mdl-28025125

There is an urgent need for Ebola virus (EBOV) proteins, EBOV-specific antibodies and recombinant antigens to be used in diagnostics and as potential vaccine candidates. Our objective was to produce and purify recombinant proteins for immunological assays and for the production of polyclonal EBOV specific antibodies. In addition, a limited comparison of the adjuvant effects of Freund's complete adjuvant (FCA) and adjuvant system 03 (AS03) was carried out. Recombinant EBOV GST-VP24, -VP30, -VP35, -VP40 and -NP were produced in E. coli and purified with affinity chromatography followed by preparative gel electrophoresis. Recombinant EBOV GP-His was produced in Sf9 insect cells and purified by preparative gel electrophoresis. To compare the adjuvant effect of FCA and AS03, 12 rabbits were immunized four times with one of the six recombinant EBOV proteins using FCA or AS03. In addition, three guinea pigs were immunized with EBOV VP24 using FCA. With the exception of sera from two rabbits immunized with GST-VP24, the antisera against all other EBOV proteins showed very high and specific antibody responses after three to four immunizations. The adjuvant effect of AS03 was comparable to that of FCA. The produced antibodies recognized the corresponding EBOV proteins in wild type EBOV-infected cells.


Adjuvants, Immunologic , Ebolavirus/immunology , Freund's Adjuvant , Polysorbates , Squalene , Viral Proteins/immunology , Viral Proteins/isolation & purification , alpha-Tocopherol , Animals , Antibodies, Viral/biosynthesis , Antibodies, Viral/blood , Antibodies, Viral/immunology , Baculoviridae/genetics , Drug Combinations , Ebolavirus/chemistry , Ebolavirus/isolation & purification , Freund's Adjuvant/supply & distribution , Guinea Pigs , Rabbits , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Viral Proteins/genetics
4.
Open Forum Infect Dis ; 3(4): ofw198, 2016 Oct.
Article En | MEDLINE | ID: mdl-27975074

We describe an imported case of Lassa fever with both encephalopathy and bilateral sensorineural hearing deficit. Absence of fever during hospitalization, initially nonspecific symptoms, and onset of hearing deficit in a late stage of disease probably contributed to delayed diagnosis (14 days after admittance to hospital). The pathogenesis of neurological manifestations of Lassa fever is poorly understood and no specific treatment was given. A total of 118 personnel had close contact with the patient, but no secondary cases occurred. This case highlights the importance of considering Lassa fever as a differential diagnosis in patients with recent travel to endemic areas.

5.
J Clin Microbiol ; 54(10): 2521-9, 2016 10.
Article En | MEDLINE | ID: mdl-27466385

Rapid bedside inactivation of Ebola virus would be a solution for the safety of medical and technical staff, risk containment, sample transport, and high-throughput or rapid diagnostic testing during an outbreak. We show that the commercially available Magna Pure lysis/binding buffer used for nucleic acid extraction inactivates Ebola virus. A rapid bedside inactivation method for nucleic acid tests is obtained by simply adding Magna Pure lysis/binding buffer directly into vacuum blood collection EDTA tubes using a thin needle and syringe prior to sampling. The ready-to-use inactivation vacuum tubes are stable for more than 4 months, and Ebola virus RNA is preserved in the Magna Pure lysis/binding buffer for at least 5 weeks independent of the storage temperature. We also show that Ebola virus RNA can be manually extracted from Magna Pure lysis/binding buffer-inactivated samples using the QIAamp viral RNA minikit. We present an easy and convenient method for bedside inactivation using available blood collection vacuum tubes and reagents. We propose to use this simple method for fast, safe, and easy bedside inactivation of Ebola virus for safe transport and routine nucleic acid detection.


Disinfection/methods , Hemorrhagic Fever, Ebola/diagnosis , Hemorrhagic Fever, Ebola/virology , Point-of-Care Systems , RNA, Viral/isolation & purification , Specimen Handling/methods , Virus Inactivation , Humans , Temperature , Time Factors
6.
J Biol Chem ; 291(2): 582-92, 2016 Jan 08.
Article En | MEDLINE | ID: mdl-26574543

Viruses have developed distinct strategies to overcome the host defense system. Regulation of apoptosis in response to viral infection is important for virus survival and dissemination. Like other viruses, Crimean-Congo hemorrhagic fever virus (CCHFV) is known to regulate apoptosis. This study, for the first time, suggests that the non-structural protein NSs of CCHFV, a member of the genus Nairovirus, induces apoptosis. In this report, we demonstrated the expression of CCHFV NSs, which contains 150 amino acid residues, in CCHFV-infected cells. CCHFV NSs undergoes active degradation during infection. We further demonstrated that ectopic expression of CCHFV NSs induces apoptosis, as reflected by caspase-3/7 activity and cleaved poly(ADP-ribose) polymerase, in different cell lines that support CCHFV replication. Using specific inhibitors, we showed that CCHFV NSs induces apoptosis via both intrinsic and extrinsic pathways. The minimal active region of the CCHFV NSs protein was determined to be 93-140 amino acid residues. Using alanine scanning, we demonstrated that Leu-127 and Leu-135 are the key residues for NSs-induced apoptosis. Interestingly, CCHFV NSs co-localizes in mitochondria and also disrupts the mitochondrial membrane potential. We also demonstrated that Leu-127 and Leu-135 are important residues for disruption of the mitochondrial membrane potential by NSs. Therefore, these results indicate that the C terminus of CCHFV NSs triggers mitochondrial membrane permeabilization, leading to activation of caspases, which, ultimately, leads to apoptosis. Given that multiple factors contribute to apoptosis during CCHFV infection, further studies are needed to define the involvement of CCHFV NSs in regulating apoptosis in infected cells.


Apoptosis/drug effects , Hemorrhagic Fever Virus, Crimean-Congo/chemistry , Membrane Potential, Mitochondrial/drug effects , Viral Nonstructural Proteins/pharmacology , Amino Acid Sequence , Animals , Cell Line, Tumor , Hemorrhagic Fever, Crimean/virology , Humans , Leucine/metabolism , Mice, Inbred BALB C , Molecular Sequence Data , Mutant Proteins/pharmacology , Signal Transduction/drug effects , Transfection , Viral Nonstructural Proteins/chemistry , Virus Replication/drug effects
7.
Euro Surveill ; 20(44)2015.
Article En | MEDLINE | ID: mdl-26558690

In the absence of a vaccine or specific treatments for Ebola virus disease (EVD), early identification of cases is crucial for the control of EVD epidemics. We evaluated a new extraction kit (SpeedXtract (SE), Qiagen) on sera and swabs in combination with an improved diagnostic reverse transcription recombinase polymerase amplification assay for the detection of Ebola virus (EBOV-RT-RPA). The performance of combined extraction and detection was best for swabs. Sensitivity and specificity of the combined SE and EBOV-RT-RPA were tested in a mobile laboratory consisting of a mobile glovebox and a Diagnostics-in-a-Suitcase powered by a battery and solar panel, deployed to Matoto Conakry, Guinea as part of the reinforced surveillance strategy in April 2015 to reach the goal of zero cases. The EBOV-RT-RPA was evaluated in comparison to two real-time PCR assays. Of 928 post-mortem swabs, 120 tested positive, and the combined SE and EBOV-RT-RPA yielded a sensitivity and specificity of 100% in reference to one real-time RT-PCR assay. Another widely used real-time RT-PCR was much less sensitive than expected. Results were provided very fast within 30 to 60 min, and the field deployment of the mobile laboratory helped improve burial management and community engagement.


Ebolavirus/isolation & purification , Hemorrhagic Fever, Ebola/diagnosis , Recombinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Disease Outbreaks , Early Diagnosis , Ebolavirus/genetics , Guinea , Hemorrhagic Fever, Ebola/virology , Humans , RNA, Viral/analysis , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Sensitivity and Specificity , Time Factors
8.
J Med Virol ; 87(4): 686-93, 2015 Apr.
Article En | MEDLINE | ID: mdl-25649667

Crimean-Congo hemorrhagic fever (CCHF) is an arthropod-borne disease of humans associated with a severe clinical picture, including hemorrhagic syndrome and a high mortality rate. CCHF virus is widely distributed throughout large areas of the world. To characterize the serological status in CCHF patients, paired clinical samples were collected from suspected CCHF patients and analyzed by microbiological and other laboratory analyses with the aim of: determining the presence of neutralizing antibodies against CCHF virus; investigating the cross-reactivity of these neutralizing antibodies against virus isolated from the same outbreak and against other available laboratory strain; and studying the relationship between the isolated virus with other virus by whole genome sequencing. Patients at Boo-Ali Hospital, Zahedan, Iran, with clinical symptoms ranging from mild to severe hemorrhagic fever were included in the study. Two serum samples were taken from each patient, the first as soon as the patient matched the criteria for CCHF notification and the second when the patient was discharged from hospital (2 weeks later). Commercial and in-house assays revealed a positive IgM signal in acute serum samples from six patients. A novel finding was that CCHF patients develop neutralizing antibodies soon after infection. Interestingly these antibodies were able to neutralize other CCHF virus strains too. The complete sequence of the Zahedan 2007 isolate, including the hitherto unknown first L-segment sequence, was identified using an original clinical sample from one patient with confirmed CCHF infection.


Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Disease Outbreaks , Hemorrhagic Fever Virus, Crimean-Congo/immunology , Hemorrhagic Fever Virus, Crimean-Congo/isolation & purification , Hemorrhagic Fever, Crimean/immunology , Hemorrhagic Fever, Crimean/virology , Adolescent , Adult , Cluster Analysis , Cross Reactions , Female , Genome, Viral , Hemorrhagic Fever, Crimean/epidemiology , Hemorrhagic Fever, Crimean/pathology , Humans , Immunoglobulin M/blood , Iran/epidemiology , Male , Middle Aged , Molecular Sequence Data , Sequence Analysis, DNA , Sequence Homology , Young Adult
9.
J Gen Virol ; 96(Pt 3): 538-546, 2015 Mar.
Article En | MEDLINE | ID: mdl-25481756

Pathogenesis of viral haemorrhagic fevers is associated with alteration of vascular barrier function and haemorrhage. To date, the specific mechanism behind this is unknown. Programmed cell death and regulation of apoptosis in response to viral infection is an important factor for host or virus survival but this has not been well-studied in the case of Crimean-Congo hemorrhagic fever virus (CCHFV). In this study, we demonstrated that CCHFV infection suppresses cleavage of poly(ADP-ribose) polymerase (PARP), triggered by staurosporine early post-infection. We also demonstrated that CCHFV infection suppresses activation of caspase-3 and caspase-9. Most interestingly, we found that CCHFV N can suppress induction of apoptosis by Bax and inhibit the release of cytochrome c from the inner membrane of mitochondria to cytosol. However, CCHFV infection induces activation of Bid late post-infection, suggesting activation of extrinsic apoptotic signalling. Consistently, supernatant from cells stimulated late post-infection was found to induce PARP cleavage, most probably through the TNF-α death receptor pathway. In summary, we found that CCHFV has strategies to interplay with apoptosis pathways and thereby regulate caspase cascades. We suggest that CCHFV suppresses caspase activation at early stages of the CCHFV replication cycle, which perhaps benefits the establishment of infection. Furthermore, we suggest that the host cellular response at late stages post-infection induces host cellular pro-apoptotic molecules through the death receptor pathway.


Apoptosis/physiology , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Virus Replication/physiology , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Cytokines/genetics , Cytokines/metabolism , Enzyme Activation , Gene Expression Regulation, Viral/physiology , Humans , Nucleoproteins/genetics , Nucleoproteins/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
10.
Emerg Infect Dis ; 18(12): 1958-65, 2012 Dec.
Article En | MEDLINE | ID: mdl-23171700

Crimean-Congo hemorrhagic fever (CCHF) is a highly contagious viral tick-borne disease with case-fatality rates as high as 50%. We describe a collaborative evaluation of the characteristics, performance, and on-site applicability of serologic and molecular assays for diagnosis of CCHF. We evaluated ELISA, immunofluorescence, quantitative reverse transcription PCR, and low-density macroarray assays for detection of CCHF virus using precharacterized archived patient serum samples. Compared with results of local, in-house methods, test sensitivities were 87.8%-93.9% for IgM serology, 80.4%-86.1% for IgG serology, and 79.6%-83.3% for genome detection. Specificity was excellent for all assays; molecular test results were influenced by patient country of origin. Our findings demonstrate that well-characterized, reliable tools are available for CCHF diagnosis and surveillance. The on-site use of such assays by health laboratories would greatly diminish the time, costs, and risks posed by the handling, packaging, and shipping of highly infectious biologic material.


Hemorrhagic Fever Virus, Crimean-Congo/isolation & purification , Hemorrhagic Fever, Crimean/diagnosis , Enzyme-Linked Immunosorbent Assay , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever Virus, Crimean-Congo/immunology , Humans , Microarray Analysis , Polymerase Chain Reaction , Reagent Kits, Diagnostic , Reproducibility of Results , Sensitivity and Specificity , Serologic Tests
11.
J Virol ; 86(22): 12294-303, 2012 Nov.
Article En | MEDLINE | ID: mdl-22951837

Crimean-Congo hemorrhagic fever, a severe hemorrhagic disease found throughout Africa, Europe, and Asia, is caused by the tick-borne Crimean-Congo hemorrhagic fever virus (CCHFV). CCHFV is a negative-sense single-stranded RNA (ssRNA) virus belonging to the Nairovirus genus of the Bunyaviridae family. Its genome of three single-stranded RNA segments is encapsidated by the nucleocapsid protein (CCHFV N) to form the ribonucleoprotein complex. This ribonucleoprotein complex is required during replication and transcription of the viral genomic RNA. Here, we present the crystal structures of the CCHFV N in two distinct forms, an oligomeric form comprised of double antiparallel superhelices and a monomeric form. The head-to-tail interaction of the stalk region of one CCHFV N subunit with the base of the globular body of the adjacent subunit stabilizes the helical organization of the oligomeric form of CCHFV N. It also masks the conserved caspase-3 cleavage site present at the tip of the stalk region from host cell caspase-3 interaction and cleavage. By incubation with primer-length ssRNAs, we also obtained the crystal structure of CCHFV N in its monomeric form, which is similar to a recently published structure. The conformational change of CCHFV N upon deoligomerization results in the exposure of the caspase-3 cleavage site and subjects CCHFV N to caspase-3 cleavage. Mutations of this cleavage site inhibit cleavage by caspase-3 and result in enhanced viral polymerase activity. Thus, cleavage of CCHFV N by host cell caspase-3 appears to be crucial for controlling viral RNA synthesis and represents an important host defense mechanism against CCHFV infection.


Caspase 3/metabolism , Hemorrhagic Fever Virus, Crimean-Congo/metabolism , Nucleoproteins/chemistry , Binding Sites , Cloning, Molecular , Crystallography, X-Ray/methods , Genome, Viral , Hemorrhagic Fever Virus, Crimean-Congo/chemistry , Humans , Models, Molecular , Molecular Conformation , Protein Binding , Protein Conformation , Protein Structure, Tertiary , RNA/metabolism , RNA, Viral/metabolism , Transcription, Genetic
12.
Vaccine ; 30(44): 6225-9, 2012 Sep 28.
Article En | MEDLINE | ID: mdl-22902680

Crimean-Congo hemorrhagic fever virus (CCHFV) poses a great threat to public health due to its high mortality and transmission rate and wide geographical distribution. There is currently no specific antiviral therapy for CCHF. This study provides the first in-depth analysis of the cellular and humoral immune response in healthy individuals following injection of inactivated Bulgarian vaccine, the only CCHFV vaccine available at present. Vaccinated individuals developed robust, anti-CCHFV-specific T-cell activity as measured by IFN-γ ELISpot assay. The frequency of IFN-γ secreting T-cells was 10-fold higher in individuals after vaccination with four doses than after one single dose. High levels of CCHFV antibodies were observed following the first dose, but repeated doses were required to achieve antibodies with neutralizing activity against CCHFV. However, the neutralizing activity in these groups was low.


Hemorrhagic Fever Virus, Crimean-Congo/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology , Adult , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Enzyme-Linked Immunospot Assay , Female , Humans , Interferon-gamma/metabolism , Male , Middle Aged , Neutralization Tests , T-Lymphocytes/immunology , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
13.
J Virol ; 85(15): 7766-74, 2011 Aug.
Article En | MEDLINE | ID: mdl-21632768

Crimean-Congo hemorrhagic fever virus (CCHFV) causes viral hemorrhagic fever with high case-fatality rates and is geographically widely distributed. Due to the requirement for a biosafety level 4 (BSL-4) laboratory and the lack of an animal model, knowledge of the viral pathogenesis is limited. Crimean-Congo hemorrhagic fever (CCHF) is characterized by hemorrhage and vascular permeability, indicating the involvement of endothelial cells (ECs). The interplay between ECs and CCHFV is therefore important for understanding the pathogenesis of CCHF. In a previous study, we found that CCHFV-infected monocyte-derived dendritic cells (moDCs) activated ECs; however, the direct effect of CCHFV on ECs was not investigated. Here, we report that ECs are activated upon infection, as demonstrated by upregulation of mRNA levels for E-selectin, vascular cell adhesion molecule 1 (VCAM1), and intercellular adhesion molecule 1 (ICAM1). Protein levels and cell surface expression of ICAM1 responded in a dose-dependent manner to increasing CCHFV titers with concomitant increase in leukocyte adhesion. Furthermore, we examined vascular endothelial (VE) cadherin in CCHFV-infected ECs by different approaches. Infected ECs released higher levels of interleukin 6 (IL-6) and IL-8; however, stimulation of resting ECs with supernatants derived from infected ECs did not result in increased ICAM1 expression. Interestingly, the moDC-mediated activation of ECs was abrogated by addition of neutralizing tumor necrosis factor alpha (TNF-α) antibody to moDC supernatants, thereby identifying this soluble mediator as the key cytokine causing EC activation. We conclude that CCHFV can exert both direct and indirect effects on ECs.


Endothelium, Vascular/virology , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Animals , Antigens, CD/metabolism , Blotting, Western , Cadherins/metabolism , Cells, Cultured , Chlorocebus aethiops , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Intercellular Adhesion Molecule-1/metabolism , Polymerase Chain Reaction , Vero Cells
14.
J Biol Chem ; 286(5): 3227-34, 2011 Feb 04.
Article En | MEDLINE | ID: mdl-21123175

Regulation of apoptosis during infection has been observed for several viral pathogens. Programmed cell death and regulation of apoptosis in response to a viral infection are important factors for host or virus survival. It is not known whether Crimean-Congo hemorrhagic fever virus (CCHFV) infection regulates the apoptosis process in vitro. This study for the first time suggests that CCHFV induces apoptosis, which may be dependent on caspase-3 activation. This study also shows that the coding sequence of the S segment of CCHFV contains a proteolytic cleavage site, DEVD, which is conserved in all CCHFV strains. By using different recombinant expression systems and site-directed mutagenesis, we demonstrated that this motif is subject to caspase cleavage. We also demonstrate that CCHFV nucleocapsid protein (NP) is cleaved into a 30-kDa fragment at the same time as caspase activity is induced during infection. Using caspase inhibitors and cells lacking caspase-3, we clearly demonstrate that the cleavage of NP is caspase-3-dependent. We also show that the inhibition of apoptosis induced progeny viral titers of ∼80-90%. Thus, caspase-3-dependent cleavage of NP may represent a host defense mechanism against lytic CCHFV infection. Taken together, these data suggest that the most abundant protein of CCHFV, which has several essential functions such as protection of viral RNA and participation in various processes in the replication cycle, can be subjected to cleavage by host cell caspases.


Apoptosis , Caspase 3/metabolism , Hemorrhagic Fever, Crimean/pathology , Nucleocapsid Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Enzyme Activation , Hemorrhagic Fever Virus, Crimean-Congo/pathogenicity , Hemorrhagic Fever, Crimean/immunology , Host-Pathogen Interactions/immunology , Humans , Hydrolysis , Peptide Hydrolases , Vero Cells
15.
J Virol ; 84(16): 8275-86, 2010 Aug.
Article En | MEDLINE | ID: mdl-20519402

The spread of the recently emerged, highly pathogenic H5N1 avian influenza virus has raised concern. Preclinical studies suggest that passive immunotherapy could be a new form of treatment for H5N1 virus infection. Here, a neutralizing monoclonal antibody (MAb) against the hemagglutinin (HA) of the influenza A/chicken/Hatay/2004 H5N1 virus, MAb 9F4, was generated and characterized. MAb 9F4 binds both the denatured and native forms of HA. It was shown to recognize the HA proteins of three heterologous strains of H5N1 viruses belonging to clades 1, 2.1, and 2.2, respectively. By use of lentiviral pseudotyped particles carrying HA on the surface, MAb 9F4 was shown to effectively neutralize the homologous strain, Hatay04, and another clade 1 strain, VN04, at a neutralization titer of 8 ng/ml. Furthermore, MAb 9F4 also neutralized two clade 2 viruses at a neutralizing titer of 40 ng/ml. The broad cross-neutralizing activity of MAb 9F4 was confirmed by its ability to neutralize live H5N1 viruses of clade 2.2.2. Epitope-mapping analysis revealed that MAb 9F4 binds a previously uncharacterized epitope below the globular head of the HA1 subunit. Consistently, this epitope is well conserved among the different clades of H5N1 viruses. MAb 9F4 does not block the interaction between HA and its receptor but prevents the pH-mediated conformational change of HA. MAb 9F4 was also found to be protective, both prophylactically and therapeutically, against a lethal viral challenge of mice. Taken together, our results showed that MAb 9F4 is a neutralizing MAb that binds a novel and well-conserved epitope in the HA1 subunit of H5N1 viruses.


Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Epitopes/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H5N1 Subtype/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/isolation & purification , Antibodies, Viral/therapeutic use , Body Weight , Conserved Sequence , Cross Protection , Cross Reactions , Epitope Mapping , Female , Mice , Mice, Inbred BALB C , Neutralization Tests , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/prevention & control , Survival Analysis , Virus Internalization/drug effects
16.
Open Virol J ; 4: 38-41, 2010 Apr 22.
Article En | MEDLINE | ID: mdl-20517491

As a first line of defence against a virus infection, mammalian cells elicit an innate immune response, characterized by secretion of type I interferons (IFN) and up-regulation of interferon stimulated genes (ISGs). We have previously included Crimean Congo Hemorrhagic Fever Virus (CCHFV) in the list of type I IFN-sensitive viruses. In this in vitro study, we have compared the antiviral activity of two recombinant IFN-alpha preparations (Roferon A and Intron A) with a natural IFN-alpha produced in human leukocytes (Multiferon). Our results clearly demonstrate that these commercially available IFNs have significant antiviral activities against CCHFV. However, we could show that Multiferon inhibits viral replication more efficiently than the two recombinant IFN alpha preparations.

17.
J Gen Virol ; 91(Pt 6): 1473-7, 2010 Jun.
Article En | MEDLINE | ID: mdl-20164263

Crimean-Congo hemorrhagic fever virus (CCHFV) poses a great threat to public health due to its high mortality, transmission and geographical distribution. To date, there is no vaccine or specific treatment available and the knowledge regarding its pathogenesis is highly limited. Using a small-animal model system, this study showed that adult mice missing the type I interferon (IFN) receptor (IFNAR(-/-)) were susceptible to CCHFV and developed an acute disease with fatal outcome. In contrast, infection of wild-type mice (129 Sv/Ew) was asymptomatic. Viral RNA was found in all analysed organs of the infected mice, but the amount of CCHFV RNA was significantly higher in the IFNAR(-/-) mice than in the wild-type mice. Furthermore, the liver of IFNAR(-/-) mice was enlarged significantly, showing that IFN is important for limiting virus spread and protecting against liver damage in mice.


Hemorrhagic Fever Virus, Crimean-Congo/immunology , Hemorrhagic Fever Virus, Crimean-Congo/pathogenicity , Hemorrhagic Fever, Crimean/immunology , Hemorrhagic Fever, Crimean/mortality , Receptor, Interferon alpha-beta/deficiency , Animal Structures/virology , Animals , Female , Hemorrhagic Fever, Crimean/pathology , Hemorrhagic Fever, Crimean/virology , Mice , Mice, Knockout , Survival Analysis
18.
J Med Virol ; 80(8): 1397-404, 2008 Aug.
Article En | MEDLINE | ID: mdl-18551619

As a first line of defence against virus infection, mammalian cells elicit an innate immune response, characterized by secretion of type I interferons and the up-regulation of interferon stimulated genes. Many viruses down-regulate the innate immune responses in order to enhance their virulence. Crimean-Congo hemorrhagic fever virus (CCHFV), a Nairovirus of the family Bunyaviridae is the causative agent of severe hemorrhagic fever in humans with high mortality. Knowledge regarding the innate immune response against CCHFV is most limited. Interestingly, in this study it is shown that replicating CCHFV delays substantially the IFN response, possibly by interfering with the activation pathway of IRF-3. In addition, it is demonstrated that CCHFV replication is almost insensitive to subsequent treatment with interferon-alpha. Once the virus is replicating, virus replication is more or less insensitive to the antiviral effects induced by the interferon. By using an interferon bioassay, it is shown that infected cells secrete interferon relatively late after infection, that is, 48 hr post-infection. In summary, the results suggest the presence of a virulence factor encoded by CCHFV that delays the host defence in order to allow rapid viral spread in the host.


Antiviral Agents , Hemorrhagic Fever Virus, Crimean-Congo/pathogenicity , Immunity, Innate , Interferon-alpha , Interferon-beta , Animals , Antiviral Agents/metabolism , Antiviral Agents/pharmacology , Cell Line , Chlorocebus aethiops , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Humans , Interferon-alpha/metabolism , Interferon-alpha/pharmacology , Interferon-beta/metabolism , Interferon-beta/pharmacology , Time Factors , Vero Cells , Virulence Factors/metabolism , Virus Replication/drug effects
...