Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Nat Commun ; 15(1): 4542, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38806525

ABSTRACT

The Crimean-Congo hemorrhagic fever virus (CCHFV) is an emerging pathogen of the Orthonairovirus genus that can cause severe and often lethal hemorrhagic diseases in humans. CCHFV has a broad tropism and can infect a variety of species and tissues. Here, by using gene silencing, blocking antibodies or soluble receptor fragments, we identify the low-density lipoprotein receptor (LDL-R) as a CCHFV entry factor. The LDL-R facilitates binding of CCHFV particles but does not allow entry of Hazara virus (HAZV), another member of the genus. In addition, we show that apolipoprotein E (apoE), an exchangeable protein that mediates LDL/LDL-R interaction, is incorporated on CCHFV particles, though not on HAZV particles, and enhances their specific infectivity by promoting an LDL-R dependent entry. Finally, we show that molecules that decrease LDL-R from the surface of target cells could inhibit CCHFV infection. Our study highlights that CCHFV takes advantage of a lipoprotein receptor and recruits its natural ligand to promote entry into cells.


Subject(s)
Apolipoproteins E , Hemorrhagic Fever Virus, Crimean-Congo , Receptors, LDL , Virus Internalization , Humans , Receptors, LDL/metabolism , Apolipoproteins E/metabolism , Apolipoproteins E/genetics , Hemorrhagic Fever Virus, Crimean-Congo/metabolism , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Animals , HEK293 Cells , Chlorocebus aethiops , Hemorrhagic Fever, Crimean/virology , Hemorrhagic Fever, Crimean/metabolism , Virion/metabolism , Vero Cells
2.
Nat Microbiol ; 9(6): 1499-1512, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38548922

ABSTRACT

Climate change and population densities accelerated transmission of highly pathogenic viruses to humans, including the Crimean-Congo haemorrhagic fever virus (CCHFV). Here we report that the Low Density Lipoprotein Receptor (LDLR) is a critical receptor for CCHFV cell entry, playing a vital role in CCHFV infection in cell culture and blood vessel organoids. The interaction between CCHFV and LDLR is highly specific, with other members of the LDLR protein family failing to bind to or neutralize the virus. Biosensor experiments demonstrate that LDLR specifically binds the surface glycoproteins of CCHFV. Importantly, mice lacking LDLR exhibit a delay in CCHFV-induced disease. Furthermore, we identified the presence of Apolipoprotein E (ApoE) on CCHFV particles. Our findings highlight the essential role of LDLR in CCHFV infection, irrespective of ApoE presence, when the virus is produced in tick cells. This discovery holds profound implications for the development of future therapies against CCHFV.


Subject(s)
Apolipoproteins E , Hemorrhagic Fever Virus, Crimean-Congo , Hemorrhagic Fever, Crimean , Receptors, LDL , Virus Internalization , Animals , Humans , Mice , Apolipoproteins E/metabolism , Apolipoproteins E/genetics , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Hemorrhagic Fever, Crimean/virology , Hemorrhagic Fever, Crimean/metabolism , Mice, Knockout , Receptors, LDL/metabolism , Receptors, LDL/genetics , Receptors, Virus/metabolism , Ticks/virology , Ticks/metabolism
3.
Nat Commun ; 14(1): 7365, 2023 11 14.
Article in English | MEDLINE | ID: mdl-37963884

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) is a biosafety level-4 pathogen requiring urgent research and development efforts. The glycoproteins of CCHFV, Gn and Gc, are considered to play multiple roles in the viral life cycle by interactions with host cells; however, these interactions remain largely unclear to date. Here, we analyzed the cellular interactomes of CCHFV glycoproteins and identified 45 host proteins as high-confidence Gn/Gc interactors. These host molecules are involved in multiple cellular biological processes potentially associated with the physiological actions of the viral glycoproteins. Then, we elucidated the role of a representative cellular protein, HAX1. HAX1 interacts with Gn by its C-terminus, while its N-terminal region leads to mitochondrial localization. By the strong interaction, HAX1 sequestrates Gn to mitochondria, thus depriving Gn of its normal Golgi localization that is required for functional glycoprotein-mediated progeny virion packaging. Consistently, the inhibitory activity of HAX1 against viral packaging and hence propagation was further elucidated in the contexts of pseudotyped and authentic CCHFV infections in cellular and animal models. Together, the findings provide a systematic CCHFV Gn/Gc-cell protein-protein interaction map, but also unravel a HAX1/mitochondrion-associated host antiviral mechanism, which may facilitate further studies on CCHFV biology and therapeutic approaches.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo , Hemorrhagic Fever, Crimean , Animals , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever Virus, Crimean-Congo/metabolism , Hemorrhagic Fever, Crimean/metabolism , Glycoproteins/genetics , Glycoproteins/metabolism
4.
Virol Sin ; 38(1): 34-46, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36075566

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) is a biosafety level-4 (BSL-4) pathogen that causes Crimean-Congo hemorrhagic fever (CCHF) characterized by hemorrhagic manifestation, multiple organ failure and high mortality rate, posing great threat to public health. Despite the recently increasing research efforts on CCHFV, host cell responses associated with CCHFV infection remain to be further characterized. Here, to better understand the cellular response to CCHFV infection, we performed a transcriptomic analysis in human kidney HEK293 â€‹cells by high-throughput RNA sequencing (RNA-seq) technology. In total, 496 differentially expressed genes (DEGs), including 361 up-regulated and 135 down-regulated genes, were identified in CCHFV-infected cells. These regulated genes were mainly involved in host processes including defense response to virus, response to stress, regulation of viral process, immune response, metabolism, stimulus, apoptosis and protein catabolic process. Therein, a significant up-regulation of type III interferon (IFN) signaling pathway as well as endoplasmic reticulum (ER) stress response was especially remarkable. Subsequently, representative DEGs from these processes were well validated by RT-qPCR, confirming the RNA-seq results and the typical regulation of IFN responses and ER stress by CCHFV. Furthermore, we demonstrate that not only type I but also type III IFNs (even at low dosages) have substantial anti-CCHFV activities. Collectively, the data may provide new and comprehensive insights into the virus-host interactions and particularly highlights the potential role of type III IFNs in restricting CCHFV, which may help inform further mechanistic delineation of the viral infection and development of anti-CCHFV strategies.


Subject(s)
Biological Phenomena , Hemorrhagic Fever Virus, Crimean-Congo , Hemorrhagic Fever, Crimean , Humans , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever, Crimean/metabolism , Interferon Lambda , HEK293 Cells , Antiviral Agents/metabolism
5.
Sci Rep ; 11(1): 2324, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33504869

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) causes a zoonotic disease, Crimean-Congo hemorrhagic fever (CCHF) endemic in Africa, Asia, the Middle East, and Southeastern Europe. However, the prevalence of CCHF is not monitored in most of the endemic countries due to limited availability of diagnostic assays and biosafety regulations required for handling infectious CCHFV. In this study, we established a protocol to purify the recombinant CCHFV nucleoprotein (NP), which is antigenically highly conserved among multiple lineages/clades of CCHFVs and investigated its utility in an enzyme-linked immunosorbent assay (ELISA) to detect CCHFV-specific antibodies. The NP gene was cloned into the pCAGGS mammalian expression plasmid and human embryonic kidney 293 T cells were transfected with the plasmid. The expressed NP molecule was purified from the cell lysate using cesium-chloride gradient centrifugation. Purified NP was used as the antigen for the ELISA to detect anti-CCHFV IgG. Using the CCHFV NP-based ELISA, we efficiently detected CCHFV-specific IgG in anti-NP rabbit antiserum and CCHFV-infected monkey serum. When compared to the commercially available Blackbox CCHFV IgG ELISA kit, our assay showed equivalent performance in detecting CCHFV-specific IgG in human sera. These results demonstrate the usefulness of our CCHFV NP-based ELISA for seroepidemiological studies.


Subject(s)
Hemorrhagic Fever, Crimean/metabolism , Nucleoproteins/metabolism , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Hemorrhagic Fever, Crimean/blood , Hemorrhagic Fever, Crimean/genetics , Humans , Nucleoproteins/blood , Nucleoproteins/genetics , Plasmids/genetics , Seroepidemiologic Studies
6.
Sci Rep ; 10(1): 12378, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32704046

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne pathogen causing a febrile illness in humans, which can progress to hemorrhagic manifestations, multi-organ failure, and death. Current mouse models of CCHFV infection reliably succumb to virus challenge but vary in their ability to reflect signs of disease similar to humans. In this study, we established a signal transducer and activator of transcription 2 (STAT2) knockout hamster model to expand the repertoire of animal models of CCHFV pathogenesis that can be used for therapeutic development. These hamsters demonstrated a systemic and lethal disease in response to infection. Hallmarks of human disease were observed including petechial rash, blood coagulation dysfunction, and various biochemistry and blood cell count abnormalities. Furthermore, we also demonstrated the utility of this model for anti-CCHFV therapeutic evaluation. The STAT2 knock-out hamster model of CCHFV infection may provide some further insights into clinical disease, viral pathogenesis, and pave the way for testing of potential drug and vaccine candidates.


Subject(s)
Animals, Genetically Modified , Disease Models, Animal , Hemorrhagic Fever Virus, Crimean-Congo/metabolism , Hemorrhagic Fever, Crimean , STAT2 Transcription Factor/deficiency , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Animals, Genetically Modified/virology , Cell Line , Cricetinae , Female , Gene Knockout Techniques , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever, Crimean/genetics , Hemorrhagic Fever, Crimean/metabolism , Hemorrhagic Fever, Crimean/pathology , Male , STAT2 Transcription Factor/metabolism
7.
PLoS Negl Trop Dis ; 14(4): e0008105, 2020 04.
Article in English | MEDLINE | ID: mdl-32251473

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne virus that can cause a hemorrhagic fever in humans, with a case fatality rate of up to 40%. Cases of CCHFV have been reported in Africa, Asia, and southern Europe; and recently, due to the expanding range of its vector, autochthonous cases have been reported in Spain. Although it was discovered over 70 years ago, our understanding of the pathogenesis of this virus remains limited. We used RNA-Seq in two human liver cell lines (HepG2 and Huh7) infected with CCHFV (strain IbAr10200), to examine kinetic changes in host expression and viral replication simultaneously at 1 and 3 days post infection. Through this, numerous host pathways were identified that were modulated by the virus including: antiviral response and endothelial cell leakage. Notably, the genes encoding DDX60, a cytosolic component of the RIG-I signalling pathway and OAS2 were both shown to be dysregulated. Interestingly, PTPRR was induced in Huh7 cells but not HepG2 cells. This has been associated with the TLR9 signalling cascade, and polymorphisms in TLR9 have been associated with poor outcomes in patients. Additionally, we performed whole-genome sequencing on CCHFV to assess viral diversity over time, and its relationship to the host response. As a result, we have demonstrated that through next-generation mRNA deep-sequencing it is possible to not only examine mRNA gene expression, but also to examine viral quasispecies and typing of the infecting strain. This demonstrates a proof-of-principle that CCHFV specimens can be analyzed to identify both the virus and host biomarkers that may have implications for prognosis.


Subject(s)
Gene Expression , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever, Crimean/genetics , Host-Pathogen Interactions/genetics , Liver/metabolism , RNA-Seq/methods , 2',5'-Oligoadenylate Synthetase/genetics , Cell Line , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , Gene Regulatory Networks , Hemorrhagic Fever, Crimean/metabolism , Hemorrhagic Fever, Crimean/virology , Hep G2 Cells , Host-Pathogen Interactions/physiology , Humans , RNA, Messenger , Receptors, Immunologic , Signal Transduction , Toll-Like Receptor 9 , Virus Replication , Exome Sequencing
8.
Autophagy ; 16(10): 1858-1870, 2020 10.
Article in English | MEDLINE | ID: mdl-31905032

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) is a virus that causes severe liver dysfunctions and hemorrhagic fever, with high mortality rate. Here, we show that CCHFV infection caused a massive lipidation of LC3 in hepatocytes. This lipidation was not dependent on ATG5, ATG7 or BECN1, and no signs for recruitment of the alternative ATG12-ATG3 pathway for lipidation was found. Both virus replication and protein synthesis were required for the lipidation of LC3. Despite an augmented transcription of SQSTM1, the amount of proteins did not show a massive and sustained increase in infected cells, indicating that degradation of SQSTM1 by macroautophagy/autophagy was still occurring. The genetic alteration of autophagy did not influence the production of CCHFV particles demonstrating that autophagy was not required for CCHFV replication. Thus, the results indicate that CCHFV multiplication imposes an overtly elevated level of LC3 mobilization that involves a possibly novel type of non-canonical lipidation. Abbreviations: BECN1: Beclin 1; CCHF: Crimean-Congo hemorrhagic fever; CCHFV: Crimean-Congo hemorrhagic fever virus; CHX: cycloheximide; ER: endoplasmic reticulum; GFP: green fluorescent protein; GP: glycoproteins; MAP1LC3: microtubule associated protein 1 light chain 3; MOI: multiplicity of infection; n.i.: non-infected; NP: nucleoprotein; p.i.: post-infection; SQSTM1: sequestosome 1.


Subject(s)
Autophagy , Epithelial Cells/virology , Hemorrhagic Fever Virus, Crimean-Congo/metabolism , Hemorrhagic Fever, Crimean/virology , Virus Replication , Animals , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 7/metabolism , Beclin-1/metabolism , Chlorocebus aethiops , HeLa Cells , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever, Crimean/diagnosis , Hemorrhagic Fever, Crimean/metabolism , Hep G2 Cells , Hepatocytes/virology , Humans , Lipids/chemistry , Microtubule-Associated Proteins/metabolism , Protein Biosynthesis , Sequestosome-1 Protein/metabolism , Vero Cells
9.
J Virol ; 93(18)2019 09 15.
Article in English | MEDLINE | ID: mdl-31292241

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) is a cause of severe hemorrhagic fever. Its tick reservoir and vector are widely distributed throughout Africa, Southern and Eastern Europe, the Middle East, and Asia. Serological evidence suggests that CCHFV can productively infect a wide variety of species, but only humans develop severe, sometimes fatal disease. The role of the host adaptive immunity in control or contribution to the severe pathology seen in CCHF cases is largely unknown. Studies of adaptive immune responses to CCHFV have been limited due to lack of suitable small animal models. Wild-type mice are resistant to CCHFV infection, and type I interferon-deficient mice typically develop a rapid-onset fatal disease prior to development of adaptive immune responses. We report here a mouse model in which type I interferon-deficient mice infected with a clinical isolate of CCHFV develop a severe inflammatory disease but ultimately recover. Recovery was coincident with development of CCHFV-specific B- and T-cell responses that were sustained for weeks postinfection. We also found that recovery from a primary CCHFV infection could protect against disease following homologous or heterologous reinfection. Together this model enables study of multiple aspects of CCHFV pathogenesis, including convalescence, an important aspect of CCHF disease that existing mouse models have been unsuitable for studying.IMPORTANCE The role of antibody or virus-specific T-cell responses in control of acute Crimean-Congo hemorrhagic fever virus infection is largely unclear. This is a critical gap in our understanding of CCHF, and investigation of convalescence following severe acute CCHF has been limited by the lack of suitable small animal models. We report here a mouse model of CCHF in which infected mice develop severe disease but ultimately recover. Although mice developed an inflammatory immune response along with severe liver and spleen pathology, these mice also developed CCHFV-specific B- and T-cell responses and were protected from reinfection. This model provides a valuable tool to investigate how host immune responses control acute CCHFV infection and how these responses may contribute to the severe disease seen in CCHFV-infected humans in order to develop therapeutic interventions that promote protective immune responses.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo/immunology , Hemorrhagic Fever Virus, Crimean-Congo/metabolism , Interferon Type I/genetics , Adaptive Immunity/immunology , Animals , Convalescence , Disease Models, Animal , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Hemorrhagic Fever, Crimean/metabolism , Hemorrhagic Fever, Crimean/virology , Interferon Type I/metabolism , Liver/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Spleen/virology
10.
J Biol Chem ; 294(13): 5023-5037, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30723154

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne Nairovirus that causes severe hemorrhagic fever with a mortality rate of up to 30% in certain outbreaks worldwide. The virus has wide endemic distribution. There is no effective antiviral therapeutic or FDA approved vaccine for this zoonotic viral illness. The multifunctional CCHFV nucleocapsid protein (N protein) plays a crucial role in the establishment of viral infection and is an important structural component of the virion. Here we show that CCHFV N protein has a distant RNA-binding site in the stalk domain that specifically recognizes the vRNA panhandle, formed by the base pairing of complementary nucleotides at the 5' and 3' termini of the vRNA genome. Using multiple approaches, including filter-bonding analysis, GFP reporter assay, and biolayer interferometry we observed an N protein-panhandle interaction both in vitro and in vivo The purified WT CCHFV N protein and the stalk domain also recognize the vRNA panhandle of hazara virus, another Nairovirus in the family Bunyaviridae, demonstrating the genus-specific nature of N protein-panhandle interaction. Another RNA-binding site was identified at the head domain of CCHFV N protein that nonspecifically recognizes the single strand RNA (ssRNA) of viral or nonviral origin. Expression of CCHFV N protein stalk domain active in panhandle binding, dramatically inhibited the hazara virus replication in cell culture, illustrating the role of N protein-panhandle interaction in Nairovirus replication. Our findings reveal the stalk domain of N protein as a potential target in therapeutic interventions to manage CCHFV disease.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo/physiology , Hemorrhagic Fever, Crimean/virology , Nucleocapsid Proteins/metabolism , RNA/metabolism , Binding Sites , Hemorrhagic Fever Virus, Crimean-Congo/chemistry , Hemorrhagic Fever, Crimean/metabolism , Humans , Models, Molecular , Nairovirus/chemistry , Nairovirus/physiology , Nucleocapsid Proteins/chemistry , Protein Domains , Virus Replication
11.
PLoS Pathog ; 13(5): e1006372, 2017 May.
Article in English | MEDLINE | ID: mdl-28542609

ABSTRACT

The recent Middle East respiratory syndrome coronavirus (MERS-CoV), Ebola and Zika virus outbreaks exemplify the continued threat of (re-)emerging viruses to human health, and our inability to rapidly develop effective therapeutic countermeasures. Many viruses, including MERS-CoV and the Crimean-Congo hemorrhagic fever virus (CCHFV) encode deubiquitinating (DUB) enzymes that are critical for viral replication and pathogenicity. They bind and remove ubiquitin (Ub) and interferon stimulated gene 15 (ISG15) from cellular proteins to suppress host antiviral innate immune responses. A variety of viral DUBs (vDUBs), including the MERS-CoV papain-like protease, are responsible for cleaving the viral replicase polyproteins during replication, and are thereby critical components of the viral replication cycle. Together, this makes vDUBs highly attractive antiviral drug targets. However, structural similarity between the catalytic cores of vDUBs and human DUBs complicates the development of selective small molecule vDUB inhibitors. We have thus developed an alternative strategy to target the vDUB activity through a rational protein design approach. Here, we report the use of phage-displayed ubiquitin variant (UbV) libraries to rapidly identify potent and highly selective protein-based inhibitors targeting the DUB domains of MERS-CoV and CCHFV. UbVs bound the vDUBs with high affinity and specificity to inhibit deubiquitination, deISGylation and in the case of MERS-CoV also viral replicative polyprotein processing. Co-crystallization studies further revealed critical molecular interactions between UbVs and MERS-CoV or CCHFV vDUBs, accounting for the observed binding specificity and high affinity. Finally, expression of UbVs during MERS-CoV infection reduced infectious progeny titers by more than four orders of magnitude, demonstrating the remarkable potency of UbVs as antiviral agents. Our results thereby establish a strategy to produce protein-based inhibitors that could protect against a diverse range of viruses by providing UbVs via mRNA or protein delivery technologies or through transgenic techniques.


Subject(s)
Antiviral Agents/pharmacology , Coronavirus Infections/virology , Enzyme Inhibitors/pharmacology , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Hemorrhagic Fever, Crimean/virology , Middle East Respiratory Syndrome Coronavirus/drug effects , Ubiquitin/metabolism , Viral Proteins/antagonists & inhibitors , Antiviral Agents/chemistry , Coronavirus Infections/metabolism , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Hemorrhagic Fever Virus, Crimean-Congo/enzymology , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever, Crimean/metabolism , Humans , Middle East Respiratory Syndrome Coronavirus/enzymology , Middle East Respiratory Syndrome Coronavirus/genetics , Ubiquitination/drug effects , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
12.
Redox Rep ; 22(6): 241-245, 2017 Nov.
Article in English | MEDLINE | ID: mdl-27159644

ABSTRACT

OBJECTIVES: Crimean Congo hemorrhagic fever (CCHF) is the second most common hemorrhagic fever worldwide. This study aimed to evaluate the oxidant-antioxidant balance of patients with CCHF by detecting dynamic thiol disulfide homeostasis (TDH), which is a novel oxidative stress marker, and other molecules, including paraoxonase (PON), arylesterase (ARES), ceruloplasmin (CLP), myeloperoxidase (MPO), and catalase. METHODS: This retrospective, cross-sectional, controlled study, which involved patients with CCHF and healthy volunteers, measured dynamic TDH using a novel automated method developed by Erel. RESULTS: We recruited 69 adult patients with CCHF (31 females, 38 males, median age 46 years). The case fatality rate was 1.49% (1/69). Increased disulfide/native thiol and disulfide/total thiol ratios, decreased total antioxidant status (TAS), and increased total oxidant status (TOS) were found in patients with CCHF. TAS, PON, and ARES values were found to be positively correlated with both native and total thiol levels, whereas TOS and CLP were negatively correlated with both, at a significant level. MPO activity was similar in both groups. DISCUSSION: This is the first study in the literature to evaluate dynamic TDH in CCHF. TDH shifts to the oxidative side in patients with CCHF, leading to an increase in TOS.


Subject(s)
Disulfides/metabolism , Hemorrhagic Fever, Crimean/metabolism , Adult , Antioxidants/metabolism , Aryldialkylphosphatase/metabolism , Carboxylic Ester Hydrolases/metabolism , Catalase/metabolism , Ceruloplasmin/metabolism , Cross-Sectional Studies , Female , Homeostasis/physiology , Humans , Male , Middle Aged , Oxidative Stress/physiology , Peroxidase/metabolism , Retrospective Studies , Sulfhydryl Compounds/metabolism
13.
J Virol ; 90(20): 9305-16, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27512070

ABSTRACT

UNLABELLED: The Nairovirus genus of the Bunyaviridae family contains serious human and animal pathogens classified within multiple serogroups and species. Of these serogroups, the Crimean-Congo hemorrhagic fever virus (CCHFV) serogroup comprises sole members CCHFV and Hazara virus (HAZV). CCHFV is an emerging zoonotic virus that causes often-fatal hemorrhagic fever in infected humans for which preventative or therapeutic strategies are not available. In contrast, HAZV is nonpathogenic to humans and thus represents an excellent model to study aspects of CCHFV biology under conditions of more-accessible biological containment. The three RNA segments that form the nairovirus genome are encapsidated by the viral nucleocapsid protein (N) to form ribonucleoprotein (RNP) complexes that are substrates for RNA synthesis and packaging into virus particles. We used quantitative proteomics to identify cellular interaction partners of CCHFV N and identified robust interactions with cellular chaperones. These interactions were validated using immunological methods, and the specific interaction between native CCHFV N and cellular chaperones of the HSP70 family was confirmed during live CCHFV infection. Using infectious HAZV, we showed for the first time that the nairovirus N-HSP70 association was maintained within both infected cells and virus particles, where N is assembled as RNPs. Reduction of active HSP70 levels in cells by the use of small-molecule inhibitors significantly reduced HAZV titers, and a model for chaperone function in the context of high genetic variability is proposed. These results suggest that chaperones of the HSP70 family are required for nairovirus replication and thus represent a genetically stable cellular therapeutic target for preventing nairovirus-mediated disease. IMPORTANCE: Nairoviruses compose a group of human and animal viruses that are transmitted by ticks and associated with serious or fatal disease. One member is Crimean-Congo hemorrhagic fever virus (CCHFV), which is responsible for fatal human disease and is recognized as an emerging threat within Europe in response to climate change. No preventative or therapeutic strategies against nairovirus-mediated disease are currently available. Here we show that the N protein of CCHFV and the related Hazara virus interact with a cellular protein, HSP70, during both the intracellular and extracellular stages of the virus life cycle. The use of inhibitors that block HSP70 function reduces virus titers by up to 1,000-fold, suggesting that this interaction is important within the context of the nairovirus life cycle and may represent a potent target for antinairovirus therapies against which the virus cannot easily develop resistance.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever Virus, Crimean-Congo/metabolism , Nairovirus/genetics , Nairovirus/metabolism , Nucleocapsid Proteins/metabolism , Virus Replication/genetics , A549 Cells , Cell Line , Cell Line, Tumor , Climate Change , Europe , HEK293 Cells , Hemorrhagic Fever, Crimean/metabolism , Hemorrhagic Fever, Crimean/virology , Humans , RNA/genetics
14.
J Vector Borne Dis ; 52(1): 36-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25815865

ABSTRACT

BACKGROUND & OBJECTIVES: Crimean-Congo hemorrhagic fever (CCHF) is a tick-borne viral disease, causing severe viral hemorrhagic fever outbreaks. This study aimed at determining the serum vitamin D levels and investigated the association between Crimean-Congo hemorrhagic fever (CCHF) and serum vitamin D levels in children with CCHF. METHODS: A total of 45 children aged between 5 and 15 yr, viz. 15 healthy control (HC) and 30 pediatric patients diagnosed with CCHF with real-time polymerase chain reaction (PCR) (patient group) were selected for the study. RESULTS: Analysis of the blood serum samples taken from the said individuals revealed that vitamin D, parathyroid hormone and calcium levels of the patients and the control groups were statistically different. INTERPRETATION & CONCLUSION: It was found that the serum vitamin D levels of the pediatric patients with CCHF were lower when compared to those of the controls, and that a low vitamin D level could negatively affect the reaction of the body to infections in children having CCHF.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo/isolation & purification , Hemorrhagic Fever, Crimean/metabolism , Vitamin D/blood , Adolescent , Calcium/blood , Child , Child, Preschool , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever, Crimean/diagnosis , Humans , Parathyroid Hormone/blood , Real-Time Polymerase Chain Reaction
15.
PLoS One ; 9(3): e91516, 2014.
Article in English | MEDLINE | ID: mdl-24621656

ABSTRACT

Crimean-Congo Haemorrhagic Fever (CCHF) is a severe tick-borne disease, endemic in many countries in Africa, the Middle East, Eastern Europe and Asia. Between 15-70% of reported cases are fatal. There is no approved vaccine available, and preclinical protection in vivo by an experimental vaccine has not been demonstrated previously. In the present study, the attenuated poxvirus vector, Modified Vaccinia virus Ankara, was used to develop a recombinant candidate vaccine expressing the CCHF virus glycoproteins. Cellular and humoral immunogenicity was confirmed in two mouse strains, including type I interferon receptor knockout mice, which are susceptible to CCHF disease. This vaccine protected all recipient animals from lethal disease in a challenge model adapted to represent infection via a tick bite. Histopathology and viral load analysis of protected animals confirmed that they had been exposed to challenge virus, even though they did not exhibit clinical signs. This is the first demonstration of efficacy of a CCHF vaccine.


Subject(s)
Hemorrhagic Fever, Crimean/prevention & control , Viral Vaccines/immunology , Animals , Cell Line , Cricetinae , DNA, Recombinant/genetics , Disease Models, Animal , Female , Glycoproteins/genetics , Glycoproteins/immunology , Hemorrhagic Fever Virus, Crimean-Congo/immunology , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Hemorrhagic Fever, Crimean/immunology , Hemorrhagic Fever, Crimean/metabolism , Hemorrhagic Fever, Crimean/pathology , Immunity, Cellular , Immunity, Humoral , Mice , Plasmids/genetics , Receptor, Interferon alpha-beta/deficiency , Receptors, Interferon/deficiency , Viral Load , Viral Proteins/genetics , Viral Proteins/immunology , Viral Vaccines/genetics
16.
Virus Res ; 179: 187-203, 2014 Jan 22.
Article in English | MEDLINE | ID: mdl-24184319

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne virus responsible for hemorrhagic manifestations and multiple organ failure, with a high mortality rate. In infected humans, damage to endothelial cells and vascular leakage may be a direct result of virus infection or an immune response-mediated indirect effect. The main target cells are mononuclear phagocytes, endothelial cells and hepatocytes; the liver being a key target for the virus, which was described as susceptible to interferon host response and to induce apoptosis. To better understand the early liver cell alterations due to virus infection, the protein profile of in vitro CCHFV-infected HepG2 cells was analyzed using two quantitative proteomic approaches, 2D-DIGE and iTRAQ. A set of 243 differentially expressed proteins was identified. Bioinformatics analysis (Ingenuity Pathways Analysis) revealed multiple host cell pathways and functions altered after CCHFV infection, with notably 106 proteins related to cell death, including 79 associated with apoptosis. Different protein networks emerged with associated pathways involved in inflammation, oxidative stress and apoptosis, ubiquitination/sumoylation, regulation of the nucleo-cytoplasmic transport, and virus entry. Collectively, this study revealed host liver protein abundances that were modified at the early stages of CCHFV infection, offering an unparalleled opportunity of the description of the potential pathogenesis processes and of possible targets for antiviral research.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo/physiology , Hemorrhagic Fever, Crimean/metabolism , Hemorrhagic Fever, Crimean/virology , Hepatocytes/virology , Animals , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever, Crimean/genetics , Hepatocytes/chemistry , Hepatocytes/metabolism , Humans , Molecular Sequence Data , Proteins/chemistry , Proteins/genetics , Proteins/metabolism , Proteomics
18.
J Virol ; 86(2): 773-85, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22072774

ABSTRACT

The innate immune response constitutes the first line of defense against viral infection and is extensively regulated through ubiquitination. The removal of ubiquitin from innate immunity signaling factors by deubiquitinating enzymes (DUBs) therefore provides a potential opportunity for viruses to evade this host defense system. It was previously found that specific proteases encoded by the unrelated arteri- and nairoviruses resemble the ovarian tumor domain-containing (OTU) family of DUBs. In arteriviruses, this domain has been characterized before as a papain-like protease (PLP2) that is also involved in replicase polyprotein processing. In nairoviruses, the DUB resides in the polymerase protein but is not essential for RNA replication. Using both in vitro and cell-based assays, we now show that PLP2 DUB activity is conserved in all members of the arterivirus family and that both arteri- and nairovirus DUBs inhibit RIG-I-mediated innate immune signaling when overexpressed. The potential relevance of RIG-I-like receptor (RLR) signaling for the innate immune response against arterivirus infection is supported by our finding that in mouse embryonic fibroblasts, the production of beta interferon primarily depends on the recognition of arterivirus RNA by the pattern-recognition receptor MDA5. Interestingly, we also found that both arteri- and nairovirus DUBs inhibit RIG-I ubiquitination upon overexpression, suggesting that both MDA5 and RIG-I have a role in countering infection by arteriviruses. Taken together, our results support the hypothesis that arteri- and nairoviruses employ their deubiquitinating potential to inactivate cellular proteins involved in RLR-mediated innate immune signaling, as exemplified by the deubiquitination of RIG-I.


Subject(s)
Arterivirus Infections/immunology , Arterivirus/enzymology , DEAD-box RNA Helicases/immunology , Endopeptidases/immunology , Hemorrhagic Fever, Crimean/immunology , Immunity, Innate , Nairovirus/enzymology , Viral Proteins/immunology , Animals , Arterivirus/chemistry , Arterivirus/genetics , Arterivirus Infections/enzymology , Arterivirus Infections/virology , Cell Line , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Endopeptidases/genetics , Endopeptidases/metabolism , Hemorrhagic Fever, Crimean/enzymology , Hemorrhagic Fever, Crimean/metabolism , Hemorrhagic Fever, Crimean/virology , Humans , Mice , Mice, Transgenic , Nairovirus/chemistry , Nairovirus/genetics , Protein Structure, Tertiary , Signal Transduction , Ubiquitin/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
19.
Int J Infect Dis ; 16(2): e89-93, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22154082

ABSTRACT

BACKGROUND: Crimean-Congo hemorrhagic fever (CCHF) is one of the viral hemorrhagic fevers caused by tick bites. Common symptoms of the infection are fatigue, high fever, headache, and myalgia. In some patients hemorrhage may accompany these symptoms and is a sign of a poor prognosis. Typical laboratory changes are thrombocytopenia, leukopenia, elevation of aspartate aminotransferase (AST), alanine aminotransferase (ALT), creatine phosphokinase (CPK), and lactate dehydrogenase (LDH), and prolongation of prothrombin time (PT) and activated partial thromboplastin time (aPTT). Mortality rates vary between 3% and 30%. The aim of this study was to determine the factors affecting the prognosis of CCHF. METHODS: A total of 70 patients with a diagnosis of CCHF who were followed at our clinic between 2005 and 2008 were included in this study. As well as patient clinical history, biochemical parameters tested during the first 5 days and the prognosis were evaluated. Findings were compared between patients who died and those who recovered. Non-parametric statistical tests were used for the statistical analysis. RESULTS: When the laboratory parameters of patients who died and recovered were compared, PT, aPTT, international normalized ratio (INR), AST, LDH, fibrinogen, C-reactive protein (CRP), high-sensitivity CRP (hs-CRP), D-dimer, IgM, IgG, C3 and C4 levels, and platelet count were found to be positively related with fatality. On the other hand, there was no significant difference between groups regarding ALT, CPK, prealbumin, ceruloplasmin, protein C, protein S, and antithrombin III levels, and white blood cell counts. CONCLUSIONS: It is essential to determine the possibility of a fatal prognosis in CCHF patients using clinical history and biochemical parameters so that the necessary precautions can be taken.


Subject(s)
Hemorrhagic Fever, Crimean/blood , Hemorrhagic Fever, Crimean/diagnosis , Adult , Alanine Transaminase/blood , Antithrombin III/metabolism , Aspartate Aminotransferases/blood , C-Reactive Protein/metabolism , Ceruloplasmin/metabolism , Complement C3/metabolism , Complement C4/metabolism , Female , Fibrin Fibrinogen Degradation Products/metabolism , Fibrinogen/metabolism , Hemorrhagic Fever, Crimean/metabolism , Humans , Immunoglobulin G/blood , Immunoglobulin M/blood , International Normalized Ratio/methods , L-Lactate Dehydrogenase/blood , Leukocyte Count/methods , Male , Middle Aged , Partial Thromboplastin Time , Platelet Count/methods , Prealbumin/metabolism , Prognosis , Protein C/metabolism , Protein S/metabolism , Prothrombin Time/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...