首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.

Background

Crimean-Congo hemorrhagic fever (CCHF) is a severe tick-borne disease well recognized through Europe and Asia where diagnostic tests and medical surveillance are available. However, it is largely neglected in Africa, especially in the tropical rainforest of Central Africa where only sporadic human cases have been reported and date back to more than 30 years. We describe here an isolated human case that occurred in the Democratic Republic of the Congo in 2008 and performed phylogenetic analysis to investigate whether it resulted from a regional re-emergence or from the introduction of a novel virus in the area.

Methods and Findings

Near complete segment S and partial segment M sequences were characterized. Bayesian phylogenetic analysis and datation were performed to investigate the relationship between this new strain and viral strains from Africa, Europe and Asia. The new strain is phylogenetically close to the previously described regional genotype (II) that appears to be specific to Central Africa. Phylogenetic discrepancy between segment S and M suggested genetic exchange among local sublineages, which was dated back to 130–590 years before present.

Conclusions

The phylogenetic analyses presented here suggest ongoing CCHF virus circulation in Central Africa for a long time despite the absence of reported human cases. Many infections have most probably been overlooked, due to the weakness of healthcare structures and the absence of available diagnostic procedure. However, despite the lack of accurate ecological data, the sporadic reporting of human cases could also be partly associated with a specific sylvatic cycle in Central Africa where deforestation may raise the risks of re-emergence. For these reasons, together with the high risk of nosocomial transmission, public health authorities'' attention should be drawn to this etiological agent.  相似文献   

2.
3.
Crimean-Congo hemorrhagic fever virus (CCHFV) is an important human pathogen. In cell culture, CCHFV is sensed by the cytoplasmic RNA sensor retinoic acid-inducible gene I (RIG-I) molecule and its adaptor molecule mitochondrial antiviral signaling (MAVS) protein. MAVS initiates both type I interferon (IFN-I) and proinflammatory responses. Here, we studied the role MAVS plays in CCHFV infection in mice in both the presence and absence of IFN-I activity. MAVS-deficient mice were not susceptible to CCHFV infection when IFN-I signaling was active and showed no signs of disease. When IFN-I signaling was blocked by antibody, MAVS-deficient mice lost significant weight, but were uniformly protected from lethal disease, whereas all control mice succumbed to infection. Cytokine activity in the infected MAVS-deficient mice was markedly blunted. Subsequent investigation revealed that CCHFV infected mice lacking TNF-α receptor signaling (TNFA-R-deficient), but not IL-6 or IL-1 activity, had more limited liver injury and were largely protected from lethal outcomes. Treatment of mice with an anti-TNF-α neutralizing antibody also conferred partial protection in a post-virus exposure setting. Additionally, we found that a disease causing, but non-lethal strain of CCHFV produced more blunted inflammatory cytokine responses compared to a lethal strain in mice. Our work reveals that MAVS activation and cytokine production both contribute to CCHFV pathogenesis, potentially identifying new therapeutic targets to treat this disease.  相似文献   

4.
Crimean-Congo hemorrhagic fever virus (CCHFV) is a highly pathogenic tick-borne virus with a fatality rate of up to 50% in humans. CCHFV is widely distributed in countries around the world. Outbreaks of CCHFV infection in humans have occurred in prior years in Xinjiang Province, China. Epidemiological surveys have detected CCHFV RNA in ticks and animals; however, few isolates were identified. In this study, we identified and isolated a new CCHFV strain from Hyalomma asiaticum asiaticum ticks collected from north of Tarim Basin in Xinjiang, China. A preliminary investigation of infection and antigens expression of CCHFV was performed in newborn mice. The target tissues for CCHFV replication in newborn mice were identified. The analysis of the phylogenetic relationships with other Chinese strains suggested that diverse genotypes of CCHFV have circulated in Xinjiang for years. These findings provide important insights into our understanding of CCHFV infection and evolution as well as disease prevention and control for local residents.
  相似文献   

5.
6.
Two isolates of a virus of the genus Orthobunyavirus (family Bunyaviridae) were obtained from hemorrhagic fever cases during a large disease outbreak in East Africa in 1997 and 1998. Sequence analysis of regions of the three genomic RNA segments of the virus (provisionally referred to as Garissa virus) suggested that it was a genetic reassortant virus with S and L segments derived from Bunyamwera virus but an M segment from an unidentified virus of the genus Orthobunyavirus. While high genetic diversity (52%) was revealed by analysis of virus M segment nucleotide sequences obtained from 21 members of the genus Orthobunyavirus, the Garissa and Ngari virus M segments were almost identical. Surprisingly, the Ngari virus L and S segments showed high sequence identity with those of Bunyamwera virus, showing that Garissa virus is an isolate of Ngari virus, which in turn is a Bunyamwera virus reassortant. Ngari virus should be considered when investigating hemorrhagic fever outbreaks throughout sub-Saharan Africa.  相似文献   

7.
The results of the isolation and identification of the causative agent of a haemorrhagic fever outbreak in the Stavropol Territory are presented. The virus isolated from blood of haemorrhagic fever patients by virological methods was identified in serological and molecular tests as Crimean haemorrhagic fever virus. This epidemiological analysis testify to increased activity of the natural focus of Crimean-Congo haemorrhagic fever in this area due to a number of natural and other factors leading to intensification of its epidemic realization.  相似文献   

8.
After the prolonged (about 30 years) absence of Crimean-Congo hemorrhagic fever (CCHF) morbidity in the Stavropol territory cases of this infection were registered, and received laboratory confirmation, in summer of 1999-2000. At the end of the 1999 season antibodies to CCHF virus were detected among cattle-breeders in all 7 inspected regions of the territory. According to the data of the determination of virus contamination of Ixodes ticks (the season of 2000), the circulation of CCHF on the territory of 14 regions out of 24 expected was established. An essential factor of the exacerbation of the epidemic situation was a rise in the number of Hyalomma marginatum ticks, the main vector of the causative agent of CCHF in the south of Russia.  相似文献   

9.
Crimean-Congo hemorrhagic fever virus (CCHFV) is an etiological agent of a disease with mortality rates in patients averaging 30%. The disease is characterized by fever, myalgia, and hemorrhage. Mechanisms underlying the hemorrhage have to our knowledge not been elucidated for CCHFV. Possibly, a direct or indirect viral effect on tight junctions (TJ) could cause the hemorrhage observed in patients, as TJ play a crucial role in vascular homeostasis and can cause leakage upon deregulation. Moreover, there is no knowledge regarding the site of entry and release of CCHFV in polarized epithelial cells. Such cells represent a barrier to virus dissemination within the host, and as a site of viral entry and release, they could play a key role in further spread. For the first time, we have shown preferential basolateral entry of CCHFV in Madin-Darby canine kidney 1 (MDCK-1) epithelial cells. Furthermore, we demonstrated basolateral release of CCHFV in polarized epithelial cells. Interestingly, by measuring transepithelial electrical resistance, we found no effect of CCHFV replication on the function of TJ in this study. Neither did we observe any difference in the localization of the TJ proteins ZO-1 and occludin in CCHFV-infected cells compared to mock-infected cells.  相似文献   

10.

Background

In January 2011, human cases with hemorrhagic manifestations in the hospital staff were reported from a tertiary care hospital in Ahmadabad, India. This paper reports a detailed epidemiological investigation of nosocomial outbreak from the affected area of Ahmadabad, Gujarat, India.

Principal Findings

Samples from 3 suspected cases, 83 contacts, Hyalomma ticks and livestock were screened for Crimean-Congo hemorrhagic fever (CCHF) virus by qRT-PCR of which samples of two medical professionals (case C and E) and the husband of the index case (case D) were positive for CCHFV. The sensitivity and specificity of indigenous developed IgM ELISA to screen CCHFV specific antibodies in human serum was 75.0% and 97.5% respectively as compared to commercial kit. About 17.0% domestic animals from Kolat, Ahmadabad were positive for IgG antibodies while only two cattle and a goat showed positivity by qRT-PCR. Surprisingly, 43.0% domestic animals (Buffalo, cattle, sheep and goat) showed IgG antibodies in the adjoining village Jivanpara but only one of the buffalo was positive for CCHFV. The Hyalomma anatolicum anatolicum ticks were positive in PCR and virus isolation. CCHFV was isolated from the blood sample of case C, E in Vero E-6 cells and Swiss albino mice. In partial nucleocapsid gene phylogeny from CCHFV positive human samples of the years 2010 and 2011, livestock and ticks showed this virus was similar to Tajikistan (strain TAJ/H08966), which belongs in the Asian/middle east genetic lineage IV.

Conclusions

The likely source of CCHFV was identified as virus infected Hyalomma ticks and livestock at the rural village residence of the primary case (case A). In addition, retrospective sample analysis revealed the existence of CCHFV in Gujarat and Rajasthan states before this outbreak. An indigenous developed IgM ELISA kit will be of great use for screening this virus in India.  相似文献   

11.
12.
13.
Jogaejeot, seasoned Venerupis philippinarum, is a traditional Korean fermented food, and hepatitis A virus (HAV) can be transmitted through contaminated food, especially bivalve shellfish, causing acute gastroenteritis worldwide. Here, we carried out a phylogenetic analysis to identify and characterize HAV strains in jogaejeot samples associated with hepatitis A (HA) outbreaks in Seoul, South Korea, in 2019. The HAV strains were identified using blast and molecular analysis of the amplified HAV VP1-P2B genome region. The HAV strains identified in the five jogaejeot samples shared at least 99% sequence identity, were all classified as genotype IA and were most closely related to strains that are widespread in East Asia. These results support a link between the consumption of jogaejeot and the HA outbreaks observed in 2019 in Seoul. In addition, they indicate a need for more stringent enforcement of food safety regulations for the shellfish industry, especially against HAV, and the value of widespread vaccination.  相似文献   

14.
Crimean-Congo hemorrhagic fever (CCHF) is a tick-borne zoonosis with a high case fatality rate in humans. Although the disease is widely found in Africa, Europe, and Asia, the distribution and genetic diversity of CCHF virus (CCHFV) are poorly understood in African countries. To assess the risks of CCHF in Zambia, where CCHF has never been reported, epidemiologic studies in cattle and ticks were conducted. Through an indirect immunofluorescence assay, CCHFV nucleoprotein-specific serum IgG was detected in 8.4% (88/1,047) of cattle. Among 290 Hyalomma ticks, the principal vector of CCHFV, the viral genome was detected in 11 ticks. Phylogenetic analyses of the CCHFV S and M genome segments revealed that one of the detected viruses was a genetic reassortant between African and Asian strains. This study provides compelling evidence for the presence of CCHFV in Zambia and its transmission to vertebrate hosts.  相似文献   

15.
16.
17.
18.
Crimean-Congo hemorrhagic fever (CCHF) is an increasing health concern in Turkey since 2002. There were also some recent human cases from the South Marmara region of Turkey; thus, a tick survey was performed, and possible vector tick species for the CCHF virus were determined in the region. A total of 740 adult ticks were collected from infested livestock from five locations: Çanakkale-Biga, Bursa-Orhaneli, Bursa-Keles, Bal?kesir and Bilecik. Total of 11 tick species from the genera Hyalomma, Rhipicephalus, Dermacentor, Ixodes and Haemaphysalis were identified. Rhipicephalus ticks were dominant in the region; the most frequently observed tick species was R. turanicus, (53.1 %), and only 15.4 % of the identified ticks were H. marginatum. The occurrence of H. rufipes infestation in the region fort he first time. A total of 73 pools of adult ticks were tested with both an antigen-detecting ELISA and RT real-time PCR (RT rt PCR). The presence of the CCHF virus was demonstrated in 9 (12.3 %) of the tested tick pools. Although seven of the tick pools were positive for the CCHF virus with both of the methods, one pool was positive only with RT rt PCR and the other pool was only positive with the ELISA. Positive results were obtained from ticks collected from cattle, sheep and goats from two locations, Bursa-Orhaneli and Bilecik. The CCHF virus was detected in R. turanicus (n = 3), R. bursa (n = 2), H. marginatum (n = 2) and D. marginatus (n = 2) ticks. The results of this study confirm the presence of the CCHF virus and present preliminary data on the vector tick species in the southern Marmara region of Turkey.  相似文献   

19.
Crimean-Congo hemorrhagic fever (CCHF) virus is a tick-borne, negative-sense, single-stranded RNA [ssRNA(−)] nairovirus that produces fever, prostration, and severe hemorrhages in humans. With fatality rates for CCHF ranging up to 70% based on several factors, CCHF is considered a dangerous emerging disease. Originally identified in the former Soviet Union and the Congo, CCHF has rapidly spread across large sections of Europe, Asia, and Africa. Recent reports have identified a viral homologue of the ovarian tumor protease superfamily (vOTU) within its L protein. This protease has subsequently been implicated in downregulation of the type I interferon immune response through cleavage of posttranslational modifying proteins ubiquitin (Ub) and the Ub-like interferon-simulated gene 15 (ISG15). Additionally, homologues of vOTU have been suggested to perform similar roles in the positive-sense, single-stranded RNA [ssRNA(+)] arteriviruses. By utilizing X-ray crystallographic techniques, the structure of vOTU covalently bound to ubiquitin propylamine, a suicide substrate of the enzyme, was elucidated to 1.7 Å, revealing unique structural elements that define this new subclass of the OTU superfamily. In addition, kinetic studies were carried out with aminomethylcoumarin (AMC) conjugates of monomeric Ub, ISG15, and NEDD8 (neural precursor cell expressed, developmentally downregulated 8) substrates in order to provide quantitative insights into vOTU''s preference for Ub and Ub-like substrates.Crimean-Congo hemorrhagic fever (CCHF) is characterized in humans by the sudden onset of fever, myalgia, headache, dizziness, sore eyes, photophobia, and hyperanemia as well as severe hemorrhages (28, 43, 46). The causative agent of CCHF is the CCHF virus, which is a tick-borne, negative-sense, single-stranded RNA [ssRNA(−)] virus of the genus Nairovirus, belonging to the viral family Bunyaviridae. Originally named after outbreaks in the former Soviet Union and in the Congo during the mid-20th century, the affected area of this disease has rapidly spread to large areas of sub-Saharan Africa, the Balkans, Northern Greece, European Russia, Pakistan, the Arabian Peninsula, Iran, Afghanistan, Iraq, Turkey, and recently, the Xinjiang province of China (43, 46). The CCHF viral genome, as well as those of the closely related Dugbe and Nairobi viruses, consists of three negative-sense RNA segments, small (S), medium (M), and large (L). Incubation of CCHF is 5 to 6 days, with fatalities occurring less than 7 days after signs of infection. Fatality rates for patients infected with the CCHF virus ranged from 5% to 70%, depending on phylogenetic variation of the virus, transmission route, treatment facility, and the reporting and confirmation of the case statistics (19, 32, 43, 47).The innate immune system serves as the human''s first line of defense from invading pathogens, including CCHF virus. The type I interferon (IFN) response comprises a key component of this system by upregulating more than 300 IFN-stimulated genes (ISGs) whose products detect viral molecules, promote amplification of the type I IFN response, modulate other signaling pathways, and directly provide antiviral activity (34). Regulation of the type I IFN response has been shown to rely on posttranslational modification by ubiquitin (Ub) and the Ub-like interferon-simulated gene 15 (ISG15) (14, 23). Both Ub and ISG15 are expressed in a proform and cleaved to leave a double-glycine C terminus that forms an isopeptide bond with predominantly the ɛ-NH2 of lysine residues of a target protein through a three-step enzymatic process. In addition to forming isopeptide bonds with target proteins, Ub, which contains seven lysine residues, has been observed to form poly-Ub chains. The most studied of these moieties are K29-linked, K48-linked, and K63-linked poly-Ub. While K29-linked and K48-linked polyubiquitination of proteins leads to their degradation in the lysosome and proteasome, respectively, conjugation of K63-linked poly-Ub to proteins has an activating effect, resulting in an enhanced type I IFN response (2, 7, 18, 33, 40). Currently, more than 150 proteins have been identified as forming conjugates with ISG15, with the number of proteins forming Ub conjugates far exceeding that number (12, 48). A subset of type I IFN signaling and effector proteins that Ub and ISG15 have been shown to stabilize includes JAK1, STAT1/2, double-stranded RNA-dependent protein kinase (PKR), myxovirus-resistant protein A (MxA), and RIG-I (17). MxA has particularly shown to be important in type I IFN response to CCHF infection. RIG-I and several other proteins have also been shown to be targets for K63-linked poly-Ub (4).Recently, investigators have identified a cysteine viral ovarian tumor domain (vOTU) protease colocated with the RNA-dependent RNA polymerase in the L protein of the CCHF virus (14). Interestingly, as CCHF is an ssRNA(−) virus, no protease is required to cleave a viral polypeptide to facilitate viral replication as in positive-sense ssRNA [ssRNA(+)] viruses. Furthermore, recent reports have observed that vOTU is not required for RNA-dependent RNA polymerase activity and for vOTU protease activity linked to impairment of the type I IFN response through its deubiquitinating and deISGylating activity (6, 14). Additional studies have also tentatively identified the presence of vOTU homologues in the Arterivirus genus of the Arteriviridae family, suggesting that they too may facilitate impairment of the type I IFN response (14). Since the discovery of the first ovarian tumor domain (OTU) protease in Drosophila oogenesis and prior to the identification of vOTU, OTU superfamily members could be divided into three subclasses according to their sequence homology, otubains, A20-like OTUs, and ubiquitin thioesterase ZRANB1 (22). With the addition of the viral OTU subclass, OTU superfamily members in more than 100 eukaryotic, bacterial, and viral proteins have now been identified (6, 27). Predominantly, OTU proteases have been linked to ubiquitin (Ub) removal and/or remodeling of Ub-conjugated proteins, placing them among five protease superfamilies that facilitate signal transduction cascades and play key roles in protein stability (22). However, vOTU is unique in that it is the only OTU to have shown both deubiquitinating and deISGylating activity (14). Instead, Otubain1/2 (OTUB1/2) plays a key role in T cell response and prefers K48-linked poly-Ub or NEDD8 (neural precursor cell expressed, developmentally downregulated 8) as a substrate (12). A20 and A20-like Cezanne OTU proteases are negative regulators of the NF-κB-mediated inflammation response, selectively cleaving K63-linked poly-Ub targets. DUBA also shows preference for K63-linked poly-Ub (20). In attempts to better understand the OTU superfamily, structures of OTUB and A20-like OTU domains have been elucidated (12, 21, 30). An X-ray structure of the yeast ovarian tumor 1 (yOTU1) domain, which interacts with Cdc48 and has a preference for K48-linked poly-Ub, was achieved in complex with mono-Ub (27). However, since yOTU1 has a preference for K48-linked Ub and possesses low sequence identity to vOTU and other OTU domain proteases, only limited information on vOTU could be obtained. In addition to vOTU, several other viral proteases, such as papain-like protease (PLpro) from the severe acute respiratory syndrome (SARS) coronavirus, have also shown deubiquitinating and deISGylating activity to evade the innate immune system (6, 8, 43, 49). However, no viral proteases that are known to possess deISGylating activity have been visualized as being bound to Ub or Ub-like substrates. To address this issue and elucidate the atomic-level structure of a member from the viral OTU superfamily subclass, we have obtained the X-ray crystal structure of vOTU bound with Ub (vOTU-Ub). We also have characterized the vOTU substrate specificity for mono-Ub, ISG15, and NEDD8 and compared the results with those from human OTUB2 (hOTUB2). Additionally, we assessed vOTU''s deubiquitinating activity toward K48- and K63-linked poly-Ub.  相似文献   

20.
Recombinant vesicular stomatitis virus (VSV) vectors expressing homologous filoviral glycoproteins can completely protect rhesus monkeys against Marburg virus when administered after exposure and can partially protect macaques after challenge with Zaire ebolavirus. Here, we administered a VSV vector expressing the Sudan ebolavirus (SEBOV) glycoprotein to four rhesus macaques shortly after exposure to SEBOV. All four animals survived SEBOV challenge, while a control animal that received a nonspecific vector developed fulminant SEBOV hemorrhagic fever and succumbed. This is the first demonstration of complete postexposure protection against an Ebola virus in nonhuman primates and provides further evidence that postexposure vaccination may have utility in treating exposures to filoviruses.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号