Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
Add more filters

Publication year range
1.
Cell ; 164(3): 392-405, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26806128

ABSTRACT

Recent studies have suggested that antibody-mediated protection against the Ebolaviruses may be achievable, but little is known about whether or not antibodies can confer cross-reactive protection against viruses belonging to diverse Ebolavirus species, such as Ebola virus (EBOV), Sudan virus (SUDV), and Bundibugyo virus (BDBV). We isolated a large panel of human monoclonal antibodies (mAbs) against BDBV glycoprotein (GP) using peripheral blood B cells from survivors of the 2007 BDBV outbreak in Uganda. We determined that a large proportion of mAbs with potent neutralizing activity against BDBV bind to the glycan cap and recognize diverse epitopes within this major antigenic site. We identified several glycan cap-specific mAbs that neutralized multiple ebolaviruses, including SUDV, and a cross-reactive mAb that completely protected guinea pigs from the lethal challenge with heterologous EBOV. Our results provide a roadmap to develop a single antibody-based treatment effective against multiple Ebolavirus infections.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/immunology , Survivors , Animals , Cross Reactions , Disease Models, Animal , Epitope Mapping , Guinea Pigs , Humans , Mice , Mice, Inbred BALB C , Microscopy, Electron , Models, Molecular , Mutagenesis , Uganda
2.
Cell ; 160(5): 893-903, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25723164

ABSTRACT

The mechanisms by which neutralizing antibodies inhibit Marburg virus (MARV) are not known. We isolated a panel of neutralizing antibodies from a human MARV survivor that bind to MARV glycoprotein (GP) and compete for binding to a single major antigenic site. Remarkably, several of the antibodies also bind to Ebola virus (EBOV) GP. Single-particle EM structures of antibody-GP complexes reveal that all of the neutralizing antibodies bind to MARV GP at or near the predicted region of the receptor-binding site. The presence of the glycan cap or mucin-like domain blocks binding of neutralizing antibodies to EBOV GP, but not to MARV GP. The data suggest that MARV-neutralizing antibodies inhibit virus by binding to infectious virions at the exposed MARV receptor-binding site, revealing a mechanism of filovirus inhibition.


Subject(s)
Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Antigen-Antibody Complex/ultrastructure , Marburg Virus Disease/immunology , Marburgvirus/chemistry , Viral Envelope Proteins/chemistry , Adult , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/metabolism , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Antibodies, Viral/metabolism , B-Lymphocytes/immunology , Female , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Marburgvirus/genetics , Marburgvirus/immunology , Models, Molecular , Mutation , Protein Structure, Tertiary , Viral Envelope Proteins/metabolism
3.
PLoS Pathog ; 20(6): e1011777, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38913740

ABSTRACT

COVID-associated coagulopathy seemly plays a key role in post-acute sequelae of SARS- CoV-2 infection. However, the underlying pathophysiological mechanisms are poorly understood, largely due to the lack of suitable animal models that recapitulate key clinical and pathological symptoms. Here, we fully characterized AC70 line of human ACE2 transgenic (AC70 hACE2 Tg) mice for SARS-CoV-2 infection. We noted that this model is highly permissive to SARS-CoV-2 with values of 50% lethal dose and infectious dose as ~ 3 and ~ 0.5 TCID50 of SARS-CoV-2, respectively. Mice infected with 105 TCID50 of SARS-CoV-2 rapidly succumbed to infection with 100% mortality within 5 days. Lung and brain were the prime tissues harboring high viral titers, accompanied by histopathology. However, viral RNA and inflammatory mediators could be detectable in other organs, suggesting the nature of a systemic infection. Lethal challenge of AC70 hACE2 Tg mice caused acute onset of leukopenia, lymphopenia, along with an increased neutrophil-to-lymphocyte ratio (NLR). Importantly, infected animals recapitulated key features of COVID-19-associated coagulopathy. SARS-CoV-2 could induce the release of circulating neutrophil extracellular traps (NETs), along with activated platelet/endothelium marker. Immunohistochemical staining with anti-platelet factor-4 (PF4) antibody revealed profound platelet aggregates especially within blocked veins of the lungs. We showed that acute SARS-CoV-2 infection triggered a hypercoagulable state coexisting with ill-regulated fibrinolysis. Finally, we highlighted the potential role of Annexin A2 (ANXA2) in fibrinolytic failure. ANXA2 is a calcium-dependent phospholipid-binding protein that forms a heterotertrameric complexes localized at the extracellular membranes with two S100A10 small molecules acting as a co-receptor for tissue-plasminogen activator (t-PA), tightly involved in cell surface fibrinolysis. Thus, our results revealing elevated IgG type anti-ANXA2 antibody production, downregulated de novo ANXA2/S100A10 synthesis, and reduced ANXA2/S100A10 association in infected mice, this protein might serve as druggable targets for development of antithrombotic and/or anti-fibrinolytic agents to attenuate pathogenesis of COVID-19.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Disease Models, Animal , Mice, Transgenic , SARS-CoV-2 , Animals , COVID-19/pathology , COVID-19/complications , COVID-19/virology , COVID-19/metabolism , Mice , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Humans , Blood Coagulation Disorders/virology , Blood Coagulation Disorders/pathology , Pneumonia, Viral/virology , Pneumonia, Viral/pathology , Pneumonia, Viral/metabolism , Betacoronavirus , Lung/virology , Lung/pathology , Lung/metabolism , Coronavirus Infections/virology , Coronavirus Infections/pathology , Coronavirus Infections/complications , Pandemics , Extracellular Traps/metabolism
4.
Clin Infect Dis ; 74(10): 1821-1830, 2022 05 30.
Article in English | MEDLINE | ID: mdl-34463715

ABSTRACT

BACKGROUND: Lassa fever is a zoonotic, acute viral illness first identified in Nigeria in 1969. An estimate shows that the "at risk" seronegative population (in Sierra Leone, Guinea, and Nigeria) may be as high as 59 million, with an annual incidence of all illnesses of 3 million, and fatalities up to 67 000, demonstrating the serious impact of the disease on the region and global health. METHODS: Histopathologic evaluation, immunohistochemical assay, and electron microscopic examination were performed on postmortem tissue samples from 12 confirmed Lassa fever cases. RESULTS: Lassa fever virus antigens and viral particles were observed in multiple organ systems and cells, including cells in the mononuclear phagocytic system and other specialized cells where it had not been described previously. CONCLUSIONS: The immunolocalization of Lassa fever virus antigens in fatal cases provides novel insightful information with clinical and pathogenetic implications. The extensive involvement of the mononuclear phagocytic system, including tissue macrophages and endothelial cells, suggests participation of inflammatory mediators from this lineage with the resulting vascular dilatation and increasing permeability. Other findings indicate the pathogenesis of Lassa fever is multifactorial and additional studies are needed.


Subject(s)
Lassa Fever , Virus Diseases , Endothelial Cells , Humans , Incidence , Lassa Fever/epidemiology , Lassa virus
5.
Emerg Infect Dis ; 26(12): 3030-3033, 2020 12.
Article in English | MEDLINE | ID: mdl-33219787

ABSTRACT

We report a unique outbreak of Rift Valley fever in the Eldamar area, Sudan, May-July 2019, that resulted in 1,129 case-patients and 19 (1.7%) deaths. Patients exhibited clinical signs including fever (100%), headache (79%), and bleeding (4%). Most (98%) patients also reported death and abortions among their livestock.


Subject(s)
Abortion, Spontaneous , Rift Valley Fever , Rift Valley fever virus , Animals , Disease Outbreaks , Female , Humans , Livestock , Pregnancy , Rift Valley Fever/diagnosis , Rift Valley Fever/epidemiology , Rift Valley fever virus/genetics , Sudan/epidemiology
6.
Emerg Infect Dis ; 26(6): 1266-1273, 2020 06.
Article in English | MEDLINE | ID: mdl-32160149

ABSTRACT

The etiologic agent of an outbreak of pneumonia in Wuhan, China, was identified as severe acute respiratory syndrome coronavirus 2 in January 2020. A patient in the United States was given a diagnosis of infection with this virus by the state of Washington and the US Centers for Disease Control and Prevention on January 20, 2020. We isolated virus from nasopharyngeal and oropharyngeal specimens from this patient and characterized the viral sequence, replication properties, and cell culture tropism. We found that the virus replicates to high titer in Vero-CCL81 cells and Vero E6 cells in the absence of trypsin. We also deposited the virus into 2 virus repositories, making it broadly available to the public health and research communities. We hope that open access to this reagent will expedite development of medical countermeasures.


Subject(s)
Betacoronavirus/isolation & purification , Coronavirus Infections/diagnosis , Pneumonia, Viral/diagnosis , Animals , Betacoronavirus/genetics , Betacoronavirus/physiology , COVID-19 , Cell Line , Chlorocebus aethiops , Genome, Viral , Humans , Nasopharynx/virology , Oropharynx/virology , Pandemics , SARS-CoV-2 , Vero Cells , Viral Tropism , Virus Replication , Washington
7.
J Infect Dis ; 218(suppl_5): S418-S422, 2018 11 22.
Article in English | MEDLINE | ID: mdl-30060231

ABSTRACT

Screening of monoclonal antibodies against ebolaviruses requires small-animal models. Wild-type mice require adaptation of ebolaviruses, whereas immunodeficient mice are still resistant to nonadapted Bundibugyo ebolavirus. Swapping of Ebola virus glycoprotein with that from Bundibugyo virus resulted in a replication-competent chimeric virus, which caused 100% lethal infection in STAT1 knockout mice. Monoclonal antibody BDBV223 isolated from a human survivor of Bundibugyo virus infection protected mice from challenge with the chimeric virus. These data demonstrate the suitability of the approach for in vivo screening of antibodies and suggest the greater contribution of internal Ebola proteins in pathogenesis compared to Bundibugyo virus proteins.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Ebolavirus/immunology , Animals , Antibodies, Neutralizing/immunology , Chlorocebus aethiops , Disease Models, Animal , Hemorrhagic Fever, Ebola/immunology , Mice , Mice, Knockout , Vero Cells , Viral Envelope Proteins/immunology
8.
J Virol ; 91(8)2017 04 15.
Article in English | MEDLINE | ID: mdl-28122983

ABSTRACT

Marburg (MARV) and Ebola (EBOV) viruses are zoonotic pathogens that cause severe hemorrhagic fever in humans. The natural reservoir of MARV is the Egyptian rousette bat (Rousettus aegyptiacus); that of EBOV is unknown but believed to be another bat species. The Egyptian rousette develops subclinical productive infection with MARV but is refractory to EBOV. Interaction of filoviruses with hosts is greatly affected by the viral interferon (IFN)-inhibiting domains (IID). Our study was aimed at characterization of innate immune responses to filoviruses and the role of filovirus IID in bat and human cells. The study demonstrated that EBOV and MARV replicate to similar levels in all tested cell lines, indicating that permissiveness for EBOV at cell and organism levels do not necessarily correlate. Filoviruses, particularly MARV, induced a potent innate immune response in rousette cells, which was generally stronger than that in human cells. Both EBOV VP35 and VP24 IID were found to suppress the innate immune response in rousette cells, but only VP35 IID appeared to promote virus replication. Along with IFN-α and IFN-ß, IFN-γ was demonstrated to control filovirus infection in bat cells but not in human cells, suggesting host species specificity of the antiviral effect. The antiviral effects of bat IFNs appeared not to correlate with induction of IFN-stimulated genes 54 and 56, which were detected in human cells ectopically expressing bat IFN-α and IFN-ß. As bat IFN-γ induced the type I IFN pathway, its antiviral effect is likely to be partially induced via cross talk.IMPORTANCE Bats serve as reservoirs for multiple emerging viruses, including filoviruses, henipaviruses, lyssaviruses, and zoonotic coronaviruses. Although there is no evidence for symptomatic disease caused by either Marburg or Ebola viruses in bats, spillover of these viruses into human populations causes deadly outbreaks. The reason for the lack of symptomatic disease in bats infected with filoviruses remains unknown. The outcome of a virus-host interaction depends on the ability of the host immune system to suppress viral replication and the ability of a virus to counteract the host defenses. Our study is a comparative analysis of the host innate immune response to either MARV or EBOV infection in bat and human cells and the role of viral interferon-inhibiting domains in the host innate immune responses. The data are useful for understanding the interactions of filoviruses with natural and accidental hosts and for identification of factors that influence filovirus evolution.


Subject(s)
Ebolavirus/immunology , Immunity, Innate , Marburgvirus/immunology , Animals , Cell Line , Chiroptera , Ebolavirus/physiology , Humans , Immune Tolerance , Interferons/analysis , Marburgvirus/physiology , Protein Domains , Viral Proteins/immunology , Virus Replication
10.
J Virol ; 90(8): 3890-3901, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26819310

ABSTRACT

UNLABELLED: Recent experiments suggest that some glycoprotein (GP)-specific monoclonal antibodies (MAbs) can protect experimental animals against the filovirus Ebola virus (EBOV). There is a need for isolation of MAbs capable of neutralizing multiple filoviruses. Antibody neutralization assays for filoviruses frequently use surrogate systems such as the rhabdovirus vesicular stomatitis Indiana virus (VSV), lentiviruses or gammaretroviruses with their envelope proteins replaced with EBOV GP or pseudotyped with EBOV GP. It is optimal for both screening and in-depth characterization of newly identified neutralizing MAbs to generate recombinant filoviruses that express a reporter fluorescent protein in order to more easily monitor and quantify the infection. Our study showed that unlike neutralization-sensitive chimeric VSV, authentic filoviruses are highly resistant to neutralization by MAbs. We used reverse genetics techniques to replace EBOV GP with its counterpart from the heterologous filoviruses Bundibugyo virus (BDBV), Sudan virus, and even Marburg virus and Lloviu virus, which belong to the heterologous genera in the filovirus family. This work resulted in generation of multiple chimeric filoviruses, demonstrating the ability of filoviruses to tolerate swapping of the envelope protein. The sensitivity of chimeric filoviruses to neutralizing MAbs was similar to that of authentic biologically derived filoviruses with the same GP. Moreover, disabling the expression of the secreted GP (sGP) resulted in an increased susceptibility of an engineered virus to the BDBV52 MAb isolated from a BDBV survivor, suggesting a role for sGP in evasion of antibody neutralization in the context of a human filovirus infection. IMPORTANCE: The study demonstrated that chimeric rhabdoviruses in which G protein is replaced with filovirus GP, widely used as surrogate targets for characterization of filovirus neutralizing antibodies, do not accurately predict the ability of antibodies to neutralize authentic filoviruses, which appeared to be resistant to neutralization. However, a recombinant EBOV expressing a fluorescent protein tolerated swapping of GP with counterparts from heterologous filoviruses, allowing high-throughput screening of B cell lines to isolate MAbs of any filovirus specificity. Human MAb BDBV52, which was isolated from a survivor of BDBV infection, was capable of partially neutralizing a chimeric EBOV carrying BDBV GP in which expression of sGP was disabled. In contrast, the parental virus expressing sGP was resistant to the MAb. Thus, the ability of filoviruses to tolerate swapping of GP can be used for identification of neutralizing MAbs specific to any filovirus and for the characterization of MAb specificity and mechanism of action.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Filoviridae/immunology , Animals , Antibodies, Neutralizing/immunology , Chlorocebus aethiops , Filoviridae/genetics , Humans , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Reverse Genetics , Vero Cells , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
11.
Proc Natl Acad Sci U S A ; 110(48): 19615-20, 2013 Nov 26.
Article in English | MEDLINE | ID: mdl-24218580

ABSTRACT

Rickettsiae are responsible for some of the most devastating human infections. A high infectivity and severe illness after inhalation make some rickettsiae bioterrorism threats. We report that deletion of the exchange protein directly activated by cAMP (Epac) gene, Epac1, in mice protects them from an ordinarily lethal dose of rickettsiae. Inhibition of Epac1 suppresses bacterial adhesion and invasion. Most importantly, pharmacological inhibition of Epac1 in vivo using an Epac-specific small-molecule inhibitor, ESI-09, completely recapitulates the Epac1 knockout phenotype. ESI-09 treatment dramatically decreases the morbidity and mortality associated with fatal spotted fever rickettsiosis. Our results demonstrate that Epac1-mediated signaling represents a mechanism for host-pathogen interactions and that Epac1 is a potential target for the prevention and treatment of fatal rickettsioses.


Subject(s)
Bacterial Adhesion/drug effects , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Host-Pathogen Interactions/physiology , Hydrazones/pharmacology , Isoxazoles/pharmacology , Rickettsia Infections/drug therapy , Signal Transduction/physiology , Animals , Bacterial Adhesion/physiology , Guanine Nucleotide Exchange Factors/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Hydrazones/therapeutic use , Immunohistochemistry , Isoxazoles/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rickettsia Infections/metabolism
12.
Clin Infect Dis ; 61(11): 1648-54, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26179011

ABSTRACT

BACKGROUND: Sierra Leone has the most cases of Ebola virus disease (EVD) ever reported. Trends in laboratory-confirmed EVD, symptom presentation, and risk factors have not been fully described. METHODS: EVD cases occurring from 23 May 2014 to 31 January 2015 are presented by geography, demographics, and risk factors for all persons who had laboratory-confirmed EVD, which was identified by Ebola virus-specific reverse-transcription polymerase chain reaction-based testing. RESULTS: During the study period, 8056 persons had laboratory-confirmed EVD. Their median age was 28 years; 51.7% were female. Common symptoms included fever (90.4%), fatigue (88.3%), loss of appetite (87.0%), headache (77.9%), joint pain (73.7%), vomiting (71.2%), and diarrhea (70.6%). Among persons with confirmed cases, 47.9% reported having had contact with someone with suspected EVD or any sick person, and 25.5% reported having attended a funeral, of whom 66.2% reported touching the body. The incidence of EVD was highest during 1-30 November 2014, at 7.5 per 100 000 population per week, and decreased to 2.1 per week during 1-31 January 2015. Between 23 May and 30 August 2014, two districts had the highest incidence of 3.8 and 7.0 per 100 000 population per week which decreased >97% by 1-31 January 2015. In comparison, the districts that include the capital city reported a 10-fold increase in incidence per week during the same time periods. CONCLUSIONS: Almost half of patients with EVD in Sierra Leone reported physical contact with a person ill with EVD or a dead body, highlighting prevention opportunities.


Subject(s)
Disease Outbreaks/prevention & control , Epidemiological Monitoring , Hemorrhagic Fever, Ebola/epidemiology , Adolescent , Adult , Child , Diarrhea/epidemiology , Epidemics , Female , Fever , Hemorrhagic Fever, Ebola/diagnosis , Hemorrhagic Fever, Ebola/prevention & control , Hemorrhagic Fever, Ebola/transmission , Humans , Incidence , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors , Sierra Leone/epidemiology , Time Factors , Young Adult
13.
J Virol ; 88(7): 3902-10, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24453361

ABSTRACT

The outbreak of Middle East respiratory syndrome coronavirus (MERS-CoV) infections and diseases represents a potential threat for worldwide spread and requires development of effective therapeutic strategies. In this study, we revealed a novel positive function of an exchange protein directly activated by cyclic AMP 1 (cAMP-1; Epac-1) on MERS-CoV replication. Specifically, we have shown that Epac-specific inhibitor treatment or silencing Epac-1 gene expression rendered cells resistant to viral infection. We believe Epac-1 inhibitors deserve further study as potential therapeutic agents for MERS-CoV infection.


Subject(s)
Coronavirus/drug effects , Coronavirus/physiology , Cyclic AMP/metabolism , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Host-Pathogen Interactions , Virus Replication/drug effects , Animals , Cell Line , Chlorocebus aethiops , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/genetics , Humans
14.
J Virol ; 87(17): 9953-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23824802

ABSTRACT

The newly emerged Middle East respiratory syndrome coronavirus (MERS-CoV) infects human bronchial epithelial Calu-3 cells. Unlike severe acute respiratory syndrome (SARS)-CoV, which exclusively infects and releases through the apical route, this virus can do so through either side of polarized Calu-3 cells. Infection results in profound apoptosis within 24 h irrespective of its production of titers that are lower than those of SARS-CoV. Together, our results provide new insights into the dissemination and pathogenesis of MERS-CoV and may indicate that the virus differs markedly from SARS-CoV.


Subject(s)
Bronchi/virology , Coronavirus/physiology , Coronavirus/pathogenicity , Apoptosis , Bronchi/pathology , Cell Line , Cell Polarity , Cytopathogenic Effect, Viral/physiology , Epithelial Cells/pathology , Epithelial Cells/virology , Humans , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Severe acute respiratory syndrome-related coronavirus/physiology , Species Specificity , Virus Internalization , Virus Release/physiology
15.
PLoS Pathog ; 8(10): e1002877, 2012.
Article in English | MEDLINE | ID: mdl-23055920

ABSTRACT

Marburg virus (family Filoviridae) causes sporadic outbreaks of severe hemorrhagic disease in sub-Saharan Africa. Bats have been implicated as likely natural reservoir hosts based most recently on an investigation of cases among miners infected in 2007 at the Kitaka mine, Uganda, which contained a large population of Marburg virus-infected Rousettus aegyptiacus fruit bats. Described here is an ecologic investigation of Python Cave, Uganda, where an American and a Dutch tourist acquired Marburg virus infection in December 2007 and July 2008. More than 40,000 R. aegyptiacus were found in the cave and were the sole bat species present. Between August 2008 and November 2009, 1,622 bats were captured and tested for Marburg virus. Q-RT-PCR analysis of bat liver/spleen tissues indicated ~2.5% of the bats were actively infected, seven of which yielded Marburg virus isolates. Moreover, Q-RT-PCR-positive lung, kidney, colon and reproductive tissues were found, consistent with potential for oral, urine, fecal or sexual transmission. The combined data for R. aegyptiacus tested from Python Cave and Kitaka mine indicate low level horizontal transmission throughout the year. However, Q-RT-PCR data show distinct pulses of virus infection in older juvenile bats (~six months of age) that temporarily coincide with the peak twice-yearly birthing seasons. Retrospective analysis of historical human infections suspected to have been the result of discrete spillover events directly from nature found 83% (54/65) events occurred during these seasonal pulses in virus circulation, perhaps demonstrating periods of increased risk of human infection. The discovery of two tags at Python Cave from bats marked at Kitaka mine, together with the close genetic linkages evident between viruses detected in geographically distant locations, are consistent with R. aegyptiacus bats existing as a large meta-population with associated virus circulation over broad geographic ranges. These findings provide a basis for developing Marburg hemorrhagic fever risk reduction strategies.


Subject(s)
Chiroptera/virology , Marburg Virus Disease/epidemiology , Marburg Virus Disease/transmission , Marburgvirus/isolation & purification , Animals , Base Sequence , Caves , Chiroptera/classification , Disease Reservoirs , Female , Humans , Male , Marburgvirus/genetics , Nuclear Proteins/genetics , Phylogeny , RNA, Viral/analysis , Retrospective Studies , Seasons , Sequence Analysis, RNA , Uganda/epidemiology , Viral Regulatory and Accessory Proteins/genetics
16.
J Med Virol ; 86(7): 1176-80, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24249525

ABSTRACT

Alkhumra hemorrhagic fever virus (AHFV) is an emerging flavivirus that was isolated originally from Saudi Arabia in 1994-1995. The main tests used for the detection of AHFV are the real time (rt) RT-PCR and virus isolation in cell culture. In the present study the detection of AHFV by rtRT-PCR was compared with virus isolation in BHK-21, HEp-2, and LLC-MK2 cell lines. AHFV suspensions grown in BHK-21, HEp-2, and LLC-MK2 cell lines were serially diluted 10-fold from 10(-1) to 10(-11) . Samples from each dilution were used to inoculate four cell culture tubes and were also examined by the rtRT-PCR for AHFV RNA. Fifteen non-inoculated cell culture samples (five from each cell line) were included blindly in both tests. Thus, a total of 132 AHFV-positive and 15 negative control samples were tested. The rtRT-PCR could detect the viral RNA in all diluted specimens up to and including the 10(-10) dilution (40 specimens for each cell line), whereas, cell cultures were positive in 70% of specimens for BHK-21, 65% for LLC-MK2, and 45% for HEp-2 at this dilution. None of the three cell cultures nor the rtRT-PCR was positive at 10(-11) dilution. The specificity and positive predictive values of virus isolation compared to rtRT-PCR were each 100%, whereas the negative predictive values were 29.4% for BHK-21, 26.3% for LLC-MK2, and 18.5% for HEp-2. In conclusion, the rtRT-PCR is more sensitive than virus isolation for detecting AHFV.


Subject(s)
Clinical Laboratory Techniques/methods , Encephalitis Viruses, Tick-Borne/isolation & purification , Encephalitis, Tick-Borne/diagnosis , Real-Time Polymerase Chain Reaction/methods , Reverse Transcriptase Polymerase Chain Reaction/methods , Animals , Cell Line , Encephalitis Viruses, Tick-Borne/genetics , Encephalitis Viruses, Tick-Borne/growth & development , Humans , Predictive Value of Tests , Saudi Arabia , Sensitivity and Specificity , Virus Cultivation/methods
17.
Intervirology ; 57(5): 300-10, 2014.
Article in English | MEDLINE | ID: mdl-25096447

ABSTRACT

BACKGROUND: Alkhumra hemorrhagic fever virus (AHFV) is a newly described flavivirus first isolated in 1994-1995 from the Alkhumra district south of Jeddah, Saudi Arabia. Subsequently, the virus was also isolated from Makkah (2001-2003) and Najran (2008-2009), Saudi Arabia. METHODS: The full-length genome of an AHFV strain isolated from patients in Najran (referred to as AHFV/997/NJ/09/SA) was PCR amplified and sequenced, and compared with the sequences of 18 other AHFV strains previously isolated from Jeddah and Makkah, dengue virus (DENV), Kyasanur forest disease virus (KFDV), Langat virus, Omsk hemorrhagic fever virus (OHFV), and tick-borne encephalitis virus (TBEV). RESULTS: The RNA of the AHFV/997/NJ/09/SA strain was found to have 10,546 nucleotides encoding for a single 3,416-amino acid polyprotein, whereas the previously reported AHFV strains were composed of 10,685-10,749 nucleotides. The AHFV/997/NJ/09/SA strain showed about 99% homology with the previously reported AHFV strains. The KFDV, Langat virus, TBEV, and OHFV isolates formed a separate cluster with a variable homology. The most important variations were observed in the core protein and NS4a gene sequences of two AHFV isolates. CONCLUSION: The variation in the number of nucleotides and phylogenetic analysis with the other AHFV isolates could have resulted from recombination of circulating virus strains.


Subject(s)
Encephalitis Viruses, Tick-Borne/genetics , Encephalitis, Tick-Borne/virology , Genome, Viral , RNA, Viral/genetics , Sequence Analysis, DNA , Cluster Analysis , Encephalitis Viruses, Tick-Borne/isolation & purification , Humans , Phylogeny , Polyproteins/genetics , Saudi Arabia , Sequence Homology
18.
Emerg Infect Dis ; 19(6): 886-91, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23731788

ABSTRACT

During outbreaks of infectious diseases or in cases of severely ill patients, it is imperative to identify the causative agent. This report describes several events in which virus isolation and identification by electron microscopy were critical to initial recognition of the etiologic agent, which was further analyzed by additional laboratory diagnostic assays. Examples include severe acute respiratory syndrome coronavirus, and Nipah, lymphocytic choriomeningitis, West Nile, Cache Valley, and Heartland viruses. These cases illustrate the importance of the techniques of cell culture and electron microscopy in pathogen identification and recognition of emerging diseases.


Subject(s)
Virus Diseases/diagnosis , Viruses/isolation & purification , Viruses/ultrastructure , Arenaviridae/isolation & purification , Arenaviridae/ultrastructure , Bunyaviridae/isolation & purification , Bunyaviridae/ultrastructure , Cell Culture Techniques , Coronaviridae/isolation & purification , Coronaviridae/ultrastructure , Flaviviridae/isolation & purification , Flaviviridae/ultrastructure , Humans , Microscopy, Electron , Paramyxoviridae/isolation & purification , Paramyxoviridae/ultrastructure , United States/epidemiology , Virus Diseases/epidemiology , Virus Diseases/virology
19.
Virus Genes ; 47(1): 168-72, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23584722

ABSTRACT

An unknown virus was isolated from a mosquito pool collected in Jakarta during routine surveillance in 1979. Analysis of the sample using the Illumina platform resulted in the identification of a Newcastle disease virus (NDV) isolate. The sequence of the isolate indicated that it is an ancestral lineage of class II, genotype XIII. The source of the isolate is unusual, as newcastle disease virus is not believed to be vector-borne, although this mosquito pool was processed in a laboratory also handling samples for avian influenza surveillance and it is possible that this resulted in cross-contamination. This NDV isolate is still ancestral to most extant genotype XIII strains and provides a useful insight into historic NDV evolution.


Subject(s)
Culex/virology , Insect Vectors/virology , Newcastle Disease/virology , Newcastle disease virus/classification , Newcastle disease virus/isolation & purification , Phylogeny , Poultry Diseases/virology , Animals , Chickens , Genotype , Indonesia , Molecular Sequence Data , Newcastle Disease/transmission , Newcastle disease virus/genetics , Poultry Diseases/transmission
20.
BMC Infect Dis ; 13: 285, 2013 Jun 23.
Article in English | MEDLINE | ID: mdl-23800282

ABSTRACT

BACKGROUND: Microvascular endothelial barrier dysfunction is the central enigma in spotted fever group (SFG) rickettsioses. Angiogenin (ANG) is one of the earliest identified angiogenic factors, of which some are relevant to the phosphorylation of VE-cadherins that serve as endothelial adherens proteins. Although exogenous ANG is known to translocate into the nucleus of growing endothelial cells (ECs) where it plays a functional role, nuclear ANG is not detected in quiescent ECs. Besides its nuclear role, ANG is thought to play a cytoplasmic role, owing to its RNase activity that cleaves tRNA to produce small RNAs. Recently, such tRNA-derived RNA fragments (tRFs) have been shown to be induced under stress conditions. All these observations raise an intriguing hypothesis about a novel cytoplasmic role of ANG, which is induced upon infection with Rickettsia and generates tRFs that may play roles in SFG rickettsioses. METHODS: C3H/HeN mice were infected intravenously with a sublethal dose of R. conorii. At days 1, 3, and 5 post infection (p.i.), liver, lung and brain were collected for immunofluorescence (IF) studies of R. conorii and angiogenin (ANG). Human umbilical vein endothelial cells (HUVECs) were infected with R. conorii for 24, 48, and 72 hrs before incubation with 1µg/ml recombinant human ANG (rANG) in normal medium for 2 hrs. HUVEC samples were subjected to IF, exogenous ANG translocation, endothelial permeability, and immunoprecipitation phosphorylation assays. To identify small non-coding RNAs (sncRNAs) upon rickettsial infection, RNAs from pulverized mouse lung tissues and HUVECs were subjected to library preparation and deep sequencing analysis using an Illumina 2000 instrument. Identified sncRNAs were confirmed by Northern hybridization, and their target mRNAs were predicted in silico using BLAST and RNA hybrid programs. RESULTS: In the present study, we have demonstrated endothelial up-regulation of ANG, co-localized with SFG rickettsial infection in vivo. We also have provided direct evidence that rickettsial infection sensitizes human ECs to the translocation of exogenous ANG in a compartmentalized pattern at different times post-infection. Typically, exogenous ANG translocates into the nucleus at 24 hrs and to the cytoplasm at 72 hrs post-infection. The ANG cytoplasmic translocation enhances phosphorylation and destabilization of VE-cadherin and attenuates endothelial barrier function. Of note, deep sequencing analysis detected tRFs, mostly derived from the 5'-halves of host tRNAs, that are induced by ANG. Northern hybridization validates the two most abundantly cloned tRFs derived from tRNA-ValGTG and tRNA-GlyGCC, in both mouse tissues and human cells. Bioinformatics analysis predicted that these tRFs may interact with transcripts associated with the endothelial barrier, the host cell inflammatory response, and autophagy. CONCLUSIONS: Our data provide new insight into the role of compartmentalized ANG during SFG rickettsioses, and highlight its possible mediation through tRFs.


Subject(s)
Endothelial Cells/pathology , RNA, Small Untranslated/metabolism , Ribonuclease, Pancreatic/metabolism , Rickettsia conorii/physiology , Animals , Base Sequence , Boutonneuse Fever/metabolism , Boutonneuse Fever/microbiology , Boutonneuse Fever/pathology , Brain/metabolism , Brain Chemistry , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Endothelium, Vascular/metabolism , Endothelium, Vascular/microbiology , Female , Host-Pathogen Interactions , Human Umbilical Vein Endothelial Cells , Humans , Immunohistochemistry , Intracellular Space/chemistry , Intracellular Space/metabolism , Liver/chemistry , Liver/metabolism , Lung/chemistry , Lung/metabolism , Mice , Mice, Inbred C3H , Molecular Sequence Data , RNA, Small Untranslated/genetics , RNA, Transfer/genetics , RNA, Transfer/metabolism , Recombinant Proteins , Reproducibility of Results , Ribonuclease, Pancreatic/genetics , Rickettsia conorii/pathogenicity , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL