Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
1.
PLoS Pathog ; 20(6): e1011777, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38913740

RESUMEN

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.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Modelos Animales de Enfermedad , Ratones Transgénicos , SARS-CoV-2 , Animales , COVID-19/patología , COVID-19/complicaciones , COVID-19/virología , COVID-19/metabolismo , Ratones , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Humanos , Trastornos de la Coagulación Sanguínea/virología , Trastornos de la Coagulación Sanguínea/patología , Neumonía Viral/virología , Neumonía Viral/patología , Neumonía Viral/metabolismo , Betacoronavirus , Pulmón/virología , Pulmón/patología , Pulmón/metabolismo , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/complicaciones , Pandemias , Trampas Extracelulares/metabolismo
2.
Nat Commun ; 14(1): 5603, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-37699929

RESUMEN

Lassa virus is a member of the Arenaviridae family, which causes human infections ranging from asymptomatic to severe hemorrhagic disease with a high case fatality rate. We have designed and generated lipid nanoparticle encapsulated, modified mRNA vaccines that encode for the wild-type Lassa virus strain Josiah glycoprotein complex or the prefusion stabilized conformation of the Lassa virus glycoprotein complex. Hartley guinea pigs were vaccinated with two 10 µg doses, 28 days apart, of either construct. Vaccination induced strong binding antibody responses, specific to the prefusion conformation of glycoprotein complex, which were significantly higher in the prefusion stabilized glycoprotein complex construct group and displayed strong Fc-mediated effects. However, Lassa virus-neutralizing antibody activity was detected in some but not all animals. Following the challenge with a lethal dose of the Lassa virus, all vaccinated animals were protected from death and severe disease. Although the definitive mechanism of protection is still unknown, and assessment of the cell-mediated immune response was not investigated in this study, these data demonstrate the promise of mRNA as a vaccine platform against the Lassa virus and that protection against Lassa virus can be achieved in the absence of virus-neutralizing antibodies.


Asunto(s)
Arenaviridae , Virus Lassa , Humanos , Cobayas , Animales , Virus Lassa/genética , Anticuerpos Neutralizantes , Vacunas de ARNm , Glicoproteínas
3.
Emerg Microbes Infect ; 12(2): 2239950, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37470396

RESUMEN

Taï Forest virus (TAFV) is a lesser-known ebolavirus that causes lethal infections in chimpanzees and is responsible for a single human case. Limited research has been done on this human pathogen; however, with the recent emergence of filoviruses in West Africa, further investigation and countermeasure development against this virus is warranted. We developed a vesicular stomatitis virus (VSV)-based vaccine expressing the TAFV glycoprotein as the viral antigen and assessed it for protective efficacy in nonhuman primates (NHPs). Following a single high-dose vaccination, NHPs developed antigen-specific binding and neutralizing antibodies as well as modest T cell responses. Importantly, all vaccinated NHPs were uniformly protected from disease after lethal TAFV challenge while the naïve control group succumbed to the disease. Histopathologic lesions consistent with filovirus disease were present in control NHPs but were not observed in vaccinated NHPs. Transcriptional analysis of whole blood samples obtained after vaccination and challenge was performed to gain insight into molecular underpinnings conferring protection. Differentially expressed genes (DEG) detected 7 days post-vaccination were enriched to processes associated with innate immunity and antiviral responses. Only a small number of DEG was detected in vaccinated NHPs post-challenge while over 1,000 DEG were detected in control NHPs at end-stage disease which mapped to gene ontology terms indicative of defense responses and inflammation. Taken together, this data demonstrates the effective single-dose protection of the VSV-TAFV vaccine, and its potential for use in outbreaks.


Asunto(s)
Ebolavirus , Fiebre Hemorrágica Ebola , Vacunas Virales , Animales , Humanos , Macaca fascicularis , Anticuerpos Antivirales , Bosques
5.
Clin Infect Dis ; 74(10): 1821-1830, 2022 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-34463715

RESUMEN

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.


Asunto(s)
Fiebre de Lassa , Virosis , Células Endoteliales , Humanos , Incidencia , Fiebre de Lassa/epidemiología , Virus Lassa
6.
Elife ; 102021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34581669

RESUMEN

High-throughput genomics of SARS-CoV-2 is essential to characterize virus evolution and to identify adaptations that affect pathogenicity or transmission. While single-nucleotide variations (SNVs) are commonly considered as driving virus adaption, RNA recombination events that delete or insert nucleic acid sequences are also critical. Whole genome targeting sequencing of SARS-CoV-2 is typically achieved using pairs of primers to generate cDNA amplicons suitable for next-generation sequencing (NGS). However, paired-primer approaches impose constraints on where primers can be designed, how many amplicons are synthesized and requires multiple PCR reactions with non-overlapping primer pools. This imparts sensitivity to underlying SNVs and fails to resolve RNA recombination junctions that are not flanked by primer pairs. To address these limitations, we have designed an approach called 'Tiled-ClickSeq', which uses hundreds of tiled-primers spaced evenly along the virus genome in a single reverse-transcription reaction. The other end of the cDNA amplicon is generated by azido-nucleotides that stochastically terminate cDNA synthesis, removing the need for a paired-primer. A sequencing adaptor containing a Unique Molecular Identifier (UMI) is appended to the cDNA fragment using click-chemistry and a PCR reaction generates a final NGS library. Tiled-ClickSeq provides complete genome coverage, including the 5'UTR, at high depth and specificity to the virus on both Illumina and Nanopore NGS platforms. Here, we analyze multiple SARS-CoV-2 isolates and clinical samples to simultaneously characterize minority variants, sub-genomic mRNAs (sgmRNAs), structural variants (SVs) and D-RNAs. Tiled-ClickSeq therefore provides a convenient and robust platform for SARS-CoV-2 genomics that captures the full range of RNA species in a single, simple assay.


Asunto(s)
Secuencia de Bases , Coronavirus/genética , Genoma Viral , ARN , SARS-CoV-2/genética , COVID-19/virología , ADN Complementario , Biblioteca de Genes , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Nanoporos , Reacción en Cadena de la Polimerasa , ARN Mensajero , ARN Viral/genética , Recombinación Genética , Secuenciación Completa del Genoma
8.
Cell Rep ; 35(5): 109086, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33951434

RESUMEN

New World hantaviruses (NWHs) are endemic in North and South America and cause hantavirus cardiopulmonary syndrome (HCPS), with a case fatality rate of up to 40%. Knowledge of the natural humoral immune response to NWH infection is limited. Here, we describe human monoclonal antibodies (mAbs) isolated from individuals previously infected with Sin Nombre virus (SNV) or Andes virus (ANDV). Most SNV-reactive antibodies show broad recognition and cross-neutralization of both New and Old World hantaviruses, while many ANDV-reactive antibodies show activity for ANDV only. mAbs ANDV-44 and SNV-53 compete for binding to a distinct site on the ANDV surface glycoprotein and show potently neutralizing activity to New and Old World hantaviruses. Four mAbs show therapeutic efficacy at clinically relevant doses in hamsters. These studies reveal a convergent and potently neutralizing human antibody response to NWHs and suggest therapeutic potential for human mAbs against HCPS.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Infecciones por Hantavirus/genética , Orthohantavirus/patogenicidad , Animales , Cricetinae , Infecciones por Hantavirus/mortalidad , Humanos , Análisis de Supervivencia
9.
Emerg Infect Dis ; 26(12): 3030-3033, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33219787

RESUMEN

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.


Asunto(s)
Aborto Espontáneo , Fiebre del Valle del Rift , Virus de la Fiebre del Valle del Rift , Animales , Brotes de Enfermedades , Femenino , Humanos , Ganado , Embarazo , Fiebre del Valle del Rift/diagnóstico , Fiebre del Valle del Rift/epidemiología , Virus de la Fiebre del Valle del Rift/genética , Sudán/epidemiología
10.
bioRxiv ; 2020 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-32511316

RESUMEN

The etiologic agent of the outbreak of pneumonia in Wuhan China was identified as severe acute respiratory syndrome associated coronavirus 2 (SARS-CoV-2) in January, 2020. The first US patient was diagnosed 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, 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 two virus repositories, making it broadly available to the public health and research communities. We hope that open access to this important reagent will expedite development of medical countermeasures.

11.
Vaccine X ; 5: 100060, 2020 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-32337506

RESUMEN

In November 2019, The World Health Organization (WHO) issued a draft set of Target Product Profiles (TPPs) describing optimal and minimally acceptable targets for vaccines against Rift Valley fever (RVF), a Phlebovirus with a three segmented genome, in both humans and ruminants. The TPPs contained rigid requirements to protect against genomic reassortment of live, attenuated vaccines (LAVs) with wild-type RVF virus (RVFV), which place undue constraints on development and regulatory approval of LAVs. We review the current LAVs in use and in development, and conclude that there is no evidence that reassortment between LAVs and wild-type RVFV has occurred during field use, that such a reassortment event if it occurred would have no untoward consequence, and that the TPPs should be revised to provide a more balanced assessment of the benefits versus the theoretical risks of reassortment.

12.
Emerg Infect Dis ; 26(6): 1266-1273, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32160149

RESUMEN

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.


Asunto(s)
Betacoronavirus/aislamiento & purificación , Infecciones por Coronavirus/diagnóstico , Neumonía Viral/diagnóstico , Animales , Betacoronavirus/genética , Betacoronavirus/fisiología , COVID-19 , Línea Celular , Chlorocebus aethiops , Genoma Viral , Humanos , Nasofaringe/virología , Orofaringe/virología , Pandemias , SARS-CoV-2 , Células Vero , Tropismo Viral , Replicación Viral , Washingtón
13.
J Infect Dis ; 218(suppl_5): S418-S422, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-30060231

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Ebolavirus/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Chlorocebus aethiops , Modelos Animales de Enfermedad , Fiebre Hemorrágica Ebola/inmunología , Ratones , Ratones Noqueados , Células Vero , Proteínas del Envoltorio Viral/inmunología
14.
Nat Microbiol ; 3(6): 670-677, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29736037

RESUMEN

Ebola virus (EBOV) in humans causes a severe illness with high mortality rates. Several strategies have been developed in the past to treat EBOV infection, including the antibody cocktail ZMapp, which has been shown to be effective in nonhuman primate models of infection 1 and has been used under compassionate-treatment protocols in humans 2 . ZMapp is a mixture of three chimerized murine monoclonal antibodies (mAbs)3-6 that target EBOV-specific epitopes on the surface glycoprotein7,8. However, ZMapp mAbs do not neutralize other species from the genus Ebolavirus, such as Bundibugyo virus (BDBV), Reston virus (RESTV) or Sudan virus (SUDV). Here, we describe three naturally occurring human cross-neutralizing mAbs, from BDBV survivors, that target an antigenic site in the canonical heptad repeat 2 (HR2) region near the membrane-proximal external region (MPER) of the glycoprotein. The identification of a conserved neutralizing antigenic site in the glycoprotein suggests that these mAbs could be used to design universal antibody therapeutics against diverse ebolavirus species. Furthermore, we found that immunization with a peptide comprising the HR2-MPER antigenic site elicits neutralizing antibodies in rabbits. Structural features determined by conserved residues in the antigenic site described here could inform an epitope-based vaccine design against infection caused by diverse ebolavirus species.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Ebolavirus/inmunología , Epítopos/inmunología , Fiebre Hemorrágica Ebola/inmunología , Glicoproteínas de Membrana/química , Animales , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Anticuerpos Antivirales/farmacología , Sitios de Unión/efectos de los fármacos , Chlorocebus aethiops , Reacciones Cruzadas , Hurones , Cobayas , Fiebre Hemorrágica Ebola/metabolismo , Humanos , Glicoproteínas de Membrana/inmunología , Glicoproteínas de Membrana/metabolismo , Ratones , Modelos Moleculares , Unión Proteica , Conejos , Células Vero , Proteínas Virales/química , Proteínas Virales/metabolismo
15.
NPJ Vaccines ; 3: 11, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29581897

RESUMEN

Lassa fever (LF) is a zoonotic disease associated with acute and potentially fatal hemorrhagic illness caused by the Lassa virus (LASV), a member of the family Arenaviridae. It is generally assumed that a single infection with LASV will produce life-long protective immunity. This suggests that protective immunity induced by vaccination is an achievable goal and that cell-mediated immunity may play a more important role in protection, at least following natural infection. Seropositive individuals in endemic regions have been shown to have LASV-specific T cells recognizing epitopes for nucleocapsid protein (NP) and glycoprotein precursor (GPC), suggesting that these will be important vaccine immunogens. The role of neutralizing antibodies in protective immunity is still equivocal as recent studies suggest a role for neutralizing antibodies. There is extensive genetic heterogeneity among LASV strains that is of concern in the development of assays to detect and identify all four LASV lineages. Furthermore, the gene disparity may complicate the synthesis of effective vaccines that will provide protection across multiple lineages. Non-human primate models of LASV infection are considered the gold standard for recapitulation of human LF. The most promising vaccine candidates to date are the ML29 (a live attenuated reassortant of Mopeia and LASV), vesicular stomatitis virus (VSV) and vaccinia-vectored platforms based on their ability to induce protection following single doses, high rates of survival following challenge, and the use of live virus platforms. To date no LASV vaccine candidates have undergone clinical evaluation.

16.
Genome Announc ; 5(42)2017 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-29051241

RESUMEN

We report here the full-length sequences of 16 historical isolates of Crimean-Congo hemorrhagic fever orthonairovirus (CCHFV) obtained in Turkey, Namibia, and South Africa. The strains may be useful for future work to develop molecular diagnostics or viral evolution studies.

17.
Vector Borne Zoonotic Dis ; 17(10): 714-719, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28836897

RESUMEN

Crimean-Congo hemorrhagic fever (CCHF) is a severe tick-borne disease caused by CCHF virus (CCHFV). Ticks in the genus Hyalomma are the main vectors and reservoirs of CCHFV. In Spain, CCHFV was first detected in Hyalomma ticks from Cáceres in 2010. Subsequently, two autochthonous CCHF cases were reported in August 2016. In this study, we describe the characterization of the CCHFV genome directly from Hyalomma lusitanicum collected in Cáceres in 2014. Phylogenetic analyses reveal a close relationship with clade III strains from West Africa, with an estimated divergence time of 50 years. The results of this work suggest that CCHFV has been circulating in Spain for some time, and most likely originated from West Africa.


Asunto(s)
Virus de la Fiebre Hemorrágica de Crimea-Congo/genética , Virus de la Fiebre Hemorrágica de Crimea-Congo/aislamiento & purificación , Ixodidae/virología , Animales , Femenino , Genoma Viral , España , Zoonosis
18.
Methods Mol Biol ; 1628: 353-362, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28573634

RESUMEN

Immunologic assays such as ELISA allow detection of either virus antigens or the host's immune response to antigens associated with prior infections and offer a powerful means to approach understanding the epidemiology and epizootiology of these agents. However, the success of these assays is highly dependent on the production of high-quality materials both to establish the assays (i.e., antigen target for antibody) and to serve as controls for establishing assay parameters and sensitivity. Here we describe methods for preparing ebolavirus antigens suitable for use as either reagents or controls in a variety of ELISA formats. Considering work with filoviruses is typically restricted to maximum containment laboratories; thus, we also provide methods for inactivating and safety testing of these antigens for safe use in properly established field laboratories.


Asunto(s)
Antígenos Virales/inmunología , Ebolavirus/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Fiebre Hemorrágica Ebola/inmunología , Anticuerpos Antivirales/inmunología , Reacciones Cruzadas/inmunología , Ebolavirus/aislamiento & purificación , Fiebre Hemorrágica Ebola/virología , Humanos
19.
Methods Mol Biol ; 1628: 363-372, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28573635

RESUMEN

Ebola viruses are high-priority pathogens first discovered in rural Africa associated with sporadic outbreaks of severe hemorrhagic disease in humans and nonhuman primates. Little is known about the disease ecology or the prevalence of past exposure of human populations to any of the five species of the genus Ebolavirus. The use of immunologic means of detection for either virus antigens or the host's immune response to antigen associated with prior infections offers a powerful approach at understanding the epidemiology and epizootiology of these agents. Here we describe methods for preparing antigen detection sandwich enzyme-linked immunosorbent assays (ELISAs) as well as IgG and IgM ELISAs for the detection of ebolavirus antigens or antibodies in biological samples.


Asunto(s)
Reacciones Cruzadas/inmunología , Ebolavirus/inmunología , Ensayo de Inmunoadsorción Enzimática/métodos , Fiebre Hemorrágica Ebola/diagnóstico , Animales , Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Antígenos Virales/aislamiento & purificación , Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/virología , Humanos
20.
PLoS One ; 12(5): e0178224, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28542463

RESUMEN

Reston virus (family Filoviridae) is unique among the viruses of the Ebolavirus genus in that it is considered non-pathogenic in humans, in contrast to the other members which are highly virulent. The virus has however, been associated with several outbreaks of highly lethal hemorrhagic fever in non-human primates (NHPs), specifically cynomolgus monkeys (Macaca fascicularis) originating in the Philippines. In addition, Reston virus has been isolated from domestic pigs in the Philippines. To better understand virus spillover events and potential adaption to new hosts, the whole genome sequences of representative Reston virus isolates were obtained using a next generation sequencing (NGS) approach and comparative genomic analysis and virus fitness analyses were performed. Nine virus genome sequences were completed for novel and previously described isolates obtained from a variety of hosts including a human case, non-human primates and pigs. Results of phylogenetic analysis of the sequence differences are consistent with multiple independent introductions of RESTV from a still unknown natural reservoir into non-human primates and swine farming operations. No consistent virus genetic markers were found specific for viruses associated with primate or pig infections, but similar to what had been seen with some Ebola viruses detected in the large Western Africa outbreak in 2014-2016, a truncated version of VP30 was identified in a subgroup of Reston viruses obtained from an outbreak in pigs 2008-2009. Finally, the genetic comparison of two closely related viruses, one isolated from a human case and one from an NHP, showed amino acid differences in the viral polymerase and detectable differences were found in competitive growth assays on human and NHP cell lines.


Asunto(s)
Filoviridae/genética , Genoma Viral/genética , Animales , Brotes de Enfermedades/veterinaria , Ebolavirus/genética , Ebolavirus/patogenicidad , Filoviridae/patogenicidad , Infecciones por Filoviridae/veterinaria , Infecciones por Filoviridae/virología , Marcadores Genéticos/genética , Fiebre Hemorrágica Ebola/veterinaria , Fiebre Hemorrágica Ebola/virología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Macaca fascicularis/virología , Porcinos/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA