Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
1.
Viruses ; 14(9)2022 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-36146824

RESUMO

Arenaviruses include important zoonotic pathogens that cause hemorrhagic fever (e.g., Junín virus; JUNV) as well as other viruses that are closely related but apathogenic (e.g., Tacaribe virus; TCRV). We have found that, while TCRV and JUNV differ in their ability to induce apoptosis in infected cells, due to active inhibition of caspase activation by the JUNV nucleoprotein, both viruses trigger similar upstream pro-apoptotic signaling events, including the activation/phosphorylation of p53. In the case of TCRV, the pro-apoptotic factor Bad is also phosphorylated (leading to its inactivation). These events clearly implicate upstream kinases in regulating the induction of apoptosis. Consistent with this, here we show activation in TCRV-infected cells of the stress-activated protein kinases p38 and JNK, which are known to regulate p53 activation, as well as the downstream kinase MK2 and transcription factor c-Jun. We also observed the early transient activation of Akt, but not Erk. Importantly, the chemical inhibition of Akt, p38, JNK and c-Jun all dramatically reduced viral growth, even though we have shown that inhibition of apoptosis itself does not. This indicates that kinase activation is crucial for viral infection, independent of its downstream role in apoptosis regulation, a finding that has the potential to shed further light on the determinants of arenavirus pathogenesis, as well as to inform future therapeutic approaches.


Assuntos
Infecções por Arenaviridae , Arenavirus do Novo Mundo , Apoptose , Arenavirus do Novo Mundo/fisiologia , Caspases , Humanos , Nucleoproteínas , Proteínas Proto-Oncogênicas c-akt , Fatores de Transcrição , Proteína Supressora de Tumor p53 , Replicação Viral , Proteínas Quinases p38 Ativadas por Mitógeno
2.
Salud(i)ciencia (Impresa) ; 25(1): 24-29, 2022. tab.
Artigo em Espanhol | LILACS | ID: biblio-1436265

RESUMO

Chapare hemorrhagic fever is an emerging zoonotic disease caused by a Mammarenavirus, of the Arenaviridae family, called Chapare virus. Its name refers to the region in which it first appeared. This virus is transmitted through direct contact with the excreta of wild rodents that interact with humans by invading homes close to a rural area and when humans enter the jungle for various reasons such as agriculture and tourism, among others. This virus has been classified by the World Health Organization as biosafety level IV, where pathogens with the highest risk for humans are included and for which there are no vaccines available. It produces a febrile and hemorrhagic picture that leads to death two weeks after the onset of symptoms. There is no vaccine or standardized treatment to treat this disease. The bio ecological aspects of the reservoir are not known, nor are the factors that could be related to the emergency in the area. An investigation should be carried out focused mainly on knowing the circulation of the Chapare virus in the Bolivian tropics, knowing the bioecological characteristics of the reservoir to produce and contribute with reliable information for the design of future surveillance and control strategies with community participation, as well as to strengthen laboratory diagnostic capacities in health establishments in the municipality of Villa Tunari.


La fiebre hemorrágica Chapare es una enfermedad zoonótica emergente causada por un Mammarenavirus de la familia Arenaviridae, llamado virus Chapare; su nombre se debe a la región en la cual apareció por primera vez. Este virus es transmitido por medio del contacto directo con las excretas de los roedores silvestres que interactúan con el ser humano, al invadir las viviendas que se encuentran cercanas al área silvestre y cuando el hombre incursiona a la selva por diversas razones, como las actividades de agricultura o turismo, entre otras. Este virus ha sido catalogado por la Organización Mundial de la Salud como nivel de bioseguridad IV, en el cual se ubican los patógenos de mayor riesgo para el ser humano y para los cuales no existen vacunas disponibles. Produce un cuadro febril y hemorrágico que lleva al fallecimiento a las dos semanas de haber iniciado los síntomas. No existe vacuna ni tratamiento estandarizado para tratar esta enfermedad, no se conocen los aspectos bioecológicos del reservorio y tampoco los factores que podrían estar relacionados con la emergencia en la zona. Se deberá realizar una investigación enfocada principalmente en conocer la circulación del virus Chapare en la región del trópico boliviano, conocer las características bioecológicas del reservorio con el propósito de producir y aportar con información fehaciente para el diseño de futuras estrategias de vigilancia y control con participación comunitaria, así como fortalecer las capacidades de diagnóstico de laboratorio en los establecimientos de salud del municipio de Villa Tunari.


Assuntos
Arenavirus do Novo Mundo , Zoonoses , Arenavirus , Hemorragia
3.
Antiviral Res ; 193: 105125, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34197863

RESUMO

Several arenaviruses, including Lassa and Lujo viruses in Africa and five New World arenavirus (NWA) species in the Americas, cause life-threatening viral hemorrhagic fevers. In the absence of licensed antiviral therapies, these viruses pose a significant public health risk. The envelope glycoprotein complex (GPC) mediates arenavirus entry through a pH-dependent fusion of the viral and host endosomal membranes. It thus is recognized as a viable target for small-molecule fusion inhibitors. Here, we report on the antiviral activity and pre-clinical development of the novel broad-spectrum arenavirus fusion inhibitors, ARN-75039 and ARN-75041. In Tacaribe virus (TCRV) pseudotyped and native virus assays, the ARN compounds were active in the low to sub-nanomolar range with selectivity indices exceeding 1000. Pharmacokinetic analysis of the orally administered compounds revealed an extended half-life in mice supporting once-daily dosing, and the compounds were well tolerated at the highest tested dose of 100 mg/kg. In a proof-of-concept prophylactic efficacy study, doses of 10 and 35 mg/kg of either compound dramatically improved survival outcome and potently inhibited TCRV replication in serum and various tissues. Additionally, in contrast to surviving mice that received ribavirin or placebo, animals treated with ARN-75039 or ARN-75041 were cured of TCRV infection. In a follow-up study with ARN-75039, impressive therapeutic efficacy was demonstrated under conditions where treatment was withheld until after the onset of disease. Taken together, the data strongly support the continued development of ARN-75039 as a candidate therapeutic for the treatment of severe arenaviral diseases.


Assuntos
Antivirais/farmacologia , Infecções por Arenaviridae/tratamento farmacológico , Arenavirus do Novo Mundo/efeitos dos fármacos , Fusão de Membrana/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Administração Oral , Animais , Antivirais/farmacocinética , Chlorocebus aethiops , Masculino , Camundongos , Ribavirina/farmacologia , Bibliotecas de Moléculas Pequenas/farmacocinética , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus/efeitos dos fármacos
4.
Proteins ; 89(3): 311-321, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33068039

RESUMO

Machupo virus, known to cause hemorrhagic fevers, enters human cells via binding with its envelope glycoprotein to transferrin receptor 1 (TfR). Similarly, the receptor interactions have been explored in biotechnological applications as a molecular system to ferry therapeutics across the cellular membranes and through the impenetrable blood-brain barrier that effectively blocks any such delivery into the brain. Study of the experimental structure of Machupo virus glycoprotein 1 (MGP1) in complex with TfR and glycoprotein sequence homology has identified some residues at the interface that influence binding. There are, however, no studies that have attempted to optimize the binding potential between MGP1 and TfR. In pursuits for finding therapeutic solutions for the New World arenaviruses, and to gain a greater understanding of MGP1 interactions with TfR, it is crucial to understand the structure-sequence relationship driving the interface formation. By displaying MGP1 on yeast surface we have examined the contributions of individual residues to the binding of solubilized ectodomain of TfR. We identified MGP1 binding hot spot residues, assessed the importance of posttranslational N-glycan modifications, and used a selection with random mutagenesis for affinity maturation. We show that the optimized MGP1 variants can bind more strongly to TfR than the native MGP1, and there is an MGP1 sequence that retains binding in the absence of glycosylation, but with the addition of further amino acid substitutions. The engineered variants can be used to probe cellular internalization or the blood-brain barrier crossing to achieve greater understanding of TfR mediated internalization.


Assuntos
Antígenos CD , Arenavirus do Novo Mundo/química , Receptores da Transferrina , Proteínas do Envelope Viral , Antígenos CD/química , Antígenos CD/genética , Antígenos CD/metabolismo , Humanos , Modelos Moleculares , Mutação , Engenharia de Proteínas , Receptores da Transferrina/química , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
5.
PLoS Negl Trop Dis ; 14(12): e0009004, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33370288

RESUMO

A detailed understanding of the mechanisms underlying the capacity of a virus to break the species barrier is crucial for pathogen surveillance and control. New World (NW) mammarenaviruses constitute a diverse group of rodent-borne pathogens that includes several causative agents of severe viral hemorrhagic fever in humans. The ability of the NW mammarenaviral attachment glycoprotein (GP) to utilize human transferrin receptor 1 (hTfR1) as a primary entry receptor plays a key role in dictating zoonotic potential. The recent isolation of Tacaribe and lymphocytic choriominingitis mammarenaviruses from host-seeking ticks provided evidence for the presence of mammarenaviruses in arthropods, which are established vectors for numerous other viral pathogens. Here, using next generation sequencing to search for other mammarenaviruses in ticks, we identified a novel replication-competent strain of the NW mammarenavirus Tamiami (TAMV-FL), which we found capable of utilizing hTfR1 to enter mammalian cells. During isolation through serial passaging in mammalian immunocompetent cells, the quasispecies of TAMV-FL acquired and enriched mutations leading to the amino acid changes N151K and D156N, within GP. Cell entry studies revealed that both substitutions, N151K and D156N, increased dependence of the virus on hTfR1 and binding to heparan sulfate proteoglycans. Moreover, we show that the substituted residues likely map to the sterically constrained trimeric axis of GP, and facilitate viral fusion at a lower pH, resulting in viral egress from later endosomal compartments. In summary, we identify and characterize a naturally occurring TAMV strain (TAMV-FL) within ticks that is able to utilize hTfR1. The TAMV-FL significantly diverged from previous TAMV isolates, demonstrating that TAMV quasispecies exhibit striking genetic plasticity that may facilitate zoonotic spillover and rapid adaptation to new hosts.


Assuntos
Antígenos CD/metabolismo , Infecções por Arenaviridae/transmissão , Arenaviridae/genética , Receptores da Transferrina/metabolismo , Receptores Virais/metabolismo , Proteínas do Envelope Viral/genética , Sequência de Aminoácidos/genética , Animais , Arenaviridae/isolamento & purificação , Arenavirus do Novo Mundo , Linhagem Celular , Chlorocebus aethiops , Células HEK293 , Humanos , Insetos Vetores/virologia , Alinhamento de Sequência , Carrapatos/virologia , Células Vero , Envelope Viral/metabolismo , Zoonoses/transmissão , Zoonoses/virologia
6.
PLoS Pathog ; 16(10): e1008948, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33045019

RESUMO

Pathogenicity often differs dramatically among even closely related arenavirus species. For instance, Junín virus (JUNV), the causative agent of Argentine hemorrhagic fever (AHF), is closely related to Tacaribe virus (TCRV), which is normally avirulent in humans. While little is known about how host cell pathways are regulated in response to arenavirus infection, or how this contributes to virulence, these two viruses have been found to differ markedly in their ability to induce apoptosis. However, details of the mechanism(s) governing the apoptotic response to arenavirus infections are unknown. Here we confirm that TCRV-induced apoptosis is mitochondria-regulated, with associated canonical hallmarks of the intrinsic apoptotic pathway, and go on to identify the pro- and anti-apoptotic Bcl-2 factors responsible for regulating this process. In particular, levels of the pro-apoptotic BH3-only proteins Noxa and Puma, as well as their canonical transcription factor p53, were strongly increased. Interestingly, TCRV infection also led to the accumulation of the inactive phosphorylated form of another pro-apoptotic BH3-only protein, Bad (i.e. as phospho-Bad). Knockout of Noxa or Puma suppressed apoptosis in response to TCRV infection, whereas silencing of Bad increased apoptosis, confirming that these factors are key regulators of apoptosis induction in response to TCRV infection. Further, we found that while the highly pathogenic JUNV does not induce caspase activation, it still activated upstream pro-apoptotic factors, consistent with current models suggesting that JUNV evades apoptosis by interfering with caspase activation through a nucleoprotein-mediated decoy function. This new mechanistic insight into the role that individual BH3-only proteins and their regulation play in controlling apoptotic fate in arenavirus-infected cells provides an important experimental framework for future studies aimed at dissecting differences in the apoptotic responses between arenaviruses, their connection to other cell signaling events and ultimately the relationship of these processes to pathogenesis.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , Infecções por Arenaviridae/patologia , Arenavirus do Novo Mundo/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Replicação Viral , Proteína de Morte Celular Associada a bcl/metabolismo , Proteínas Reguladoras de Apoptose/genética , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/metabolismo , Infecções por Arenaviridae/virologia , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Domínios Proteicos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteína de Morte Celular Associada a bcl/genética
7.
Nat Commun ; 10(1): 1121, 2019 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-30850661

RESUMO

Human transferrin receptor 1 (CD71) guarantees iron supply by endocytosis upon binding of iron-loaded transferrin and ferritin. Arenaviruses and the malaria parasite exploit CD71 for cell invasion and epitopes on CD71 for interaction with transferrin and pathogenic hosts were identified. Here, we provide the molecular basis of the CD71 ectodomain-human ferritin interaction by determining the 3.9 Å resolution single-particle cryo-electron microscopy structure of their complex and by validating our structural findings in a cellular context. The contact surfaces between the heavy-chain ferritin and CD71 largely overlap with arenaviruses and Plasmodium vivax binding regions in the apical part of the receptor ectodomain. Our data account for transferrin-independent binding of ferritin to CD71 and suggest that select pathogens may have adapted to enter cells by mimicking the ferritin access gate.


Assuntos
Antígenos CD/química , Apoferritinas/química , Proteínas de Protozoários/química , Receptores da Transferrina/química , Receptores Virais/química , Transferrina/química , Proteínas do Envelope Viral/química , Antígenos CD/genética , Antígenos CD/metabolismo , Apoferritinas/genética , Apoferritinas/metabolismo , Arenavirus do Novo Mundo/genética , Arenavirus do Novo Mundo/metabolismo , Sítios de Ligação , Clonagem Molecular , Microscopia Crioeletrônica , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Células HeLa , Proteína da Hemocromatose/química , Proteína da Hemocromatose/genética , Proteína da Hemocromatose/metabolismo , Humanos , Plasmodium vivax/genética , Plasmodium vivax/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Proteínas de Protozoários/genética , Proteínas de Protozoários/metabolismo , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo , Receptores Virais/genética , Receptores Virais/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Transferrina/genética , Transferrina/metabolismo , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
8.
PLoS Biol ; 17(2): e3000137, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30726215

RESUMO

Tripartite motif (TRIM) proteins belong to a large family with many roles in host biology, including restricting virus infection. Here, we found that TRIM2, which has been implicated in cases of Charcot-Marie-Tooth disease (CMTD) in humans, acts by blocking hemorrhagic fever New World arenavirus (NWA) entry into cells. We show that Trim2-knockout mice, as well as primary fibroblasts from a CMTD patient with mutations in TRIM2, are more highly infected by the NWAs Junín and Tacaribe virus than wild-type mice or cells are. Using mice with different Trim2 gene deletions and TRIM2 mutant constructs, we demonstrate that its antiviral activity is uniquely independent of the RING domain encoding ubiquitin ligase activity. Finally, we show that one member of the TRIM2 interactome, signal regulatory protein α (SIRPA), a known inhibitor of phagocytosis, also restricts NWA infection and conversely that TRIM2 limits phagocytosis of apoptotic cells. In addition to demonstrating a novel antiviral mechanism for TRIM proteins, these studies suggest that the NWA entry and phagocytosis pathways overlap.


Assuntos
Antígenos de Diferenciação/genética , Arenavirus do Novo Mundo/genética , Doença de Charcot-Marie-Tooth/genética , Interações Hospedeiro-Patógeno/genética , Proteínas Nucleares/genética , Receptores Imunológicos/genética , Animais , Antígenos de Diferenciação/imunologia , Antígenos de Diferenciação/metabolismo , Apoptose , Arenavirus do Novo Mundo/crescimento & desenvolvimento , Arenavirus do Novo Mundo/patogenicidade , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/virologia , Linhagem Celular Tumoral , Doença de Charcot-Marie-Tooth/metabolismo , Doença de Charcot-Marie-Tooth/patologia , Chlorocebus aethiops , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Regulação da Expressão Gênica , Células HEK293 , Interações Hospedeiro-Patógeno/imunologia , Humanos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/imunologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/imunologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/imunologia , Proteínas de Neurofilamentos/metabolismo , Proteínas Nucleares/imunologia , Proteínas Nucleares/metabolismo , Osteoblastos/imunologia , Osteoblastos/metabolismo , Osteoblastos/virologia , Cultura Primária de Células , Receptores Imunológicos/imunologia , Receptores Imunológicos/metabolismo , Transdução de Sinais , Células Vero , Internalização do Vírus
9.
J Virol ; 93(1)2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30305351

RESUMO

The emergence of Old and New World arenaviruses from rodent reservoirs persistently threatens human health. The GP1 subunit of the envelope-displayed arenaviral glycoprotein spike complex (GPC) mediates host cell recognition and is an important determinant of cross-species transmission. Previous structural analyses of Old World arenaviral GP1 glycoproteins, alone and in complex with a cognate GP2 subunit, have revealed that GP1 adopts two distinct conformational states distinguished by differences in the orientations of helical regions of the molecule. Here, through comparative study of the GP1 glycoprotein architectures of Old World Loei River virus and New World Whitewater Arroyo virus, we show that these rearrangements are restricted to Old World arenaviruses and are not induced solely by the pH change that is associated with virus endosomal trafficking. Our structure-based phylogenetic analysis of arenaviral GP1s provides a blueprint for understanding the discrete structural classes adopted by these therapeutically important targets.IMPORTANCE The genetically and geographically diverse group of viruses within the family Arenaviridae includes a number of zoonotic pathogens capable of causing fatal hemorrhagic fever. The multisubunit GPC glycoprotein spike complex displayed on the arenavirus envelope is a key determinant of species tropism and a primary target of the host humoral immune response. Here, we show that the receptor-binding GP1 subcomponent of the GPC spike from Old World but not New World arenaviruses adopts a distinct, pH-independent conformation in the absence of the cognate GP2. Our analysis provides a structure-based approach to understanding the discrete conformational classes sampled by these therapeutically important targets, informing strategies to develop arenaviral glycoprotein immunogens that resemble GPC as presented on the mature virion surface.


Assuntos
Arenavirus do Novo Mundo/classificação , Arenavirus do Velho Mundo/classificação , Proteínas do Envelope Viral/química , Arenavirus do Novo Mundo/química , Arenavirus do Novo Mundo/metabolismo , Arenavirus do Velho Mundo/química , Arenavirus do Velho Mundo/metabolismo , Endossomos/virologia , Evolução Molecular , Concentração de Íons de Hidrogênio , Modelos Moleculares , Filogenia , Estrutura Secundária de Proteína
10.
Enferm Infecc Microbiol Clin (Engl Ed) ; 37(9): 602-608, 2019 Nov.
Artigo em Inglês, Espanhol | MEDLINE | ID: mdl-29907366

RESUMO

Ribavirin is a molecule with antiviral activity against different viruses. In clinical practice, it has made its niche almost exclusively for the treatment of the hepatitisC virus. However, there are other diseases in which it could be of benefit and it has the advantage of being suitable for oral, intravenous and inhaled administration. We conducted a review of the indications of the main drug agencies (Spanish, European and American) and other possible indications, mainly haemorrhagic fevers and coronavirus.


Assuntos
Antivirais/uso terapêutico , Ribavirina/uso terapêutico , Viroses/tratamento farmacológico , Vírus/efeitos dos fármacos , Infecções por Adenoviridae/tratamento farmacológico , Adenovírus Humanos/efeitos dos fármacos , Antivirais/farmacologia , Arenavirus do Novo Mundo/efeitos dos fármacos , Ensaios Clínicos como Assunto , Infecções por Coronavirus/tratamento farmacológico , Orthohantavírus/efeitos dos fármacos , Infecções por Hantavirus/tratamento farmacológico , Vírus da Febre Hemorrágica da Crimeia-Congo/efeitos dos fármacos , Febre Hemorrágica Americana/tratamento farmacológico , Febre Hemorrágica da Crimeia/tratamento farmacológico , Humanos , Febre Lassa/tratamento farmacológico , Vírus Lassa/efeitos dos fármacos , Metanálise como Assunto , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Vírus Sinciciais Respiratórios/efeitos dos fármacos
11.
mSphere ; 3(3)2018.
Artigo em Inglês | MEDLINE | ID: mdl-29720525

RESUMO

Arenaviruses pose a major public health threat and cause numerous infections in humans each year. Although most viruses belonging to this family do not cause disease in humans, some arenaviruses, such as Lassa virus and Machupo virus, are the etiological agents of lethal hemorrhagic fevers. The absence of a currently licensed vaccine and the highly pathogenic nature of these viruses both make the necessity of developing viable vaccines and therapeutics all the more urgent. Arenaviruses have a single glycoprotein on the surface of virions, the glycoprotein complex (GPC), and this protein can be used as a target for vaccine development. Here, we describe immunization strategies to generate monoclonal antibodies (MAbs) that cross-react between the glycoprotein complexes of both Old World and New World arenaviruses. Several monoclonal antibodies isolated from immunized mice were highly cross-reactive, binding a range of Old World arenavirus glycoproteins, including that of Lassa virus. One such monoclonal antibody, KL-AV-2A1, bound to GPCs of both New World and Old World viruses, including Lassa and Machupo viruses. These cross-reactive antibodies bound to epitopes present on the glycoprotein 2 subunit of the glycoprotein complex, which is relatively conserved among arenaviruses. Monoclonal antibodies binding to these epitopes, however, did not inhibit viral entry as they failed to neutralize a replication-competent vesicular stomatitis virus pseudotyped with the Lassa virus glycoprotein complex in vitro In addition, no protection from virus challenge was observed in in vivo mouse models. Even so, these monoclonal antibodies might still prove to be useful in the development of clinical and diagnostic assays.IMPORTANCE Several viruses in the Arenaviridae family infect humans and cause severe hemorrhagic fevers which lead to high case fatality rates. Due to their pathogenicity and geographic tropisms, these viruses remain very understudied. As a result, an effective vaccine or therapy is urgently needed. Here, we describe efforts to produce cross-reactive monoclonal antibodies that bind to both New and Old World arenaviruses. All of our MAbs seem to be nonneutralizing and nonprotective and target subunit 2 of the glycoprotein. Due to the lack of reagents such as recombinant glycoproteins and antibodies for rapid detection assays, our MAbs could be beneficial as analytic and diagnostic tools.


Assuntos
Anticorpos Antivirais/imunologia , Arenavirus do Novo Mundo/imunologia , Arenavirus do Velho Mundo/imunologia , Reações Cruzadas , Glicoproteínas/imunologia , Proteínas Estruturais Virais/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/isolamento & purificação , Infecções por Arenaviridae/imunologia , Infecções por Arenaviridae/prevenção & controle , Modelos Animais de Doenças , Mapeamento de Epitopos , Epitopos de Linfócito B/imunologia , Camundongos
12.
Viruses ; 10(5)2018 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-29724035

RESUMO

New World arenaviruses cause fatal hemorrhagic disease in South America. Pirital virus (PIRV), a mammarenavirus hosted by Alston’s cotton rat (Sigmodon alstoni), causes a disease in Syrian golden hamsters (Mesocricetus auratus) (biosafety level-3, BSL-3) that has many pathologic similarities to the South American hemorrhagic fevers (BSL-4) and, thus, is considered among the best small-animal models for human arenavirus disease. Here, we extend in greater detail previously described clinical and pathological findings in Syrian hamsters and provide evidence for a pro-inflammatory macrophage response during PIRV infection. The liver was the principal target organ of the disease, and signs of Kupffer cell involvement were identified in mortally infected hamster histopathology data. Differential expression analysis of liver mRNA revealed signatures of the pro-inflammatory response, hematologic dysregulation, interferon pathway and other host response pathways, including 17 key transcripts that were also reported in two non-human primate (NHP) arenavirus liver-infection models, representing both Old and New World mammarenavirus infections. Although antigen presentation may differ among rodent and NHP species, key hemostatic and innate immune-response components showed expression parallels. Signatures of pro-inflammatory macrophage involvement in PIRV-infected livers included enrichment of Ifng, Nfkb2, Stat1, Irf1, Klf6, Il1b, Cxcl10, and Cxcl11 transcripts. Together, these data indicate that pro-inflammatory macrophage M1 responses likely contribute to the pathogenesis of acute PIRV infection.


Assuntos
Infecções por Arenaviridae/imunologia , Arenavirus do Novo Mundo/patogenicidade , Fígado/imunologia , Macrófagos/imunologia , Animais , Cricetinae , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno , Imunidade Inata , Células de Kupffer/virologia , Fígado/patologia , Fígado/virologia
13.
Nat Commun ; 9(1): 1884, 2018 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-29760382

RESUMO

While five arenaviruses cause human hemorrhagic fevers in the Western Hemisphere, only Junin virus (JUNV) has a vaccine. The GP1 subunit of their envelope glycoprotein binds transferrin receptor 1 (TfR1) using a surface that substantially varies in sequence among the viruses. As such, receptor-mimicking antibodies described to date are type-specific and lack the usual breadth associated with this mode of neutralization. Here we isolate, from the blood of a recipient of the live attenuated JUNV vaccine, two antibodies that cross-neutralize Machupo virus with varying efficiency. Structures of GP1-Fab complexes explain the basis for efficient cross-neutralization, which involves avoiding receptor mimicry and targeting a conserved epitope within the receptor-binding site (RBS). The viral RBS, despite its extensive sequence diversity, is therefore a target for cross-reactive antibodies with activity against New World arenaviruses of public health concern.


Assuntos
Anticorpos Neutralizantes/química , Anticorpos Antivirais/química , Arenavirus do Novo Mundo/imunologia , Febre Hemorrágica Americana/prevenção & controle , Fragmentos Fab das Imunoglobulinas/química , Vírus Junin/imunologia , Proteínas do Envelope Viral/química , Sequência de Aminoácidos , Anticorpos Neutralizantes/isolamento & purificação , Anticorpos Antivirais/isolamento & purificação , Antígenos CD/química , Antígenos CD/genética , Antígenos CD/imunologia , Antígenos Virais/química , Antígenos Virais/genética , Antígenos Virais/imunologia , Arenavirus do Novo Mundo/genética , Sítios de Ligação de Anticorpos , Reações Cruzadas , Epitopos/química , Epitopos/genética , Epitopos/imunologia , Células HEK293 , Febre Hemorrágica Americana/imunologia , Febre Hemorrágica Americana/virologia , Humanos , Soros Imunes/química , Fragmentos Fab das Imunoglobulinas/isolamento & purificação , Vírus Junin/genética , Modelos Moleculares , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/imunologia , Receptores da Transferrina/química , Receptores da Transferrina/genética , Receptores da Transferrina/imunologia , Receptores Virais/química , Receptores Virais/genética , Receptores Virais/imunologia , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem
14.
Medicina (B.Aires) ; 77(5): 353-357, oct. 2017. tab
Artigo em Espanhol | LILACS | ID: biblio-894499

RESUMO

Candid#1 es la primera vacuna a virus vivo atenuado producida y registrada en Argentina. Se produce en el INEVH desde 2003 para prevenir la fiebre hemorrágica argentina y se obtiene mediante cosecha de sobrenadantes de cultivos de células diploides infectadas con una cepa atenuada del virus Junín, formulación y posterior liofilización. Su estabilidad es crucial para asegurar su efectividad. El objetivo de este trabajo fue evaluar la estabilidad de Candid#1 exponiéndola a distintas condiciones de temperatura y tiempo. Tres lotes producidos en 2003 fueron sometidos al siguiente esquema de almacenamiento: (a) vacuna reconstituida conservada entre 2 °C y 8 °C durante 8 días, (b) vacuna liofilizada conservada entre 2 °C y 8 °C durante 6 meses, y (c) vacuna liofilizada conservada entre -18 °C y -20 °C durante 10 años. La potencia fue evaluada en monocapa de células Vero bajo agar. Los resultados fueron: (a) Candid#1 reconstituida fue estable 8 días entre 2 °C y 8 °C, (b) Candid#1 liofilizada fue estable 2 meses entre 2 °C y 8 °C y (c) Candid#1 liofilizada fue estable 9 años entre -18 °C y -20 °C manteniendo todos sus atributos. Estos resultados permitieron establecer las siguientes condiciones de almacenamiento: reconstituida 12 horas entre 2 °C y 8 °C, liofilizada 30 días entre 2 °C y 8 °C y 9 años entre -18 °C y -20 °C. A la luz de estos resultados, se generaron cambios favorables en las condiciones de transporte, almacenamiento y distribución de la vacuna. Se implementó la instalación de freezers domésticos en centros estratégicamente distribuidos, permitiendo preservar stocks de vacuna y distribuir las dosis necesarias a vacunatorios.


Candid#1 is the first live attenuated vaccine produced and registered in Argentina. Produced since 2003 at the INEVH to prevent Argentine hemorrhagic fever, it is obtained by harvesting supernatants of diploid cells infected with an attenuated strain of Junin virus and subsequent lyophilization. The stability of this vaccine is crucial to ensure its effectiveness. This study was aimed to evaluate the stability of Candid#1 by exposing it to different time and temperature conditions. Three vaccine batches produced in 2003 were analysed according to the following storage scheme: (a) reconstituted vaccine at 2 °C to 8°C for 8 days; (b) lyophilized vaccine at 2 °C to 8 °C for 6 months; (c) lyophilized vaccine at -18 °C to -20 °C for 10 years. The potency was assessed in Vero cell monolayers under agar. The results were: (a) reconstituted vaccine was stable between 2 °C and 8 °C for 8 days, (b) lyophilized vaccine was stable between 2 °C and 8 °C for 2 months, and (c) lyophilized vaccine was stable 9 years between -18 °C and -20 °C, keeping all its properties. These results allowed us to establish the following storage conditions and expiration times for Candid#1: (a) reconstituted: 12 hours between 2 °C and 8 °C, (b) lyophilized: 30 days between 2 °C and 8 °C and (c) lyophilized: 9 years between -18 °C and -20 °C. Based on our results, favorable changes were made in the conditions of transport, storage and distribution of the vaccine. Domestic freezers in strategically located centers were installed, allowing the preservation of vaccine stocks for distribution to secondary vaccination centers.


Assuntos
Humanos , Vacinas Virais/imunologia , Arenavirus do Novo Mundo/imunologia , Armazenamento de Medicamentos/métodos , Febre Hemorrágica Americana/prevenção & controle , Anticorpos Antivirais/imunologia , Argentina , Vacinas Atenuadas/imunologia , Estabilidade de Medicamentos
16.
Rev. Soc. Bras. Med. Trop ; 50(1): 3-8, Jan.-Feb. 2017. tab, graf
Artigo em Inglês | LILACS | ID: biblio-842821

RESUMO

Abstract Emerging infectious diseases are a global threat. In countries like Brazil, where biodiversity is high and public health conditions in terms of infrastructure and medical care are often precarious, emerging diseases are particularly worrisome. The lack of monitoring strategies to identify pathogens with the potential to cause outbreaks or epidemics is another problem in Brazil and other developing countries. In this article, we present the history of the Sabiá virus (SABV), a pathogen that was described in the 1990s in Brazil. Several aspects of the biology and ecology of the SABV remain unknown. The SABV has the potential to cause hemorrhagic fever in humans. To date, four cases of human infections have been reported worldwide; two were naturally acquired (both in Brazil), whereas the other two were linked to occupational exposure in the laboratory environment (one in Brazil and one in the USA). In this review, we summarize the basic biological and ecological characteristics of the SABV. This is the first work to gather all available data on the historical aspects involving the cases of SABV infection along with an update on its characteristic features.


Assuntos
Humanos , Masculino , Adulto , Acidentes de Trabalho , Arenavirus do Novo Mundo , Febre Hemorrágica Americana/virologia , Brasil , Pessoal de Laboratório
17.
São Paulo; s.n; 2017. ilus.
Tese em Português | LILACS, CONASS, Sec. Est. Saúde SP, SESSP-CTDPROD, Sec. Est. Saúde SP | ID: biblio-1414832

RESUMO

As febres hemorrágicas virais causadas por arenavirus são zoonoses emergentes e graves . Os humanos são acidentalmente infectado s principalmente pela exposição da mucosa a aerossóis formados a partir de excretas de roedores reservatórios. Nos reservatórios, a presença de anticorpos pode indicar a circulação viral em uma população de uma região específica, e esse dado pode ser utili zado como indicador para investigações posteriores por meio de técnicas moleculares. Até o presente momento não se detectou a espécie reservatório do vírus Sabiá agente etiológico da Febre Hemorrágica Brasileira. Com a finalidade de ampliar as regiões de busca por espécies de roedores silvestres envolvidos na transmissão de arenavírus 2.243 amostras de sangue de roedores coletados entre 1998 e 2008 selecionadas aleatoriamente, foram analisadas por meio de ELISA para detecção de anticorpos IgG específicos pa ra arenavírus. Um tota l de 2230 amostras apresent aram resultados não reagente, 10 apresentaram resultados reagentes e 3 apresentaram resultados indeterminados. As espécies soropositivas foram de Necromys lasiurus 0,6% 6/1012), Calomys callosus, 0,3% 2/94) Akodon sp 0,3% (2/ 723). Obtivemos uma amostra positiva proveniente do Estado de Goiás, cinco do Estado do Mato Grosso do Sul e seis do Estado de São Paulo. Os municípios de Campo Alegre de Goiás, Bodoquena, Nuporanga e Mogi das Cruzes, podem futurame nte ser localidades definidas para ampliar as análises com novas expedições de campo e coleta de amostras biológicas, objetivando a pesquisa de evidencias da circulação viral.


Assuntos
Imunoglobulina G , Reservatórios de Água , Arenavirus do Novo Mundo , Aerossóis , Anticorpos , Mucosa
18.
J Gen Virol ; 97(4): 855-866, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26769540

RESUMO

The Arenaviridae is a diverse and growing family of viruses that already includes more than 25 distinct species. While some of these viruses have a significant impact on public health, others appear to be non-pathogenic. At present little is known about the host cell responses to infection with different arenaviruses, particularly those found in the New World; however, apoptosis is known to play an important role in controlling infection of many viruses. Here we show that infection with Tacaribe virus (TCRV), which is widely considered the prototype for non-pathogenic arenaviruses, leads to stronger induction of apoptosis than does infection with its human-pathogenic relative Junín virus. TCRV-induced apoptosis occurred in several cell types during late stages of infection and was shown to be caspase-dependent, involving the activation of caspases 3, 7, 8 and 9. Further, UV-inactivated TCRV did not induce apoptosis, indicating that the activation of this process is dependent on active viral replication/transcription. Interestingly, when apoptosis was inhibited, growth of TCRV was not enhanced, indicating that apoptosis does not have a direct negative effect on TCRV infection in vitro. Taken together, our data identify and characterize an important virus-host cell interaction of the prototypic, non-pathogenic arenavirus TCRV, which provides important insight into the growing field of arenavirus research aimed at better understanding the diversity in responses to different arenavirus infections and their functional consequences.


Assuntos
Arenavirus do Novo Mundo/genética , Caspases/genética , Interações Hospedeiro-Patógeno , Macrófagos/virologia , Replicação Viral/genética , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Apoptose/genética , Apoptose/imunologia , Arenavirus do Novo Mundo/efeitos dos fármacos , Arenavirus do Novo Mundo/imunologia , Arenavirus do Novo Mundo/efeitos da radiação , Camptotecina/farmacologia , Caspases/imunologia , Linhagem Celular Tumoral , Chlorocebus aethiops , Regulação da Expressão Gênica , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Vírus Junin/genética , Vírus Junin/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/imunologia , Cultura Primária de Células , Transdução de Sinais , Tubulina (Proteína)/genética , Tubulina (Proteína)/imunologia , Raios Ultravioleta , Células Vero , Replicação Viral/efeitos dos fármacos , Replicação Viral/efeitos da radiação
19.
J Virol ; 90(3): 1290-7, 2016 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26581982

RESUMO

UNLABELLED: Machupo virus (MACV) is the causative agent of Bolivian hemorrhagic fever. Our previous study demonstrated that a MACV strain with a single amino acid substitution (F438I) in the transmembrane domain of glycoprotein is attenuated but genetically unstable in mice. MACV is closely related to Junin virus (JUNV), the causative agent of Argentine hemorrhagic fever. Others and our group have identified the glycoprotein to be the major viral factor determining JUNV attenuation. In this study, we tested the compatibility of the glycoprotein of the Candid#1 live-attenuated vaccine strain of JUNV in MACV replication and its ability to attenuate MACV in vivo. Recombinant MACV with the Candid#1 glycoprotein (rMACV/Cd#1-GPC) exhibited growth properties similar to those of Candid#1 and was genetically stable in vitro. In a mouse model of lethal infection, rMACV/Cd#1-GPC was fully attenuated, more immunogenic than Candid#1, and fully protective against MACV infection. Therefore, the MACV strain expressing the glycoprotein of Candid#1 is safe, genetically stable, and highly protective against MACV infection in a mouse model. IMPORTANCE: Currently, there are no FDA-approved vaccines and/or treatments for Bolivian hemorrhagic fever, which is a fatal human disease caused by MACV. The development of antiviral strategies to combat viral hemorrhagic fevers, including Bolivian hemorrhagic fever, is one of the top priorities of the Implementation Plan of the U.S. Department of Health and Human Services Public Health Emergency Medical Countermeasures Enterprise. Here, we demonstrate for the first time that MACV expressing glycoprotein of Candid#1 is a safe, genetically stable, highly immunogenic, and protective vaccine candidate against Bolivian hemorrhagic fever.


Assuntos
Arenavirus do Novo Mundo/genética , Arenavirus do Novo Mundo/imunologia , Glicoproteínas de Membrana/genética , Recombinação Genética , Proteínas do Envelope Viral/genética , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Estruturas Animais/patologia , Animais , Arenavirus do Novo Mundo/patogenicidade , Peso Corporal , Modelos Animais de Doenças , Instabilidade Genômica , Febre Hemorrágica Americana/patologia , Febre Hemorrágica Americana/prevenção & controle , Histocitoquímica , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Análise de Sequência de DNA , Análise de Sobrevida , Temperatura , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Virais/genética , Virulência
20.
J Virol ; 89(22): 11643-53, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26355089

RESUMO

UNLABELLED: Animal viruses frequently cause zoonotic disease in humans. As these viruses are highly diverse, evaluating the threat that they pose remains a major challenge, and efficient approaches are needed to rapidly predict virus-host compatibility. Here, we develop a combined computational and experimental approach to assess the compatibility of New World arenaviruses, endemic in rodents, with the host TfR1 entry receptors of different potential new host species. Using signatures of positive selection, we identify a small motif on rodent TfR1 that conveys species specificity to the entry of viruses into cells. However, we show that mutations in this region affect the entry of each arenavirus differently. For example, a human single nucleotide polymorphism (SNP) in this region, L212V, makes human TfR1 a weaker receptor for one arenavirus, Machupo virus, but a stronger receptor for two other arenaviruses, Junin and Sabia viruses. Collectively, these findings set the stage for potential evolutionary trade-offs, where natural selection for resistance to one virus may make humans or rodents susceptible to other arenavirus species. Given the complexity of this host-virus interplay, we propose a computational method to predict these interactions, based on homology modeling and computational docking of the virus-receptor protein-protein interaction. We demonstrate the utility of this model for Machupo virus, for which a suitable cocrystal structural template exists. Our model effectively predicts whether the TfR1 receptors of different species will be functional receptors for Machupo virus entry. Approaches such at this could provide a first step toward computationally predicting the "host jumping" potential of a virus into a new host species. IMPORTANCE: We demonstrate how evolutionary trade-offs may exist in the dynamic evolutionary interplay between viruses and their hosts, where natural selection for resistance to one virus could make humans or rodents susceptible to other virus species. We present an algorithm that predicts which species have cell surface receptors that make them susceptible to Machupo virus, based on computational docking of protein structures. Few molecular models exist for predicting the risk of spillover of a particular animal virus into humans or new animal populations. Our results suggest that a combination of evolutionary analysis, structural modeling, and experimental verification may provide an efficient approach for screening and assessing the potential spillover risks of viruses circulating in animal populations.


Assuntos
Antígenos CD/genética , Arenavirus do Novo Mundo/fisiologia , Especificidade de Hospedeiro , Receptores da Transferrina/genética , Receptores Virais/metabolismo , Ligação Viral , Algoritmos , Animais , Linhagem Celular Tumoral , Biologia Computacional/métodos , Resistência à Doença/genética , Cães , Células HEK293 , Humanos , Simulação de Acoplamento Molecular , Receptores da Transferrina/metabolismo , Receptores Virais/ultraestrutura , Internalização do Vírus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA