Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
1.
Viruses ; 14(9)2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-36146793

RESUMEN

Mastomys natalensis is the natural host of various arenaviruses, including the human-pathogenic Lassa virus. Homologous arenaviruses, defined here as those having M. natalensis as a natural host, can establish long-lasting infection in M. natalensis, while these animals rapidly clear arenaviruses having another rodent species as a natural host (heterologous viruses). Little is known about the mechanisms behind the underlying arenavirus-host barriers. The innate immune system, particularly the type I interferon (IFN) response, might play a role. In this study, we developed and validated RT-PCR assays to analyse the expression of M. natalensis interferon-stimulated genes (ISGs). We then used these assays to study if homologous and heterologous viruses induce different IFN responses in M. natalensis cells. Infection experiments were performed with the homologous Lassa and Morogoro viruses and the related but heterologous Mobala virus. Compared to the direct induction with IFN or Poly(I:C), arenaviruses generally induced a weak IFN response. However, the ISG-expression profiles of homologous and heterologous viruses were similar. Our data indicate that, at least in M. natalensis cells, the IFN system is not a major factor in the virus-host barrier for arenaviruses. Our system provides a valuable tool for future in vivo investigation of arenavirus host restrictions at the level of the innate immune response.


Asunto(s)
Infecciones por Arenaviridae , Arenavirus , Interferón Tipo I , Animales , Arenavirus/fisiología , Humanos , Inmunidad Innata , Murinae , Tanzanía
2.
Viruses ; 13(7)2021 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-34203149

RESUMEN

Lujo virus (LUJV), a highly pathogenic arenavirus, was first identified in 2008 in Zambia. To aid the identification of effective therapeutics for LUJV, we developed a recombinant reporter virus system, confirming reporter LUJV comparability with wild-type virus and its utility in high-throughput antiviral screening assays. Using this system, we evaluated compounds with known and unknown efficacy against related arenaviruses, with the aim of identifying LUJV-specific and potential new pan-arenavirus antivirals. We identified six compounds demonstrating robust anti-LUJV activity, including several compounds with previously reported activity against other arenaviruses. These data provide critical evidence for developing broad-spectrum antivirals against high-consequence arenaviruses.


Asunto(s)
Antivirales/farmacología , Arenavirus/efectos de los fármacos , Lujo virus/efectos de los fármacos , Animales , Infecciones por Arenaviridae/tratamiento farmacológico , Infecciones por Arenaviridae/virología , Arenavirus/fisiología , Línea Celular Tumoral , Chlorocebus aethiops , Genoma Viral , Proteínas Fluorescentes Verdes/genética , Humanos , Lujo virus/genética , Lujo virus/fisiología , Pruebas de Sensibilidad Microbiana , Proteínas Recombinantes , Células Vero , Internalización del Virus/efectos de los fármacos
3.
Viruses ; 13(5)2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-34067011

RESUMEN

Natural hosts of most arenaviruses are rodents. The human-pathogenic Lassa virus and several non-pathogenic arenaviruses such as Morogoro virus (MORV) share the same host species, namely Mastomys natalensis (M. natalensis). In this study, we investigated the history of infection and virus transmission within the natural host population. To this end, we infected M. natalensis at different ages with MORV and measured the health status of the animals, virus load in blood and organs, the development of virus-specific antibodies, and the ability of the infected individuals to transmit the virus. To explore the impact of the lack of evolutionary virus-host adaptation, experiments were also conducted with Mobala virus (MOBV), which does not share M. natalensis as a natural host. Animals infected with MORV up to two weeks after birth developed persistent infection, seroconverted and were able to transmit the virus horizontally. Animals older than two weeks at the time of infection rapidly cleared the virus. In contrast, MOBV-infected neonates neither developed persistent infection nor were able to transmit the virus. In conclusion, we demonstrate that MORV is able to develop persistent infection in its natural host, but only after inoculation shortly after birth. A related arenavirus that is not evolutionarily adapted to M. natalensis is not able to establish persistent infection. Persistently infected animals appear to be important to maintain virus transmission within the host population.


Asunto(s)
Infecciones por Arenaviridae/veterinaria , Arenavirus/fisiología , Reservorios de Enfermedades/virología , Murinae/virología , Animales , Animales Recién Nacidos , Arenavirus/clasificación , Especificidad del Huésped , Enfermedades de los Roedores/virología , Replicación Viral
4.
Viruses ; 13(6)2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-34073735

RESUMEN

Several of the human-pathogenic arenaviruses cause hemorrhagic fever and have to be handled under biosafety level 4 conditions, including Lassa virus. Rapid and safe inactivation of specimens containing these viruses is fundamental to enable downstream processing for diagnostics or research under lower biosafety conditions. We established a protocol to test the efficacy of inactivation methods using the low-pathogenic Morogoro arenavirus as surrogate for the related highly pathogenic viruses. As the validation of chemical inactivation methods in cell culture systems is difficult due to cell toxicity of commonly used chemicals, we employed filter devices to remove the chemical and concentrate the virus after inactivation and before inoculation into cell culture. Viral replication in the cells was monitored over 4 weeks by using indirect immunofluorescence and immunofocus assay. The performance of the protocol was verified using published inactivation methods including chemicals and heat. Ten additional methods to inactivate virus in infected cells or cell culture supernatant were validated and shown to reduce virus titers to undetectable levels. In summary, we provide a robust protocol for the validation of chemical and physical inactivation of arenaviruses in cell culture, which can be readily adapted to different inactivation methods and specimen matrices.


Asunto(s)
Arenavirus/fisiología , Desinfección/métodos , Inactivación de Virus , Animales , Técnicas de Cultivo de Célula , Línea Celular , Células Cultivadas , Chlorocebus aethiops , Desinfección/normas , Humanos , Reproducibilidad de los Resultados , Manejo de Especímenes/métodos , Células Vero
5.
Cell Struct Funct ; 45(2): 155-163, 2020 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-33191384

RESUMEN

The smallest arenavirus gene product, Z protein, plays critical roles in the virus life cycle. Z is the major driving force of budding and particle production because of a unique property that defines self-assembly. In addition to the roles in budding, Z also participates in the suppression of type I interferon production to evade host antiviral immunity. Therefore, Z and its assembled form are an attractive drug target for arenaviral hemorrhagic fever, such as Lassa fever. Here, we developed a biosensor that enabled the evaluation of the prototype arenavirus, lymphocytic choriomeningitis virus (LCMV), Z assembly using the principle of Förster resonance energy transfer (FRET). This FRET biosensor consisted of three tandem Z that were sandwiched between super-enhanced cyan-emitting fluorescent protein and variant of a yellow-emitting mutant of green fluorescent protein so that Z-Z intermolecular binding via the really interesting new gene finger domain increased the emission ratio. To identify novel anti-arenavirus compounds, the FRET biosensor was employed to screen the PathogenBox400 for inhibitors of Z assembly in a 96-well plate format. The assay performed well, with a Z'-factor of 0.89, and identified two compounds that decreased the emission ratio of the FRET biosensor in a dose-dependent manner. Of them, the compound, 5,6,7,8-tetrahydro-7-(benzyl)-pyrido[4',3':4,5]thieno[2,3-d]pyrimidin-2,4-diamine, was found to significantly inhibit LCMV propagation in infected cells. Thereby, the present study demonstrated that a novel FRET biosensor incorporating Z assembly built on FRET and named Zabton, was a valuable screening tool to identify anti-arenavirus compounds in the context of inhibition of Z assembly.Key words: Arenavirus, Förster resonance energy transfer, anti-viral drugs, Z protein.


Asunto(s)
Antivirales , Arenavirus/fisiología , Técnicas Biosensibles , Transferencia Resonante de Energía de Fluorescencia , Proteínas Virales/metabolismo , Ensamble de Virus/efectos de los fármacos , Antivirales/química , Antivirales/farmacología , Evaluación Preclínica de Medicamentos , Células HEK293 , Células HeLa , Humanos
6.
J Exp Clin Cancer Res ; 39(1): 34, 2020 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-32041643

RESUMEN

Odontogenic tumors (OT) are considered rare events and their epidemiologic data are scarce and under-estimated in developing countries because there is no systematic collection of clinical features including histological analyses of the tissue samples. Furthermore, there is an underestimation of the disease relevance and affected people are often marginalized in spite of severe functional impairment of aero-digestive tract. Etiology of OT in humans is still unknown and it represents an important therapeutic and diagnostic challenge.Lassa fever is an acute viral haemorrhagic illness caused by Lassa virus, a member of the arenavirus family of viruses. The disease is endemic in the rodent population in West-East Africa. Humans usually become infected with Lassa virus through exposure to the food or household items contaminated with urine or feces of infected rats. It is also reported person-to-person infections. About 80% of people infected by Lassa virus have no symptoms but the virus establishes a life-long persistent infection.The present commentary significance is to start, for the first time ever, a systematic collection of clinical features and tissue sample collection at the St. Mary's Hospital in Lacor (Gulu) North Uganda where the considered pathologies have an important frequency. The systematic collection will allow to corroborate the possible association between arenaviruses infection and pathogenesis of odontogenic tumors in humans.


Asunto(s)
Infecciones por Arenaviridae/complicaciones , Infecciones por Arenaviridae/virología , Arenavirus/fisiología , Transformación Celular Viral , Tumores Odontogénicos/etiología , Infecciones por Arenaviridae/epidemiología , Biopsia , Susceptibilidad a Enfermedades , Humanos , Fiebre de Lassa/complicaciones , Fiebre de Lassa/virología , Virus Lassa , Tumores Odontogénicos/diagnóstico , Tumores Odontogénicos/epidemiología , Uganda
7.
Viruses ; 11(3)2019 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-30909570

RESUMEN

Lassa virus (LASV) and Mopeia virus (MOPV) are two closely related Old-World mammarenaviruses. LASV causes severe hemorrhagic fever with high mortality in humans, whereas no case of MOPV infection has been reported. Comparing MOPV and LASV is a powerful strategy to unravel pathogenic mechanisms that occur during the course of pathogenic arenavirus infection. We used a yeast two-hybrid approach to identify cell partners of MOPV and LASV Z matrix protein in which two autophagy adaptors were identified, NDP52 and TAX1BP1. Autophagy has emerged as an important cellular defense mechanism against viral infections but its role during arenavirus infection has not been shown. Here, we demonstrate that autophagy is transiently induced by MOPV, but not LASV, in infected cells two days after infection. Impairment of the early steps of autophagy significantly decreased the production of MOPV and LASV infectious particles, whereas a blockade of the degradative steps impaired only MOPV infectious particle production. Our study provides insights into the role played by autophagy during MOPV and LASV infection and suggests that this process could partially explain their different pathogenicity.


Asunto(s)
Arenavirus/fisiología , Autofagia , Virus Lassa/fisiología , Animales , Arenavirus/patogenicidad , Chlorocebus aethiops , Células HEK293 , Células HeLa , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Virus Lassa/patogenicidad , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Técnicas del Sistema de Dos Híbridos , Células Vero
8.
Antiviral Res ; 160: 87-93, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30339847

RESUMEN

The family Arenaviridae consists of numerous enveloped RNA viruses with ambisense coding strategies. Eight arenaviruses, including Lassa virus, are known to cause severe and fatal viral hemorrhagic fever (VHF) in humans, yet vaccines and treatments for disease caused by arenaviruses are very limited. In this study, we screened a natural product library consisting of 131 compounds and identified tangeretin, a polymethoxylated flavone widely present in citrus fruit peels, as a Lassa virus entry inhibitor that blocks viral fusion. Further analyses demonstrated the efficacy of tangeretin against seven other VHF-causing arenaviruses, suggesting that this compound, which has a history of medical usage, could be used to develop an effective therapeutic to treat infection and disease caused by Lassa virus and related viruses.


Asunto(s)
Antivirales/farmacología , Arenavirus/efectos de los fármacos , Citrus/química , Flavonas/farmacología , Internalización del Virus/efectos de los fármacos , Antivirales/aislamiento & purificación , Arenavirus/fisiología , Productos Biológicos/aislamiento & purificación , Productos Biológicos/farmacología , Evaluación Preclínica de Medicamentos , Flavonas/aislamiento & purificación
9.
Parasit Vectors ; 11(1): 416, 2018 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-30005641

RESUMEN

BACKGROUND: Lassa fever, killing thousands of people annually, is the most reported viral zoonotic disease in Nigeria. Recently, different rodent species carrying diverse lineages of the Lassa virus (LASV) in addition to a novel Mobala-like genetic sequence were detected within the country. Here, screening 906 small mammal specimens from 11 localities for IgG antibodies and incorporating previous PCR detection data involving the same populations, we further describe arenavirus prevalence across Nigeria in relation to host species and geographical location. METHODS: Small mammals were trapped during the period 2011-2015 according to geographical location (endemic and non-endemic zones for Lassa fever), season (rainy and dry seasons between 2011 and 2012 for certain localities) and habitat (indoors, peridomestic settings and sylvatic vegetation). Identification of animal specimens from genera such as Mastomys and Mus (Nannomys) was assisted by DNA sequencing. Small mammals were tested for LASV IgG antibody using an indirect immunofluorescence assay (IFA). RESULTS: Small mammals were infected in both the endemic and non-endemic zones for Lassa fever, with a wider range of species IgG-positive (n = 8) than those which had been previously detected to be PCR-positive (n = 3). IgG-positive species, according to number of infected individuals, were Mastomys natalensis (n = 40), Mastomys erythroleucus (n = 15), Praomys daltoni (n = 6), Mus baoulei (n = 5), Rattus rattus (n = 2), Crocidura spp. (n = 2), Mus minutoides (n = 1) and Praomys misonnei (n = 1). Multimammate mice (Mastomys natalensis and M. erythroleucus) were the most ubiquitously infected, with animals testing positive by either PCR or IgG in 7 out of the 11 localities sampled. IgG prevalence in M. natalensis ranged from 1% in Abagboro, 17-36 % in Eguare Egoro, Ekpoma and Ngel Nyaki, up to 52 % in Mayo Ranewo. Prevalence according to locality, season and age was not, however, statistically significant for M. natalensis in Eguare Egoro and Ekpoma, localities that were sampled longitudinally. CONCLUSIONS: Overall, our study demonstrates that arenavirus occurrence is probably more widely distributed geographically and in extent of host taxa than is currently realized. This expanded scope should be taken into consideration in Lassa fever control efforts. Further sampling should also be carried out to isolate and characterize potential arenaviruses present in small mammal populations we found to be seropositive.


Asunto(s)
Anticuerpos Antivirales/sangre , Infecciones por Arenaviridae/sangre , Infecciones por Arenaviridae/veterinaria , Arenavirus/fisiología , Enfermedades de los Roedores/sangre , Enfermedades de los Roedores/epidemiología , Animales , Infecciones por Arenaviridae/epidemiología , Infecciones por Arenaviridae/virología , Arenavirus/inmunología , Reservorios de Enfermedades/virología , Eulipotyphla/virología , Geografía , Virus Lassa/inmunología , Virus Lassa/fisiología , Ratones , Nigeria/epidemiología , Prevalencia , ARN Viral/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Enfermedades de los Roedores/virología , Roedores/virología , Estudios Seroepidemiológicos
10.
J Mol Biol ; 430(13): 1839-1852, 2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29705070

RESUMEN

Viral hemorrhagic fevers caused by emerging RNA viruses of the Arenavirus family are among the most devastating human diseases. Climate change, global trade, and increasing urbanization promote the emergence and re-emergence of these human pathogenic viruses. Emerging pathogenic arenaviruses are of zoonotic origin and reservoir-to-human transmission is crucial for spillover into human populations. Host cell attachment and entry are the first and most fundamental steps of every virus infection and represent major barriers for zoonotic transmission. During host cell invasion, viruses critically depend on cellular factors, including receptors, co-receptors, and regulatory proteins of endocytosis. An in-depth understanding of the complex interaction of a virus with cellular factors implicated in host cell entry is therefore crucial to predict the risk of zoonotic transmission, define the tissue tropism, and assess disease potential. Over the past years, investigation of the molecular and cellular mechanisms underlying host cell invasion of human pathogenic arenaviruses uncovered remarkable viral strategies and provided novel insights into viral adaptation and virus-host co-evolution that will be covered in the present review.


Asunto(s)
Infecciones por Arenaviridae/virología , Arenavirus/fisiología , Zoonosis/virología , Animales , Infecciones por Arenaviridae/transmisión , Arenavirus/genética , Interacciones Microbiota-Huesped , Humanos , Tropismo Viral , Acoplamiento Viral , Internalización del Virus , Zoonosis/transmisión
11.
Viruses ; 10(2)2018 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-29439402

RESUMEN

Mammarenavirusesare single-stranded RNA viruses with a bisegmented ambisense genome. Ingestion has been shown as a natural route of transmission for both Lassa virus (LASV) and Lymphocytic choriomeningitis virus (LCMV). Due to the mechanism of transmission, epithelial tissues are among the first host cells to come in contact with the viruses, and as such they potentially play a role in spread of virus to naïve hosts. The role of the intestinal epithelia during arenavirus infection remains to be uncharacterized. We have utilized a well-established cell culture model, Caco-2, to investigate the role of intestinal epithelia during intragastric infection. We found that LCMV-Armstrong, LCMV-WE, and Mopeia (MOPV) release infectious progeny via similar patterns. However, the reassortant virus, ML-29, containing the L segment of MOPV and S segment of LASV, exhibits a unique pattern of viral release relative to LCMV and MOPV. Furthermore, we have determined attachment efficacy to Caco-2 cells is potentially responsible for observed replication kinetics of these viruses in a polarized Caco-2 cell model. Collectively, our data shows that viral dissemination and interaction with intestinal epithelia may be host, tissue, and viral specific.


Asunto(s)
Arenavirus/fisiología , Mucosa Intestinal/virología , Animales , Infecciones por Arenaviridae/virología , Células CACO-2 , Línea Celular , Células Cultivadas , Chlorocebus aethiops , Humanos , Virus Reordenados , Células Vero , Internalización del Virus , Replicación Viral
12.
ACS Infect Dis ; 4(5): 815-824, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29405696

RESUMEN

Several arenaviruses cause hemorrhagic fever (HF) disease in humans and represent important public health problems in their endemic regions. In addition, evidence indicates that the worldwide-distributed prototypic arenavirus lymphocytic choriomeningitis virus is a neglected human pathogen of clinical significance. There are no licensed arenavirus vaccines, and current antiarenavirus therapy is limited to an off-label use of ribavirin that is only partially effective. Therefore, there is an unmet need for novel therapeutics to combat human pathogenic arenaviruses, a task that will be facilitated by the identification of compounds with antiarenaviral activity that could serve as probes to identify arenavirus-host interactions suitable for targeting, as well as lead compounds to develop future antiarenaviral drugs. Screening of a combinatorial library of Krönhke pyridines identified compound KP-146 [(5-(5-(2,3-dihydrobenzo[ b][1,4] dioxin-6-yl)-4'-methoxy-[1,1'-biphenyl]-3-yl)thiophene-2-carboxamide] as having strong anti-lymphocytic choriomeningitis virus (LCMV) activity in cultured cells. KP-146 did not inhibit LCMV cell entry but rather interfered with the activity of the LCMV ribonucleoprotein (vRNP) responsible for directing virus RNA replication and gene transcription, as well as with the budding process mediated by the LCMV matrix Z protein. LCMV variants with increased resistance to KP-146 did not emerge after serial passages in the presence of KP-146. Our findings support the consideration of Kröhnke pyridine scaffold as a valuable source to identify compounds that could serve as tools to dissect arenavirus-host interactions, as well as lead candidate structures to develop antiarenaviral drugs.


Asunto(s)
Antivirales/farmacología , Arenavirus/efectos de los fármacos , Minería de Datos , Descubrimiento de Drogas , Piridinas/farmacología , Bibliotecas de Moléculas Pequeñas , Animales , Antivirales/síntesis química , Antivirales/química , Infecciones por Arenaviridae/tratamiento farmacológico , Infecciones por Arenaviridae/virología , Arenavirus/fisiología , Línea Celular , Técnicas de Química Sintética , Chlorocebus aethiops , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos , Virus de la Coriomeningitis Linfocítica/efectos de los fármacos , Piridinas/síntesis química , Piridinas/química , Células Vero , Replicación Viral/efectos de los fármacos
13.
J Gen Virol ; 99(2): 187-193, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29393022

RESUMEN

We report the development of recombinant New World (Junín; JUNV) and Old World (lymphocytic choriomeningitis virus; LCMV) mammarenaviruses that encode an HA-tagged matrix protein (Z). These viruses permit the robust affinity purification of Z from infected cells or virions, as well as the detection of Z by immunofluorescent microscopy. Importantly, the HA-tagged viruses grow with wild-type kinetics in a multi-cycle growth assay. Using these viruses, we report a novel description of JUNV Z localization in infected cells, as well as the first description of colocalization between LCMV Z and the GTPase Rab5c. This latter result, when combined with our previous findings that LCMV genome and glycoprotein also colocalize with Rab5c, suggest that LCMV may target Rab5c-positive membranes for preassembly of virus particles prior to budding. The recombinant viruses reported here will provide the field with new tools to better study Z protein functionality and identify key Z protein interactions with host machinery.


Asunto(s)
Arenavirus/fisiología , Proteínas Portadoras/metabolismo , Epítopos/inmunología , GTP Fosfohidrolasas/metabolismo , Interacciones Huésped-Patógeno , Virus de la Coriomeningitis Linfocítica/fisiología , Células A549 , Arenavirus/inmunología , Proteínas Portadoras/genética , Endosomas/metabolismo , Endosomas/virología , GTP Fosfohidrolasas/genética , Genes Reporteros , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/virología , Péptidos y Proteínas de Señalización Intracelular , Virus de la Coriomeningitis Linfocítica/inmunología , Microscopía Fluorescente , Ensamble de Virus
14.
Uirusu ; 68(1): 51-62, 2018.
Artículo en Japonés | MEDLINE | ID: mdl-31105135

RESUMEN

Arenavirus is a genetic term for viruses belonging to the family Arenaviridae and is presented from lymphocytic choriomeningitis virus (LCMV), which shows almost no pathogenicity to humans, to Lassa virus, Junin virus, Machupo virus, Chapare virus, Lujo virus, Sabia virus, and Guanarito virus, which shows high pathogenicity to humans. These viruses except for LCMV are risk group 4 pathogens specified by World Health Organization. Based on this designation, it is designated as Class I pathogens in Japan. Although there have been no reports excluding one imported case of the Lassa fever patient, it is not surprising whenever imported cases occur in our country. Considering the disease severity and mortality rate, it is an urgent matter to develop vaccines and therapeutic drugs in endemic areas, and maintenances of these are also important in countries other than endemic areas. However, basic research on highly pathogenic arenavirus infections and development of therapeutic drugs are not easily progressed, because handling in highly safe research facilities is indispensable. In this article, we will outline the current knowledge from the recent basic research on arenavirus to the development situation of antivirals against arenaviruses.


Asunto(s)
Antivirales , Infecciones por Arenaviridae/tratamiento farmacológico , Infecciones por Arenaviridae/virología , Arenavirus/clasificación , Arenavirus/patogenicidad , Descubrimiento de Drogas , África Occidental/epidemiología , Infecciones por Arenaviridae/epidemiología , Infecciones por Arenaviridae/prevención & control , Arenavirus/genética , Arenavirus/fisiología , Brotes de Enfermedades , Descubrimiento de Drogas/tendencias , Genoma Viral/genética , Humanos , Investigación/tendencias , Transcripción Genética , Vacunas Virales , Virión
15.
Methods Mol Biol ; 1604: 371-392, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28986849

RESUMEN

Among the members of the Arenaviridae family, Junín virus and Lassa virus represent important human health threats generating annual outbreaks of severe human hemorrhagic fever (HF) in endemic areas of Argentina and Western Africa, respectively. Given the lack of a specific and safe chemotherapy, the search for effective antiviral compounds is a continuous demanding effort. During the last two decades, academic research studies originated important results identifying novel molecules to be considered for further in vivo characterization. This chapter summarizes experimental in vitro approaches used to determine the possible mechanism of action of these antiviral agents.


Asunto(s)
Antivirales/farmacología , Arenavirus/efectos de los fármacos , Arenavirus/fisiología , Virus Hantaan/efectos de los fármacos , Virus Hantaan/fisiología
16.
Microbes Infect ; 20(2): 65-80, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29081359

RESUMEN

In recent years there has been a greatly increased interest in the interactions of arenaviruses with the apoptotic machinery, and particularly the extent to which these interactions may be an important contributor to pathogenesis. Here we summarize the current state of our knowledge on this subject and address the potential for interplay with other immunological mechanisms known to be regulated by these viruses. We also compare and contrast what is known for arenavirus-induced apoptosis with observations from other segmented hemorrhagic fever viruses.


Asunto(s)
Apoptosis , Infecciones por Arenaviridae/patología , Infecciones por Arenaviridae/virología , Arenavirus/fisiología , Evasión Inmune , Animales , Infecciones por Arenaviridae/inmunología , Humanos , Inmunidad Celular , Nucleoproteínas/metabolismo , Transducción de Señal , Replicación Viral
17.
PLoS One ; 12(9): e0185308, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28953976

RESUMEN

Tacaribe virus (TCRV) was isolated in the 1950s from artibeus bats captured on the island of Trinidad. The initial characterization of TCRV suggested that artibeus bats were natural reservoir hosts. However, nearly 60 years later experimental infections of Jamaican fruit bats (Artibeus jamaicensis) resulted in fatal disease or clearance, suggesting artibeus bats may not be a reservoir host. To further evaluate the TCRV reservoir host status of artibeus bats, we captured bats of six species in Trinidad for evidence of infection. Bats of all four fruigivorous species captured had antibodies to TCRV nucleocapsid, whereas none of the insectivore or nectarivore species did. Many flat-faced fruit-eating bats (A. planirostris) and great fruit-eating bats (A. literatus) were seropositive by ELISA and western blot to TCRV nucleocapsid antigen, as were two of four Seba's fruit bats (Carollia perspicillata) and two of three yellow-shouldered fruit bats (Sturnira lilium). Serum neutralization tests failed to detect neutralizing antibodies to TCRV from these bats. TCRV RNA was not detected in lung tissues or lung homogenates inoculated onto Vero cells. These data indicate that TCRV or a similar arenavirus continues to circulate among fruit bats of Trinidad but there was no evidence of persistent infection, suggesting artibeus bats are not reservoir hosts.


Asunto(s)
Arenavirus/fisiología , Quirópteros/sangre , Quirópteros/virología , Pruebas Serológicas , Animales , Arenavirus/aislamiento & purificación , Geografía , Estudios Seroepidemiológicos , Trinidad y Tobago
18.
Nat Commun ; 8: 14447, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28248314

RESUMEN

Immune-mediated effector molecules can limit cancer growth, but lack of sustained immune activation in the tumour microenvironment restricts antitumour immunity. New therapeutic approaches that induce a strong and prolonged immune activation would represent a major immunotherapeutic advance. Here we show that the arenaviruses lymphocytic choriomeningitis virus (LCMV) and the clinically used Junin virus vaccine (Candid#1) preferentially replicate in tumour cells in a variety of murine and human cancer models. Viral replication leads to prolonged local immune activation, rapid regression of localized and metastatic cancers, and long-term disease control. Mechanistically, LCMV induces antitumour immunity, which depends on the recruitment of interferon-producing Ly6C+ monocytes and additionally enhances tumour-specific CD8+ T cells. In comparison with other clinically evaluated oncolytic viruses and to PD-1 blockade, LCMV treatment shows promising antitumoural benefits. In conclusion, therapeutically administered arenavirus replicates in cancer cells and induces tumour regression by enhancing local immune responses.


Asunto(s)
Arenavirus/fisiología , Vigilancia Inmunológica , Interferón Tipo I/metabolismo , Neoplasias/inmunología , Neoplasias/virología , Replicación Viral/fisiología , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Humanos , Activación de Linfocitos/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Ratones Endogámicos C57BL , Monocitos/metabolismo , Neoplasias/irrigación sanguínea , Virus Oncolíticos/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo
19.
PLoS Pathog ; 13(1): e1006073, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28076397

RESUMEN

Many emerging infections are RNA virus spillovers from animal reservoirs. Reservoir identification is necessary for predicting the geographic extent of infection risk, but rarely are taxonomic levels below the animal species considered as reservoir, and only key circumstances in nature and methodology allow intrinsic virus-host associations to be distinguished from simple geographic (co-)isolation. We sampled and genetically characterized in detail a contact zone of two subtaxa of the rodent Mastomys natalensis in Tanzania. We find two distinct arenaviruses, Gairo and Morogoro virus, each spatially confined to a single M. natalensis subtaxon, only co-occurring at the contact zone's centre. Inter-subtaxon hybridization at this centre and a continuum of quality habitat for M. natalensis show that both viruses have the ecological opportunity to spread into the other substaxon's range, but do not, strongly suggesting host-intrinsic barriers. Such barriers could explain why human cases of another M. natalensis-borne arenavirus, Lassa virus, are limited to West Africa.


Asunto(s)
Arenavirus/clasificación , Arenavirus/metabolismo , Reservorios de Enfermedades/virología , Murinae/virología , Enfermedades de los Roedores/virología , Animales , Arenavirus/fisiología , Humanos , Fiebre de Lassa/virología , Virus Lassa/fisiología , Filogeografía , Especificidad de la Especie , Tanzanía
20.
Cell Mol Immunol ; 14(1): 90-107, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27569560

RESUMEN

Neonates are at increased risk of viral encephalopathies that can result in neurological dysfunction, seizures, permanent disability and even death. The neurological damage results from the combined effect of the virus and the immune response it elicits, thus finding tools to facilitate viral clearance from central nervous system (CNS) while minimizing neuron damage remains a critical challenge. Neonatal mice inoculated intraperitoneally with Tacaribe virus (TCRV) develop seizures, hindlimb paralysis and death within 15 days of inoculation. TCRV localizes to the CNS within days of challenge, primarily infecting astrocytes in the cerebellum and brain stem. We show that infection leads to inflammation, T cell and monocyte infiltration into the cerebellar parenchyma, apoptosis of astrocytes, neuronal degeneration and loss of Purkinje cells. Infiltrating antigen-specific T cells fail to clear the virus but drive the disease, as T-cell-deficient CD3ɛ KO mice survive TCRV infection with minimal inflammation or clinical manifestations despite no difference in CNS viral loads in comparison with T-cell sufficient mice. CD8+ T cells drive the pathology, which even in the absence of CD4+ T-cell help, infiltrate the parenchyma and mediate the apoptotic loss of cerebellar astrocytes, neurodegeneration and loss of Purkinje cells resulting in loss of balance, paralysis and death. CD4+ T cells are also pathogenic inducing gliosis and inflammation in the cerebellum and cerebrum that are associated with wasting and death several weeks after CD4+ T-cell transfer. These data demonstrate distinct pathogenic effects of CD4+ and CD8+ T cells and identify them as possible therapeutic targets.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/virología , Arenavirus/fisiología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Meningoencefalitis/inmunología , Meningoencefalitis/virología , Animales , Animales Recién Nacidos , Apoptosis , Infecciones por Arenaviridae/patología , Astrocitos/patología , Sistema Nervioso Central/patología , Sistema Nervioso Central/virología , Gliosis/patología , Meningoencefalitis/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/patología , Degeneración Nerviosa/patología , Neuronas/patología , Células de Purkinje/patología , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...