Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Viruses ; 14(10)2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36298848

RESUMEN

Immunological memory to a previously encountered pathogen can influence the outcome of a sequential infection, which is called heterologous immunity. Lymphocytic choriomeningitis virus (LCMV) immune mice develop a NP205-specific T cell response that is cross-reactive to Pichinde virus infection (PICV). So far, limited data are available if cross-reactive T cell responses appear also during chronic infections with exhausted T cell responses. Exhaustion in chronic viral infections can be treated with checkpoint inhibitors, which might affect heterologous outcomes unexpectedly. The aim of this study was to investigate the cross-reactive immune response in chronic LCMV clone 13 (LCMVcl13) infection during primary PICV infection at phenotypic, functional, and T cell receptor (TCR) level. Moreover, the influence of checkpoint inhibitor therapy with αPD-L1 was investigated. Cross-reactive NP205-specific responses were present and functional in the chronic environment. Additionally, chronically infected mice were also protected from PICV mediated weight loss compared to naive PICV mice. An altered phenotype of NP205-specific T cells was detectable, but no major differences in the clonality and diversity of their TCR repertoire were observed. Checkpoint inhibitor treatment with αPD-L1 did alter chronic LCMV infection but had no major effect on heterologous immunity to PICV. Our study demonstrated that cross-reactive CD8+ T cells also exist in the setting of chronic infection, indicating a clinically relevant role of cross-reactive T cells in chronic infections.


Asunto(s)
Infecciones por Arenaviridae , Coriomeningitis Linfocítica , Virus Pichinde , Ratones , Animales , Virus de la Coriomeningitis Linfocítica , Linfocitos T CD8-positivos , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/genética
2.
J Virol ; 96(16): e0075422, 2022 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-35913216

RESUMEN

Lassa virus (LASV) is a mammarenavirus that can cause lethal Lassa fever disease with no FDA-approved vaccine and limited treatment options. Fatal LASV infections are associated with innate immune suppression. We have previously shown that the small matrix Z protein of LASV, but not of a nonpathogenic arenavirus Pichinde virus (PICV), can inhibit the cellular RIG-I-like receptors (RLRs), but its biological significance has not been evaluated in an infectious virus due to the multiple essential functions of the Z protein required for the viral life cycle. In this study, we developed a stable HeLa cell line (HeLa-iRIGN) that could be rapidly and robustly induced by doxycycline (Dox) treatment to express RIG-I N-terminal effector, with concomitant production of type I interferons (IFN-Is). We also generated recombinant tri-segmented PICVs, rP18tri-LZ, and rP18tri-PZ, which encode LASV Z and PICV Z, respectively, as an extra mScarlet fusion protein that is nonessential for the viral life cycle. Upon infection, rP18tri-LZ consistently expressed viral genes at a higher level than rP18tri-PZ. rP18tri-LZ also showed a higher level of a viral infection than rP18tri-PZ did in HeLa-iRIGN cells, especially upon Dox induction. The heterologous Z gene did not alter viral growth in Vero and A549 cells by growth curve analysis, while LASV Z strongly increased and prolonged viral gene expression, especially in IFN-competent A549 cells. Our study provides important insights into the biological role of LASV Z-mediated RIG-I inhibition and implicates LASV Z as a potential virulence factor. IMPORTANCE Lassa virus (LASV) can cause lethal hemorrhagic fever disease in humans but other arenaviruses, such as Pichinde virus (PICV), do not cause obvious disease. We have previously shown that the Z protein of LASV but not of PICV can inhibit RIG-I, a cytosolic innate immune receptor. In this study, we developed a stable HeLa cell line that can be induced to express the RIG-I N-terminal effector domain, which allows for timely control of RIG-I activation. We also generated recombinant PICVs encoding LASV Z or PICV Z as an extra gene that is nonessential for the viral life cycle. Compared to PICV Z, LASV Z could increase viral gene expression and viral infection in an infectious arenavirus system, especially when RIG-I signaling is activated. Our study presented a convenient cell system to characterize RIG-I signaling and its antagonists and revealed LASV Z as a possible virulence factor and a potential antiviral target.


Asunto(s)
Virus Lassa , Proteínas Virales/metabolismo , Células HeLa , Humanos , Fiebre de Lassa/virología , Virus Lassa/patogenicidad , Virus Lassa/fisiología , Virus Pichinde/genética , Factores de Virulencia
3.
Virulence ; 13(1): 1049-1061, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35758052

RESUMEN

Lassa fever (LF) is a neglected tropical disease that is caused by Lassa virus (LASV), a human hemorrhagic fever-causing mammarenavirus. A notable sequela of LF is sensorineural hearing loss (SNHL) that can develop in about 33% of the patients. Animal models of LF-associated SNHL have been limited in size and scope because LASV is a biosafety level 4 (BSL4) pathogen that requires its handling in a high biocontainment laboratory. In this report, we describe the development of an alternative arenavirus hearing loss model by infecting outbred Hartley guinea pigs with a virulent strain (rP18) of the Pichinde virus (PICV), which is a guinea pig-adapted mammarenavirus that has been used as a surrogate model of mammarenaviral hemorrhagic fevers in a conventional (BSL2) laboratory. By measuring auditory brainstem response (ABR) throughout the course of the virulent rP18 PICV infection, we noticed that some of the animals experienced an acute but transient level of hearing loss. Cochleae of hearing-impaired animals, but not of controls, had demonstrable viral RNA by quantitative RT-PCR, indicating the presence of virus in the affected inner ear with no overt histopathological changes. In contrast, neither the outbred Hartley guinea pigs infected with a known avirulent strain (rP2) of PICV nor those that were mock-infected showed any evidence of hearing loss or viral infection of the inner ear. This is the first report of an immunocompetent small animal model of mammarenavirus-induced hearing loss that can be used to evaluate potential therapeutics against virus-induced hearing impairment under a conventional laboratory setting.


Asunto(s)
Pérdida Auditiva , Fiebre de Lassa , Virus Pichinde , Animales , Modelos Animales de Enfermedad , Fiebre , Cobayas , Pérdida Auditiva/complicaciones , Humanos , Virus Lassa , Virus Pichinde/genética
4.
Front Immunol ; 12: 801811, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925387

RESUMEN

RIG-I and MDA5 are major cytoplasmic innate-immune sensor proteins that recognize aberrant double-stranded RNAs generated during virus infection to activate type 1 interferon (IFN-I) and IFN-stimulated gene (ISG) expressions to control virus infection. The roles of RIG-I and MDA5 in controlling replication of Pichinde virus (PICV), a mammarenavirus, in mice have not been examined. Here, we showed that MDA5 single knockout (SKO) and RIG-I/MDA5 double knockout (DKO) mice are highly susceptible to PICV infection as evidenced by their significant reduction in body weights during the course of the infection, validating the important roles of these innate-immune sensor proteins in controlling PICV infection. Compared to the wildtype mice, SKO and DKO mice infected with PICV had significantly higher virus titers and lower IFN-I expressions early in the infection but appeared to exhibit a late and heightened level of adaptive immune responses to clear the infection. When a recombinant rPICV mutant virus (rPICV-NPmut) that lacks the ability to suppress IFN-I was used to infect mice, as expected, there were heightened levels of IFN-I and ISG expressions in the wild-type mice, whereas infected SKO and DKO mice showed delayed mouse growth kinetics and relatively low, delayed, and transient levels of innate and adaptive immune responses to this viral infection. Taken together, our data suggest that PICV infection triggers activation of immune sensors that include but might not be necessarily limited to RIG-I and MDA5 to stimulate effective innate and adaptive immune responses to control virus infection in mice.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Helicasa Inducida por Interferón IFIH1/inmunología , Receptores de Superficie Celular/inmunología , Animales , Ratones , Ratones Noqueados , Virus Pichinde/inmunología , Replicación Viral/inmunología
5.
Cell Rep Med ; 2(3): 100209, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33763654

RESUMEN

Therapeutic vaccination regimens inducing clinically effective tumor-specific CD8+ T lymphocyte (CTL) responses are an unmet medical need. We engineer two distantly related arenaviruses, Pichinde virus and lymphocytic choriomeningitis virus, for therapeutic cancer vaccination. In mice, life-replicating vector formats of these two viruses delivering a self-antigen in a heterologous prime-boost regimen induce tumor-specific CTL responses up to 50% of the circulating CD8 T cell pool. This CTL attack eliminates established solid tumors in a significant proportion of animals, accompanied by protection against tumor rechallenge. The magnitude of CTL responses is alarmin driven and requires combining two genealogically distantly related arenaviruses. Vector-neutralizing antibodies do not inhibit booster immunizations by the same vector or by closely related vectors. Rather, CTL immunodominance hierarchies favor vector backbone-targeted responses at the expense of self-reactive CTLs. These findings establish an arenavirus-based immunotherapy regimen that allows reshuffling of immunodominance hierarchies and breaking self-directed tolerance for efficient tumor control.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Inmunoterapia/métodos , Virus de la Coriomeningitis Linfocítica/inmunología , Mastocitoma/terapia , Virus Pichinde/inmunología , Linfocitos T Citotóxicos/inmunología , Alarminas/genética , Alarminas/inmunología , Animales , Anticuerpos Neutralizantes/farmacología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Femenino , Expresión Génica , Ingeniería Genética/métodos , Vectores Genéticos/clasificación , Vectores Genéticos/inmunología , Cobayas , Inmunización Secundaria , Virus de la Coriomeningitis Linfocítica/clasificación , Virus de la Coriomeningitis Linfocítica/genética , Mastocitoma/genética , Mastocitoma/inmunología , Mastocitoma/mortalidad , Ratones , Ratones Endogámicos C57BL , Filogenia , Virus Pichinde/clasificación , Virus Pichinde/genética , Autotolerancia , Análisis de Supervivencia , Vacunación/métodos
6.
Virulence ; 11(1): 1131-1141, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32799623

RESUMEN

Arenaviruses, such as Lassa virus (LASV), can cause severe and fatal hemorrhagic fevers (e.g., Lassa fever, LF) in humans with no vaccines or therapeutics. Research on arenavirus-induced hemorrhagic fevers (AHFs) has been hampered by the highly virulent nature of these viral pathogens, which require high biocontainment laboratory, and the lack of an immune-competent small animal model that can recapitulate AHF disease and pathological features. Guinea pig infected with Pichinde virus (PICV), an arenavirus that does not cause disease in humans, has been established as a convenient surrogate animal model for AHFs as it can be handled in a conventional laboratory. The PICV strain P18, derived from sequential passaging of the virus 18 times in strain 13 inbred guinea pigs, causes severe febrile illness in guinea pigs that is reminiscent of lethal LF in humans. As inbred guinea pigs are not readily available and are difficult to maintain, outbred Hartley guinea pigs have been used but they show a high degree of disease heterogeneity upon virulent P18 PICV infection. Here, we describe an improved outbred guinea-pig infection model using recombinant rP18 PICV generated by reverse genetics technique followed by plaque purification, which consistently shows >90% mortality and virulent infection. Comprehensive virological, histopathological, and immunohistochemical analyses of the rP18-virus infected animals show similar features of human LASV infection. Our data demonstrate that this improved animal model can serve as a safe, affordable, and convenient surrogate small animal model for studying human LF pathogenesis and for evaluating efficacy of preventative or therapeutic approaches.


Asunto(s)
Modelos Animales de Enfermedad , Cobayas , Fiebre de Lassa/patología , Fiebre de Lassa/virología , Virus Pichinde/genética , Virus Pichinde/patogenicidad , Animales , Animales no Consanguíneos , Infecciones por Arenaviridae/virología , Línea Celular , Chlorocebus aethiops , Cricetinae , Humanos , Recombinación Genética , Genética Inversa , Células Vero , Virulencia
7.
J Virol ; 93(22)2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31462569

RESUMEN

Several mammarenaviruses can cause deadly hemorrhagic fever infections in humans, with limited preventative and therapeutic measures available. Arenavirus cell entry is mediated by the viral glycoprotein (GP) complex, which consists of the stable signal peptide (SSP), the receptor-binding subunit GP1, and the transmembrane subunit GP2. The GP2 cytoplasmic tail (CT) is relatively conserved among arenaviruses and is known to interact with the SSP to regulate GP processing and membrane fusion, but its biological role in the context of an infectious virus has not been fully characterized. Using a Pichinde virus (PICV) GP expression vector and a PICV reverse genetics system, we systematically characterized the functional roles of 12 conserved residues within the GP2 CT in GP processing, trafficking, assembly, and fusion, as well as in viral replication. Except for P478A and K505A R508A, alanine substitutions at conserved residues abolished GP processing and membrane fusion in plasmid-transfected cells. Six invariant H and C residues and W503 are essential for viral replication, as evidenced by the fact that their mutant viruses could not be rescued. Both P480A and R482A mutant viruses were rescued, grew similarly to wild-type (WT) virus, and produced evidently processed GP1 and GP2 subunits in virus-infected cells, despite the fact that the same mutations abolished GP processing and membrane fusion in a plasmid-based protein expression system, illustrating the importance of using an infectious-virus system for analyzing viral glycoprotein function. In summary, our results demonstrate an essential biological role of the GP2 CT in arenavirus replication and suggest it as a potential novel target for developing antivirals and/or attenuated viral vaccine candidates.IMPORTANCE Several arenaviruses, such as Lassa virus (LASV), can cause severe and lethal hemorrhagic fever diseases with high mortality and morbidity, for which no FDA-approved vaccines or therapeutics are available. Viral entry is mediated by the arenavirus GP complex, which consists of the stable signal peptide (SSP), the receptor-binding subunit GP1, and the transmembrane subunit GP2. The cytoplasmic tail (CT) of GP2 is highly conserved among arenaviruses, but its functional role in viral replication is not completely understood. Using a reverse genetics system of a prototypic arenavirus, Pichinde virus (PICV), we show that the GP2 CT contains certain conserved residues that are essential for virus replication, implicating it as a potentially good target for developing antivirals and live-attenuated viral vaccines against deadly arenavirus pathogens.


Asunto(s)
Glicoproteínas/metabolismo , Virus Pichinde/genética , Proteínas del Envoltorio Viral/genética , Células A549 , Sustitución de Aminoácidos/genética , Animales , Arenaviridae , Infecciones por Arenaviridae/genética , Infecciones por Arenaviridae/metabolismo , Arenavirus/genética , Arenavirus/metabolismo , Línea Celular , Chlorocebus aethiops , Glicoproteínas/genética , Células HEK293 , Humanos , Fusión de Membrana/genética , Mutación/genética , Virus Pichinde/metabolismo , Señales de Clasificación de Proteína/genética , Células Vero , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Replicación Viral
8.
J Virol ; 92(13)2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29669840

RESUMEN

RIG-I is a major cytoplasmic sensor of viral pathogen-associated molecular pattern (PAMP) RNA and induces type I interferon (IFN) production upon viral infection. A double-stranded RNA (dsRNA)-binding protein, PACT, plays an important role in potentiating RIG-I function. We have shown previously that arenaviral nucleoproteins (NPs) suppress type I IFN production via their RNase activity to degrade PAMP RNA. We report here that NPs of arenaviruses block the PACT-induced enhancement of RIG-I function to mediate type I IFN production and that this inhibition is dependent on the RNase function of NPs, which is different from that of a known mechanism of other viral proteins to abolish the interaction between PACT and RIG-I. To understand the biological roles of PACT and RIG-I in authentic arenavirus infection, we analyze growth kinetics of recombinant Pichinde virus (PICV), a prototypical arenavirus, in RIG-I knockout (KO) and PACT KO mouse embryonic fibroblast (MEF) cells. Wild-type (WT) PICV grew at higher titers in both KO MEF lines than in normal MEFs, suggesting the important roles of these cellular proteins in restricting virus replication. PICV carrying the NP RNase catalytically inactive mutation could not grow in normal MEFs but could replicate to some extent in both KO MEF lines. The level of virus growth was inversely correlated with the amount of type I IFNs produced. These results suggest that PACT plays an important role in potentiating RIG-I function to produce type I IFNs in order to restrict arenavirus replication and that viral NP RNase activity is essential for optimal viral replication by suppressing PACT-induced RIG-I activation.IMPORTANCE We report here a new role of the nucleoproteins of arenaviruses that can block type I IFN production via their specific inhibition of the cellular protein sensors of virus infection (RIG-I and PACT). Our results suggest that PACT plays an important role in potentiating RIG-I function to produce type I IFNs in order to restrict arenavirus replication. This new knowledge can be exploited for the development of novel antiviral treatments and/or vaccines against some arenaviruses that can cause severe and lethal hemorrhagic fever diseases in humans.


Asunto(s)
Arenavirus/patogenicidad , Interacciones Huésped-Patógeno , Interferón Tipo I/metabolismo , Nucleoproteínas/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptores de Ácido Retinoico/metabolismo , Proteínas Virales/metabolismo , Infecciones por Arenaviridae/genética , Infecciones por Arenaviridae/metabolismo , Infecciones por Arenaviridae/virología , Células HEK293 , Humanos , Nucleoproteínas/genética , Virus Pichinde/fisiología , Proteínas de Unión al ARN/genética , Receptores de Ácido Retinoico/genética , Proteínas Virales/genética , Replicación Viral
9.
Methods Mol Biol ; 1604: 169-178, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28986832

RESUMEN

Arenaviruses, such as Lassa virus (LASV) and Pichindé virus (PICV), are enveloped viruses with a bi-segmented ambisense RNA genome. The large (L) genomic segment encodes the Z matrix protein and the L RNA-dependent RNA polymerase, whereas the small (S) genomic segment encodes the nucleoprotein (NP) and the glycoprotein precursor complex (GPC). GPC is processed by signal peptidase in the endoplasmic reticulum into the stable signal peptide (SSP) and GP1/GP2, which is further cleaved by the Golgi-resident subtilisin kexin isozyme-1 (SKI-1)/site-1 protease (S1P) into the cellular receptor-recognition subunit GP1 and the transmembrane subunit GP2, which helps promote the membrane fusion reaction to allow virus entry into the cell. This article describes assays to assess PICV GPC expression, proteolytic processing, fusion function, and GPC-mediated virus-like particle (VLP) entry into cells under tissue-culture conditions.


Asunto(s)
Arenavirus/metabolismo , Bioensayo/métodos , Glicoproteínas/metabolismo , Animales , Arenavirus/genética , Glicoproteínas/genética , Humanos , Virus Lassa/genética , Virus Lassa/metabolismo , Virus Pichinde/genética , Virus Pichinde/metabolismo , Proproteína Convertasas/genética , Proproteína Convertasas/metabolismo , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo
10.
Methods Mol Biol ; 1604: 217-227, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28986837

RESUMEN

The smallest arenaviral protein is the zinc-finger protein (Z) that belongs to the RING finger protein family. Z serves as a main component required for virus budding from the membrane of the infected cells through self-oligomerization, a process that can be aided by the viral nucleoprotein (NP) to form the viral matrix of progeny virus particles. Z has also been shown to be essential for mediating viral transcriptional repression activity by locking the L polymerase onto the viral promoter in a catalytically inactive state, thus limiting viral replication. The Z protein has also recently been shown to inhibit the type I interferon-induction pathway by directly binding to the intracellular pathogen-sensor proteins RIG-I and MDA5, and thus inhibiting their normal functions. This chapter describes several assays used to examine the important roles of the arenaviral Z protein in mediating virus budding (i.e., either Z self-budding or NP-Z budding activities), viral transcriptional inhibition in a viral minigenome (MG) assay, and type I IFN suppression in an IFN-ß promoter-mediated luciferase reporter assay.


Asunto(s)
Arenavirus/metabolismo , Liberación del Virus/fisiología , Replicación Viral/fisiología , Arenavirus/genética , Interferón beta/metabolismo , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Virus Pichinde/genética , Virus Pichinde/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Liberación del Virus/genética , Replicación Viral/genética
11.
Methods Mol Biol ; 1604: 247-253, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28986840

RESUMEN

Pichindé virus (PICV), isolated from rice rats in Colombia, South America, is an enveloped arenavirus with a bisegmented RNA genome. The large (L) genomic segment encodes the Z matrix protein and the L RNA-dependent RNA polymerase, whereas the small (S) genomic segment encodes the nucleoprotein (NP) and the glycoprotein (GPC). This article describes the successful development of reverse genetics systems to generate recombinant PICV with either a bisegmented or trisegmented genome. We have successfully demonstrated that these systems can generate high-titered and genetically stable replication-competent viruses from plasmid transfection into appropriate cell lines. These systems demonstrate the power and versatility of reverse genetic technology to generate recombinant arenaviruses for use in pathogenesis studies and as new viral vaccine vectors.


Asunto(s)
Virus Pichinde/genética , Animales , Arenavirus/genética , Genoma Viral/genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Ratas , Genética Inversa , Replicación Viral/genética
12.
Methods Mol Biol ; 1581: 169-179, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28374249

RESUMEN

Pichinde virus (PICV) is a nonpathogenic arenavirus with a bi-segmented RNA genome (L and S segments) that encodes four viral genes. We have developed a reverse genetics system to generate recombinant tri-segmented PICV (rP18tri) that packages three RNA segments (L, S1, and S2) and can encode up to two foreign genes. Using influenza virus HA and NP as model antigens, we show that the rP18tri vector can induce strong humoral and cell-mediated immunity, which further increases upon a booster dose. We propose that this novel rP18tri vector can be developed into a useful vaccine platform for other antigens, particularly when strong cellular immunity and prime-boost vaccination strategy are desired.


Asunto(s)
Antígenos Virales/inmunología , Virus Pichinde/genética , Vacunas Atenuadas/administración & dosificación , Animales , Antígenos Virales/genética , Línea Celular , Chlorocebus aethiops , Perros , Inmunidad Celular , Células de Riñón Canino Madin Darby , Ratones , Virus Pichinde/inmunología , Genética Inversa , Vacunación , Vacunas Atenuadas/inmunología , Células Vero , Ensamble de Virus
13.
J Virol ; 90(22): 10390-10397, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27630230

RESUMEN

Arenaviruses can cause lethal hemorrhagic fevers in humans with few preventative and therapeutic measures. The arenaviral glycoprotein stable signal peptide (SSP) is unique among signal peptides in that it is an integral component of the mature glycoprotein complex (GPC) and plays important roles not only in GPC expression and processing but also in the membrane fusion process during viral entry. Using the Pichinde virus (PICV) reverse genetics system, we analyzed the effects of alanine substitutions at many conserved residues within the SSP on viral replication in cell culture and in a guinea pig infection model. Our data showed that the K33A, F49A, and C57A mutations abolished GPC-mediated cell entry and therefore could not allow for the generation of viable recombinant viruses, demonstrating that these residues are essential for the PICV life cycle. The G2A mutation caused a marked reduction of cell entry at the membrane fusion step, and while this mutant virus was viable, it was significantly attenuated in vitro and in vivo The N20A mutation also reduced membrane fusion activity and viral virulence in guinea pigs, but it did not significantly affect cell entry or viral growth in cell culture. Two other mutations (N37A and R55A) did not affect membrane fusion or viral growth in vitro but significantly reduced viral virulence in vivo Taken together, our data suggest that the GPC SSP plays an essential role in mediating viral entry and also contributes to viral virulence in vivo IMPORTANCE: Several arenaviruses, such as Lassa fever virus, can cause severe and lethal hemorrhagic fever diseases with high mortality and morbidity, and no FDA-approved vaccines or therapies are currently available. Viral entry into cells is mediated by arenavirus GPC that consists of an SSP, the receptor-binding GP1, and transmembrane GP2 protein subunits. Using a reverse genetics system of a prototypic arenavirus, Pichinde virus (PICV), we have shown for the first time in the context of virus infections of cell culture and of guinea pigs that the SSP plays an essential role in mediating the membrane fusion step as well as in other yet-to-be-determined processes during viral infection. Our study provides important insights into the biological roles of GPC SSP and implicates it as a good target for the development of antivirals against deadly human arenavirus pathogens.


Asunto(s)
Glicoproteínas/genética , Virus Pichinde/genética , Señales de Clasificación de Proteína/genética , Virulencia/genética , Células A549 , Animales , Infecciones por Arenaviridae/virología , Arenavirus/genética , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Replicación del ADN/genética , Cobayas , Células HEK293 , Humanos , Fusión de Membrana/genética , Mutación/genética , Subunidades de Proteína/genética , Células Vero , Proteínas del Envoltorio Viral/genética , Internalización del Virus , Replicación Viral/genética
14.
Virol J ; 13: 54, 2016 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-27029488

RESUMEN

BACKGROUND: Research with high biocontainment pathogens such as Rift Valley fever virus (RVFV) and Lassa virus (LASV) is expensive, potentially hazardous, and limited to select institutions. Surrogate pathogens such as Punta Toro virus (PTV) for RVFV infection and Pichinde virus (PICV) for LASV infection allow research to be performed under more permissive BSL-2 conditions. Although used as infection models, PTV and PICV have no standard real-time RT-qPCR assays to detect and quantify pathogenesis. PTV is also a human pathogen, making a standardized detection assay essential for biosurveillance. Here, we developed and characterized two real-time RT-qPCR assays for PICV and PTV by optimizing assay conditions and measuring the limit of detection (LOD) and performance in multiple clinical matrices. METHODS: Total nucleic acid from virus-infected Vero E6 cells was used to optimize TaqMan-minor groove binder (MGB) real-time RT-qPCR assays. A 10-fold dilution series of nucleic acid was used to perform analytical experiments with 60 replicates used to confirm assay LODs. Serum and whole blood spiked with 10-fold dilutions of PTV and PICV virus were assessed as matrices in a mock clinical context. The Cq, or cycle at which the fluoresce of each sample first crosses a threshold line, was determined using the second derivative method using Roche LightCycler 480 software version 1.5.1. Digital droplet PCR (ddPCR) was utilized to quantitatively determine RNA target counts/µl for PTV and PICV. RESULTS: Optimized PTV and PICV assays had LODs of 1000 PFU/ml and 100 PFU/ml, respectively, and this LOD was confirmed in 60/60 (PTV) and 58/60 (PICV) positive replicates. Preliminary mock clinical LODs remained consistent in serum and whole blood for PTV and PICV at 1000 PFU/ml and 100 PFU/ml. An exclusivity panel showed no cross reaction with near neighbors. CONCLUSIONS: PTV and PICV Taq-man MGB based real-time RT-qPCR assays developed here showed relevant sensitivity and reproducibility in samples extracted from a variety of clinical matrices. These assays will be useful as a standard by researchers for future experiments utilizing PTV and PICV as infection models, offering the ability to track infection and viral replication kinetics during research studies.


Asunto(s)
Infecciones por Arenaviridae/diagnóstico , Infecciones por Bunyaviridae/diagnóstico , Phlebovirus/aislamiento & purificación , Virus Pichinde/aislamiento & purificación , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Virología/métodos , Humanos , Técnicas de Diagnóstico Molecular/métodos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Factores de Tiempo
15.
J Virol ; 90(5): 2551-60, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26676795

RESUMEN

UNLABELLED: Pichinde virus (PICV) is a bisegmented enveloped RNA virus that targets macrophages and dendritic cells (DCs) early in infection and induces strong innate and adaptive immunity in mice. We have developed a reverse genetics system to produce live recombinant PICV (strain P18) with a trisegmented RNA genome (rP18tri), which encodes all four PICV gene products and as many as two foreign genes. We have engineered the vector to express the green fluorescent protein (GFP) reporter gene (abbreviated as G in virus designations) and either the hemagglutination (HA [H]) or the nucleoprotein (NP [P]) gene of the influenza A/PR8 virus. The trisegmented viruses rP18tri-G/H and rP18tri-G/P showed slightly reduced growth in vitro and expressed HA and NP, respectively. Mice immunized with rP18tri-G/H were completely protected against lethal influenza virus challenge even 120 days after immunization. These rP18tri-based vectors could efficiently induce both neutralizing antibodies and antigen-specific T cell responses via different immunization routes. Interestingly, the immune responses were significantly increased upon a booster dose and remained at high levels even after three booster doses. In summary, we have developed a novel PICV-based live vaccine vector that can express foreign antigens to induce strong humoral and cell-mediated immunity and is ideal for a prime-and-boost vaccination strategy. IMPORTANCE: We have developed a novel Pichinde virus (PICV)-based live viral vector, rP18tri, that packages three RNA segments and encodes as many as two foreign genes. Using the influenza virus HA and NP genes as model antigens, we show that this rP18tri vector can induce strong humoral and cellular immunity via different immunization routes and can lead to protection in mice. Interestingly, a booster dose further enhances the immune responses, a feature that distinguishes this from other known live viral vectors. In summary, our study demonstrates a unique feature of this live rP18tri vector to be used as a novel vaccine platform for a prime-and-boost vaccination strategy.


Asunto(s)
Portadores de Fármacos , Vectores Genéticos , Vacunas contra la Influenza/inmunología , Virus Pichinde/genética , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Cobayas , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Masculino , Ratones Endogámicos C57BL , Proteínas de la Nucleocápside , Infecciones por Orthomyxoviridae/prevención & control , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Análisis de Supervivencia , Linfocitos T/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/inmunología
16.
J Virol ; 89(24): 12513-7, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26423945

RESUMEN

Several arenavirus pathogens, such as Lassa and Junin viruses, inhibit macrophage activation, the molecular mechanism of which is unclear. We show that lymphocytic choriomeningitis virus (LCMV) can also inhibit macrophage activation, in contrast to Pichinde and Tacaribe viruses, which are not known to naturally cause human diseases. Using a recombinant Pichinde virus system, we show that the LCMV Z N-terminal domain (NTD) mediates the inhibition of macrophage activation and immune functions.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Proteínas Portadoras/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Activación de Macrófagos , Macrófagos/inmunología , Virus Pichinde/inmunología , Animales , Humanos , Péptidos y Proteínas de Señalización Intracelular , Macrófagos/virología , Ratones , Estructura Terciaria de Proteína
17.
J Virol ; 89(13): 6595-607, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25878103

RESUMEN

UNLABELLED: Arenaviruses cause severe hemorrhagic fever diseases in humans, and there are limited preventative and therapeutic measures against these diseases. Previous structural and functional analyses of arenavirus nucleoproteins (NPs) revealed a conserved DEDDH exoribonuclease (RNase) domain that is important for type I interferon (IFN) suppression, but the biological roles of the NP RNase in viral replication and host immune suppression have not been well characterized. Infection of guinea pigs with Pichinde virus (PICV), a prototype arenavirus, can serve as a surrogate small animal model for arenavirus hemorrhagic fevers. In this report, we show that mutation of each of the five RNase catalytic residues of PICV NP diminishes the IFN suppression activity and slightly reduces the viral RNA replication activity. Recombinant PICVs with RNase catalytic mutations can induce high levels of IFNs and barely grow in IFN-competent A549 cells, in sharp contrast to the wild-type (WT) virus, while in IFN-deficient Vero cells, both WT and mutant viruses can replicate at relatively high levels. Upon infection of guinea pigs, the RNase mutant viruses stimulate strong IFN responses, fail to replicate productively, and can become WT revertants. Serial passages of the RNase mutants in vitro can also generate WT revertants. Thus, the NP RNase function is essential for the innate immune suppression that allows the establishment of a productive early viral infection, and it may be partly involved in the process of viral RNA replication. IMPORTANCE: Arenaviruses, such as Lassa, Lujo, and Machupo viruses, can cause severe and deadly hemorrhagic fever diseases in humans, and there are limited preventative and treatment options against these diseases. Development of broad-spectrum antiviral drugs depends on a better mechanistic understanding of the conserved arenavirus proteins in viral infection. The nucleoprotein (NPs) of all arenaviruses carry a unique exoribonuclease (RNase) domain that has been shown to be critical for the suppression of type I interferons. However, the functional roles of the NP RNase in arenavirus replication and host immune suppression have not been characterized systematically. Using a prototype arenavirus, Pichinde virus (PICV), we characterized the viral growth and innate immune suppression of recombinant RNase-defective mutants in both cell culture and guinea pig models. Our study suggests that the NP RNase plays an essential role in the suppression of host innate immunity, and possibly in viral RNA replication, and that it can serve as a novel target for developing antiviral drugs against arenavirus pathogens.


Asunto(s)
Exorribonucleasas/metabolismo , Interacciones Huésped-Patógeno , Evasión Inmune , Nucleoproteínas/metabolismo , Virus Pichinde/enzimología , Virus Pichinde/fisiología , Replicación Viral , Secuencias de Aminoácidos , Sustitución de Aminoácidos , Animales , Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/virología , Línea Celular , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Exorribonucleasas/genética , Cobayas , Humanos , Masculino , Nucleoproteínas/genética , Virus Pichinde/genética , Virus Pichinde/inmunología
18.
J Virol ; 89(5): 2944-55, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25552708

RESUMEN

UNLABELLED: Arenavirus pathogens cause a wide spectrum of diseases in humans ranging from central nervous system disease to lethal hemorrhagic fevers with few treatment options. The reason why some arenaviruses can cause severe human diseases while others cannot is unknown. We find that the Z proteins of all known pathogenic arenaviruses, lymphocytic choriomeningitis virus (LCMV) and Lassa, Junin, Machupo, Sabia, Guanarito, Chapare, Dandenong, and Lujo viruses, can inhibit retinoic acid-inducible gene 1 (RIG-i) and Melanoma Differentiation-Associated protein 5 (MDA5), in sharp contrast to those of 14 other nonpathogenic arenaviruses. Inhibition of the RIG-i-like receptors (RLRs) by pathogenic Z proteins is mediated by the protein-protein interactions of Z and RLRs, which lead to the disruption of the interactions between RLRs and mitochondrial antiviral signaling (MAVS). The Z-RLR interactive interfaces are located within the N-terminal domain (NTD) of the Z protein and the N-terminal CARD domains of RLRs. Swapping of the LCMV Z NTD into the nonpathogenic Pichinde virus (PICV) genome does not affect virus growth in Vero cells but significantly inhibits the type I interferon (IFN) responses and increases viral replication in human primary macrophages. In summary, our results show for the first time an innate immune-system-suppressive mechanism shared by the diverse pathogenic arenaviruses and thus shed important light on the pathogenic mechanism of human arenavirus pathogens. IMPORTANCE: We show that all known human-pathogenic arenaviruses share an innate immune suppression mechanism that is based on viral Z protein-mediated RLR inhibition. Our report offers important insights into the potential mechanism of arenavirus pathogenesis, provides a convenient way to evaluate the pathogenic potential of known and/or emerging arenaviruses, and reveals a novel target for the development of broad-spectrum therapies to treat this group of diverse pathogens. More broadly, our report provides a better understanding of the mechanisms of viral immune suppression and host-pathogen interactions.


Asunto(s)
ARN Helicasas DEAD-box/antagonistas & inhibidores , Interacciones Huésped-Patógeno , Interferones/antagonistas & inhibidores , Virus de la Coriomeningitis Linfocítica/fisiología , Virus Pichinde/fisiología , Proteínas Virales/metabolismo , Replicación Viral , Animales , Células Cultivadas , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , Humanos , Tolerancia Inmunológica , Interferones/biosíntesis , Virus de la Coriomeningitis Linfocítica/genética , Virus Pichinde/genética , Unión Proteica , Mapeo de Interacción de Proteínas , Receptores Inmunológicos , Recombinación Genética , Proteínas Virales/genética
19.
Mol Cell Proteomics ; 14(3): 646-57, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25573744

RESUMEN

The Syrian golden hamster has been increasingly used to study viral hemorrhagic fever (VHF) pathogenesis and countermeasure efficacy. As VHFs are a global health concern, well-characterized animal models are essential for both the development of therapeutics and vaccines as well as for increasing our understanding of the molecular events that underlie viral pathogenesis. However, the paucity of reagents or platforms that are available for studying hamsters at a molecular level limits the ability to extract biological information from this important animal model. As such, there is a need to develop platforms/technologies for characterizing host responses of hamsters at a molecular level. To this end, we developed hamster-specific kinome peptide arrays to characterize the molecular host response of the Syrian golden hamster. After validating the functionality of the arrays using immune agonists of defined signaling mechanisms (lipopolysaccharide (LPS) and tumor necrosis factor (TNF)-α), we characterized the host response in a hamster model of VHF based on Pichinde virus (PICV(1)) infection by performing temporal kinome analysis of lung tissue. Our analysis revealed key roles for vascular endothelial growth factor (VEGF), interleukin (IL) responses, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, and Toll-like receptor (TLR) signaling in the response to PICV infection. These findings were validated through phosphorylation-specific Western blot analysis. Overall, we have demonstrated that hamster-specific kinome arrays are a robust tool for characterizing the species-specific molecular host response in a VHF model. Further, our results provide key insights into the hamster host response to PICV infection and will inform future studies with high-consequence VHF pathogens.


Asunto(s)
Fiebre Hemorrágica Americana/virología , Pulmón/enzimología , Virus Pichinde/fisiología , Proteínas Quinasas/aislamiento & purificación , Proteoma/análisis , Animales , Modelos Animales de Enfermedad , Femenino , Fiebre Hemorrágica Americana/enzimología , Interleucinas/aislamiento & purificación , Pulmón/virología , Mesocricetus , FN-kappa B/aislamiento & purificación , Fosforilación , Transducción de Señal , Especificidad de la Especie , Receptores Toll-Like/aislamiento & purificación , Factor A de Crecimiento Endotelial Vascular/aislamiento & purificación
20.
Virology ; 464-465: 213-217, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25094042

RESUMEN

Having a history of infection with one pathogen may sometimes provide a level of T cell-dependent protective heterologous immunity to another pathogen. This immunity was initially thought due to cross-reactive T cell epitopes, but recent work has suggested that such protective immunity can be initiated nonspecifically by the action of cytokines on memory T cells. We retested this concept using two small and well-defined arenaviruses, lymphocytic choriomeningitis virus (LCMV) and Pichinde virus (PV), and found that heterologous immunity in these systems was indeed linked to T cell epitopes and the major histocompatibility complex.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/virología , Inmunidad Heteróloga , Virus de la Coriomeningitis Linfocítica/inmunología , Complejo Mayor de Histocompatibilidad , Virus Pichinde/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Protección Cruzada , Reacciones Cruzadas , Epítopos de Linfocito T/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA