Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Virology ; 587: 109867, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37633192

RESUMEN

Lujo virus (LUJV), which belongs to Mammarenavirus, family Arenaviridae, has emerged as a pathogen causing severe hemorrhagic fever with high mortality. Currently, there are no effective treatments for arenaviruses, including LUJV. Here, we screened chemical compound libraries of Food and Drug Administration (FDA)-approved drugs and G protein-coupled receptor-associated drugs to identify effective antivirals against LUJV targeting cell entry using a vesicular stomatitis virus-based pseudotyped virus bearing the LUJV envelope glycoprotein (GP). Cannabinoid receptor 1 (CB1) antagonists, such as rimonabant, AM251 and AM281, have been identified as robust inhibitors of LUJV entry. The IC50 of rimonabant was 0.26 and 0.53 µM in Vero and Huh7 cells, respectively. Analysis of the cell fusion activity of the LUJV GP in the presence of CB1 inhibitors revealed that these inhibitors suppressed the fusion activity of the LUJV GP. Moreover, rimonabant, AM251 and AM281 reduced the infectivity of authentic LUJV in vitro, suggesting that the antiviral activity of CB1 antagonists against LUJV is mediated, at least in part, by inhibition of the viral entry, especially, membrane fusion. These findings suggest promising candidates for developing new therapies against LUJV infections.


Asunto(s)
Infecciones por Arenaviridae , Arenaviridae , Lujo virus , Humanos , Chlorocebus aethiops , Animales , Lujo virus/metabolismo , Rimonabant/farmacología , Rimonabant/metabolismo , Infecciones por Arenaviridae/metabolismo , Internalización del Virus , Receptores de Cannabinoides/metabolismo , Células Vero
2.
Biomed Pharmacother ; 166: 115196, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37586116

RESUMEN

Mammarenaviruses are enveloped RNA viruses that can be associated with rodent-transmitted diseases in humans. Their virions are composed of a nucleocapsid surrounded by a lipid bilayer with glycoprotein (GP) spikes interacting with receptors on target cells. Both the GP and receptors are highly glycosylated, with glycosylation patterns being crucial for virus binding and cell entry, viral tropism, immune responses, or therapy strategies. These effects have been previously described for several different viruses. In case of arenaviruses, they remain insufficiently understood. Thus, it is important to determine the mechanisms of glycosylation of viral proteins and receptors responsible for infection, in order to fully understand the biology of arenaviruses. In this article, we have summarized and critically evaluated the available literature data on the glycosylation of mammarenavirus-associated proteins to facilitate further research in this field.


Asunto(s)
Infecciones por Arenaviridae , Internalización del Virus , Humanos , Glicosilación , Receptores de Superficie Celular/metabolismo , Infecciones por Arenaviridae/metabolismo , Glicoproteínas
3.
Front Immunol ; 13: 782441, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35185882

RESUMEN

CD8+ T cells play a crucial role in the control and resolution of viral infections and can adopt a wide range of phenotypes and effector functions depending on the inflammatory context and the duration and extent of antigen exposure. Similarly, viral infections can exert diverse selective pressures on populations of clonally related T cells. Technical limitations have nevertheless made it challenging to investigate the relationship between clonal selection and transcriptional phenotypes of virus-specific T cells. We therefore performed single-cell T cell receptor (TCR) repertoire and transcriptome sequencing of virus-specific CD8 T cells in murine models of acute, chronic and latent infection. We observed clear infection-specific populations corresponding to memory, effector, exhausted, and inflationary phenotypes. We further uncovered a mouse-specific and polyclonal T cell response, despite all T cells sharing specificity to a single viral epitope, which was accompanied by stereotypic TCR germline gene usage in all three infection types. Persistent antigen exposure during chronic and latent viral infections resulted in a higher proportion of clonally expanded T cells relative to acute infection. We furthermore observed a relationship between transcriptional heterogeneity and clonal expansion for all three infections, with highly expanded clones having distinct transcriptional phenotypes relative to less expanded clones. Together our work relates clonal selection to gene expression in the context of viral infection and further provides a dataset and accompanying software for the immunological community.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Receptores de Antígenos de Linfocitos T/genética , Transcriptoma , Enfermedad Aguda , Animales , Infecciones por Arenaviridae/genética , Infecciones por Arenaviridae/metabolismo , Infecciones por Arenaviridae/virología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Enfermedad Crónica , Células Clonales/metabolismo , Modelos Animales de Enfermedad , Femenino , Infección Latente , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones , Ratones Endogámicos C57BL , Fenotipo , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Virosis
4.
Sci Adv ; 8(6): eabk2691, 2022 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-35138904

RESUMEN

Upon virus infection, CD8+ T cell accumulation is tightly controlled by simultaneous proliferation and apoptosis. However, it remains unclear how TCR signal coordinates these events to achieve expansion and effector cell differentiation. We found that T cell-specific deletion of nuclear helicase Dhx9 led to impaired CD8+ T cell survival, effector differentiation, and viral clearance. Mechanistically, Dhx9 acts as the key regulator to ensure LCK- and CD3ε-mediated ZAP70 phosphorylation and ERK activation to protect CD8+ T cells from apoptosis before proliferative burst. Dhx9 directly regulates Id2 transcription to control effector CD8+ T cell differentiation. The DSRM and OB_Fold domains are required for LCK binding and Id2 transcription, respectively. Dhx9 expression is predominantly increased in effector CD8+ T cells of COVID-19 patients. Therefore, we revealed a previously unknown regulatory mechanism that Dhx9 protects activated CD8+ T cells from apoptosis and ensures effector differentiation to promote antiviral immunity independent of nuclear sensor function.


Asunto(s)
Antivirales/farmacología , Infecciones por Arenaviridae/prevención & control , Linfocitos T CD8-positivos/inmunología , COVID-19/prevención & control , ARN Helicasas DEAD-box/metabolismo , Inmunidad Innata , Proteínas de Neoplasias/metabolismo , Animales , Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/metabolismo , Infecciones por Arenaviridae/patología , COVID-19/inmunología , COVID-19/metabolismo , COVID-19/patología , Diferenciación Celular , ARN Helicasas DEAD-box/genética , Humanos , Activación de Linfocitos , Virus de la Coriomeningitis Linfocítica/fisiología , Ratones , Proteínas de Neoplasias/genética , SARS-CoV-2/fisiología , Replicación Viral
6.
Commun Biol ; 4(1): 508, 2021 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-33927339

RESUMEN

Immune evasion of pathogens can modify the course of infection and impact viral persistence and pathology. Here, using different strains of the lymphocytic choriomeningitis virus (LCMV) model system, we show that slower propagation results in limited type I interferon (IFN-I) production and viral persistence. Specifically, cells infected with LCMV-Docile exhibited reduced viral replication when compared to LCMV-WE and as a consequence, infection with LCMV-Docile resulted in reduced activation of bone marrow derived dendritic cells (BMDCs) and IFN-I production in vitro in comparison with LCMV-WE. In vivo, we observed a reduction of IFN-I, T cell exhaustion and viral persistence following infection of LCMV-Docile but not LCMV-WE. Mechanistically, block of intracellular protein transport uncovered reduced propagation of LCMV-Docile when compared to LCMV-WE. This reduced propagation was critical in blunting the activation of the innate and adaptive immune system. When mice were simultaneously infected with LCMV-Docile and LCMV-WE, immune function was restored and IFN-I production, T cell effector functions as well as viral loads were similar to that of mice infected with LCMV-WE alone. Taken together, this study suggests that reduced viral propagation can result in immune evasion and viral persistence.


Asunto(s)
Infecciones por Arenaviridae/virología , Células Dendríticas/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Receptor de Interferón alfa y beta/fisiología , Linfocitos T/virología , Replicación Viral , Animales , Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/inmunología , Linfocitos T/metabolismo
7.
Nat Immunol ; 22(4): 434-448, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33649580

RESUMEN

T cells dynamically interact with multiple, distinct cellular subsets to determine effector and memory differentiation. Here, we developed a platform to quantify cell location in three dimensions to determine the spatial requirements that direct T cell fate. After viral infection, we demonstrated that CD8+ effector T cell differentiation is associated with positioning at the lymph node periphery. This was instructed by CXCR3 signaling since, in its absence, T cells are confined to the lymph node center and alternatively differentiate into stem-like memory cell precursors. By mapping the cellular sources of CXCR3 ligands, we demonstrated that CXCL9 and CXCL10 are expressed by spatially distinct dendritic and stromal cell subsets. Unlike effector cells, retention of stem-like memory precursors in the paracortex is associated with CCR7 expression. Finally, we demonstrated that T cell location can be tuned, through deficiency in CXCL10 or type I interferon signaling, to promote effector or stem-like memory fates.


Asunto(s)
Infecciones por Arenaviridae/metabolismo , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/metabolismo , Memoria Inmunológica , Ganglios Linfáticos/metabolismo , Células Precursoras de Linfocitos T/metabolismo , Receptores CXCR3/metabolismo , Animales , Infecciones por Arenaviridae/genética , Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Linaje de la Célula , Células Cultivadas , Quimiocina CXCL10/genética , Quimiocina CXCL9/genética , Quimiotaxis de Leucocito , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno , Interferón Tipo I/metabolismo , Ligandos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Células Precursoras de Linfocitos T/inmunología , Células Precursoras de Linfocitos T/virología , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Receptores CCR7/metabolismo , Receptores CXCR3/genética , Transducción de Señal , Nicho de Células Madre , Células del Estroma/inmunología , Células del Estroma/metabolismo
8.
Nat Immunol ; 22(3): 370-380, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33574619

RESUMEN

During chronic infection and cancer, a self-renewing CD8+ T cell subset maintains long-term immunity and is critical to the effectiveness of immunotherapy. These stem-like CD8+ T cells diverge from other CD8+ subsets early after chronic viral infection. However, pathways guarding stem-like CD8+ T cells against terminal exhaustion remain unclear. Here, we show that the gene encoding transcriptional repressor BACH2 is transcriptionally and epigenetically active in stem-like CD8+ T cells but not terminally exhausted cells early after infection. BACH2 overexpression enforced stem-like cell fate, whereas BACH2 deficiency impaired stem-like CD8+ T cell differentiation. Single-cell transcriptomic and epigenomic approaches revealed that BACH2 established the transcriptional and epigenetic programs of stem-like CD8+ T cells. In addition, BACH2 suppressed the molecular program driving terminal exhaustion through transcriptional repression and epigenetic silencing. Thus, our study reveals a new pathway that enforces commitment to stem-like CD8+ lineage and prevents an alternative terminally exhausted cell fate.


Asunto(s)
Infecciones por Arenaviridae/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Epigénesis Genética , Células Precursoras de Linfocitos T/metabolismo , Transcripción Genética , Animales , Infecciones por Arenaviridae/genética , Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/virología , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/deficiencia , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Linaje de la Célula , Células Cultivadas , Enfermedad Crónica , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Células Precursoras de Linfocitos T/inmunología , Células Precursoras de Linfocitos T/virología , Transducción de Señal
9.
PLoS Pathog ; 16(10): e1008948, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33045019

RESUMEN

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.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Infecciones por Arenaviridae/patología , Arenavirus del Nuevo Mundo/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Replicación Viral , Proteína Letal Asociada a bcl/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Infecciones por Arenaviridae/genética , Infecciones por Arenaviridae/metabolismo , Infecciones por Arenaviridae/virología , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Dominios Proteicos , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteína Letal Asociada a bcl/genética
10.
Front Immunol ; 11: 986, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32547546

RESUMEN

CD8 T cells play a crucial role in providing protection from viral infections. It has recently been established that a subset of CD8 T cells expressing Tcf1 are responsible for sustaining exhausted T cells during chronic lymphocytic choriomeningitis virus (LCMV) infection. Many of these studies, however, have been performed using T cell receptor (TCR) transgenic mice, in which CD8 T cells express a monoclonal TCR specific for the LCMV glycoprotein. To investigate whether the Tcf1+ and Tcf1- repertoires are naturally composed of similar or different clones in wild-type mice exposed to acute or chronic LCMV infection, we performed TCR repertoire sequencing of virus-specific CD8 T cells, including Tcf1+ and Tcf1- populations. Our analysis revealed that the Tcf1+ TCR repertoire is maintained at an equal or higher degree of clonal diversity despite harboring fewer cells. Additionally, within the same animal, there was extensive clonal overlap between the Tcf1+ and Tcf1- repertoires in both chronic and acute LCMV infection. We could further detect these virus-specific clones in longitudinal blood samples earlier in the infection. With respect to common repertoire parameters (clonal overlap, germline gene usage, and clonal expansion), we found minor differences between the virus-specific TCR repertoire of acute and chronic LCMV infection 40 days post infection. Overall, our results indicate that the Tcf1+ population emerging during chronic LCMV infection is not clonally distinct from the Tcf1- population, supporting the notion that the Tcf1+ pool is indeed a fuel for the more exhausted Tcf1- population within the heterogenous repertoire of LCMV-specific CD8 T cells.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Perfilación de la Expresión Génica , Virus de la Coriomeningitis Linfocítica/inmunología , Receptores de Antígenos de Linfocitos T/genética , Transcriptoma , Enfermedad Aguda , Animales , Infecciones por Arenaviridae/genética , Infecciones por Arenaviridae/metabolismo , Infecciones por Arenaviridae/virología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Enfermedad Crónica , Modelos Animales de Enfermedad , Femenino , Factor Nuclear 1-alfa del Hepatocito/deficiencia , Factor Nuclear 1-alfa del Hepatocito/genética , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones Noqueados , Fenotipo , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Factores de Tiempo
12.
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
13.
Cell Rep ; 28(7): 1758-1772.e4, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31412245

RESUMEN

Following infection, inflammatory cues upregulate core transcriptional programs to establish pathogen-specific protection. In viral infections, T follicular helper (TFH) cells express the prototypical T helper 1 transcription factor T-bet. Several studies have demonstrated essential but conflicting roles for T-bet in TFH biology. Understanding the basis of this controversy is crucial, as modulation of T-bet expression instructs TFH differentiation and ultimately protective antibody responses. Comparing influenza and LCMV viral infections, we demonstrate that the role of T-bet is contingent on the environmental setting of TFH differentiation, IL-2 signaling, and T cell competition. Furthermore, we demonstrate that T-bet expression by either TFH or GC B cells independently drives antibody isotype class switching. Specifically, T cell-specific loss of T-bet promotes IgG1, whereas B cell-specific loss of T-bet inhibits IgG2a/c switching. Combined, this work highlights that the context-dependent induction of T-bet instructs the development of protective, neutralizing antibodies following viral infection or vaccination.


Asunto(s)
Formación de Anticuerpos/inmunología , Diferenciación Celular , Centro Germinal/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Orthomyxoviridae/inmunología , Proteínas de Dominio T Box/fisiología , Linfocitos T Colaboradores-Inductores/citología , Animales , Anticuerpos Antivirales/inmunología , Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/metabolismo , Infecciones por Arenaviridae/virología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/virología , Femenino , Centro Germinal/metabolismo , Centro Germinal/virología , Inmunoglobulina G/metabolismo , Activación de Linfocitos , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/metabolismo , Coriomeningitis Linfocítica/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Transducción de Señal , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/virología
14.
PLoS Pathog ; 14(7): e1007125, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30001425

RESUMEN

Several arenaviruses cause hemorrhagic fever (HF) diseases that are associated with high morbidity and mortality in humans. Accordingly, HF arenaviruses have been listed as top-priority emerging diseases for which countermeasures are urgently needed. Because arenavirus nucleoprotein (NP) plays critical roles in both virus multiplication and immune-evasion, we used an unbiased proteomic approach to identify NP-interacting proteins in human cells. DDX3, a DEAD-box ATP-dependent-RNA-helicase, interacted with NP in both NP-transfected and virus-infected cells. Importantly, DDX3 deficiency compromised the propagation of both Old and New World arenaviruses, including the HF arenaviruses Lassa and Junin viruses. The DDX3 role in promoting arenavirus multiplication associated with both a previously un-recognized DDX3 inhibitory role in type I interferon production in arenavirus infected cells and a positive DDX3 effect on arenavirus RNA synthesis that was dependent on its ATPase and Helicase activities. Our results uncover novel mechanisms used by arenaviruses to exploit the host machinery and subvert immunity, singling out DDX3 as a potential host target for developing new therapies against highly pathogenic arenaviruses.


Asunto(s)
Infecciones por Arenaviridae/metabolismo , ARN Helicasas DEAD-box/metabolismo , Interacciones Huésped-Patógeno/fisiología , Evasión Inmune/inmunología , Replicación Viral/fisiología , Infecciones por Arenaviridae/inmunología , Arenavirus , Línea Celular , ARN Helicasas DEAD-box/inmunología , Humanos , Interferón Tipo I/biosíntesis , Interferón Tipo I/inmunología , Proteínas del Núcleo Viral/inmunología , Proteínas del Núcleo Viral/metabolismo
15.
PLoS Pathog ; 14(4): e1006985, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29672594

RESUMEN

Regulatory T cells (Tregs) play a cardinal role in the immune system by suppressing detrimental autoimmune responses, but their role in acute, chronic infectious diseases and tumor microenvironment remains unclear. We recently demonstrated that IFN-α/ß receptor (IFNAR) signaling promotes Treg function in autoimmunity. Here we dissected the functional role of IFNAR-signaling in Tregs using Treg-specific IFNAR deficient (IFNARfl/flxFoxp3YFP-Cre) mice in acute LCMV Armstrong, chronic Clone-13 viral infection, and in tumor models. In both viral infection and tumor models, IFNARfl/flxFoxp3YFP-Cre mice Tregs expressed enhanced Treg associated activation antigens. LCMV-specific CD8+ T cells and tumor infiltrating lymphocytes from IFNARfl/flxFoxp3YFP-Cre mice produced less antiviral and antitumor IFN-γ and TNF-α. In chronic viral model, the numbers of antiviral effector and memory CD8+ T cells were decreased in IFNARfl/flxFoxp3YFP-Cre mice and the effector CD4+ and CD8+ T cells exhibited a phenotype compatible with enhanced exhaustion. IFNARfl/flxFoxp3YFP-Cre mice cleared Armstrong infection normally, but had higher viral titers in sera, kidneys and lungs during chronic infection, and higher tumor burden than the WT controls. The enhanced activated phenotype was evident through transcriptome analysis of IFNARfl/flxFoxp3YFP-Cre mice Tregs during infection demonstrated differential expression of a unique gene signature characterized by elevated levels of genes involved in suppression and decreased levels of genes mediating apoptosis. Thus, IFN signaling in Tregs is beneficial to host resulting in a more effective antiviral response and augmented antitumor immunity.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Neoplasias del Colon/inmunología , Interferón Tipo I/farmacología , Coriomeningitis Linfocítica/inmunología , Melanoma Experimental/inmunología , Linfocitos T Reguladores/inmunología , Microambiente Tumoral/inmunología , Animales , Antivirales/farmacología , Infecciones por Arenaviridae/tratamiento farmacológico , Infecciones por Arenaviridae/metabolismo , Infecciones por Arenaviridae/virología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Neoplasias del Colon/virología , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/inmunología , Interferón gamma/metabolismo , Coriomeningitis Linfocítica/tratamiento farmacológico , Coriomeningitis Linfocítica/metabolismo , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/efectos de los fármacos , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/metabolismo , Melanoma Experimental/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Interferón alfa y beta/fisiología , Transducción de Señal/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/virología , Microambiente Tumoral/efectos de los fármacos
16.
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
17.
J Virol ; 92(5)2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29237830

RESUMEN

Fatal infection with the highly pathogenic Lassa virus (LASV) is characterized by extensive viral dissemination, indicating broad tissue tropism. The major cellular receptor for LASV is the highly conserved extracellular matrix receptor dystroglycan (DG). Binding of LASV depends on DG's tissue-specific posttranslational modification with the unusual O-linked polysaccharide matriglycan. Interestingly, functional glycosylation of DG does not always correlate with viral tropism observed in vivo The broadly expressed phosphatidylserine (PS) receptors Axl and Tyro3 were recently identified as alternative LASV receptor candidates. However, their role in LASV entry is not entirely understood. Here, we examine LASV receptor candidates in primary human cells and found coexpression of Axl with differentially glycosylated DG. To study LASV receptor use in the context of productive arenavirus infection, we employed recombinant lymphocytic choriomeningitis virus expressing LASV glycoprotein (rLCMV-LASV GP) as a validated biosafety level 2 (BSL2) model. We confirm and extend previous work showing that Axl can contribute to LASV entry in the absence of functional DG using "apoptotic mimicry" in a way similar to that of other enveloped viruses. We further show that Axl-dependent LASV entry requires receptor activation and involves a pathway resembling macropinocytosis. Axl-mediated LASV entry is facilitated by heparan sulfate and critically depends on the late endosomal protein LAMP-1 as an intracellular entry factor. In endothelial cells expressing low levels of functional DG, both receptors are engaged by the virus and can contribute to productive entry. In sum, we characterize the role of Axl in LASV entry and provide a rationale for targeting Axl in antiviral therapy.IMPORTANCE The highly pathogenic arenavirus Lassa virus (LASV) represents a serious public health problem in Africa. Although the principal LASV receptor, dystroglycan (DG), is ubiquitously expressed, virus binding critically depends on DG's posttranslational modification, which does not always correlate with tissue tropism. The broadly expressed phosphatidylserine receptor Axl was recently identified as an alternative LASV receptor candidate, but its role in LASV entry is unclear. Here, we investigate the exact role of Axl in LASV entry as a function of DG's posttranslational modification. We found that in the absence of functional DG, Axl can mediate LASV entry via apoptotic mimicry. Productive entry requires virus-induced receptor activation, involves macropinocytosis, and critically depends on LAMP-1. In endothelial cells that express low levels of glycosylated DG, both receptors can promote LASV entry. In sum, our study defines the roles of Axl in LASV entry and provides a rationale for targeting Axl in antiviral therapy.


Asunto(s)
Distroglicanos/metabolismo , Virus Lassa/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores Virales/metabolismo , Acoplamiento Viral , Internalización del Virus , Células A549 , Antivirales/farmacología , Infecciones por Arenaviridae/metabolismo , Línea Celular Tumoral , Distroglicanos/genética , Endosomas/metabolismo , Expresión Génica , Glicosilación , Células HEK293 , Células HeLa , Heparitina Sulfato/farmacología , Humanos , Virus Lassa/efectos de los fármacos , Virus Lassa/patogenicidad , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/metabolismo , Proteínas de Membrana de los Lisosomas/metabolismo , Pinocitosis/fisiología , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Proteínas Tirosina Quinasas Receptoras/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal , Tropismo , Tirosina Quinasa del Receptor Axl
18.
Nat Immunol ; 18(9): 1046-1057, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28714979

RESUMEN

Translation is a critical process in protein synthesis, but translational regulation in antigen-specific T cells in vivo has not been well defined. Here we have characterized the translatome of virus-specific CD8+ effector T cells (Teff cells) during acute infection of mice with lymphocytic choriomeningitis virus (LCMV). Antigen-specific T cells exerted dynamic translational control of gene expression that correlated with cell proliferation and stimulation via the T cell antigen receptor (TCR). The translation of mRNAs that encode translation machinery, including ribosomal proteins, was upregulated during the T cell clonal-expansion phase, followed by inhibition of the translation of those transcripts when the CD8+ Teff cells stopped dividing just before the contraction phase. That translational suppression was more pronounced in terminal effector cells than in memory precursor cells and was regulated by antigenic stimulation and signals from the kinase mTOR. Our studies show that translation of transcripts encoding ribosomal proteins is regulated during the differentiation of CD8+ Teff cells and might have a role in fate 'decisions' involved in the formation of memory cells.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Biosíntesis de Proteínas/inmunología , Animales , Infecciones por Arenaviridae/genética , Infecciones por Arenaviridae/metabolismo , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/genética , Citometría de Flujo , Regulación de la Expresión Génica , Memoria Inmunológica/inmunología , Interferón gamma/inmunología , Virus de la Coriomeningitis Linfocítica , Ratones , Biosíntesis de Proteínas/genética , ARN Mensajero/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Serina-Treonina Quinasas TOR/inmunología
19.
Microb Pathog ; 104: 17-27, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28062291

RESUMEN

Boid inclusion body disease (BIBD) is a viral disease of boid snakes believed to be caused by reptarenavirus belonging to the family Arenaviridae. Unlike most mammalian arenaviruses, the reservoir host for reptarenavirus is still unknown. In this study, the pathological responses were evaluated in a mouse model for a period of 28 days. Blood and tissue samples (lung, liver, spleen, heart, kidney and brain) were collected for evaluation of hematology, biochemistry, histopathology and oxidative enzyme levels at six time points (1, 3, 7, 14, 21 and 28 days), after viral infection (2.0 × 106 pfu/mL) in the infected and normal saline in the control groups. An initial increase (p < 0.05) in white blood cell (WBC), neutrophil and lymphocyte counts were observed in the infected group at day 3 post infection, and a decline (p < 0.05) on day 7 and 4 post infection. Significant (p < 0.05) increases in alanine transaminase (ALT), aspartate transaminase (AST), creatinine, total protein and globulin levels were also observed in the infected group. An increased (p < 0.05) level of hydrogen peroxide, total antioxidant capacity (TAC), superoxide dismutase (SOD) activity and catalase activity (CAT) were frequently observed on different days in the infected group. The MDA activity was increased (p < 0.05) in the infected group on day 7 and 14. Histopathological changes observed in the liver, kidney, spleen, brain and lungs were mainly associated with degeneration, necrosis and infiltration of lymphocytes. Viral counts were low on days 7 and 14 but surged in both the liver and spleen on day 21 and 28. This study has shown that reptarenavirus replicates in mammalian host and induces oxidative stress. Furthermore, the resultant hematobiochemical and histopathological changes observed in infected mice were similar to what has been reported in mammarenavirus infections. This suggests that rodents may serve as potential reservoir hosts for reptarenavirus.


Asunto(s)
Infecciones por Arenaviridae/metabolismo , Arenaviridae , Estrés Oxidativo , Alanina Transaminasa , Enfermedades de los Animales/genética , Enfermedades de los Animales/metabolismo , Enfermedades de los Animales/patología , Enfermedades de los Animales/virología , Animales , Antioxidantes/metabolismo , Infecciones por Arenaviridae/genética , Infecciones por Arenaviridae/patología , Infecciones por Arenaviridae/virología , Biomarcadores , Catalasa , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Peróxido de Hidrógeno/metabolismo , Peroxidación de Lípido , Hígado/patología , Hígado/virología , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Especies Reactivas de Oxígeno , Bazo/patología , Bazo/virología , Superóxido Dismutasa/metabolismo , Células Vero , Carga Viral
20.
Immunity ; 45(5): 1024-1037, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27836431

RESUMEN

Extensive metabolic changes accompany T cell activation, including a switch to glycolytic energy production and increased biosynthesis. Recent studies suggest that subsequent return to reliance on oxidative phosphorylation and increasing spare respiratory capacity are essential for the differentiation of memory CD8+ T cells. In contrast, we found that constitutive glycolytic metabolism and suppression of oxidative phosphorylation in CD8+ T cells, achieved by conditional deletion of hypoxia-inducible factor regulator Vhl, accelerated CD8+ memory cell differentiation during viral infection. Despite sustained glycolysis, CD8+ memory cells emerged that upregulated key memory-associated cytokine receptors and transcription factors and showed a heightened response to secondary challenge. In addition, increased glycolysis not only permitted memory formation, but it also favored the formation of long-lived effector-memory CD8+ T cells. These data redefine the role of cellular metabolism in memory cell differentiation, showing that reliance on glycolytic metabolism does not hinder formation of a protective memory population.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Linfocitos T CD8-positivos/inmunología , Glucólisis/inmunología , Memoria Inmunológica/inmunología , Activación de Linfocitos/inmunología , Animales , Infecciones por Arenaviridae/metabolismo , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Separación Celular , Modelos Animales de Enfermedad , Citometría de Flujo , Virus de la Coriomeningitis Linfocítica , Ratones , Ratones Transgénicos , Fosforilación Oxidativa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA