Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 15(4): e1007674, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30958867

RESUMEN

Viral myocarditis is a serious disease, commonly caused by type B coxsackieviruses (CVB). Here we show that innate immune protection against CVB3 myocarditis requires the IFIT (IFN-induced with tetratricopeptide) locus, which acts in a biphasic manner. Using IFIT locus knockout (IFITKO) cardiomyocytes we show that, in the absence of the IFIT locus, viral replication is dramatically increased, indicating that constitutive IFIT expression suppresses CVB replication in this cell type. IFNß pre-treatment strongly suppresses CVB3 replication in wild type (wt) cardiomyocytes, but not in IFITKO cardiomyocytes, indicating that other interferon-stimulated genes (ISGs) cannot compensate for the loss of IFITs in this cell type. Thus, in isolated wt cardiomyocytes, the anti-CVB3 activity of IFITs is biphasic, being required for protection both before and after T1IFN signaling. These in vitro findings are replicated in vivo. Using novel IFITKO mice we demonstrate accelerated CVB3 replication in pancreas, liver and heart in the hours following infection. This early increase in virus load in IFITKO animals accelerates the induction of other ISGs in several tissues, enhancing virus clearance from some tissues, indicating that-in contrast to cardiomyocytes-other ISGs can offset the loss of IFITs from those cell types. In contrast, CVB3 persists in IFITKO hearts, and myocarditis occurs. Thus, cardiomyocytes have a specific, biphasic, and near-absolute requirement for IFITs to control CVB infection.


Asunto(s)
Proteínas Portadoras/fisiología , Infecciones por Coxsackievirus/prevención & control , Enterovirus Humano B/patogenicidad , Miocarditis/prevención & control , Miocitos Cardíacos/enzimología , Proteínas Adaptadoras Transductoras de Señales , Animales , Células Cultivadas , Infecciones por Coxsackievirus/metabolismo , Infecciones por Coxsackievirus/virología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocarditis/metabolismo , Miocarditis/virología , Proteínas de Unión al ARN , Replicación Viral
2.
PLoS Pathog ; 12(8): e1005861, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27580079

RESUMEN

Innate immune responses in general, and type I interferons (T1IFNs) in particular, play an important and often essential role during primary viral infections, by directly combatting the virus and by maximizing the primary adaptive immune response. Several studies have suggested that T1IFNs also contribute very substantially to the secondary (recall) response; they are thought (i) to be required to drive the early attrition of memory T cells, (ii) to support the subsequent expansion of surviving virus-specific memory cells, and (iii) to assist in the suppression and clearance of the infectious agent. However, many of these observations were predicated upon models in which T1IFN signaling was interrupted prior to a primary immune response, raising the possibility that the resulting memory cells might be intrinsically abnormal. We have directly addressed this by using an inducible-Cre model system in which the host remains genetically-intact during the primary response to infection, and in which T1IFN signaling can be effectively ablated prior to secondary viral challenge. We report that, in stark contrast to primary infection, T1IFN signaling is not required during the recall response. IFNαßR-deficient memory CD8+ and CD4+ memory T cells undergo attrition and expansion with kinetics that are indistinguishable from those of receptor-sufficient cells. Moreover, even in the absence of functional T1IFN signaling, the host's immune capacity to rapidly suppress, and then to eradicate, a secondary infection remains intact. Thus, this study shows that T1IFN signaling is dispensable during the recall response to a virus infection. Moreover, two broader implications may be drawn. First, a T cell's requirement for a cytokine is highly dependent on the cell's maturation / differentiation status. Consequently, second, these data underscore the importance of evaluating a gene's impact by modulating its expression or function in a temporally-controllable manner.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Interferón Tipo I/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Transducción de Señal/inmunología , Animales , Interferón Tipo I/genética , Coriomeningitis Linfocítica/genética , Ratones , Ratones Transgénicos , Transducción de Señal/genética
3.
PLoS Pathog ; 10(4): e1004045, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24722773

RESUMEN

Coxsackievirus B3 (CVB3), a member of the picornavirus family and enterovirus genus, causes viral myocarditis, aseptic meningitis, and pancreatitis in humans. We genetically engineered a unique molecular marker, "fluorescent timer" protein, within our infectious CVB3 clone and isolated a high-titer recombinant viral stock (Timer-CVB3) following transfection in HeLa cells. "Fluorescent timer" protein undergoes slow conversion of fluorescence from green to red over time, and Timer-CVB3 can be utilized to track virus infection and dissemination in real time. Upon infection with Timer-CVB3, HeLa cells, neural progenitor and stem cells (NPSCs), and C2C12 myoblast cells slowly changed fluorescence from green to red over 72 hours as determined by fluorescence microscopy or flow cytometric analysis. The conversion of "fluorescent timer" protein in HeLa cells infected with Timer-CVB3 could be interrupted by fixation, suggesting that the fluorophore was stabilized by formaldehyde cross-linking reactions. Induction of a type I interferon response or ribavirin treatment reduced the progression of cell-to-cell virus spread in HeLa cells or NPSCs infected with Timer-CVB3. Time lapse photography of partially differentiated NPSCs infected with Timer-CVB3 revealed substantial intracellular membrane remodeling and the assembly of discrete virus replication organelles which changed fluorescence color in an asynchronous fashion within the cell. "Fluorescent timer" protein colocalized closely with viral 3A protein within virus replication organelles. Intriguingly, infection of partially differentiated NPSCs or C2C12 myoblast cells induced the release of abundant extracellular microvesicles (EMVs) containing matured "fluorescent timer" protein and infectious virus representing a novel route of virus dissemination. CVB3 virions were readily observed within purified EMVs by transmission electron microscopy, and infectious virus was identified within low-density isopycnic iodixanol gradient fractions consistent with membrane association. The preferential detection of the lipidated form of LC3 protein (LC3 II) in released EMVs harboring infectious virus suggests that the autophagy pathway plays a crucial role in microvesicle shedding and virus release, similar to a process previously described as autophagosome-mediated exit without lysis (AWOL) observed during poliovirus replication. Through the use of this novel recombinant virus which provides more dynamic information from static fluorescent images, we hope to gain a better understanding of CVB3 tropism, intracellular membrane reorganization, and virus-associated microvesicle dissemination within the host.


Asunto(s)
Micropartículas Derivadas de Células/virología , Enterovirus Humano B/fisiología , Infecciones por Enterovirus/metabolismo , Fagosomas/virología , Esparcimiento de Virus/fisiología , Animales , Micropartículas Derivadas de Células/genética , Micropartículas Derivadas de Células/metabolismo , Infecciones por Enterovirus/genética , Células HeLa , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Fagosomas/genética , Fagosomas/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo
4.
Transgenic Res ; 25(5): 639-48, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27165291

RESUMEN

Transgenic mice expressing a tamoxifen-inducible Cre recombinase specifically in cardiomyocytes were generated in 2001 and are in widespread use, having been employed in >150 published studies. However, several groups recently have reported that tamoxifen administration to these mice can have off-target effects that include cardiac dysfunction, fibrosis, and death. For this reason, among others, we considered it important to better characterize the transgene (termed herein, CM-MCM) and its chromosomal location(s). Cytogenetic analysis positioned the CM-MCM transgene within the C band of chromosome 19, and more precise mapping, using genome walking and DNA sequencing, showed that transgene insertion is in the C1 region. Using the genome walking data, we have developed PCR assays that not only identify mice that carry the transgene, but also distinguish homozygous animals (CM-MCM(Tg/Tg)) from hemizygous (CM-MCM(Tg/0)), permitting the rapid assessment of transgene zygosity and, thereby, helping to minimize off-target tamoxifen-induced effects. Substantial rearrangement/duplication of transgene elements is present, and transgene integration was accompanied by the deletion of a 19,500 bp fragment of genomic DNA that contains the promoter, exon 1 and part of intron 1 of the APOBEC1 complementation factor (A1cf) gene, as well as several elements that are predicted to regulate chromosomal architecture. A1cf protein expression is ablated by the deletion and, therefore, homozygous mice are functionally A1cf knockout. The implications of this unexpected finding are discussed.


Asunto(s)
Desaminasas APOBEC-1/genética , Genoma/efectos de los fármacos , Genómica , Integrasas/genética , Animales , Mapeo Cromosómico , Ratones , Ratones Transgénicos , Proteínas de Mantenimiento de Minicromosoma/genética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Regiones Promotoras Genéticas , Análisis de Secuencia de ADN , Eliminación de Secuencia/genética , Tamoxifeno/farmacología
5.
J Immunol ; 193(4): 1873-85, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25015828

RESUMEN

In vitro studies have shown that naive CD8(+) T cells are unable to express most of their effector proteins until after at least one round of cell division has taken place. We have reassessed this issue in vivo and find that naive CD8(+) T cells mount Ag-specific responses within hours of infection, before proliferation has commenced. Newly activated naive Ag-specific CD8(+) T cells produce a rapid pulse of IFN-γ in vivo and begin to accumulate granzyme B and perforin. Later, in vivo cytolytic activity is detectable, coincident with the initiation of cell division. Despite the rapid development of these functional attributes, no antiviral effect was observed early during infection, even when the cells are present in numbers similar to those of virus-specific memory cells. The evolutionary reason for the pulse of IFN-γ synthesis by naive T cells is uncertain, but the lack of antiviral impact suggests that it may be regulatory.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interferón gamma/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/trasplante , Diferenciación Celular/inmunología , División Celular/inmunología , Granzimas/biosíntesis , Memoria Inmunológica/inmunología , Interferón gamma/biosíntesis , Activación de Linfocitos/inmunología , Coriomeningitis Linfocítica/virología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Perforina/biosíntesis , Proteínas de Dominio T Box/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis , Replicación Viral/inmunología
6.
J Virol ; 88(9): 5087-99, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24574394

RESUMEN

UNLABELLED: Acute coxsackievirus B3 (CVB3) infection is one of the most prevalent causes of acute myocarditis, a disease that frequently is identified only after the sudden death of apparently healthy individuals. CVB3 infects cardiomyocytes, but the infection is highly focal, even in the absence of a strong adaptive immune response, suggesting that virus spread within the heart may be tightly constrained by the innate immune system. Type I interferons (T1IFNs) are an obvious candidate, and T1IFN receptor (T1IFNR) knockout mice are highly susceptible to CVB3 infection, succumbing within a few days of challenge. Here, we investigated the role of T1IFNs in the heart using a mouse model in which the T1IFNR gene can be ablated in vivo, specifically in cardiomyocytes. We found that T1IFN signaling into cardiomyocytes contributed substantially to the suppression of viral replication and infectious virus yield in the heart; in the absence of such signaling, virus titers were markedly elevated by day 3 postinfection (p.i.) and remained high at day 12 p.i., a time point at which virus was absent from genetically intact littermates, suggesting that the T1IFN-unresponsive cardiomyocytes may act as a safe haven for the virus. Nevertheless, in these mice the myocardial infection remained highly focal, despite the cardiomyocytes' inability to respond to T1IFN, indicating that other factors, as yet unidentified, are sufficient to prevent the more widespread dissemination of the infection throughout the heart. The absence of T1IFN signaling into cardiomyocytes also was accompanied by a profound acceleration and exacerbation of myocarditis and by a significant increase in mortality. IMPORTANCE: Acute coxsackievirus B3 (CVB3) infection is one of the most common causes of acute myocarditis, a serious and sometimes fatal disease. To optimize treatment, it is vital that we identify the immune factors that limit virus spread in the heart and other organs. Type I interferons play a key role in controlling many virus infections, but it has been suggested that they may not directly impact CVB3 infection within the heart. Here, using a novel line of transgenic mice, we show that these cytokines signal directly into cardiomyocytes, limiting viral replication, myocarditis, and death.


Asunto(s)
Infecciones por Coxsackievirus/inmunología , Infecciones por Coxsackievirus/virología , Enterovirus Humano B/inmunología , Miocarditis/inmunología , Miocarditis/virología , Miocitos Cardíacos/virología , Receptor de Interferón alfa y beta/inmunología , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Ratones Transgénicos , Miocarditis/patología , Miocitos Cardíacos/fisiología , Receptor de Interferón alfa y beta/deficiencia , Análisis de Supervivencia
7.
J Immunol ; 191(8): 4211-22, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24026080

RESUMEN

CD8(+) memory T cells are abundant and are activated in a near-synchronous manner by infection, thereby providing a unique opportunity to evaluate the coordinate functional and phenotypic changes that occur in vivo within hours of viral challenge. Using two disparate virus challenges of mice, we show that splenic CD8(+) memory T cells rapidly produced IFN-γ in vivo; however, within 18-24 h, IFN-γ synthesis was terminated and remained undetectable for ≥ 48 h. A similar on/off response was observed in CD8(+) memory T cells in the peritoneal cavity. Cessation of IFN-γ production in vivo occurred despite the continued presence of immunostimulatory viral Ag, indicating that the initial IFN-γ response had been actively downregulated and that the cells had been rendered refractory to subsequent in vivo Ag contact. Downregulation of IFN-γ synthesis was accompanied by the upregulation of inhibitory receptor expression on the T cells, and ex vivo analyses using synthetic peptides revealed a concurrent hierarchical loss of cytokine responsiveness (IL-2, then TNF, then IFN-γ) taking place during the first 24 h following Ag contact. Thus, within hours of virus challenge, CD8(+) memory T cells display the standard hallmarks of T cell exhaustion, a phenotype that previously was associated only with chronic diseases and that is generally viewed as a gradually developing and pathological change in T cell function. Our data suggest that, instead, the "exhaustion" phenotype is a rapid and normal physiological T cell response.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Receptores Coestimuladores e Inhibidores de Linfocitos T/biosíntesis , Receptores Coestimuladores e Inhibidores de Linfocitos T/inmunología , Regulación hacia Abajo , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Poxviridae/inmunología , Bazo/citología , Bazo/inmunología , Factores de Necrosis Tumoral/biosíntesis , Regulación hacia Arriba , Virus Vaccinia/genética , Virus Vaccinia/inmunología
8.
J Immunol ; 191(11): 5655-68, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24146043

RESUMEN

The contraction phase of the T cell response is a poorly understood period after the resolution of infection when virus-specific effector cells decline in number and memory cells emerge with increased frequencies. CD8(+) T cells plummet in number and quickly reach stable levels of memory following acute lymphocytic choriomeningitis virus infection in mice. In contrast, virus-specific CD4(+) T cells gradually decrease in number and reach homeostatic levels only after many weeks. In this study, we provide evidence that MHCII-restricted viral Ag persists during the contraction phase following this prototypical acute virus infection. We evaluated whether the residual Ag affected the cell division and number of virus-specific naive and memory CD4(+) T cells and CD8(+) T cells. We found that naive CD4(+) T cells underwent cell division and accumulated in response to residual viral Ag for >2 mo after the eradication of infectious virus. Surprisingly, memory CD4(+) T cells did not undergo cell division in response to the lingering Ag, despite their heightened capacity to recognize Ag and make cytokine. In contrast to CD4(+) T cells, CD8(+) T cells did not undergo cell division in response to the residual Ag. Thus, CD8(+) T cells ceased division within days after the infection was resolved, indicating that CD8(+) T cell responses are tightly linked to endogenous processing of de novo synthesized virus protein. Our data suggest that residual viral Ag delays the contraction of CD4(+) T cell responses by recruiting new populations of CD4(+) T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Animales , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/virología , Proliferación Celular , Células Cultivadas , Reactividad Cruzada , Antígenos de Histocompatibilidad Clase II/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Unión Proteica
9.
J Virol ; 87(5): 2823-34, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23269810

RESUMEN

Coxsackievirus B3 (CVB3) is known to infect stem cells in the neonatal central nervous system. Here, we evaluated the effects of CVB3 infection on the major source and repository of stem cells, the bone marrow (BM). Viral genome was detectable in BM within 24 h of infection, and productive infection of BM cells was evident, peaking at 48 h postinfection (p.i.), when ∼1 to 2% of BM cells produced infectious virus particles. Beginning at 2 to 3 days p.i., a dramatic and persistent loss of immature erythroid cells, B and T lymphocytes, and neutrophils was observed in BM and, by day 3 to 4 p.i., the femoral BM stroma was largely destroyed. Analysis of peripheral blood revealed a modest neutrophilia, a loss of reticulocytes, and a massive lymphopenia. The abundance of multipotent progenitor cells (Lin(-)/c-kit(+)/Flt3(+)) in BM declined ∼10-fold during CVB3 infection and, consistent with a deficiency of primitive hematopoietic progenitors, serum levels of the hematopoietic growth factor Flt3 ligand were dramatically elevated. Therefore, we analyzed the regenerative capacity of BM from CVB3-infected mice. Granulocyte/macrophage progenitors displayed a relatively normal proliferative ability, consistent with the fact that the peripheral blood level of neutrophils-which are very short-lived cells-remained high throughout infection. However, erythroid and lymphoid hematopoietic progenitors in BM from CVB3-infected mice showed a markedly reduced colony-forming capacity, consonant with the observed loss of both lymphocytes and immature erythroid cells/reticulocytes from the BM and peripheral blood. In summary, CVB3 infects the BM and exerts differential effects on the various hematopoietic progenitor populations.


Asunto(s)
Células de la Médula Ósea/virología , Infecciones por Coxsackievirus/virología , Enterovirus Humano B/patogenicidad , Células Precursoras Eritroides/fisiología , Células Progenitoras Linfoides/fisiología , Traslado Adoptivo , Animales , Linfocitos B , Recuento de Células Sanguíneas , Médula Ósea/patología , Médula Ósea/virología , Proliferación Celular , Ensayo de Unidades Formadoras de Colonias , Infecciones por Coxsackievirus/sangre , Citocinas/sangre , Hematopoyesis , Recuento de Linfocitos , Linfopenia/virología , Macrófagos/virología , Masculino , Proteínas de la Membrana/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos , Reticulocitos , Linfocitos T
10.
Am J Pathol ; 180(3): 1107-1120, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22214838

RESUMEN

Coxsackieviruses are significant human pathogens causing myocarditis, meningitis, and encephalitis. We previously demonstrated the ability of coxsackievirus B3 (CVB3) to persist within the neonatal central nervous system (CNS) and to target neural stem cells. Given that CVB3 is a cytolytic virus and may therefore damage target cells, we characterized the potential reduction in neurogenesis within the developing brain and the subsequent developmental defects that occurred after the loss of these essential neural stem cells. Neonatal mice were inoculated with a recombinant CVB3 expressing eGFP (eGFP-CVB3), and alterations in neurogenesis and brain development were evaluated over time. We observed a reduction in proliferating cells in CNS neurogenic regions simultaneously with the presence of nestin(+) cells undergoing apoptosis. The size of the brain appeared smaller by histology, and a permanent decrease in brain wet weight was observed after eGFP-CVB3 infection. We also observed an inverse relationship between the amount of virus material and brain wet weight up to day 30 postinfection. In addition, signs of astrogliosis and a compaction of the cortical layers were observed at 90 days postinfection. Intriguingly, partial brain wet weight recovery was observed in mice treated with the antiviral drug ribavirin during the persistent stage of infection. Hence, long-term neurological sequelae might be expected after neonatal enteroviral infections, yet antiviral treatment initiated long after the end of acute infection might limit virus-mediated neuropathology.


Asunto(s)
Sistema Nervioso Central/virología , Infecciones por Coxsackievirus/complicaciones , Enterovirus Humano B , Células-Madre Neurales/virología , Neurogénesis/fisiología , Animales , Animales Recién Nacidos , Antivirales/farmacología , Apoptosis/fisiología , Astrocitos/virología , Encéfalo/crecimiento & desarrollo , Encéfalo/virología , División Celular , Proliferación Celular , Sistema Nervioso Central/crecimiento & desarrollo , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Endogámicos BALB C , Tamaño de los Órganos , Proteínas Recombinantes , Carga Viral
11.
J Neurosci ; 31(16): 6247-54, 2011 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-21508247

RESUMEN

Tissue inhibitor of metalloproteinase-1 (TIMP-1) is an extracellular protein and endogenous regulator of matrix metalloproteinases (MMPs) secreted by astrocytes in response to CNS myelin injury. We have previously reported that adult TIMP-1 knock-out (KO) mice exhibit poor myelin repair following demyelinating injury. This observation led us to hypothesize a role for TIMP-1 in oligodendrogenesis and CNS myelination. Herein, we demonstrate that compact myelin formation is significantly delayed in TIMP-1 KO mice, a situation that coincided with dramatically reduced numbers of white matter astrocytes in the developing CNS. Analysis of differentiation in CNS progenitor cells (neurosphere) cultures from TIMP-1 KO mice revealed a specific deficit of NG2(+) oligodendrocyte progenitor cells. Application of recombinant murine TIMP-1 (rmTIMP-1) to TIMP-1 KO neurosphere cultures evoked a dose-dependent increase in NG2(+) cell numbers, while treatment with GM6001, a potent broad-spectrum MMP inhibitor did not. Similarly, administration of rmTIMP-1 to A2B5(+) immunopanned oligodendrocyte progenitors significantly increased the number of differentiated O1(+) oligodendrocytes, while antisera to TIMP-1 reduced oligodendrocyte numbers. We also determined that A2B5(+) oligodendrocyte progenitors grown in conditioned media derived from TIMP-1 KO primary glial cultures resulted in reduced differentiation of mature O1(+) oligodendrocytes. Finally, we report that addition of rmTIMP-1 to primary glial cultures resulted in a dose-dependent proliferative response of astrocytes. Together, these findings describe a previously uncharacterized role for TIMP-1 in the regulation of oligodendrocytes and astrocytes during development and provide a novel function for TIMP-1 on myelination in the developing CNS.


Asunto(s)
Astrocitos/metabolismo , Diferenciación Celular/fisiología , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Análisis de Varianza , Animales , Astrocitos/citología , Células Cultivadas , Técnicas de Cocultivo , Ensayo de Inmunoadsorción Enzimática , Inmunohistoquímica , Ratones , Ratones Noqueados , Microscopía Electrónica , Oligodendroglía/citología , Inhibidor Tisular de Metaloproteinasa-1/genética
12.
J Virol ; 85(12): 5718-32, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21471247

RESUMEN

Enteroviruses, including coxsackieviruses, exhibit significant tropism for the central nervous system, and these viruses are commonly associated with viral meningitis and encephalitis. Previously, we described the ability of coxsackievirus B3 (CVB3) to infect proliferating neuronal progenitor cells located in the neonatal subventricular zone and persist in the adult murine central nervous system (CNS). Here, we demonstrate that cultured murine neurospheres, which comprise neural stem cells and their progeny at different stages of development, were highly susceptible to CVB3 infection. Neurospheres, or neural progenitor and stem cells (NPSCs), isolated from neonatal C57BL/6 mice, supported high levels of infectious virus production and high viral protein expression levels following infection with a recombinant CVB3 expressing enhanced green fluorescent protein (eGFP) protein. Similarly, NPSCs isolated from neonatal actin-promoter-GFP transgenic mice (actin-GFP NPSCs) were highly susceptible to infection with a recombinant CVB3 expressing DsRed (Discosoma sp. red fluorescent protein). Both nestin-positive and NG2(+) progenitor cells within neurospheres were shown to preferentially express high levels of viral protein as soon as 24 h postinfection (p.i.). By day 3 p.i., viral protein expression and viral titers increased dramatically in NPSCs with resultant cytopathic effects (CPE) and eventual cell death. In contrast, reduced viral replication, lower levels of CPE, and diminished viral protein expression levels were observed in NPSCs differentiated for 5 or 16 days in the presence of fetal bovine serum (FBS). Despite the presence of CPE and high levels of cell death following early CVB3 infection, surviving neurospheres were readily observed and continued to express detectable levels of viral protein as long as 37 days after initial infection. Also, CVB3 infection of actin-GFP NPSCs increased the percentage of cells expressing neuronal class III ß-tubulin following their differentiation in the presence of FBS. These results suggest that neural stem cells may be preferentially targeted by CVB3 and that neurogenic regions of the CNS may support persistent viral replication in the surviving host. In addition, normal progenitor cell differentiation may be altered in the host following infection.


Asunto(s)
Diferenciación Celular , Enterovirus Humano B/fisiología , Enterovirus Humano B/patogenicidad , Células-Madre Neurales/virología , Animales , Células Cultivadas , Efecto Citopatogénico Viral , Enterovirus Humano B/genética , Enterovirus Humano B/ultraestructura , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Células-Madre Neurales/citología , Células-Madre Neurales/ultraestructura , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral , Proteína Fluorescente Roja
13.
J Neurosci ; 30(25): 8676-91, 2010 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-20573913

RESUMEN

Enterovirus infection in newborn infants is a significant cause of aseptic meningitis and encephalitis. Using a neonatal mouse model, we previously determined that coxsackievirus B3 (CVB3) preferentially targets proliferating neural stem cells located in the subventricular zone within 24 h after infection. At later time points, immature neuroblasts, and eventually mature neurons, were infected as determined by expression of high levels of viral protein. Here, we show that blood-derived Mac3(+) mononuclear cells were rapidly recruited to the CNS within 12 h after intracranial infection with CVB3. These cells displayed a myeloid-like morphology, were of a peripheral origin based on green fluorescent protein (GFP)-tagged adoptive cell transplant examination, and were highly susceptible to CVB3 infection during their migration into the CNS. Serial immunofluorescence images suggested that the myeloid cells enter the CNS via the choroid plexus, and that they may be infected during their extravasation and passage through the choroid plexus epithelium; these infected myeloid cells ultimately penetrate into the parenchyma of the brain. Before their migration through the ependymal cell layer, a subset of these infected myeloid cells expressed detectable levels of nestin, a marker for neural stem and progenitor cells. As these nestin(+) myeloid cells infected with CVB3 migrated through the ependymal cell layer, they revealed distinct morphological characteristics typical of type B neural stem cells. The recruitment of these novel myeloid cells may be specifically set in motion by the induction of a unique chemokine profile in the CNS induced very early after CVB3 infection, which includes upregulation of CCL12. We propose that intracranial CVB3 infection may lead to the recruitment of nestin(+) myeloid cells into the CNS which might represent an intrinsic host CNS repair response. In turn, the proliferative and metabolic status of recruited myeloid cells may render them attractive targets for CVB3 infection. Moreover, the migratory ability of these myeloid cells may point to a productive method of virus dissemination within the CNS.


Asunto(s)
Infecciones por Coxsackievirus/virología , Células Mieloides/virología , Animales , Animales Recién Nacidos , Plexo Coroideo/inmunología , Plexo Coroideo/virología , Infecciones por Coxsackievirus/inmunología , Técnica del Anticuerpo Fluorescente , Hibridación in Situ , Ratones , Microscopía Confocal , Células Mieloides/inmunología , Neuronas/inmunología , Neuronas/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/inmunología , Células Madre/virología
14.
J Exp Med ; 201(7): 1053-9, 2005 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-15809350

RESUMEN

Interferon-gamma (IFNgamma) is important in regulating the adaptive immune response, and most current evidence suggests that it exerts a negative (proapoptotic) effect on CD8(+) T cell responses. We have developed a novel technique of dual adoptive transfer, which allowed us to precisely compare, in normal mice, the in vivo antiviral responses of two T cell populations that differ only in their expression of the IFNgamma receptor. We use this technique to show that, contrary to expectations, IFNgamma strongly stimulates the development of CD8(+) T cell responses during an acute viral infection. The stimulatory effect is abrogated in T cells lacking the IFNgamma receptor, indicating that the cytokine acts directly upon CD8(+) T cells to increase their abundance during acute viral infection.


Asunto(s)
Traslado Adoptivo/métodos , Infecciones por Arenaviridae/inmunología , Linfocitos T CD8-positivos/metabolismo , Interferón gamma/metabolismo , Virus de la Coriomeningitis Linfocítica , Animales , Linfocitos T CD8-positivos/inmunología , Pruebas Inmunológicas de Citotoxicidad , Citometría de Flujo , Fluorescencia , Interferón gamma/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Bazo/citología , Bazo/inmunología
15.
Eur J Neurosci ; 33(2): 197-204, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21138487

RESUMEN

Autophagy is emerging as a central regulator of cellular health and disease and, in the central nervous system (CNS), this homeostatic process appears to influence synaptic growth and plasticity. Herein, we review the evidence that dysregulation of autophagy may contribute to several neurodegenerative diseases of the CNS. Up-regulation of autophagy may prevent, delay or ameliorate at least some of these disorders, and - based on recent findings from our laboratory - we speculate that this goal may be achieved using a safe, simple and inexpensive approach.


Asunto(s)
Autofagia/fisiología , Inflamación/fisiopatología , Enfermedades Neurodegenerativas/fisiopatología , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/fisiopatología , Restricción Calórica , Infecciones del Sistema Nervioso Central/patología , Infecciones del Sistema Nervioso Central/fisiopatología , Humanos , Inflamación/patología , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Neuronas/fisiología
16.
J Virol ; 84(19): 9947-56, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20668086

RESUMEN

Arenaviruses cause severe human disease ranging from aseptic meningitis following lymphocytic choriomeningitis virus (LCMV) infection to hemorrhagic fever syndromes following infection with Guanarito virus (GTOV), Junin virus (JUNV), Lassa virus (LASV), Machupo virus (MACV), Sabia virus (SABV), or Whitewater Arroyo virus (WWAV). Cellular immunity, chiefly the CD8(+) T-cell response, plays a critical role in providing protective immunity following infection with the Old World arenaviruses LASV and LCMV. In the current study, we evaluated whether HLA class I-restricted epitopes that are cross-reactive among pathogenic arenaviruses could be identified for the purpose of developing an epitope-based vaccination approach that would cross-protect against multiple arenaviruses. We were able to identify a panel of HLA-A*0201-restricted peptides derived from the same region of the glycoprotein precursor (GPC) of LASV (GPC spanning residues 441 to 449 [GPC(441-449)]), LCMV (GPC(447-455)), JUNV (GPC(429-437)), MACV (GPC(444-452)), GTOV (GPC(427-435)), and WWAV (GPC(428-436)) that displayed high-affinity binding to HLA-A*0201 and were recognized by CD8(+) T cells in a cross-reactive manner following LCMV infection or peptide immunization of HLA-A*0201 transgenic mice. Immunization of HLA-A*0201 mice with the Old World peptide LASV GPC(441-449) or LCMV GPC(447-455) induced high-avidity CD8(+) T-cell responses that were able to kill syngeneic target cells pulsed with either LASV GPC(441-449) or LCMV GPC(447-455) in vivo and provided significant protection against viral challenge with LCMV. Through this study, we have demonstrated that HLA class I-restricted, cross-reactive epitopes exist among diverse arenaviruses and that individual epitopes can be utilized as effective vaccine determinants for multiple pathogenic arenaviruses.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/prevención & control , Arenavirus del Viejo Mundo , Vacunas Virales/administración & dosificación , Secuencia de Aminoácidos , Animales , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/virología , Antígenos Virales/genética , Infecciones por Arenaviridae/genética , Arenavirus del Nuevo Mundo/genética , Arenavirus del Nuevo Mundo/inmunología , Arenavirus del Nuevo Mundo/patogenicidad , Arenavirus del Viejo Mundo/genética , Arenavirus del Viejo Mundo/inmunología , Arenavirus del Viejo Mundo/patogenicidad , Linfocitos T CD8-positivos/inmunología , Reacciones Cruzadas , Citotoxicidad Inmunológica , Epítopos/administración & dosificación , Epítopos/genética , Antígenos HLA-A/genética , Antígeno HLA-A2 , Humanos , Virus Lassa/genética , Virus Lassa/inmunología , Virus Lassa/patogenicidad , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones , Ratones Transgénicos , Vacunas Virales/genética , Vacunas Virales/inmunología
17.
J Virol ; 84(23): 12110-24, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20861268

RESUMEN

Autophagy can play an important part in protecting host cells during virus infection, and several viruses have developed strategies by which to evade or even exploit this homeostatic pathway. Tissue culture studies have shown that poliovirus, an enterovirus, modulates autophagy. Herein, we report on in vivo studies that evaluate the effects on autophagy of coxsackievirus B3 (CVB3). We show that in pancreatic acinar cells, CVB3 induces the formation of abundant small autophagy-like vesicles and permits amphisome formation. However, the virus markedly, albeit incompletely, limits the fusion of autophagosomes (and/or amphisomes) with lysosomes, and, perhaps as a result, very large autophagy-related structures are formed within infected cells; we term these structures megaphagosomes. Ultrastructural analyses confirmed that double-membraned autophagy-like vesicles were present in infected pancreatic tissue and that the megaphagosomes were related to the autophagy pathway; they also revealed a highly organized lattice, the individual components of which are of a size consistent with CVB RNA polymerase; we suggest that this may represent a coxsackievirus replication complex. Thus, these in vivo studies demonstrate that CVB3 infection dramatically modifies autophagy in infected pancreatic acinar cells.


Asunto(s)
Autofagia/fisiología , Infecciones por Coxsackievirus/fisiopatología , Enterovirus Humano B , Páncreas/citología , Fagosomas/virología , Análisis de Varianza , Animales , Western Blotting , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Microscopía Electrónica de Transmisión , Microscopía Inmunoelectrónica , Páncreas/virología , Fagosomas/ultraestructura
18.
PLoS Pathog ; 5(10): e1000618, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19834548

RESUMEN

Many viruses encode proteins whose major function is to evade or disable the host T cell response. Nevertheless, most viruses are readily detected by host T cells, and induce relatively strong T cell responses. Herein, we employ transgenic CD4(+) and CD8(+) T cells as sensors to evaluate in vitro and in vivo antigen presentation by coxsackievirus B3 (CVB3), and we show that this virus almost completely inhibits antigen presentation via the MHC class I pathway, thereby evading CD8(+) T cell immunity. In contrast, the presentation of CVB3-encoded MHC class II epitopes is relatively unencumbered, and CVB3 induces in vivo CD4(+) T cell responses that are, by several criteria, phenotypically normal. The cells display an effector phenotype and mature into multi-functional CVB3-specific memory CD4(+) T cells that expand dramatically following challenge infection and rapidly differentiate into secondary effector cells capable of secreting multiple cytokines. Our findings have implications for the efficiency of antigen cross-presentation during coxsackievirus infection.


Asunto(s)
Presentación de Antígeno/inmunología , Enterovirus Humano B/fisiología , Antígenos de Histocompatibilidad Clase I/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Células Cultivadas , Infecciones por Coxsackievirus/inmunología , Enterovirus Humano B/inmunología , Epítopos/inmunología , Células HeLa , Antígenos de Histocompatibilidad Clase I/fisiología , Humanos , Memoria Inmunológica/efectos de los fármacos , Memoria Inmunológica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/inmunología
19.
Autophagy ; 17(2): 402-419, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32019403

RESUMEN

Almost a billion people worldwide are chronically undernourished. Herein, using a mouse model of coxsackievirus B3 (CVB3) infection, we report that a single day of food restriction (FR) markedly increases susceptibility to attenuated enterovirus infection, replication, and disease. These "pro-viral" effects, which are rapidly-reversed by the restoration of food, are mediated by several genes whose expression is altered by FR, and which support CVB3 replication. Central to this is TFEB, a protein whose expression and activation status are rapidly increased by FR. TFEB, which regulates the transcription of >100 genes involved in macroautophagy/autophagy and lysosomal biogenesis, responds similarly to both FR and CVB3 infection and plays a pivotal role in determining host susceptibility to CVB3. We propose that, by upregulating TFEB, FR generates an intracellular environment that is more hospitable to the incoming virus, facilitating its replication. This interplay between nutritional status and enterovirus replication has implications for human health and, perhaps, for the evolution of these viruses.Abbreviations: Atg/ATG: autophagy-related; CAR: Coxsackievirus and adenovirus receptor; Cas9: CRISPR associated protein 9; Cre: recombinase that causes recombination; CRISPR: clustered regularly interspaced short palindromic repeats; Ctsb/CTSB: cathepsin B; CVB3: coxsackievirus B3; DsRedCVB3: a recombinant CVB3 that encodes the Discosoma red fluorescent protein; EL: elastase; FR: food restriction; GFP: green fluorescent protein; gRNA: guide RNA; HBSS: Hanks Buffered Salt Solution; LYNUS: lysosomal nutrient sensing machinery; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MFI: mean fluorescence intensity; MOI: multiplicity of infection; MTOR: mechanistic target of rapamycin kinase; Nluc: nanoluciferase; NlucCVB3: a recombinant CVB3 encoding nanoluciferase; pfu: plaque-forming unit(s); p.i.: post infection; rCVB: recombinant coxsackievirus B3; RPS6KB/p70S6K: ribosomal protein S6 kinase; RT: room temperature; siRNA: small interfering RNA; TFEB: transcription factor EB; tg: transgenic; TUBB: ß-tubulin; UNINF: uninfected; wrt: with respect to; WT: wild type.


Asunto(s)
Autofagia/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Infecciones por Coxsackievirus/virología , Pancreatitis/virología , Animales , Autofagia/fisiología , Enterovirus/aislamiento & purificación , Células HeLa , Humanos , Lisosomas/metabolismo , Ratones Endogámicos C57BL , Pancreatitis/metabolismo , Replicación Viral/genética
20.
Eur J Immunol ; 39(6): 1494-504, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19384870

RESUMEN

Stem cell antigen-1 (Sca1, Ly-6A/E) is a well-established marker of murine hematopoietic stem cells, and also is expressed on memory T cells. It has been suggested that the functional maintenance of T-cell memory requires the expression of Sca1 on a specialized population of memory T cells termed "memory stem cells". Here, we evaluate the requirement for Sca1 in the primary T-cell response to virus infection, and in the establishment and maintenance of T-cell memory. We find that Sca1 expression increases on almost all CD4(+) and CD8(+) T cells during virus infection, and remains high on virus-specific memory cells. However, Sca1-deficient (Sca1KO) mice generate normal primary T-cell responses to infection; the kinetics, the immunodominance hierarchy, and the absolute numbers of CD4(+) and CD8(+) T cells are essentially indistinguishable from those observed in WT mice. Furthermore, by several criteria, primary and memory T cells in Sca1-deficient mice are phenotypically and functionally normal. These data indicate that Sca1, although perhaps a useful marker of virus-specific memory T cells, is not required for the regulation of T-cell quantity or quality, or for the development of a competent pool of memory cells.


Asunto(s)
Infecciones por Arenaviridae/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica/inmunología , Proteínas de la Membrana/deficiencia , Animales , Antígenos Ly/fisiología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Recuento de Células , Receptores de Hialuranos/metabolismo , Inmunidad Celular/inmunología , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Cinética , Virus de la Coriomeningitis Linfocítica/inmunología , Proteínas de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Interferón/genética , Bazo/citología , Bazo/inmunología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Citotóxicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA