Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
Clin Cancer Res ; 15(19): 6167-76, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19789304

RESUMEN

PURPOSE: The goal of this study was to investigate the therapeutic potential of a novel immunotherapy strategy resulting in immunity to localized or metastatic human papillomavirus 16-transformed murine tumors. EXPERIMENTAL DESIGN: Animals bearing E7-expressing tumors were coimmunized by lymph node injection with E7 49-57 antigen and TLR3-ligand (synthetic dsRNA). Immune responses were measured by flow cytometry and antitumor efficacy was evaluated by tumor size and survival. In situ cytotoxicity assays and identification of tumor-infiltrating lymphocytes and T regulatory cells were used to assess the mechanisms of treatment resistance in bulky disease. Chemotherapy with cyclophosphamide was explored to augment immunotherapy in late-stage disease. RESULTS: In therapeutic and prophylactic settings, immunization resulted in a considerable expansion of E7 49-57 antigen-specific T lymphocytes in the range of 1/10 CD8(+) T cells. The resulting immunity was effective in suppressing disease progression and mortality in a pulmonary metastatic disease model. Therapeutic immunization resulted in control of isolated tumors up to a certain volume, and correlated with antitumor immune responses measured in blood. In situ analysis showed that within bulky tumors, T-cell function was affected by negative regulatory mechanisms linked to an increase in T regulatory cells and could be overcome by cyclophosphamide treatment in conjunction with immunization. CONCLUSIONS: This study highlights a novel cancer immunotherapy platform with potential for translatability to the clinic and suggests its potential usefulness for controlling metastatic disease, solid tumors of limited size, or larger tumors when combined with cytotoxic agents that reduce the number of tumor-infiltrating T regulatory cells.


Asunto(s)
Papillomavirus Humano 16/fisiología , Inmunidad Celular/fisiología , Inmunoterapia/métodos , Ganglios Linfáticos/inmunología , Neoplasias/patología , Neoplasias/terapia , Animales , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/uso terapéutico , Transformación Celular Viral/inmunología , Terapia Combinada , Citotoxinas/administración & dosificación , Femenino , Papillomavirus Humano 16/inmunología , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Neoplasias/inmunología , Neoplasias/prevención & control , Proteínas E7 de Papillomavirus/metabolismo , Carga Tumoral/inmunología
3.
Immunol Lett ; 127(1): 60-7, 2009 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-19751765

RESUMEN

Elevated Programmed Death-1 (PD-1) expression can inhibit T cell activity and is a potential barrier to achieving persisting and optimal immunity via therapeutic vaccination. Using a direct lymph node-targeted vaccination procedure that enabled uncoupling of synthetic peptide (signal 1, TCR-mediated) and adjuvant (signal 2, non-TCR-mediated), we evaluated the impact of varied doses of Toll-like receptor (TLR)-9 ligand CpG oligodeoxynucleotide (ODN) adjuvant on epitope-specific CD8(+) T cell-associated PD-1 expression. Peptide vaccination without adjuvant yielded CD8(+) T cells with significantly elevated PD-1 expression. This conferred impaired function ex vivo, but was reversible by antibody-mediated PD-1 blockade. By comparison, peptide vaccination with escalating doses of CpG ODN adjuvant yielded higher magnitudes of CD8(+) T cells with progressively lower PD-1 expression and greater ex vivo function. CpG ODN adjuvant in context of titrated peptide doses for vaccination yielded the lowest overall PD-1 expression levels, demonstrating that fine-tuning both TCR-independent (adjuvant dose) and -dependent (antigen dose) stimuli can synergize to co-regulate PD-1 expression on epitope-specific CD8(+) T cells. These data hint at strategies to elicit PD-1(low) CD8(+) T cells using TLR-9 ligand adjuvants, and also shed light on the PD-1-regulated homeostasis of CD8(+) T cells.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Regulación de la Expresión Génica , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Inmunológicos/biosíntesis , Receptor Toll-Like 9/metabolismo , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , ADN/administración & dosificación , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Antígenos HLA-A/genética , Antígeno HLA-A2 , Inmunización , Ligandos , Antígeno MART-1 , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/inmunología , Oligodesoxirribonucleótidos , Fragmentos de Péptidos/inmunología , Receptor de Muerte Celular Programada 1 , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Receptor Toll-Like 9/inmunología
4.
J Virol ; 83(18): 9356-69, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19570873

RESUMEN

Coxsackieviruses are significant human pathogens, and the neonatal central nervous system (CNS) is a major target for infection. Despite the extreme susceptibility of newborn infants to coxsackievirus infection and viral tropism for the CNS, few studies have been aimed at determining the long-term consequences of infection on the developing CNS. We previously described a neonatal mouse model of coxsackievirus B3 (CVB3) infection and determined that proliferating stem cells in the CNS were preferentially targeted. Here, we describe later stages of infection, the ensuing inflammatory response, and subsequent lesions which remain in the adult CNS of surviving animals. High levels of type I interferons and chemokines (in particular MCP-5, IP10, and RANTES) were upregulated following infection and remained at high levels up to day 10 postinfection (p.i). Chronic inflammation and lesions were observed in the hippocampus and cortex of surviving mice for up to 9 months p.i. CVB3 RNA was detected in the CNS up to 3 months p.i at high abundance ( approximately 10(6) genomes/mouse brain), and viral genomic material remained detectable in culture after two rounds of in vitro passage. These data suggest that CVB3 may persist in the CNS as a low-level, noncytolytic infection, causing ongoing inflammatory lesions. Thus, the effects of a relatively common infection during the neonatal period may be long lasting, and the prognosis for newborn infants recovering from acute infection should be reexplored.


Asunto(s)
Sistema Nervioso Central/virología , Infecciones por Coxsackievirus/patología , Enterovirus/patogenicidad , Animales , Animales Recién Nacidos , Corteza Cerebral/patología , Corteza Cerebral/virología , Quimiocinas/análisis , Enfermedad Crónica , Infecciones por Coxsackievirus/inmunología , Enterovirus/genética , Enterovirus Humano B , Genoma Viral , Hipocampo/patología , Hipocampo/virología , Humanos , Inflamación , Interferón Tipo I/análisis , Ratones , ARN Viral/sangre , Factores de Tiempo , Regulación hacia Arriba
5.
Vaccine ; 27(19): 2603-15, 2009 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-19428867

RESUMEN

DNA vaccines or peptides are capable of inducing specific immunity; however, their translation to the clinic has generally been problematic, primarily due to the reduced magnitude of immune response and poor pharmacokinetics. Herein, we demonstrate that a novel immunization strategy, encompassing sequential exposure of the lymph node milieu to plasmid and peptide in a heterologous prime-boost fashion, results in considerable MHC class I-restricted immunity in mice. Plasmid-primed antigen expression was essential for the generation of a population of central memory T cells, expressing CD62L and low in PD-1, with substantial capability to expand and differentiate to peripheral memory and effector cells, following subsequent exposure to peptide. These vaccine-induced T cells dominated the T cell repertoire, were able to produce large amounts of chemokines and pro-inflammatory cytokines, and recognized tumor cells effectively. In addition to outlining a feasible and effective method to transform plasmid DNA vaccination into a potentially viable immunotherapeutic approach for cancer, this study sheds light on the mechanism of heterologous prime-boost and the considerable heterogeneity of MHC class I-restricted T cell responses.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Inmunización Secundaria/métodos , Ganglios Linfáticos/inmunología , Vacunación/métodos , Vacunas de ADN/inmunología , Animales , Citocinas/metabolismo , Femenino , Memoria Inmunológica , Pulmón/inmunología , Ratones , Ratones Transgénicos , Bazo/inmunología , Subgrupos de Linfocitos T/inmunología , Vacunas de Subunidad/inmunología
6.
J Neurosci ; 25(9): 2434-44, 2005 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-15745971

RESUMEN

Type B coxsackieviruses (CVB) frequently infect the CNS and, together with other enteroviruses, are the most common cause of viral meningitis in humans. Newborn infants are particularly vulnerable, and CVB also can infect the fetus, leading to mortality, or to neurodevelopmental defects in surviving infants. Using a mouse model of neonatal CVB infection, we previously demonstrated that coxsackievirus B3 (CVB3) could infect neuronal progenitor cells in the subventricular zone (SVZ). Here we extend these findings, and we show that CVB3 targets actively proliferating (bromodeoxyuridine+, Ki67+) cells in the SVZ, including type B and type A stem cells. However, infected cells exiting the SVZ have lost their proliferative capacity, in contrast to their uninfected companions. Despite being proliferation deficient, the infected neuronal precursors could migrate along the rostral migratory stream and radial glia, to reach their final destinations in the olfactory bulb or cerebral cortex. Furthermore, infection did not prevent cell differentiation, as determined by cellular morphology and the expression of maturation markers. These data lead us to propose a model of CVB infection of the developing CNS, which may explain the neurodevelopmental defects that result from fetal infection.


Asunto(s)
Proliferación Celular , Sistema Nervioso Central/patología , Infecciones por Coxsackievirus/patología , Enterovirus Humano B/patogenicidad , Neuronas/virología , Células Madre/virología , Animales , Animales Recién Nacidos , Bromodesoxiuridina/metabolismo , Recuento de Células/métodos , Sistema Nervioso Central/crecimiento & desarrollo , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/virología , Ventrículos Cerebrales/citología , Ventrículos Cerebrales/metabolismo , Ventrículos Cerebrales/virología , Infecciones por Coxsackievirus/metabolismo , Infecciones por Coxsackievirus/fisiopatología , Infecciones por Coxsackievirus/virología , Modelos Animales de Enfermedad , Enterovirus Humano B/genética , Técnica del Anticuerpo Fluorescente/métodos , Proteínas Fluorescentes Verdes/metabolismo , Hibridación in Situ/métodos , Indoles/metabolismo , Proteínas de Filamentos Intermediarios/metabolismo , Antígeno Ki-67/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteínas del Tejido Nervioso/metabolismo , Nestina , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Neuronas/fisiología , Fosfopiruvato Hidratasa/metabolismo , Receptores Virales/genética , Receptores Virales/metabolismo , Ácidos Siálicos/metabolismo , Células Madre/fisiología , Tubulina (Proteína)/metabolismo
7.
Med Microbiol Immunol ; 193(2-3): 83-90, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-12925877

RESUMEN

Coxsackieviruses (CV) are characterized by their ability to cause cytopathic effects in tissue culture and by their capacity to initiate acute disease by inducing apoptosis within targeted organs in vivo. These viruses are considered highly cytolytic, but can establish persistence/latency in susceptible cells, indicating that a regulatory mechanism may exist to shut off viral protein synthesis and replication under certain situations. The persistence of coxsackieviral RNA is of particular medical interest due to its association with chronic human diseases such as dilated cardiomyopathy and chronic inflammatory myopathy. Here, we discuss the potential mechanisms regulating coxsackievirus replication, and the ability of viral RNA to remain in an apparent latent state within quiescent cells.


Asunto(s)
Ciclo Celular/fisiología , Enterovirus Humano B/fisiología , Enterovirus Humano B/patogenicidad , Regulación de la Expresión Génica , Latencia del Virus , Replicación Viral , Animales , Animales Recién Nacidos , Encéfalo/virología , Enterovirus Humano B/genética , Infecciones por Enterovirus/fisiopatología , Infecciones por Enterovirus/virología , Proteínas Fluorescentes Verdes , Células HeLa , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Proteínas Virales/genética , Proteínas Virales/metabolismo
8.
Am J Pathol ; 163(4): 1379-93, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14507646

RESUMEN

Neonates are particularly susceptible to coxsackievirus infections of the central nervous system (CNS), which can cause meningitis, encephalitis, and long-term neurological deficits. However, viral tropism and mechanism of spread in the CNS have not been examined. Here we investigate coxsackievirus B3 (CVB3) tropism and pathology in the CNS of neonatal mice, using a recombinant virus expressing the enhanced green fluorescent protein (eGFP). Newborn pups were extremely vulnerable to coxsackievirus CNS infection, and this susceptibility decreased dramatically by 7 days of age. Twenty-four hours after intracranial infection of newborn mice, viral genomic RNA and viral protein expression were detected in the choroid plexus, the olfactory bulb, and in cells bordering the cerebral ventricles. Many of the infected cells bore the anatomical characteristics of type B stem cells, which can give rise to neurons and astrocytes, and expressed the intermediate filament protein nestin, a marker for progenitor cells. As the infection progressed, viral protein was identified in the brain parenchyma, first in cells expressing neuron-specific class III beta-tubulin, an early marker of neuronal differentiation, and subsequently in cells expressing NeuN, a marker of mature neurons. At later time points, viral protein expression was restricted to neurons in specific regions of the brain, including the hippocampus, the entorhinal and temporal cortex, and the olfactory bulb. Extensive neuronal death was visible, and appeared to result from virus-induced apoptosis. We propose that the increased susceptibility of the neonatal CNS to CVB infection may be explained by the virus' targeting neonatal stem cells; and that CVB is carried into the brain parenchyma by developing neurons, which continue to migrate and differentiate despite the infection. On full maturation, some or all of the infected neurons undergo apoptosis, and the resulting neuronal loss can explain the longer-term clinical picture.


Asunto(s)
Animales Recién Nacidos , Enfermedades del Sistema Nervioso Central/fisiopatología , Enfermedades del Sistema Nervioso Central/virología , Infecciones por Coxsackievirus/complicaciones , Envejecimiento , Animales , Apoptosis , Encéfalo/virología , Senescencia Celular , Enfermedades del Sistema Nervioso Central/mortalidad , Infecciones por Coxsackievirus/mortalidad , Infecciones por Coxsackievirus/virología , Progresión de la Enfermedad , Susceptibilidad a Enfermedades , Enterovirus/aislamiento & purificación , Proteínas Fluorescentes Verdes , Hibridación in Situ , Indicadores y Reactivos , Proteínas Luminiscentes , Ratones , Ratones Endogámicos BALB C , Neuronas/metabolismo , Células Madre/virología , Factores de Tiempo , Proteínas Virales/metabolismo
9.
J Clin Invest ; 111(6): 805-10, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12639986

RESUMEN

Among infectious agents, measles virus (MV) remains a scourge responsible for 1 million deaths per year and is a leading cause of childhood deaths in developing countries. Although MV infection itself is not commonly lethal, MV-induced suppression of the immune system results in a greatly increased susceptibility to opportunistic bacterial infections that are largely responsible for the morbidity and mortality associated with this disease. Despite its clinical importance, the underlying mechanisms of MV-induced immunosuppression remain unresolved. To begin to understand the basis of increased susceptibility to bacterial infections during MV infection, we inoculated transgenic mice expressing the MV receptor, CD46, with MV and Listeria monocytogenes. We found that MV-infected mice were more susceptible to infection with Listeria and that this corresponded with significantly decreased numbers of macrophages and neutrophils in the spleen and substantial defects in IFN-gamma production by CD4(+) T cells. The reduction in CD11b(+) macrophages and IFN-gamma-producing T cells was due to reduced proliferative expansion and not to enhanced apoptosis or to altered distribution of these cells between spleen, blood, and the lymphatic system. These results document that MV infection can suppress both innate and adaptive immune responses and lead to increased susceptibility to bacterial infection.


Asunto(s)
Infecciones Bacterianas/inmunología , Tolerancia Inmunológica , Sarampión/inmunología , Animales , Antígenos CD/fisiología , Antígeno CD11b/análisis , Linfocitos T CD4-Positivos/inmunología , Inmunidad Innata , Interferón gamma/biosíntesis , Listeriosis/inmunología , Proteína Cofactora de Membrana , Glicoproteínas de Membrana/fisiología , Ratones
10.
J Virol ; 76(9): 4430-40, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11932410

RESUMEN

Enteroviral persistence has been implicated in the pathogenesis of several chronic human diseases, including dilated cardiomyopathy, insulin-dependent diabetes mellitus, and chronic inflammatory myopathy. However, these viruses are considered highly cytolytic, and it is unclear what mechanisms might permit their long-term survival. Here, we describe the generation of a recombinant coxsackievirus B3 (CVB3) expressing the enhanced green fluorescent protein (eGFP), which we used to mark and track infected cells in vitro. Following exposure of quiescent tissue culture cells to either wild-type CVB3 or eGFP-CVB3, virus production was very limited but increased dramatically after cells were permitted to divide. Studies with cell cycle inhibitors revealed that cells arrested at the G(1) or G(1)/S phase could express high levels of viral polyprotein and produced abundant infectious virus. In contrast, both protein expression and virus yield were markedly reduced in quiescent cells (i.e., cells in G(0)) and in cells blocked at the G(2)/M phase. Following infection with eGFP-CVB3, quiescent cells retained viral RNA for several days in the absence of infectious virus production. Furthermore, RNA extracted from nonproductive quiescent cells was infectious when transfected into dividing cells, indicating that CVB3 appears to be capable of establishing a latent infection in G(0) cells, at least in tissue culture. Finally, wounding of infected quiescent cells resulted in viral protein expression limited to cells in and adjacent to the lesion. We suggest that (i) cell cycle status determines the distribution of CVB3 during acute infection and (ii) the persistence of CVB3 in vivo may rely on infection of quiescent (G(0)) cells incapable of supporting viral replication; a subsequent change in the cell cycle status may lead to virus reactivation, triggering chronic viral and/or immune-mediated pathology in the host.


Asunto(s)
Ciclo Celular/fisiología , Enterovirus Humano B/crecimiento & desarrollo , Enterovirus Humano B/patogenicidad , Activación Viral , Replicación Viral , Medios de Cultivo , Enterovirus Humano B/genética , Proteínas Fluorescentes Verdes , Células HeLa/citología , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Recombinación Genética , Latencia del Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA