Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 17(1): e1009066, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33400715

RESUMEN

In adult mice the severity of disease from viral infections is determined by the balance between the efficiency of the immune response and the magnitude of viral load. Here, the impact of this dynamic is examined in neonates. Newborns are highly susceptible to infections due to poor innate responses, lower numbers of T cells and Th2-prone immune responses. Eighty-percent of 7-day old mice, immunologically equivalent to human neonates, succumbed to extremely low doses (5 PFU) of the essentially non-lethal lymphocytic choriomeningitis virus (LCMV-Armstrong) given intraperitoneally. This increased lethality was determined to be dependent upon poor early viral control, as well as, T cells and perforin as assessed in knockout mice. By day 3, these neonatal mice had 400-fold higher viral loads as compared to adults receiving a 10,000-fold (5X104 PFU) higher dose of LCMV. The high viral load in combination with the subsequent immunological defect of partial CD8 T cell clonal exhaustion in the periphery led to viral entry and replication in the brain. Within the brain, CD8 T cells were protected from exhaustion, and thus were able to mediate lethal immunopathology. To further delineate the role of early viral control, neonatal mice were infected with Pichinde virus, a less virulent arenavirus, or LCMV was given to pups of LCMV-immune mothers. In both cases, peak viral load was at least 29-fold lower, leading to functional CD8 T cell responses and 100% survival.


Asunto(s)
Encéfalo/virología , Linfocitos T CD8-positivos/virología , Memoria Inmunológica/inmunología , Coriomeningitis Linfocítica/mortalidad , Virus de la Coriomeningitis Linfocítica/patogenicidad , Animales , Animales Recién Nacidos , Encéfalo/inmunología , Linfocitos T CD8-positivos/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
2.
Eur J Immunol ; 49(5): 782-789, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30793761

RESUMEN

Hyper-activated or deviated immune responses can result in immunopathological diseases. Paradoxically, immunodeficiency represents a frequent cause of such immune-mediated pathologies. Immunopathological manifestations are commonly treated by immunosuppression, but in situations in which immunodeficiency is the basis of disease development, enhancing immunity may represent an alternative treatment option. Here, we tested this counterintuitive concept in a preclinical model using infection of mice with lymphocytic choriomeningitis virus (LCMV). Firstly, we demonstrate that infection of B-cell-deficient (B-/- ) but not of wild-type (WT) mice with the LCMV strain Docile induced a rapid and fatal CD8+ T-cell-mediated immunopathological disease. Similar to WT mice, LCMV-infected B-/- mice generated a potent, functional LCMV-specific CD8+ T-cell response but exhibited prolonged viral antigen presentation and increased vascular leakage in liver and lungs. Secondly, we were able to prevent this virus-induced immunopathology in B-/- mice by active or passive T-cell immunizations or by treatment with LCMV-specific virus neutralizing or non-neutralizing monoclonal antibodies (mAb). Thus, boosting antiviral immunity did not aggravate immunopathology in this model, but prevented it by decreasing the formation of target structures for damage-causing CD8+ T cells.


Asunto(s)
Linfocitos B/inmunología , Linfocitos B/metabolismo , Susceptibilidad a Enfermedades , Inmunidad , Linfocitos T/inmunología , Linfocitos T/metabolismo , Virus/inmunología , Animales , Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Susceptibilidad a Enfermedades/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/metabolismo , Coriomeningitis Linfocítica/mortalidad , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Noqueados , Mortalidad
3.
J Immunol ; 195(1): 116-25, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25980007

RESUMEN

Patients who survive sepsis display suppressed immune functions, often manifested as an increased susceptibility to secondary infections. Recently, using a cecal-ligation and puncture (CLP) model of sepsis, we showed that sepsis induces substantial and long-lasting changes in the available naive CD8(+) T cell repertoire affecting the capacity of the host to respond to newly encountered acute infections. However, the extent to which sepsis changes the host susceptibility to chronic infection and affects CD8(+) T cell responses is currently unknown. In this study, we demonstrate that inbred and outbred mice recovering from a septic event are more susceptible to lymphocytic choriomeningitis virus (LCMV) clone-13 infection exhibited by mortality and viral burden. Primary virus-specific CD8(+) T cells in LCMV clone-13-infected septic mice displayed exacerbated CD8(+) T cell exhaustion illustrated by increased inhibitory molecule expression (e.g., programmed cell death 1, lymphocyte-activation gene 3, and 2B4) and diminished Ag-driven cytokine production (e.g., IFN-γ, TNF-α) compared with similarly infected sham-treated mice. Importantly, therapeutic inhibitory molecule dual blockade (anti-PD-L1 and anti-lymphocyte-activation gene 3) increased the number of circulating LCMV-specific CD8(+) T cells, and improved CD8(+) T cell function and pathogen control in chronically infected septic mice. Together, these results illustrate that polymicrobial sepsis compromises the overall health of the host leading to increased vulnerability to chronic infection and exacerbated CD8(+) T cell exhaustion. Collectively, our findings suggest that septic survivors may be more susceptible and at greater risk for developing exhaustible CD8(+) T cells upon encountering a subsequent chronic infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interacciones Huésped-Patógeno/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Sepsis/inmunología , Animales , Anticuerpos/farmacología , Antígenos CD/genética , Antígenos CD/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/patología , Convalecencia , Susceptibilidad a Enfermedades , Regulación de la Expresión Génica , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Recuento de Linfocitos , Coriomeningitis Linfocítica/etiología , Coriomeningitis Linfocítica/mortalidad , Coriomeningitis Linfocítica/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Cultivo Primario de Células , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Sepsis/complicaciones , Sepsis/mortalidad , Sepsis/patología , Transducción de Señal , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Análisis de Supervivencia , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Carga Viral , Proteína del Gen 3 de Activación de Linfocitos
4.
J Virol ; 88(13): 7578-88, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24760883

RESUMEN

UNLABELLED: Following systemic infection with lymphocytic choriomeningitis virus (LCMV), STAT1 knockout (KO) mice but not wild-type, STAT2 KO, IRF9 KO, or IFNAR KO mice develop lethal disease perpetrated by CD4(+) T cells. IRF7 is a key transcriptional activator of type I IFN (IFN-I) during LCMV infection. Here, the role of IRF7 in the lethal host response to LCMV infection in STAT1 KO mice was examined. In contrast to STAT1 KO mice, STAT1/IRF7 double KO (DKO) mice survived LCMV infection with a reduced immune pathology in key organs, such as the liver and spleen. However, similar to STAT1 KO mice, STAT1/IRF7 DKO mice failed to control LCMV replication and spread. LCMV infection in STAT1 KO mice was associated with a significant elevation in the levels of a number of cytokines in serum, including IFN-Is, but this was largely absent in STAT1/IRF7 DKO mice, which had a modest increase in the levels of gamma interferon and CCL2 only. Since IRF7 is known to be a key transcriptional regulator of IFN-I gene expression, the possible role of IFN-I in lethal disease was examined further. STAT1/IFNAR DKO mice, in contrast to STAT1 KO mice, all survived infection with LCMV and exhibited little tissue immune pathology. Additionally, STAT1 KO mice that were deficient for either of the two IFN-I signaling molecules, STAT2 or IRF9, also survived LCMV infection. We conclude that the lethal immune-mediated disease resulting from LCMV infection in STAT1 KO mice is (i) dependent on IRF7-induced IFN-I production and (ii) driven by noncanonical IFN-I signaling via STAT2 and IRF9. IMPORTANCE: Here we report on the basis for the novel, fatal immune-mediated disease of STAT1 KO mice infected with LCMV. Our findings show that, surprisingly, the pathogenesis of this disease is dependent on IRF7-mediated type I interferon production. Moreover, our study identifies noncanonical type I interferon signaling via STAT2 and IRF9 to be essential for the type I IFN-driven fatal disease in LCMV-infected STAT1 KO mice. These results further highlight the significance of noncanonical type I IFN signaling in the pathogenesis of host-mediated injury following viral infection.


Asunto(s)
Genes Letales/inmunología , Interferón Tipo I/metabolismo , Subunidad gamma del Factor 3 de Genes Estimulados por el Interferón/fisiología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Factor de Transcripción STAT1/fisiología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Ensayo de Inmunoadsorción Enzimática , Femenino , Coriomeningitis Linfocítica/mortalidad , Coriomeningitis Linfocítica/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tasa de Supervivencia
5.
Emerg Infect Dis ; 18(8): 1256-62, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22839997

RESUMEN

Three clusters of organ transplant-associated lymphocytic choriomeningitis virus (LCMV) transmissions have been identified in the United States; 9 of 10 recipients died. In February 2011, we identified a fourth cluster of organ transplant-associated LCMV infections. Diabetic ketoacidosis developed in the organ donor in December 2010; she died with generalized brain edema after a short hospitalization. Both kidneys, liver, and lung were transplanted to 4 recipients; in all 4, severe posttransplant illness developed; 2 recipients died. Through multiple diagnostic methods, we identified LCMV infection in all persons, including in at least 1 sample from the donor and 4 recipients by reverse transcription PCR, and sequences of a 396-bp fragment of the large segment of the virus from all 5 persons were identical. In this cluster, all recipients developed severe illness, but 2 survived. LCMV infection should be considered as a possible cause of severe posttransplant illness.


Asunto(s)
Coriomeningitis Linfocítica/transmisión , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/aislamiento & purificación , Trasplante de Órganos/efectos adversos , Adolescente , Femenino , Humanos , Riñón/patología , Riñón/virología , Hígado/patología , Hígado/virología , Pulmón/patología , Pulmón/virología , Coriomeningitis Linfocítica/diagnóstico , Coriomeningitis Linfocítica/mortalidad , Virus de la Coriomeningitis Linfocítica/genética , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Donantes de Tejidos , Estados Unidos/epidemiología
6.
Eur J Immunol ; 42(6): 1488-99, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22678903

RESUMEN

Perforin-deficient (PKO) mice serve as models for familial hemophagocytic lympho-histiocytosis, a uniformly fatal disease associated with viral infection of perforin-deficient humans. Naïve perforin-deficient BALB/c mice survive while vaccinated PKO mice containing virus-specific memory CD8(+) T cells rapidly succumb to lymphocytic choriomeningitis virus (LCMV) infection. Thus, vaccination converts a nonlethal persistent infection into a fatal disease mediated by virus-specific memory CD8(+) T cells. Here, we determine the extent to which vaccination-induced mortality in PKO mice following LCMV challenge is due to differences in vaccine modalities, the quantity or epitope specificity of memory CD8(+) T cells. We show that LCMV-induced mortality in immune PKO mice is independent of vaccine modalities and that the starting number of memory CD8(+) T cells specific to the immunodominant epitope NP(118-126) dictates the magnitude of secondary CD8(+) T-cell expansion, the inability to regulate production of CD8(+) T-cell-derived IFN-γ, and mortality in the vaccinated PKO mice. Importantly, mortality is determined by the epitope specificity of memory CD8(+) T cells and the associated degree of functional exhaustion and cytokine dysregulation but not the absolute magnitude of CD8(+) T-cell expansion. These data suggest that deeper understanding of the parameters that influence the outcome of vaccine-induced diseases would aid rational vaccine design to minimize adverse outcomes after infection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epítopos de Linfocito T , Memoria Inmunológica , Coriomeningitis Linfocítica/mortalidad , Virus de la Coriomeningitis Linfocítica/inmunología , Proteínas Citotóxicas Formadoras de Poros/deficiencia , Vacunación/efectos adversos , Vacunas Virales/efectos adversos , Traslado Adoptivo , Animales , Citocinas/biosíntesis , Ratones , Ratones Endogámicos BALB C , Proteínas Citotóxicas Formadoras de Poros/fisiología , Vacunas Virales/inmunología
7.
Virology ; 425(2): 122-32, 2012 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-22305620

RESUMEN

Wasting is a sign of various underlying disorders and is a common feature of cancer, sepsis, and AIDS. We have developed an in vivo model to study the various stages of wasting following infection of mice with lymphocytic choriomeningitis virus cl-13. Using this model we have identified four distinct stages of wasting and have discovered that all stages occur in the different groups of mice regardless of whether the virus is cleared or persists. However, the degree and extent of wasting vary between groups of mice, depending upon the dose of virus administered. Blocking IFNγ or TNFα, which are believed to take part in the wasting process, did not affect the wasting state. Finally, we found that CD4+ T cells control the maintenance stage of wasting. We believe this model will be useful in studying the regulation of wasting during a persistent viral infection, hopefully leading to improved therapies to ameliorate the disorder.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/mortalidad , Virus de la Coriomeningitis Linfocítica/fisiología , Animales , Femenino , Humanos , Coriomeningitis Linfocítica/virología , Ratones , Ratones Endogámicos C57BL
8.
J Virol ; 84(1): 312-20, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19828618

RESUMEN

Although much is known about lymphocytic choriomeningitis virus (LCMV) infection and the subsequent immune response in its natural murine host, some crucial aspects of LCMV-mediated pathogenesis remain undefined, including the underlying basis of the characteristic central nervous system disease that occurs following intracerebral (i.c.) challenge. We show that the classic seizures and paresis that occur following i.c. infection of adult, immunocompetent mice with LCMV are accompanied by anatomical and histological changes that are consistent with brain herniation, likely of the uncal subtype, as a causative basis for disease and precipitous death. Both by water weight determinations and by magnetic resonance imaging of infected brain tissues, edema was detected only at the terminal stages of disease, likely caused by the leakage of cerebrospinal fluid from the ventricles into the parenchyma. Furthermore, death was accompanied by unilateral pupillary dilation, which is indicative of uncal herniation. While immunohistochemical analysis revealed periventricular inflammation and a loss of integrity of the blood-brain barrier (BBB), these events preceded seizures by 2 to 3 days. Moreover, surviving perforin knockout mice showed barrier permeability equivalent to that of moribund, immunocompetent mice; thus, BBB damage does not appear to be the basis of LCMV-induced neuropathogenesis. Importantly, brain herniation can occur in humans as a consequence of injuries that would be predicted to increase intracranial pressure, including inflammation, head trauma, and brain tumors. Thus, a mechanistic dissection of the basis of LCMV neuropathogenesis may be informative for the development of interventive therapies to prevent this typically fatal human condition.


Asunto(s)
Edema/etiología , Encefalocele/etiología , Coriomeningitis Linfocítica/mortalidad , Virus de la Coriomeningitis Linfocítica , Animales , Barrera Hematoencefálica/patología , Edema/patología , Encefalocele/patología , Inflamación , Imagen por Resonancia Magnética , Ratones , Ratones Noqueados , Mortalidad , Paresia , Perforina/deficiencia , Convulsiones
9.
J Gen Virol ; 90(Pt 2): 414-422, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19141451

RESUMEN

It has recently been demonstrated that a recombinant replication-deficient human adenovirus 5 (Ad5) vector expressing lymphocytic choriomeningitis virus (LCMV) glycoprotein (GP) fused to the p31 invariant (Ii) chain confers broad, long-lasting T-cell immunity that completely protects C57BL/6 mice against lethal peripheral challenge. The current study questioned whether the same strategy, i.e. linkage of GP to an Ii chain, could be applied to a naked DNA vaccine. Following gene-gun immunization with the linked construct (DNA-IiGP), GP-specific CD4(+) T cells could not be detected by flow cytometry. However, inclusion of the Ii chain augmented the priming of GP-specific CD8(+) T cells directed towards both immunodominant (GP(33-41)) and subdominant (GP(276-286) and GP(92-101)) epitopes, and vaccination with DNA-IiGP conferred significantly improved protection against systemic LCMV infection compared with the unlinked construct. In contrast, substantial protection against peripheral challenge was not observed. Additional experiments with T-cell subset-depleted or perforin-deficient mice revealed that virus control in vaccinated mice depends critically on cytotoxic CD8(+) T cells. Finally, priming with the naked DNA vaccine was shown to augment the immune response raised by subsequent immunization with the Ad5 vector. In conclusion, this study showed that the immunoenhancing effect of Ii chain linkage is not limited to the Ad5 vector, but is also relevant with a DNA platform. Furthermore, given the fact that the Ii chain enhances the presentation of more than one epitope, this suggests that Ii-chain-based DNA vaccines may be promising candidates for various heterologous prime-boost regimes.


Asunto(s)
Antígenos Virales/inmunología , Biolística/métodos , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Linfocitos T/inmunología , Vacunas de ADN/administración & dosificación , Vacunas Virales/administración & dosificación , Animales , Biolística/normas , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Escherichia coli/inmunología , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/mortalidad , Coriomeningitis Linfocítica/prevención & control , Virus de la Coriomeningitis Linfocítica/genética , Ratones , Ratones Endogámicos C57BL , Tasa de Supervivencia , Subgrupos de Linfocitos T/inmunología , Vacunas de ADN/genética , Vacunas de ADN/inmunología
10.
Viral Immunol ; 21(4): 425-33, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19115931

RESUMEN

Intracranial infection of mice with lymphocytic choriomeningitis virus (LCMV) results in a lethal neurological disease termed lymphocytic choriomeningitis (LCM) that is mediated by antiviral CD8(+) T cells. Previous studies have implicated the chemokine receptor CXCR3 and its ligand CXCL10 in CD8(+) T cell trafficking in the brain and in the lethal disease following intracranial infection of mice with the LCMV-Traub strain. Here we investigated the role of CXCR3 in LCM following intracranial infection of mice with the LCMV-Armstrong strain. Significant induction of both CXCL9 and CXCL10 RNA and protein was seen in the central nervous system (CNS) in LCM. Cellular localization of the CXCL9 and CXCL10 RNA transcripts was identified predominantly in infiltrating mononuclear cells, as well as in subpial and paraventricular microglia (CXCL9) and astrocytes (CXCL10). Despite a primary role of interferon (IFN)-gamma in inducing the expression of the CXCL9 gene, and to a lesser extent the CXCL10 gene in LCM, the absence of the IFN-gamma receptor did not influence the disease outcome. This finding suggested that these chemokines may not play a major role in the pathogenesis of LCM. To evaluate this possibility further the development of LCM was examined in mice that were deficient for CXCR3. Surprisingly, in the absence of CXCR3 there was no alteration in mortality, cytokine expression, or T cell infiltration in the CNS, demonstrating that in contrast to LCMV-Traub, CXCR3 is not involved in the pathogenesis of LCMV-Armstrong-induced neurological disease in mice. Our findings indicate that despite similar immunopathogenetic mechanisms involving antiviral CD8(+) T cells, whether or not CXCR3 signaling has a role in LCM is dependent upon the infecting strain of LCMV.


Asunto(s)
Coriomeningitis Linfocítica/mortalidad , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Receptores CXCR3/inmunología , Animales , Células Cultivadas , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Quimiocinas CXC/genética , Quimiocinas CXC/inmunología , Expresión Génica , Humanos , Interferón gamma/genética , Interferón gamma/inmunología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/virología , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mortalidad , Receptores CXCR3/genética
13.
J Endotoxin Res ; 9(5): 313-6, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14577848

RESUMEN

The phenomenon of endotoxin sensitization by virus infection is well documented but not yet well understood. Infection by virtually any viral agent will quickly induce expression of type I interferons (IFN-alpha/beta), and type II IFN-gamma production will follow as NK cells and T cells are activated. It has been well established that type II IFN pretreatment can intensify the effects of endotoxin. We have recently demonstrated that type I IFN induction by lymphocytic choriomeningitis virus (LCMV) infection dramatically increases TNF-alpha production following LPS treatment, and that this sensitization by type I IFN is STAT1 dependent. Taken together these data suggest that the STAT1-mediated, MyD88-independent, arm of the LPS signaling pathway plays an important role in endotoxin toxicity, and that this pathway mediates a major component of virus-enhanced LPS sensitization.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Lipopolisacáridos/farmacología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Transducción de Señal/inmunología , Transactivadores/inmunología , Animales , Células Cultivadas , Relación Dosis-Respuesta Inmunológica , Escherichia coli/inmunología , Lipopolisacáridos/administración & dosificación , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/mortalidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción STAT1 , Factor de Necrosis Tumoral alfa/biosíntesis
14.
Immunity ; 18(4): 463-74, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12705850

RESUMEN

Perforin-mediated cytotoxicity is essential for clearance of primary LCMV infection. BALB/c-perforin-deficient (PKO) mice survived LCMV infection by deleting NP(118)-specific CD8(+) T cells whereas vaccination of PKO mice with Listeria expressing NP(118) generated a stable memory CD8(+) T cell population. However, >85% of vaccinated BALB/c-PKO mice died after LCMV infection. Mortality was associated with enormous expansion of NP(118)-specific CD8(+) T cells in both lymphoid and nonlymphoid tissues and aberrant CD8(+) T cell cytokine production. Depletion of CD8(+) T cells or treatment with anti-IFNgamma antibody rescued vaccinated mice from mortality. Thus, perforin was essential for resistance to secondary LCMV infection, and, in the absence of perforin, vaccination resulted in lethal disease mediated by dysregulated CD8(+) T cell expansion and cytokine production.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Coriomeningitis Linfocítica/inmunología , Glicoproteínas de Membrana/fisiología , Vacunación , Vacunas Virales/inmunología , Animales , Citocinas/biosíntesis , Homeostasis , Inmunofenotipificación , Listeria monocytogenes/genética , Coriomeningitis Linfocítica/mortalidad , Glicoproteínas de Membrana/deficiencia , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Nucleoproteínas/inmunología , Fragmentos de Péptidos/inmunología , Perforina , Proteínas Citotóxicas Formadoras de Poros , Vacunas Sintéticas/inmunología
15.
J Immunol ; 169(9): 4867-72, 2002 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-12391197

RESUMEN

In vivo administration of APC expressing Fas ligand (Fas-L(+) dendritic cells (DCs)) has shown promise in dampening allergic reactions and transplant rejection. Since the effect in these studies was mainly on CD4 lymphocytes, our goal was to evaluate the ability of such killer DCs to eliminate antiviral CD8 lymphocytes and in this way ameliorate viral immunopathology or, conversely, impede viral clearance. Intravenous administration of Fas-L(+) DCs resulted in a 50% reduction of lytic CD8 precursors following intracerebral infection with lymphocytic choriomeningitis virus (LCMV), and accordingly, immunopathology and survival of LCMV meningitis were improved, whereas viral clearance remained unaffected. In transfer studies the effect of the Fas-L(+) DCs was only quantifiable on experienced, not naive, CD8 lymphocytes. Importantly, loading of Fas-L(+) DCs with viral Ag before therapy was not necessary to achieve this effect, indicating that non-LCMV-infected Fas-L(+) DCs acquired viral Ag during acute LCMV infection in vivo. Our studies delineate important aspects for the clinical use of Fas-L(+) DCs in vivo. One should expect that they acquire viral Ags and suppress antiviral CD8 responses to some degree when given while an acute infection is ongoing. In terms of safety it is encouraging that resolution of the infection, at least in the case of LCMV, is not inhibited.


Asunto(s)
Apoptosis/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/mortalidad , Glicoproteínas de Membrana/biosíntesis , Receptor fas/metabolismo , Traslado Adoptivo , Animales , Línea Celular , Células Dendríticas/metabolismo , Células Dendríticas/trasplante , Células Dendríticas/virología , Proteína Ligando Fas , Inyecciones Intravenosas , Activación de Linfocitos , Recuento de Linfocitos , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Endogámicos A , Ratones Endogámicos BALB C , Análisis de Supervivencia
16.
J Immunol ; 168(12): 6022-31, 2002 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12055210

RESUMEN

During a viral response, Ag-specific effector T cells show dramatically increased binding by the mAb 1B11 and the lectin peanut agglutinin (PNA). We investigated the contribution of CD43 expression to 1B11 and PNA binding as well as its role in generation and maintenance of a CD8 T cell response. Analysis of CD43(-/-) mice revealed no increased 1B11 binding and reduced PNA binding on virus-specific CD8 T cells from -/- mice compared with +/+ mice. Furthermore, we examined the role of CD43 in the kinetics of an immune response. We show that CD43 expression modestly effects generation of a primary virus-specific CD8 T cell response in vivo but plays a more significant role in trafficking of CD8 T cells to tissues such as the brain. More interestingly, CD43 plays a role in the contraction of the immune response, with CD43(-/-) mice showing increased numbers of Ag-specific CD8 T cells following initial expansion. Following the peak of expansion, Ag-specific CD8 T cells from -/- mice show similar proliferation but demonstrate increased Bcl-2 levels and decreased apoptosis of Ag-specific effector CD8 T cells in vitro. Consistent with a delay in the down-modulation of the immune response, following chronic viral infection CD43(-/-) mice show increased morbidity. These data suggest a dynamic role of CD43 during an immune response: a positive regulatory role in costimulation and trafficking of T cells to the CNS and a negative regulatory role in the down-modulation of an immune response.


Asunto(s)
Antígenos CD , Sialoglicoproteínas/fisiología , Subgrupos de Linfocitos T/inmunología , Animales , Apoptosis/genética , Apoptosis/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , División Celular/genética , División Celular/inmunología , Movimiento Celular/genética , Movimiento Celular/inmunología , Enfermedad Crónica , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Inmunidad Celular/genética , Memoria Inmunológica/genética , Leucosialina , Activación de Linfocitos/genética , Recuento de Linfocitos , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/mortalidad , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Aglutinina de Mani/antagonistas & inhibidores , Aglutinina de Mani/metabolismo , Unión Proteica/genética , Unión Proteica/inmunología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Sialoglicoproteínas/deficiencia , Sialoglicoproteínas/genética , Subgrupos de Linfocitos T/patología , Subgrupos de Linfocitos T/virología
17.
J Immunol ; 168(12): 6047-56, 2002 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-12055213

RESUMEN

Tolerance-based stem cell transplantation using sublethal conditioning is being considered for the treatment of human disease, but safety and efficacy remain to be established. We have shown that mouse bone marrow recipients treated with sublethal irradiation plus transient blockade of the CD40-CD154 costimulatory pathway develop permanent hematopoietic chimerism across allogeneic barriers. We now report that infection with lymphocytic choriomeningitis virus at the time of transplantation prevented engraftment of allogeneic, but not syngeneic, bone marrow in similarly treated mice. Infected allograft recipients also failed to clear the virus and died. Postmortem study revealed hypoplastic bone marrow and spleens. The cause of death was virus-induced IFN-alphabeta. The rejection of allogeneic bone marrow was mediated by a radioresistant CD8(+)TCR-alphabeta(+)NK1.1(-) T cell population. We conclude that a noncytopathic viral infection at the time of transplantation can prevent engraftment of allogeneic bone marrow and result in the death of sublethally irradiated mice treated with costimulation blockade. Clinical application of stem cell transplantation protocols based on costimulation blockade and tolerance induction may require patient isolation to facilitate the procedure and to protect recipients.


Asunto(s)
Rechazo de Injerto/mortalidad , Rechazo de Injerto/virología , Trasplante de Células Madre Hematopoyéticas/mortalidad , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/mortalidad , Proteínas , Tolerancia al Trasplante/inmunología , Animales , Anticuerpos Bloqueadores/administración & dosificación , Anticuerpos Bloqueadores/uso terapéutico , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Antígenos/biosíntesis , Antígenos Ly , Antígenos de Superficie , Médula Ósea/anomalías , Trasplante de Médula Ósea/inmunología , Trasplante de Médula Ósea/mortalidad , Trasplante de Médula Ósea/patología , Ligando de CD40/inmunología , Antígenos CD8/biosíntesis , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Femenino , Rechazo de Injerto/genética , Rechazo de Injerto/inmunología , Supervivencia de Injerto/genética , Supervivencia de Injerto/inmunología , Hematopoyesis/genética , Hematopoyesis/inmunología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Inyecciones Intraperitoneales , Cinética , Lectinas Tipo C , Depleción Linfocítica/efectos adversos , Depleción Linfocítica/mortalidad , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/virología , Tejido Linfoide/anomalías , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL/genética , Ratones Endogámicos C57BL/inmunología , Ratones Endogámicos C57BL/virología , Ratones Endogámicos CBA , Ratones Noqueados/genética , Ratones Noqueados/inmunología , Ratones Noqueados/virología , Subfamilia B de Receptores Similares a Lectina de Células NK , Biosíntesis de Proteínas , Quimera por Radiación/genética , Quimera por Radiación/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/biosíntesis , Trasplante de Piel/inmunología , Subgrupos de Linfocitos T/inmunología , Factores de Tiempo , Tolerancia al Trasplante/genética
18.
J Med Virol ; 67(2): 171-86, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11992578

RESUMEN

Arenaviruses can cause hemorrhagic fever and death in primates and guinea pigs, but these viruses are not highly pathogenic for most rodent carriers. In the United States, arenaviruses precipitated outbreaks of hepatitis in captive monkeys, and they present an emerging health threat in the tropical areas of Africa and South America. We describe infection of rhesus macaques with the prototype arenavirus, lymphocytic choriomeningitis virus (LCMV), using the WE strain that has been known to cause both encephalopathy and multifocal hemorrhage. Five macaques were inoculated: two by the intravenous (i.v.) and three by the intragastric (i.g.) route. Whereas the two i.v.-inoculated monkeys developed signs and lesions consistent with fatal hemorrhagic fever, the i.g.-inoculated monkeys had an attenuated infection with no disease. Pathological signs of the primate i.v. infection differ significantly from guinea pig arenavirus infections and make this a superior model for human viral hemorrhagic disease.


Asunto(s)
Fiebres Hemorrágicas Virales/virología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Animales , Modelos Animales de Enfermedad , Cobayas , Fiebres Hemorrágicas Virales/fisiopatología , Humanos , Inyecciones Intravenosas , Coriomeningitis Linfocítica/mortalidad , Coriomeningitis Linfocítica/fisiopatología , Virus de la Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/aislamiento & purificación , Macaca mulatta , ARN Viral/análisis , Estómago/virología
19.
J Immunol ; 166(4): 2658-64, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11160329

RESUMEN

Underlying viral infections can heighten sensitivity and worsen cytokine-mediated disease following secondary inflammatory challenges. Mechanisms for this are poorly understood. The impact of the innate response to lymphocytic choriomeningitis virus (LCMV) infection on sensitivity to endotoxin (LPS) was investigated. Compared with uninfected mice, infection with LCMV for 2-days-sensitized mice to LPS by approximately 2-fold for lethality and by 2- to 6-fold for serum TNF-alpha levels. Priming for LPS-induced TNF-alpha was also seen with splenic and peritoneal leukocytes isolated from infected mice and challenged with LPS ex vivo. The effect on TNF-alpha production was present in the absence of IFN-gamma, its major producers NK and T cells, and the major pathways for its induction through IL-12 and the signal transducer and activator of transcription 4 (STAT4), and therefore was IFN-gamma independent. Early LCMV infection induces high concentrations of the type 1 IFNs, IFN-alphabeta. Administration of recombinant IFN-alpha alone heightened the TNF-alpha response to LPS. Innate IFN-alphabeta and IFN-gamma responses to LCMV exist in a delicate balance. To reduce priming for LPS-induced TNF-alpha during LCMV, deficiencies in both the IFN-alphabeta and IFN-gamma receptors or STAT1, a transcription factor downstream to both IFNs, were required. These data demonstrate that early viral infection can enhance sensitivity to bacterial products, and that this sensitization can occur in part as a result of endogenously expressed IFN-alphabeta. This work also raises issues about potential complications associated with IFN-alphabeta therapies.


Asunto(s)
Interferón Tipo I/fisiología , Lipopolisacáridos/toxicidad , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Animales , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Relación Dosis-Respuesta Inmunológica , Inmunización , Inyecciones Intraperitoneales , Interferón Tipo I/administración & dosificación , Interferón Tipo I/metabolismo , Interferón-alfa/administración & dosificación , Interferón gamma/fisiología , Lipopolisacáridos/administración & dosificación , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/mortalidad , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Receptores de Interferón/deficiencia , Receptores de Interferón/genética , Factor de Transcripción STAT1 , Transducción de Señal/genética , Transducción de Señal/inmunología , Transactivadores/deficiencia , Transactivadores/genética , Transactivadores/fisiología
20.
J Immunol ; 166(2): 982-8, 2001 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11145676

RESUMEN

LPS is the major active agent in the pathogenesis of Gram-negative septic shock. In this report we have studied the influence of concurrent viral infection on the outcome of LPS-induced shock. We find that infection with vesicular stomatitis virus sensitizes mice to LPS at an early time point following infection. Treatment of mice with the chemical IFN inducer, polyinosinic:polycytidylic acid, has a similar effect. This hypersensitivity to LPS correlated with hyperproduction of TNF-alpha in vivo. The cellular and molecular mechanisms underlying this phenomenon were investigated using Ab-depleted and gene-targeted mice. Our results revealed that while NK cell depletion and elimination of IFN-gamma partially protected against the sensitizing effects of vesicular stomatitis virus and polyinosinic:polycytidylic acid, the most striking effect was observed in IFN-alphabetaR-deficient mice. Thus hyperproduction of TNF-alpha was completely abrogated in IFN-alphabetaR-deficient mice, indicating that the principal mechanism underlying rapid virus-induced sensitization to LPS is an IFN-alphabeta-mediated priming of mice for an augmented production of TNF-alpha in response to LPS. This conclusion was further supported by the finding that pretreatment of mice with rIFN-alphabeta mimicked the effect of viral infection. In conclusion, our results reveal a previously unrecognized proinflammatory effect of IFN-alphabeta and point to a new pathway through which viral infection may influence the outcome of concurrent bacterial infection.


Asunto(s)
Interferón Tipo I/fisiología , Lipopolisacáridos/toxicidad , Infecciones por Rhabdoviridae/inmunología , Choque Séptico/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Animales , Linfocitos B/inmunología , Susceptibilidad a Enfermedades , Femenino , Humanos , Inyecciones Intraperitoneales , Inductores de Interferón/administración & dosificación , Interferón Tipo I/administración & dosificación , Interferón gamma/fisiología , Células Asesinas Naturales/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/microbiología , Coriomeningitis Linfocítica/mortalidad , Virus de la Coriomeningitis Linfocítica/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Poli I-C/administración & dosificación , Proteínas Recombinantes , Infecciones por Rhabdoviridae/microbiología , Infecciones por Rhabdoviridae/mortalidad , Choque Séptico/mortalidad , Choque Séptico/virología , Linfocitos T/inmunología , Factores de Tiempo , Factor de Necrosis Tumoral alfa/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA