Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros

Bases de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Immunol ; 205(3): 708-719, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32591391

RESUMEN

Clearance of intracellular infections caused by Salmonella Typhimurium (STm) requires IFN-γ and the Th1-associated transcription factor T-bet. Nevertheless, whereas IFN-γ-/- mice succumb rapidly to STm infections, T-bet-/- mice do not. In this study, we assess the anatomy of immune responses and the relationship with bacterial localization in the spleens and livers of STm-infected IFN-γ-/- and T-bet-/- mice. In IFN-γ-/- mice, there is deficient granuloma formation and inducible NO synthase (iNOS) induction, increased dissemination of bacteria throughout the organs, and rapid death. The provision of a source of IFN-γ reverses this, coincident with subsequent granuloma formation and substantially extends survival when compared with mice deficient in all sources of IFN-γ. T-bet-/- mice induce significant levels of IFN-γ- after challenge. Moreover, T-bet-/- mice have augmented IL-17 and neutrophil numbers, and neutralizing IL-17 reduces the neutrophilia but does not affect numbers of bacteria detected. Surprisingly, T-bet-/- mice exhibit surprisingly wild-type-like immune cell organization postinfection, including extensive iNOS+ granuloma formation. In wild-type mice, most bacteria are within iNOS+ granulomas, but in T-bet-/- mice, most bacteria are outside these sites. Therefore, Th1 cells act to restrict bacteria within IFN-γ-dependent iNOS+ granulomas and prevent dissemination.


Asunto(s)
Granuloma/inmunología , Óxido Nítrico Sintasa de Tipo II/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Proteínas de Dominio T Box/deficiencia , Células TH1/inmunología , Animales , Granuloma/genética , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-17/genética , Interleucina-17/inmunología , Ratones , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/genética , Infecciones por Salmonella/genética , Salmonella typhimurium/genética , Proteínas de Dominio T Box/inmunología
2.
Eur J Immunol ; 44(8): 2318-30, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24825601

RESUMEN

The generation of immune cells from BM precursors is a carefully regulated process. This is essential to limit the potential for oncogenesis and autoimmunity yet protect against infection. How infection modulates this is unclear. Salmonella can colonize systemic sites including the BM and spleen. This resolving infection has multiple IFN-γ-mediated acute and chronic effects on BM progenitors, and during the first week of infection IFN-γ is produced by myeloid, NK, NKT, CD4(+) T cells, and some lineage-negative cells. After infection, the phenotype of BM progenitors rapidly but reversibly alters, with a peak ∼ 30-fold increase in Sca-1(hi) progenitors and a corresponding loss of Sca-1(lo/int) subsets. Most strikingly, the capacity of donor Sca-1(hi) cells to reconstitute an irradiated host is reduced; the longer donor mice are exposed to infection, and Sca-1(hi) c-kit(int) cells have an increased potential to generate B1a-like cells. Thus, Salmonella can have a prolonged influence on BM progenitor functionality not directly related to bacterial persistence. These results reflect changes observed in leucopoiesis during aging and suggest that BM functionality can be modulated by life-long, periodic exposure to infection. Better understanding of this process could offer novel therapeutic opportunities to modulate BM functionality and promote healthy aging.


Asunto(s)
Células de la Médula Ósea/inmunología , Salmonelosis Animal/inmunología , Células Madre/inmunología , Animales , Antígenos Ly/inmunología , Células de la Médula Ósea/microbiología , Células de la Médula Ósea/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Homeostasis/inmunología , Interferón gamma/inmunología , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Salmonella/inmunología , Salmonelosis Animal/patología , Células Madre/microbiología , Células Madre/patología
3.
Blood ; 120(23): 4552-9, 2012 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-23065152

RESUMEN

Antibody-forming cells (AFCs) expressing the chemokine receptor CXCR3 are recruited to sites of inflammation where they help clear pathogens but may participate in autoimmune diseases. Here we identify a mechanism that induces CXCR3 expression by AFC and germinal center (GC) B cells. This happens when CD8 T cells are recruited into CD4 T cell-dependent B-cell responses. Ovalbumin-specific CD4 T cells (OTII) were transferred alone or with ovalbumin-specific CD8 T cells (OTI) and the response to subcutaneous alum-precipitated ovalbumin was followed in the draining lymph nodes. OTII cells alone induce T helper 2-associated class switching to IgG1, but few AFC or GC B cells express CXCR3. By contrast, OTI-derived IFN-γ induces most responding GC B cells and AFCs to express high levels of CXCR3, and diverse switching to IgG2a, IgG2b, with some IgG1. Up-regulation of CXCR3 by GC B cells and AFCs and their migration toward its ligand CXCL10 are shown to depend on B cells' intrinsic T-bet, a transcription factor downstream of the IFN-γR signaling. This model clarifies how precursors of long-lived AFCs and memory B cells acquire CXCR3 that causes their migration to inflammatory foci.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Movimiento Celular/inmunología , Receptores CXCR3/inmunología , Proteínas de Dominio T Box/inmunología , Vacunas/inmunología , Traslado Adoptivo , Compuestos de Alumbre , Animales , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/trasplante , Diferenciación Celular/inmunología , Quimiocina CXCL10/inmunología , Quimiocina CXCL10/metabolismo , Citometría de Flujo , Centro Germinal/inmunología , Centro Germinal/metabolismo , Inmunización/métodos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ligandos , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Receptores CXCR3/genética , Receptores CXCR3/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Células Th2/inmunología , Células Th2/metabolismo , Regulación hacia Arriba/genética
4.
J Immunol ; 189(12): 5745-54, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23152564

RESUMEN

Mucosal immunity is poorly activated after systemic immunization with protein Ags. Nevertheless, induction of mucosal immunity in such a manner would be an attractive and simple way to overcome the intrinsic difficulties in delivering Ag to such sites. Flagellin from Salmonella enterica serovar Typhimurium (FliC) can impact markedly on host immunity, in part via its recognition by TLR5. In this study, we show that systemic immunization with soluble FliC (sFliC) drives distinct immune responses concurrently in the mesenteric lymph nodes (MLN) and the spleen after i.p. and s.c. immunization. In the MLN, but not the spleen, sFliC drives a TLR5-dependent recruitment of CD103(+) dendritic cells (DCs), which correlates with a diminution in CD103(+) DC numbers in the lamina propria. In the MLN, CD103(+) DCs carry Ag and are the major primers of endogenous and transgenic T cell priming. A key consequence of these interactions with CD103(+) DCs in the MLN is an increase in local regulatory T cell differentiation. In parallel, systemic sFliC immunization results in a pronounced switching of FliC-specific B cells to IgA in the MLN but not elsewhere. Loss of TLR5 has more impact on MLN than splenic Ab responses, reflected in an ablation of IgA, but not IgG, serum Ab titers. Therefore, systemic sFliC immunization targets CD103(+) DCs and drives distinct mucosal T and B cell responses. This offers a potential "Trojan horse" approach to modulate mucosal immunity by systemically immunizing with sFliC.


Asunto(s)
Antígenos CD/biosíntesis , Células Dendríticas/inmunología , Flagelina/administración & dosificación , Flagelina/inmunología , Factores de Transcripción Forkhead/biosíntesis , Inmunoglobulina A/biosíntesis , Cadenas alfa de Integrinas/biosíntesis , Ganglios Linfáticos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/metabolismo , Factores de Transcripción Forkhead/fisiología , Inmunización , Ganglios Linfáticos/citología , Ganglios Linfáticos/metabolismo , Mesenterio , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Membrana Mucosa/citología , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Infecciones por Salmonella/metabolismo , Infecciones por Salmonella/patología , Infecciones por Salmonella/prevención & control , Salmonella typhimurium , Bazo/citología , Bazo/inmunología , Bazo/metabolismo , Linfocitos T Reguladores/metabolismo
5.
J Immunol ; 189(9): 4266-74, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22993205

RESUMEN

Thymic atrophy is a frequent consequence of infection with bacteria, viruses, and parasites and is considered a common virulence trait between pathogens. Multiple reasons have been proposed to explain this atrophy, including premature egress of immature thymocytes, increased apoptosis, or thymic shutdown to prevent tolerance to the pathogen from developing. The severe loss in thymic cell number can reflect an equally dramatic reduction in thymic output, potentially reducing peripheral T cell numbers. In this study, we examine the relationship between systemic Salmonella infection and thymic function. During infection, naive T cell numbers in peripheral lymphoid organs increase. Nevertheless, this occurs despite a pronounced thymic atrophy caused by viable bacteria, with a peak 50-fold reduction in thymocyte numbers. Thymic atrophy is not dependent upon homeostatic feedback from peripheral T cells or on regulation of endogenous glucocorticoids, as demonstrated by infection of genetically altered mice. Once bacterial numbers fall, thymocyte numbers recover, and this is associated with increases in the proportion and proliferation of early thymic progenitors. During atrophy, thymic T cell maturation is maintained, and single-joint TCR rearrangement excision circle analysis reveals there is only a modest fall in recent CD4(+) thymic emigrants in secondary lymphoid tissues. Thus, thymic atrophy does not necessarily result in a matching dysfunctional T cell output, and thymic homeostasis can constantly adjust to systemic infection to ensure that naive T cell output is maintained.


Asunto(s)
Recuperación de la Función/inmunología , Infecciones por Salmonella/inmunología , Timo/inmunología , Timo/patología , Animales , Atrofia , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Ratones , Infecciones por Salmonella/patología , Infecciones por Salmonella/fisiopatología , Salmonella typhimurium/inmunología , Timo/microbiología
6.
J Immunol ; 187(4): 1553-65, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21734076

RESUMEN

Hematopoietic cells constitutively express CD31/PECAM1, a signaling adhesion receptor associated with controlling responses to inflammatory stimuli. Although expressed on CD4(+) T cells, its function on these cells is unclear. To address this, we have used a model of systemic Salmonella infection that induces high levels of T cell activation and depends on CD4(+) T cells for resolution. Infection of CD31-deficient (CD31KO) mice demonstrates that these mice fail to control infection effectively. During infection, CD31KO mice have diminished numbers of total CD4(+) T cells and IFN-γ-secreting Th1 cells. This is despite a higher proportion of CD31KO CD4(+) T cells exhibiting an activated phenotype and an undiminished capacity to prime normally and polarize to Th1. Reduced numbers of T cells reflected the increased propensity of naive and activated CD31KO T cells to undergo apoptosis postinfection compared with wild-type T cells. Using adoptive transfer experiments, we show that loss of CD31 on CD4(+) T cells alone is sufficient to account for the defective CD31KO T cell accumulation. These data are consistent with CD31 helping to control T cell activation, because in its absence, T cells have a greater propensity to become activated, resulting in increased susceptibility to become apoptotic. The impact of CD31 loss on T cell homeostasis becomes most pronounced during severe, inflammatory, and immunological stresses such as those caused by systemic Salmonella infection. This identifies a novel role for CD31 in regulating CD4 T cell homeostasis.


Asunto(s)
Apoptosis/inmunología , Activación de Linfocitos/inmunología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Infecciones por Salmonella/inmunología , Salmonella/inmunología , Células TH1/inmunología , Traslado Adoptivo , Animales , Apoptosis/genética , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Inflamación/genética , Inflamación/inmunología , Inflamación/microbiología , Activación de Linfocitos/genética , Ratones , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Salmonella/genética , Infecciones por Salmonella/genética
7.
Proc Natl Acad Sci U S A ; 107(40): 17292-7, 2010 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-20855629

RESUMEN

Alum-precipitated protein (alum protein) vaccines elicit long-lasting neutralizing antibody responses that prevent bacterial exotoxins and viruses from entering cells. Typically, these vaccines induce CD4 T cells to become T helper 2 (Th2) cells that induce Ig class switching to IgG1. We now report that CD8 T cells also respond to alum proteins, proliferating extensively and producing IFN-γ, a key Th1 cytokine. These findings led us to question whether adoptive transfer of antigen-specific CD8 T cells alters the characteristic CD4 Th2 response to alum proteins and the switching pattern in responding B cells. To this end, WT mice given transgenic ovalbumin (OVA)-specific CD4 (OTII) or CD8 (OTI) T cells, or both, were immunized with alum-precipitated OVA. Cotransfer of antigen-specific CD8 T cells skewed switching patterns in responding B cells from IgG1 to IgG2a and IgG2b. Blocking with anti-IFN-γ antibody largely inhibited this altered B-cell switching pattern. The transcription factor T-bet is required in B cells for IFN-γ-dependent switching to IgG2a. By contrast, we show that this transcription factor is dispensable in B cells both for IFN-γ-induced switching to IgG2b and for inhibition of switching to IgG1. Thus, T-bet dependence identifies distinct transcriptional pathways in B cells that regulate IFN-γ-induced switching to different IgG isotypes.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD8-positivos/inmunología , Cambio de Clase de Inmunoglobulina , Interferón gamma/inmunología , Ovalbúmina/inmunología , Proteínas de Dominio T Box/metabolismo , Vacunas/inmunología , Traslado Adoptivo , Compuestos de Alumbre , Animales , Linfocitos B/citología , Linfocitos T CD8-positivos/citología , Ratones , Ratones Endogámicos C57BL
8.
Eur J Immunol ; 41(9): 2654-65, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21630252

RESUMEN

Control of intracellular Salmonella infection requires Th1 priming and IFN-γ production. Here, we show that efficient Th1 priming after Salmonella infection requires CD11c(+) CD11b(hi) F4/80(+) monocyte-derived dendritic cells (moDCs). In non-infected spleens, moDCs are absent from T-cell zones (T zones) of secondary lymphoid tissues, but by 24 h post-infection moDCs are readily discernible in these sites. The accumulation of moDCs is more dependent upon bacterial viability than bacterial virulence. Kinetic studies showed that moDCs were necessary to prime but not sustain Th1 responses, while ex vivo studies showed that antigen-experienced moDCs were sufficient to induce T-cell proliferation and IFN-γ production via a TNF-α-dependent mechanism. Importantly, moDCs and cDCs when co-cultured induced superior Th1 differentiation than either subset alone, and this activity was independent of TNF-α. Thus, optimal Th1 development to Salmonella requires the rapid accumulation of moDCs within T zones and their collaboration with cDCs.


Asunto(s)
Células Dendríticas/metabolismo , Infecciones por Salmonella/inmunología , Salmonella/inmunología , Bazo/patología , Células TH1/metabolismo , Animales , Presentación de Antígeno , Antígenos de Diferenciación/biosíntesis , Antígeno CD11b/biosíntesis , Antígeno CD11c/biosíntesis , Comunicación Celular , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Células Dendríticas/inmunología , Células Dendríticas/microbiología , Células Dendríticas/patología , Interferón gamma/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Monocitos/patología , Salmonella/patogenicidad , Células TH1/inmunología , Células TH1/microbiología , Células TH1/patología , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
9.
Proc Natl Acad Sci U S A ; 106(24): 9803-8, 2009 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-19487686

RESUMEN

Invasive nontyphoidal Salmonella (NTS), including Salmonella typhimurium (STm), are major yet poorly-recognized killers of infants in sub-Saharan Africa. Death in these children is usually associated with bacteremia, commonly in the absence of gastrointestinal symptoms. Evidence from humans and animal studies suggest that severe infection and bacteremia occur when specific Ab is lacking. Understanding how Ab responses to Salmonella are regulated will help develop vaccines against these devastating infections. STm induces atypical Ab responses characterized by prominent, accelerated, extrafollicular T-independent (TI) Ab against a range of surface antigens. These responses develop without concomitant germinal centers, which only appear as infection resolves. Here, we show STm rapidly induces a population of TI B220(+)CD5(-) B1b cells during infection and TI Ab from B1b cells targets the outer membrane protein (Omp) porins OmpC, OmpD and OmpF but not flagellin. When porins are used as immunogens they can ablate bacteremia and provide equivalent protection against STm as killed bacterial vaccine and this is wholly B cell-dependent. Furthermore Ab from porin-immunized chimeras, that have B1b cells, is sufficient to impair infection. Infecting with porin-deficient bacteria identifies OmpD, a protein absent from Salmonella Typhi, as a key target of Ab in these infections. This work broadens the recognized repertoire of TI protein antigens and highlights the importance of Ab from different B cell subsets in controlling STm infection. OmpD is a strong candidate vaccine target and may, in part, explain the lack of cross-protection between Salmonella Typhi and STm infections.


Asunto(s)
Anticuerpos Antibacterianos/biosíntesis , Porinas/inmunología , Salmonella/metabolismo , Animales , Linfocitos B/citología , Secuencia de Bases , Western Blotting , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Ratones , Cavidad Peritoneal/citología , Salmonella/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA