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












Base de datos
Intervalo de año de publicación
1.
Front Immunol ; 11: 1861, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32973768

RESUMEN

Indoleamine-2,3-dioxygenase (IDO)1 and IDO2 are two closely related tryptophan catabolizing enzymes encoded by linked genes. The IDO pathway is also immunomodulatory, with IDO1 well-characterized as a mediator of tumor immune evasion. Due to its homology with IDO1, IDO2 has been proposed to have a similar immunoregulatory function. Indeed, IDO2, like IDO1, is necessary for the differentiation of regulatory T cells in vitro. However, compared to IDO1, in vivo studies demonstrated a contrasting role for IDO2, with experiments in preclinical models of autoimmune arthritis establishing a proinflammatory role for IDO2 in mediating B and T cell activation driving autoimmune disease. Given their potentially opposing roles in inflammatory responses, interpretation of results obtained using IDO1 or IDO2 single knockout mice could be complicated by the expression of the other enzyme. Here we use IDO1 and IDO2 single and double knockout (dko) mice to define the differential roles of IDO1 and IDO2 in B cell-mediated immune responses. Autoreactive T and B cell responses and severity of joint inflammation were decreased in IDO2 ko, but not IDO1 ko arthritic mice. Dko mice had a reduction in the number of autoantibody secreting cells and severity of arthritis: however, percentages of differentiated T cells and their associated cytokines were not reduced compared to IDO1 ko or wild-type mice. These data suggest that autoreactive B cell responses are mediated by IDO2, while autoreactive T cell responses are indirectly affected by IDO1 expression in the IDO2 ko mice. IDO2 also influenced antibody responses in models of influenza infection and immunization with T cell-independent type II antigens. Taken together, these studies provide evidence for the contrasting roles IDO1 and IDO2 play in immune responses, with IDO1 mediating T cell suppressive effects and IDO2 working directly in B cells as a proinflammatory mediator of B cell responses.


Asunto(s)
Linfocitos B/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Activación de Linfocitos/inmunología , Linfocitos T/inmunología , Animales , Artritis Experimental/inmunología , Humanos , Inflamación/inmunología , Ratones , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología
2.
J Immunol ; 194(8): 3784-97, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25780041

RESUMEN

How the formation and activity of CD4(+)Foxp3(+) regulatory T cells (Tregs) are shaped by TCR recognition of the diverse array of peptide:MHC complexes that can be generated from self-antigens and/or foreign Ags in vivo remains poorly understood. We show that a self-peptide with low (but not high) stimulatory potency promotes thymic Treg formation and can induce conventional CD4(+) T cells in the periphery to become Tregs that express different levels of the transcription factor Helios according to anatomical location. When Tregs generated in response to this self-peptide subsequently encountered the same peptide derived instead from influenza virus in the lung-draining lymph nodes of infected mice, they proliferated, acquired a T-bet(+)CXCR3(+) phenotype, and suppressed the antiviral effector T cell response in the lungs. However, these self-antigen-selected Tregs were unable to suppress the antiviral immune response based on recognition of the peptide as a self-antigen rather than a viral Ag. Notably, when expressed in a more immunostimulatory form, the self-peptide inhibited the formation of T-bet(+)CXCR3(+) Tregs in response to viral Ag, and Ag-expressing B cells from these mice induced Treg division without upregulation of CXCR3. These studies show that a weakly immunostimulatory self-peptide can induce thymic and peripheral Foxp3(+) Treg formation but is unable to activate self-antigen-selected Tregs to modulate an antiviral immune response. Moreover, a strongly immunostimulatory self-peptide expressed by B cells induced Tregs to proliferate without acquiring an effector phenotype that allows trafficking from the draining lymph node to the lungs and, thereby, prevented the Tregs from suppressing the antiviral immune response.


Asunto(s)
Presentación de Antígeno , Diferenciación Celular/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Pulmón/inmunología , Infecciones por Orthomyxoviridae/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos Virales/inmunología , Autoantígenos/inmunología , Linfocitos B/inmunología , Linfocitos B/patología , Diferenciación Celular/genética , División Celular/inmunología , Pulmón/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/patología , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Proteínas de Dominio T Box/inmunología , Linfocitos T Reguladores/patología
3.
J Virol ; 88(20): 11995-2005, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25100838

RESUMEN

Pathogen-specific antibodies (Abs) protect against respiratory infection with influenza A virus (IAV) and Streptococcus pneumoniae and are the basis of effective vaccines. Sequential or overlapping coinfections with both pathogens are common, yet the impact of coinfection on the generation and maintenance of Ab responses is largely unknown. We report here that the B cell response to IAV is altered in mice coinfected with IAV and S. pneumoniae and that this response differs, depending on the order of pathogen exposure. In mice exposed to S. pneumoniae prior to IAV, the initial virus-specific germinal center (GC) B cell response is significantly enhanced in the lung-draining mediastinal lymph node and spleen, and there is an increase in CD4(+) T follicular helper (TFH) cell numbers. In contrast, secondary S. pneumoniae infection exaggerates early antiviral antibody-secreting cell formation, and at later times, levels of GCs, TFH cells, and antiviral serum IgG are elevated. Mice exposed to S. pneumoniae prior to IAV do not maintain the initially robust GC response in secondary lymphoid organs and exhibit reduced antiviral serum IgG with diminished virus neutralization activity a month after infection. Our data suggest that the history of pathogen exposures can critically affect the generation of protective antiviral Abs and may partially explain the differential susceptibility to and disease outcomes from IAV infection in humans. Importance: Respiratory tract coinfections, specifically those involving influenza A viruses and Streptococcus pneumoniae, remain a top global health burden. We sought to determine how S. pneumoniae coinfection modulates the B cell immune response to influenza virus since antibodies are key mediators of protection.


Asunto(s)
Linfocitos B/inmunología , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Infecciones Neumocócicas/inmunología , Streptococcus pneumoniae/inmunología , Animales , Femenino , Citometría de Flujo , Centro Germinal , Humanos , Gripe Humana/complicaciones , Ratones , Ratones Endogámicos BALB C , Infecciones Neumocócicas/complicaciones
4.
Virology ; 462-463: 254-65, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24999050

RESUMEN

The response to influenza virus (IAV) infection and severity of disease is highly variable in humans. We hypothesized that one factor contributing to this variability is the presence of specific respiratory tract (RT) microbes. One such microbe is Streptococcus pneumoniae (Sp) that is carried asymptomatically in the RT of many humans. In a mouse co-infection model we found that in contrast to secondary bacterial infection that exacerbates disease, Sp colonization 10 days prior to IAV protects from virus-induced morbidity and lung pathology. Using mutant Sp strains, we identified a critical role for the bacterial virulence factor pneumolysin (PLY) in mediating this protection. Colonization with the PLY-sufficient Sp strain induces expression of the immune-suppressive enzyme arginase 1 in alveolar macrophages (aMø) and correlates with attenuated recruitment and function of pulmonary inflammatory cells. Our study demonstrates a novel role for PLY in Sp-mediated protection by maintaining aMø as "gatekeepers" against virus-induced immunopathology.


Asunto(s)
Infecciones por Orthomyxoviridae/inmunología , Orthomyxoviridae/inmunología , Streptococcus pneumoniae/enzimología , Estreptolisinas/inmunología , Estreptolisinas/metabolismo , Animales , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Modelos Animales de Enfermedad , Factores Inmunológicos/inmunología , Factores Inmunológicos/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/prevención & control , Análisis de Supervivencia
6.
Immunol Rev ; 259(1): 11-22, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24712456

RESUMEN

Foxp3(+) regulatory T (Treg) cells are required to prevent the immune system from spontaneously mounting a severe autoaggressive lymphoproliferative disease and can modulate immune responses in a variety of settings, including infections. In this review, we describe studies that use transgenic mice to determine how signals through the T-cell receptor (TCR) contribute to the development, differentiation, and activity of Treg cells in in vivo settings. By varying the amount and quality of the self-peptide recognized by an autoreactive TCR, we have shown that the interplay between autoreactive thymocyte deletion and Treg cell formation leads to a Treg cell repertoire that is biased toward low abundance agonist self-peptides. In an autoimmune disease setting, we have demonstrated that diverse TCR specificities can be required in order for Treg cells to prevent disease in a mouse model of autoimmune inflammatory arthritis. Lastly, we have shown that Treg cells initially selected based on specificity for a self-peptide can be activated by TCR recognition of a viral peptide, and that they can acquire a specialized phenotype and suppress antiviral effector cell activity at the site of infection. These studies provide insights into the pivotal role that TCR specificity plays in the formation and activity of Treg cells.


Asunto(s)
Péptidos/inmunología , Péptidos/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Antígenos Virales/inmunología , Artritis/inmunología , Artritis/metabolismo , Autoantígenos/química , Autoantígenos/inmunología , Autoantígenos/metabolismo , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Supresión Clonal/inmunología , Factores de Transcripción Forkhead/metabolismo , Humanos , Complejo Mayor de Histocompatibilidad/inmunología , Fenotipo , Unión Proteica/inmunología , Timocitos/citología , Timocitos/inmunología , Timo/citología , Timo/inmunología , Virosis/inmunología , Virosis/metabolismo
7.
Cancer Res ; 74(5): 1390-1403, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24590809

RESUMEN

Proteins that communicate signals from the cytoskeleton to the nucleus are prime targets for effectors of metastasis as they often transduce signals regulating adhesion, motility, and invasiveness. LIM domain proteins shuttle between the cytoplasm and the nucleus, and bind to partners in both compartments, often coupling changes in gene expression to extracellular cues. In this work, we characterize LIMD2, a mechanistically undefined LIM-only protein originally found to be overexpressed in metastatic lesions but absent in the matched primary tumor. LIMD2 levels in fresh and archival tumors positively correlate with cell motility, metastatic potential, and grade, including bladder, melanoma, breast, and thyroid tumors. LIMD2 directly contributes to these cellular phenotypes as shown by overexpression, knockdown, and reconstitution experiments in cell culture models. The solution structure of LIMD2 that was determined using nuclear magnetic resonance revealed a classic LIM-domain structure that was highly related to LIM1 of PINCH1, a core component of the integrin-linked kinase-parvin-pinch complex. Structural and biochemical analyses revealed that LIMD2 bound directly to the kinase domain of integrin-linked kinase (ILK) near the active site and strongly activated ILK kinase activity. Cells that were null for ILK failed to respond to the induction of invasion by LIMD2. This strongly suggests that LIMD2 potentiates its biologic effects through direct interactions with ILK, a signal transduction pathway firmly linked to cell motility and invasion. In summary, LIMD2 is a new component of the signal transduction cascade that links integrin-mediated signaling to cell motility/metastatic behavior and may be a promising target for controlling tumor spread.


Asunto(s)
Movimiento Celular/genética , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Progresión de la Enfermedad , Fibroblastos/patología , Células HEK293 , Humanos , Células MCF-7 , Ratones , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Neoplasias/patología , Unión Proteica/genética , Transducción de Señal/genética
8.
J Immunol ; 192(7): 3043-56, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24591372

RESUMEN

Although therapies targeting distinct cellular pathways (e.g., anticytokine versus anti-B cell therapy) have been found to be an effective strategy for at least some patients with inflammatory arthritis, the mechanisms that determine which pathways promote arthritis development are poorly understood. We have used a transgenic mouse model to examine how variations in the CD4(+) T cell response to a surrogate self-peptide can affect the cellular pathways that are required for arthritis development. CD4(+) T cells that are highly reactive with the self-peptide induce inflammatory arthritis that affects male and female mice equally. Arthritis develops by a B cell-independent mechanism, although it can be suppressed by an anti-TNF treatment, which prevented the accumulation of effector CD4(+) Th17 cells in the joints of treated mice. By contrast, arthritis develops with a significant female bias in the context of a more weakly autoreactive CD4(+) T cell response, and B cells play a prominent role in disease pathogenesis. In this setting of lower CD4(+) T cell autoreactivity, B cells promote the formation of autoreactive CD4(+) effector T cells (including Th17 cells), and IL-17 is required for arthritis development. These studies show that the degree of CD4(+) T cell reactivity for a self-peptide can play a prominent role in determining whether distinct cellular pathways can be targeted to prevent the development of inflammatory arthritis.


Asunto(s)
Artritis/inmunología , Autoinmunidad/inmunología , Linfocitos T CD4-Positivos/inmunología , Transducción de Señal/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Artritis/genética , Artritis/prevención & control , Autoinmunidad/genética , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Femenino , Citometría de Flujo , Miembro Anterior/inmunología , Miembro Anterior/metabolismo , Miembro Anterior/patología , Factores de Transcripción Forkhead/inmunología , Factores de Transcripción Forkhead/metabolismo , Miembro Posterior/inmunología , Miembro Posterior/metabolismo , Miembro Posterior/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Factores Sexuales , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
9.
Eur J Immunol ; 44(3): 785-93, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24307208

RESUMEN

Autoreactive CD4(+) CD8(-) (CD4SP) thymocytes can be subjected to deletion when they encounter self-peptide during their development, but they can also undergo selection to become CD4SPFoxp3(+) Treg cells. We have analyzed the relationship between these distinct developmental fates using mice in which signals transmitted by the TCR have been attenuated by mutation of a critical tyrosine residue of the adapter protein SLP-76. In mice containing polyclonal TCR repertoires, the mutation caused increased frequencies of CD4SPFoxp3(+) thymocytes. CD4SP thymocytes expressing TCR Vß-chains that are subjected to deletion by endogenous retroviral superantigens were also present at increased frequencies, particularly among Foxp3(+) thymocytes. In transgenic mice in which CD4SP thymocytes expressing an autoreactive TCR undergo both deletion and Treg-cell formation in response to a defined self-peptide, SLP-76 mutation abrogated deletion of autoreactive CD4SP thymocytes. Notably, Foxp3(+) Treg-cell formation still occurred, albeit with a reduced efficiency, and the mutation was also associated with decreased Nur77 expression by the autoreactive CD4SP thymocytes. These studies provide evidence that the strength of the TCR signal can play a direct role in directing the extent of both thymocyte deletion and Treg-cell differentiation, and suggest that distinct TCR signaling thresholds and/or pathways can promote CD4SP thymocyte deletion versus Treg-cell formation.


Asunto(s)
Autoantígenos/inmunología , Supresión Clonal/inmunología , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Timocitos/inmunología , Timocitos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Presentación de Antígeno , Autoantígenos/química , Autoinmunidad , Supresión Clonal/genética , Factores de Transcripción Forkhead/metabolismo , Expresión Génica , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Mutación , Péptidos/química , Fenotipo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Transducción de Señal
10.
J Immunol ; 190(12): 6115-25, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23667113

RESUMEN

We examined the formation, participation, and functional specialization of virus-reactive Foxp3(+) regulatory T cells (Tregs) in a mouse model of influenza virus infection. "Natural" Tregs generated intrathymically, based on interactions with a self-peptide, proliferated in response to a homologous viral Ag in the lungs and, to a lesser extent, in the lung-draining mediastinal lymph nodes (medLNs) of virus-infected mice. In contrast, conventional CD4(+) T cells with identical TCR specificity underwent little or no conversion to become "adaptive" Tregs. The virus-reactive Tregs in the medLNs and the lungs of infected mice upregulated a variety of molecules associated with Treg activation, as well as acquired expression of molecules (T-bet, Blimp-1, and IL-10) that confer functional specialization to Tregs. Notably, however, the phenotypes of the T-bet(+) Tregs obtained from these sites were distinct, because Tregs isolated from the lungs expressed significantly higher levels of T-bet, Blimp-1, and IL-10 than did Tregs from the medLNs. Adoptive transfer of Ag-reactive Tregs led to decreased proliferation of antiviral CD4(+) and CD8(+) effector T cells in the lungs of infected hosts, whereas depletion of Tregs had a reciprocal effect. These studies demonstrate that thymically generated Tregs can become activated by a pathogen-derived peptide and acquire discrete T-bet(+) Treg phenotypes while participating in and modulating an antiviral immune response.


Asunto(s)
Activación de Linfocitos/inmunología , Infecciones por Orthomyxoviridae/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Antígenos Virales/inmunología , Diferenciación Celular/inmunología , Citometría de Flujo , Factores de Transcripción Forkhead/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Orthomyxoviridae/inmunología , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/citología , Linfocitos T Reguladores/citología
11.
J Immunol ; 190(7): 3134-41, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23420889

RESUMEN

We have examined mechanisms underlying the formation of pathologic Th17 cells using a transgenic mouse model in which autoreactive CD4(+) T cells recognize influenza virus hemagglutinin (HA) as a ubiquitously expressed self-Ag and induce inflammatory arthritis. The lymph nodes of arthritic mice contain elevated numbers of inflammatory monocytes (iMO) with an enhanced capacity to promote CD4(+) Th17 cell differentiation, and a regional inflammatory response develops in the paw-draining lymph nodes by an IL-17-dependent mechanism. The activation of these Th17-trophic iMO precedes arthritis development and occurs in the context of an autoreactive CD4(+) Th1 cell response. Adoptive transfer of HA-specific CD4(+) T cells into nonarthritic mice expressing HA as a self-Ag similarly led to the formation of Th1 cells and of iMO that could support Th17 cell formation, and, notably, the accumulation of these iMO in the lymph nodes was blocked by IFN-γ neutralization. These studies show that autoreactive CD4(+) Th1 cells directed to a systemically distributed self-Ag can promote the development of a regional Th17 cell inflammatory response by driving the recruitment of Th17-trophic iMO to the lymph nodes.


Asunto(s)
Artritis Experimental/inmunología , Autoinmunidad , Monocitos/inmunología , Células TH1/inmunología , Células Th17/inmunología , Animales , Autoantígenos/inmunología , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Citocinas/inmunología , Inflamación/inmunología , Interferón gamma/biosíntesis , Activación de Linfocitos/inmunología , Ratones , Ratones Congénicos , Ratones Noqueados
12.
J Immunol ; 188(9): 4171-80, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22450809

RESUMEN

CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) are required to restrain the immune system from mounting an autoaggressive systemic inflammatory response, but why their activity can prevent (or allow) organ-specific autoimmunity remains poorly understood. We have examined how TCR specificity contributes to Treg activity using a mouse model of spontaneous autoimmune arthritis, in which CD4(+) T cells expressing a clonotypic TCR induce disease by an IL-17-dependent mechanism. Administration of polyclonal Tregs suppressed Th17 cell formation and prevented arthritis development; notably, Tregs expressing the clonotypic TCR did not. These clonotypic Tregs exerted Ag-specific suppression of effector CD4(+) T cells using the clonotypic TCR in vivo, but failed to mediate bystander suppression and did not prevent Th17 cells using nonclonotypic TCRs from accumulating in joint-draining lymph nodes of arthritic mice. These studies indicate that the availability of Tregs with diverse TCR specificities can be crucial to their activity in autoimmune arthritis.


Asunto(s)
Artritis Experimental/inmunología , Enfermedades Autoinmunes/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Animales , Artritis Experimental/patología , Enfermedades Autoinmunes/patología , Interleucina-17/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Especificidad de Órganos/inmunología , Linfocitos T Reguladores/patología
13.
PLoS One ; 6(12): e28445, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22180783

RESUMEN

BACKGROUND: The extracellular domain of the influenza A virus protein matrix protein 2 (M2e) is remarkably conserved between various human isolates and thus is a viable target antigen for a universal influenza vaccine. With the goal of inducing protection in multiple mouse haplotypes, M2e-based multiple antigenic peptides (M2e-MAP) were synthesized to contain promiscuous T helper determinants from the Plasmodium falciparum circumsporozoite protein, the hepatitis B virus antigen and the influenza virus hemagglutinin. Here, we investigated the nature of the M2e-MAP-induced B cell response in terms of the distribution of antibody (Ab) secreting cells (ASCs) and Ab isotypes, and tested the protective efficacy in various mouse strains. METHODOLOGY/PRINCIPAL FINDINGS: Immunization of BALB/c mice with M2e-MAPs together with potent adjuvants, CpG 1826 oligonucleotides (ODN) and cholera toxin (CT) elicited high M2e-specific serum Ab titers that protected mice against viral challenge. Subcutaneous (s.c.) and intranasal (i.n.) delivery of M2e-MAPs resulted in the induction of IgG in serum and airway secretions, however only i.n. immunization induced anti-M2e IgA ASCs locally in the lungs, correlating with M2-specific IgA in the bronchio-alveolar lavage (BAL). Interestingly, both routes of vaccination resulted in equal protection against viral challenge. Moreover, M2e-MAPs induced cross-reactive and protective responses to diverse M2e peptides and variant influenza viruses. However, in contrast to BALB/c mice, immunization of other inbred and outbred mouse strains did not induce protective Abs. This correlated with a defect in T cell but not B cell responsiveness to the M2e-MAPs. CONCLUSION/SIGNIFICANCE: Anti-M2e Abs induced by M2e-MAPs are highly cross-reactive and can mediate protection to variant viruses. Although synthetic MAPs are promising designs for vaccines, future constructs will need to be optimized for use in the genetically heterogeneous human population.


Asunto(s)
Antígenos Virales/inmunología , Linfocitos B/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Fragmentos de Péptidos/inmunología , Vacunación/métodos , Proteínas de la Matriz Viral/inmunología , Secuencia de Aminoácidos , Animales , Animales no Consanguíneos , Especificidad de Anticuerpos , Antígenos Virales/química , Reacciones Cruzadas , Epítopos de Linfocito T/inmunología , Femenino , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Mutación , Fragmentos de Péptidos/química , Plasmodium falciparum/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/genética
14.
J Clin Invest ; 121(10): 3954-64, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21881204

RESUMEN

Antiviral Abs, for example those produced in response to influenza virus infection, are critical for virus neutralization and defense against secondary infection. While the half-life of Abs is short, Ab titers can last a lifetime due to a subset of the Ab-secreting cells (ASCs) that is long lived. However, the mechanisms governing ASC longevity are poorly understood. Here, we have identified a critical role for extrinsic cytokine signals in the survival of respiratory tract ASCs in a mouse model of influenza infection. Irradiation of mice at various time points after influenza virus infection markedly diminished numbers of lung ASCs, suggesting that they are short-lived and require extrinsic factors in order to persist. Neutralization of the TNF superfamily cytokines B lymphocyte stimulator (BLyS; also known as BAFF) and a proliferation-inducing ligand (APRIL) reduced numbers of antiviral ASCs in the lungs and bone marrow, whereas ASCs in the spleen and lung-draining lymph node were surprisingly unaffected. Mice deficient in transmembrane activator and calcium-modulator and cyclophilin ligand interactor (TACI), a receptor for BLyS and APRIL, mounted an initial antiviral B cell response similar to that generated in WT mice but failed to sustain protective Ab titers in the airways and serum, leading to increased susceptibility to secondary viral challenge. These studies highlight the importance of TACI signaling for the maintenance of ASCs and protection against influenza virus infection.


Asunto(s)
Anticuerpos Antivirales/biosíntesis , Infecciones por Orthomyxoviridae/inmunología , Proteína Activadora Transmembrana y Interactiva del CAML/inmunología , Animales , Células Productoras de Anticuerpos/inmunología , Células Productoras de Anticuerpos/patología , Células Productoras de Anticuerpos/efectos de la radiación , Factor Activador de Células B/inmunología , Linfocitos T CD8-positivos/inmunología , Supervivencia Celular , Modelos Animales de Enfermedad , Femenino , Pulmón/inmunología , Pulmón/patología , Pulmón/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal/inmunología , Proteína Activadora Transmembrana y Interactiva del CAML/deficiencia , Proteína Activadora Transmembrana y Interactiva del CAML/genética , Miembro 13 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/inmunología
15.
Proc Natl Acad Sci U S A ; 108(36): 14890-5, 2011 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-21873239

RESUMEN

CD4(+)CD25(+)Foxp3(+) regulatory T (Treg) cells are generated during thymocyte development and play a crucial role in preventing the immune system from attacking the body's cells and tissues. However, how the formation of these cells is directed by T-cell receptor (TCR) recognition of self-peptide:major histocompatibility complex (MHC) ligands remains poorly understood. We show that an agonist self-peptide with which a TCR is strongly reactive can induce a combination of thymocyte deletion and CD4(+)CD25(+)Foxp3(+) Treg cell formation in vivo. A weakly cross-reactive partial agonist self-peptide could similarly induce thymocyte deletion, but failed to induce Treg cell formation. These studies indicate that CD4(+)CD25(+)Foxp3(+) Treg cell formation can require highly stringent recognition of an agonist self-peptide by developing thymocytes. They also refine the "avidity" model of thymocyte selection by demonstrating that the quality of the signal mediated by agonist self-peptides, rather than the overall intensity of TCR signaling, can be a critical factor in directing autoreactive thymocytes to undergo CD4(+)CD25(+)Foxp3(+) Treg cell formation and/or deletion during their development.


Asunto(s)
Antígenos de Histocompatibilidad/inmunología , Modelos Inmunológicos , Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Timo/inmunología , Animales , Ratones , Ratones Endogámicos BALB C , Linfocitos T Reguladores/citología , Timo/citología
16.
Blood ; 118(23): 6209-19, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21768295

RESUMEN

Effector memory T cells (T(EM)) do not cause graft-versus-host disease (GVHD), though why this is has not been elucidated. To compare the fates of alloreactive naive (T(N)) or memory (T(M)) T cells, we developed a model of GVHD in which donor T cells express a transgene-encoded TCR specific for an antigenic peptide that is ubiquitously expressed in the recipient. Small numbers of naive TCR transgenic (Tg) T cells induced a robust syndrome of GVHD in transplanted recipients. We then used an established method to convert TCR Tg cells to T(M) and tested these for GVHD induction. This allowed us to control for the potentially different frequencies of alloreactive T cells among T(N) and T(M), and to track fates of alloreactive T cells after transplantation. T(EM) caused minimal, transient GVHD whereas central memory T cells (T(CM)) caused potent GVHD. Surprisingly, T(EM) were not inert: they, engrafted, homed to target tissues, and proliferated extensively, but they produced less IFN-γ and their expansion in target tissues was limited at later time points, and local proliferation was reduced. Thus, cell-intrinsic properties independent of repertoire explain the impairment of T(EM), which can initiate but cannot sustain expansion and tissue damage.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Memoria Inmunológica/inmunología , Trasplante de Células Madre/efectos adversos , Linfocitos T/inmunología , Animales , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Fenotipo , Trasplante Homólogo
17.
Transplantation ; 91(7): 707-13, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21386770

RESUMEN

BACKGROUND: Because CD4CD25Foxp3 regulatory T cells (Tregs) are essential for the maintenance of self-tolerance, significant interest surrounds the developmental cues for thymic-derived natural Tregs (nTregs) and periphery-generated adaptive Tregs (aTregs). In the transplant setting, the allograft may play a role in the generation of alloantigen-specific Tregs, but this role remains undefined. We examined whether the immune response to a transplant allograft results in the peripheral generation of aTregs. METHODS: To identify generation of aTregs, purified graft-reactive CD4CD25 T cells were adoptively transferred to mice-bearing skin allograft. To demonstrate that aTregs are necessary for tolerance, DBA/2 skin was transplanted onto C57BL/6-RAG-1-deficient recipients adoptively transferred with purified sorted CD4CD25 T cells; half of the recipients undergo tolerance induction treatment. RESULTS: By tracking adoptively transferred cells, we show that purified graft-reactive CD4CD25 T lymphocytes up-regulate Foxp3 in mice receiving skin allografts in the absence of any treatment. Interestingly, cotransfer of antigen-specific nTregs suppresses the up-regulation of Foxp3 by inhibiting the proliferation of allograft-responsive T cells. In vitro data are consistent with our in vivo data-Foxp3 cells are generated on antigen activation, and this generation is suppressed on coculture with antigen-specific nTregs. Finally, blocking aTreg generation in grafted, rapamycin-treated mice disrupts alloantigen-specific tolerance induction. In contrast, blocking aTreg generation in grafted mice treated with nondepleting anti-CD4 plus anti-CD40L antibodies does not disrupt graft tolerance. CONCLUSIONS: We conclude that graft alloantigen stimulates the de novo generation of aTregs, and this generation may represent a necessary step in some but not all protocols of tolerance induction.


Asunto(s)
Isoantígenos/inmunología , Linfocitos T Reguladores/fisiología , Tolerancia al Trasplante , Animales , Antígenos CD40/fisiología , Ligando de CD40/fisiología , Factores de Transcripción Forkhead/genética , Interleucina-2/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Sirolimus/farmacología , Factor de Crecimiento Transformador beta/farmacología , Trasplante Homólogo
18.
Methods Mol Biol ; 707: 55-69, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21287329

RESUMEN

Natural Regulatory T (Treg) cells are a subset of CD4(+) T cells characterized by expression of the transcription factor Foxp3 and the ability to suppress immune responses. Treg cells develop in the thymus in response to highly specific interactions between the T cell receptor (TCR) and self-antigens. These processes can be recapitulated in antigen-specific systems using transgenic mice that coexpress a TCR with its cognate peptide as a neoself-antigen. Here, we describe a method for using such a system to establish a flow cytometric profile of phenotype markers expressed by developing and mature Treg cells in the thymus. Our approach is to compare antigen-specific thymocytes developing in the presence or absence of Treg cell-selecting ligands to identify phenotypic changes that characterize thymocytes undergoing selection into the Treg cell lineage.


Asunto(s)
Citometría de Flujo/métodos , Linfocitos T Reguladores/citología , Timo/citología , Animales , Antígenos/inmunología , Ratones , Ratones Transgénicos , Linfocitos T Reguladores/inmunología , Timo/inmunología
19.
Am J Respir Cell Mol Biol ; 45(3): 480-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21148741

RESUMEN

Drugs that can rapidly inhibit respiratory infection from influenza or other respiratory pathogens are needed. One approach is to engage primary innate immune defenses against viral infection, such as activating the IFN pathway. In this study, we report that a small, cell-permeable compound called 5,6-di-methylxanthenone-4-acetic acid (DMXAA) can induce protection against vesicular stomatitis virus in vitro and H1N1 influenza A virus in vitro and in vivo through innate immune activation. Using the mouse C10 bronchial epithelial cell line and primary cultures of nasal epithelial cells, we demonstrate DMXAA activates the IFN regulatory factor-3 pathway leading to production of IFN-ß and subsequent high-level induction of IFN-ß-dependent proteins, such as myxovirus resistance 1 (Mx1) and 2',5'-oligoadenylate synthetase 1 (OAS1). Mice treated with DMXAA intranasally elevate mRNA/protein expression of Mx1 and OAS1 in the nasal mucosa, trachea, and lung. When challenged intranasally with a lethal dose of H1N1 influenza A virus, DMXAA reduced viral titers in the lungs and protected 80% of mice from death, even when given at 24 hours before infection. These data show that agents, like DMXAA, that can directly activate innate immune pathways, such as the IFN regulatory factor-3/IFN-ß system, in respiratory epithelial cells can be used to protect from influenza pneumonia and potentially in other respiratory viral infections. Development of this approach in humans could be valuable for protecting health care professionals and "first responders" in the early stages of viral pandemics or bioterror attacks.


Asunto(s)
Infecciones del Sistema Respiratorio/prevención & control , Infecciones del Sistema Respiratorio/virología , Virosis/prevención & control , Animales , Antineoplásicos/farmacología , Bronquios/virología , Células Epiteliales/virología , Femenino , Humanos , Sistema Inmunológico , Inmunidad Innata , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones del Sistema Respiratorio/inmunología , Virosis/inmunología , Xantonas/farmacología
20.
J Immunol ; 185(5): 2790-9, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20686126

RESUMEN

Regulatory T cells (Tregs) are a subset of T cells with suppressive function that protect the host from autoimmunity and prevent excessive immunopathology. Functional Tregs must be present throughout life to provide continuous protection for the host. Despite the intense study of this lineage, the mechanisms by which Tregs are maintained in the steady-state remain incompletely understood. In this study, we investigated the role of dendritic cells (DCs) in the control of Treg proliferation. In the absence of overt TCR stimulation, we found that DCs induce polyclonal Treg division in murine splenocyte cultures. In vivo expansion of DCs also correlated with polyclonal Treg expansion. DC-induced Treg division required IL-2, which was provided by conventional CD4(+) T cells through an MHC class II (MHC II)-dependent interaction with DCs. Provision of exogenous IL-2 obviated the need for conventional CD4(+) T cells in the induction of Treg proliferation, but this process still required a contact-dependent but MHC II-independent interaction between DCs and Tregs. Although Treg division could occur in the absence of MHC II expression by DCs, direct stimulation of Tregs by cognate Ag/MHC II complexes enhanced IL-2-induced Treg proliferation. These data demonstrate that DCs coordinate the interactions that are necessary to initiate polyclonal Treg proliferation.


Asunto(s)
Proliferación Celular , Células Dendríticas/inmunología , Diabetes Mellitus Experimental/inmunología , Epítopos de Linfocito T/fisiología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Antígenos CD8/metabolismo , Antígenos CD8/fisiología , Línea Celular Tumoral , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/patología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/fisiología , Activación de Linfocitos/inmunología , Melanoma Experimental , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/patología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...