Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 121(5): e2311487121, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38261611

RESUMEN

Roughly one-half of mice with partial defects in two immune tolerance pathways (AireGW/+Lyn-/- mice) spontaneously develop severe damage to their retinas due to T cell reactivity to Aire-regulated interphotoreceptor retinoid-binding protein (IRBP). Single-cell T cell receptor (TCR) sequencing of CD4+ T cells specific for a predominate epitope of IRBP showed a remarkable diversity of autoantigen-specific TCRs with greater clonal expansions in mice with disease. TCR transgenic mice made with an expanded IRBP-specific TCR (P2.U2) of intermediate affinity exhibited strong but incomplete negative selection of thymocytes. This negative selection was absent in IRBP-/- mice and greatly defective in AireGW/+ mice. Most P2.U2+/- mice and all P2.U.2+/-AireGW/+ mice rapidly developed inflammation of the retina and adjacent uvea (uveitis). Aire-dependent IRBP expression in the thymus also promoted Treg differentiation, but the niche for this fate determination was small, suggesting differences in antigen presentation leading to negative selection vs. thymic Treg differentiation and a stronger role for negative selection in preventing autoimmune disease in the retina.


Asunto(s)
Presentación de Antígeno , Receptores de Antígenos de Linfocitos T , Animales , Ratones , Autoantígenos , Modelos Animales de Enfermedad , Ratones Endogámicos , Ratones Transgénicos
2.
Immunohorizons ; 3(8): 389-401, 2019 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-31427364

RESUMEN

The germinal center (GC) is the anatomical site where humoral immunity evolves. B cells undergo cycles of proliferation and selection to produce high-affinity Abs against Ag. Direct linkage of a TLR9 agonist (CpG) to a T-dependent Ag increases the number of GC B cells. We used a T-dependent Ag complexed with CpG and a genetic model for ablating the TLR9 signaling adaptor molecule MyD88 specifically in B cells (B-MyD88- mice) together with transcriptomics to determine how this innate pathway positively regulates the GC. GC B cells from complex Ag-immunized B-MyD88- mice were defective in inducing gene expression signatures downstream of c-Myc and mTORC1. In agreement with the latter gene signature, ribosomal protein S6 phosphorylation was increased in GC B cells from wild-type mice compared with B-MyD88- mice. However, GC B cell expression of a c-Myc protein reporter was enhanced by CpG attached to Ag in both wild-type and B-MyD88- mice, indicating a B cell-extrinsic effect on c-Myc protein expression combined with a B cell-intrinsic enhancement of gene expression downstream of c-Myc. Both mTORC1 activity and c-Myc are directly induced by T cell help, indicating that TLR9 signaling in GC B cells either enhances their access to T cell help or directly influences these pathways to further enhance the effect of T cell help. Taken together, these findings indicate that TLR9 signaling in the GC could provide a surrogate prosurvival stimulus, "TLR help," thus lowering the threshold for selection and increasing the magnitude of the GC response.


Asunto(s)
Antígenos/química , Linfocitos B/inmunología , Centro Germinal/metabolismo , Ligandos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptor Toll-Like 9/agonistas , Animales , Antígenos/metabolismo , Activación de Linfocitos/inmunología , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal/inmunología , Linfocitos T/inmunología , Transcriptoma , gammaglobulinas/inmunología
3.
Front Immunol ; 9: 739, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29740430

RESUMEN

This article describes the work I did in Bill Paul's lab as a postdoctoral fellow between 1979 and 1983, and to a lesser extent puts that work in the context of other work on B cell activation and antibody responses that was going on in Bill's lab at that time and shortly beforehand, including the discovery of interleukin 4. In addition, this work describes the subsequent and continuing work in my own lab following-up on themes I began during my time working directly with Bill. A particular emphasis was on understanding the biochemical mechanisms of signaling by the B cell antigen receptor (BCR) to the interior of the B cell. Some of the studies from my lab related to the regulation of BCR signaling by Lyn are described in relationship to the lymphocyte tuning hypothesis put forth by Grossman and Paul in 1992 and subsequently.


Asunto(s)
Alergia e Inmunología/historia , Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Historia del Siglo XX , Historia del Siglo XXI , Activación de Linfocitos
4.
J Immunol ; 200(3): 937-948, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29282308

RESUMEN

Although TLR signaling in B cells has been implicated in the germinal center (GC) responses during viral infections and autoimmune diseases, the underlying mechanism is unclear. Bacterial phage Qß-derived virus-like particle (Qß-VLP) contains TLR ligands, which can enhance Qß-VLP-induced Ab response, including GC response, through TLR/MyD88 signaling in B cells. In this study, by examining Ag-specific B cell response to Qß-VLP, we found that lack of B cell MyD88 from the beginning of the immune response led to a more severe defect in the GC scale than abolishing MyD88 at later time points of the immune response. Consistently, B cell-intrinsic MyD88 signaling significantly enhanced the initial proliferation of Ag-specific B cells, which was accompanied with a dramatic increase of plasma cell generation and induction of Bcl-6+ GC B cell precursors. In addition, B cell-intrinsic MyD88 signaling promoted strong T-bet expression independent of IFN-γ and led to the preferential isotype switching to IgG2a/c. Thus, by promoting the initial Ag-specific B cell proliferation and differentiation, B cell-intrinsic MyD88 signaling enhanced both T-independent and T-dependent Ab responses elicited by Qß-VLP. This finding will provide additional insight into the role of TLR signaling in antiviral immunity, autoimmune diseases, and vaccine design.


Asunto(s)
Allolevivirus/inmunología , Linfocitos B/inmunología , Centro Germinal/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Receptores Toll-Like/inmunología , Animales , Anticuerpos Antivirales/inmunología , Diferenciación Celular/inmunología , Proliferación Celular/fisiología , Femenino , Cambio de Clase de Inmunoglobulina/inmunología , Inmunoglobulina G/inmunología , Interferón gamma/inmunología , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Transducción de Señal/inmunología , Proteínas de Dominio T Box/biosíntesis , Proteínas Estructurales Virales/inmunología
5.
J Leukoc Biol ; 102(3): 791-803, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28642279

RESUMEN

Psoriasis is a chronic skin disease associated with deregulated activation of immune cells and keratinocytes. In this study, we used the imiquimod (IMQ)-induced mouse model of psoriasis to dissect better the contribution of hematopoietic and skin-resident stromal cells to psoriasis development. The comparison of disease development in mice carrying the hematopoietic cell-specific deletion of MyD88 (Myd88fl/flVav-cre+ mice) with mice carrying the total MyD88 deficiency (Myd88-/- mice), we show that the progression of skin and systemic inflammation, as well as of epidermal thickening, was completely dependent on MyD88 expression in hematopoietic cells. However, both Myd88-/- mouse strains developed some degree of epidermal thickening during the initial stages of IMQ-induced psoriasis, even in the absence of hematopoietic cell activation and infiltration into the skin, suggesting a contribution of MyD88-independent mechanisms in skin-resident stromal cells. With the use of conditional knockout mouse strains lacking MyD88 in distinct lineages of myeloid cells (Myd88fl/flLysM-cre+ and Myd88fl/flMRP8-cre+ mice), we report that MyD88 signaling in monocytes and Mϕ, but not in neutrophils, plays an important role in disease propagation and exacerbation by modulating their ability to sustain γδ T cell effector functions via IL-1ß and IL-23 production. Overall, these findings add new insights into the specific contribution of skin-resident stromal vs. hematopoietic cells to disease initiation and progression in the IMQ-induced mouse model of psoriasis and uncover a potential novel pathogenic role for monocytes/Mϕ to psoriasis development.


Asunto(s)
Aminoquinolinas/toxicidad , Inmunidad Innata/efectos de los fármacos , Células Mieloides/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Psoriasis/inducido químicamente , Psoriasis/inmunología , Animales , Modelos Animales de Enfermedad , Imiquimod , Inmunidad Innata/genética , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-23/genética , Interleucina-23/inmunología , Ratones , Ratones Noqueados , Células Mieloides/patología , Factor 88 de Diferenciación Mieloide/genética , Psoriasis/genética , Psoriasis/patología , Piel/inmunología , Piel/patología
6.
Nat Immunol ; 18(3): 248-250, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28198824
7.
PLoS One ; 11(12): e0167693, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27977701

RESUMEN

Excessive type 2 helper T cell responses to environmental antigens can cause immunopathology such as asthma and allergy, but how such immune responses are induced remains unclear. We studied this process in the airways by immunizing mice intranasally with the antigen ovalbumin together with either of two Toll-like receptor (TLR) ligands. We found the TLR5 ligand flagellin promoted a type 2 helper T cell response, whereas, a TLR9 ligand CpG oligodeoxyribonucleotide (ODN) promoted a type 1 helper T cell response. CpG ODN induced mRNA encoding interleukin (IL)-12 p40, whereas, flagellin caused IL-33 secretion and induced mRNAs encoding IL-1 and thymic stromal lymphopoietin (TSLP). By using mice deficient in the TLR and IL-1R signaling molecule, myeloid differentiation primary response 88 (MyD88), in conventional dendritic cells (cDCs) and alveolar macrophages (AMs), and by cell sorting different lung populations after 2 hours of in vivo stimulation, we characterized the cell types that rapidly produced inflammatory cytokines in response to TLR stimulation. CpG ODN was likely recognized by TLR9 on cDCs and AMs, which made mRNA encoding IL-12. IL-12 was necessary for the subsequent innate and adaptive interferon-γ production. In contrast, flagellin stimulated multiple cells of hematopoietic and non-hematopoietic origin, including AMs, DCs, monocytes, and lung epithelial cells. AMs were largely responsible for IL-1α, whereas lung epithelial cells made TSLP. Multiple hematopoietic cells, including AMs, DCs, and monocytes contributed to other cytokines, including IL-1ß and TNFα. MyD88-dependent signals, likely through IL-1R and IL-33R, and MyD88-independent signals, likely from TSLP, were necessary in cDCs for promotion of the early IL-4 response by CD4 T cells in the draining lymph node. Thus, the cell types that responded to TLR ligands were a critical determinant of the innate cytokines produced and the character of the resulting adaptive immune response in the airways.


Asunto(s)
Inmunidad Adaptativa/efectos de los fármacos , Flagelina/farmacología , Oligonucleótidos/farmacología , Receptor Toll-Like 5/metabolismo , Receptor Toll-Like 9/metabolismo , Animales , Citocinas/metabolismo , Femenino , Interleucina-1/metabolismo , Interleucina-33/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Factor 88 de Diferenciación Mieloide/metabolismo , Receptor Toll-Like 5/agonistas , Receptor Toll-Like 9/agonistas , Linfopoyetina del Estroma Tímico
8.
PLoS Pathog ; 12(8): e1005787, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27542117

RESUMEN

Soaring rates of systemic fungal infections worldwide underscore the need for vaccine prevention. An understanding of the elements that promote vaccine immunity is essential. We previously reported that Th17 cells are required for vaccine immunity to the systemic dimorphic fungi of North America, and that Card9 and MyD88 signaling are required for the development of protective Th17 cells. Herein, we investigated where, when and how MyD88 regulates T cell development. We uncovered a novel mechanism in which MyD88 extrinsically regulates the survival of activated T cells during the contraction phase and in the absence of inflammation, but is dispensable for the expansion and differentiation of the cells. The poor survival of activated T cells in Myd88-/- mice is linked to increased caspase3-mediated apoptosis, but not to Fas- or Bim-dependent apoptotic pathways, nor to reduced expression of the anti-apoptotic molecules Bcl-2 or Bcl-xL. Moreover, TLR3, 7, and/or 9, but not TLR2 or 4, also were required extrinsically for MyD88-dependent Th17 cell responses and vaccine immunity. Similar MyD88 requirements governed the survival of virus primed T cells. Our data identify unappreciated new requirements for eliciting adaptive immunity and have implications for designing vaccines.


Asunto(s)
Vacunas Fúngicas/inmunología , Activación de Linfocitos , Micosis/inmunología , Factor 88 de Diferenciación Mieloide/inmunología , Células Th17/inmunología , Animales , Proteína 11 Similar a Bcl2/genética , Proteína 11 Similar a Bcl2/inmunología , Proteínas Adaptadoras de Señalización CARD/genética , Proteínas Adaptadoras de Señalización CARD/inmunología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Supervivencia Celular/genética , Supervivencia Celular/inmunología , Ratones , Ratones Noqueados , Micosis/genética , Micosis/prevención & control , Factor 88 de Diferenciación Mieloide/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/inmunología , Receptores Toll-Like/genética , Receptores Toll-Like/inmunología , Proteína bcl-X/genética , Proteína bcl-X/inmunología , Receptor fas/genética , Receptor fas/inmunología
9.
Immunol Cell Biol ; 94(10): 918-924, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27562062

RESUMEN

Antibodies are involved in the pathogenesis of many autoimmune diseases. Although the mechanisms underlying the antibody response to infection or vaccination are reasonably well understood, we still have a poor understanding of the nature of autoimmune antibody responses. The most well studied are the anti-nuclear antibody responses characteristic of systemic lupus erythematosus and studies over the past decade or so have demonstrated a critical role for signaling by TLR7 and/or TLR9 in B cells to promote these responses. These Toll-like receptors (TLRs) can promote T-cell-independent extrafollicular antibody responses with a heavy-chain class switch and a low degree of somatic mutation, but they can also strongly boost the germinal center response that gives rise to high-affinity antibodies and long-lived plasma cells. TLRs have been shown to enhance affinity maturation in germinal center responses to produce high-affinity neutralizing antibodies in several virus infection models of mice. Although more data are needed, it appears that anti-nuclear antibodies in mouse models of lupus and in lupus patients can be generated by either pathway, provided there are genetic susceptibility alleles that compromise B-cell tolerance at one or another stage. Limited data in other autoimmune diseases suggest that the germinal center response may be the predominant pathway leading to autoantibodies in those diseases. A better understanding of the mechanisms of autoantibody production may ultimately be helpful in the development of targeted therapeutics for lupus or other autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Centro Germinal/inmunología , Animales , Formación de Anticuerpos/inmunología , Autoanticuerpos/biosíntesis , Linfocitos B/inmunología , Humanos , Ratones , Receptores Toll-Like/metabolismo
10.
J Clin Invest ; 126(10): 3758-3771, 2016 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-27571405

RESUMEN

Studies of the genetic factors associated with human autoimmune disease suggest a multigenic origin of susceptibility; however, how these factors interact and through which tolerance pathways they operate generally remain to be defined. One key checkpoint occurs through the activity of the autoimmune regulator AIRE, which promotes central T cell tolerance. Recent reports have described a variety of dominant-negative AIRE mutations that likely contribute to human autoimmunity to a greater extent than previously thought. In families with these mutations, the penetrance of autoimmunity is incomplete, suggesting that other checkpoints play a role in preventing autoimmunity. Here, we tested whether a defect in LYN, an inhibitory protein tyrosine kinase that is implicated in systemic autoimmunity, could combine with an Aire mutation to provoke organ-specific autoimmunity. Indeed, mice with a dominant-negative allele of Aire and deficiency in LYN spontaneously developed organ-specific autoimmunity in the eye. We further determined that a small pool of retinal protein-specific T cells escaped thymic deletion as a result of the hypomorphic Aire function and that these cells also escaped peripheral tolerance in the presence of LYN-deficient dendritic cells, leading to highly destructive autoimmune attack. These findings demonstrate how 2 distinct tolerance pathways can synergize to unleash autoimmunity and have implications for the genetic susceptibility of autoimmune disease.


Asunto(s)
Autoinmunidad , Factores de Transcripción/fisiología , Familia-src Quinasas/fisiología , Animales , Presentación de Antígeno , Autoanticuerpos/metabolismo , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Proteínas del Ojo/inmunología , Microbioma Gastrointestinal/inmunología , Ratones Endogámicos C57BL , Ratones Transgénicos , Especificidad de Órganos , Proteínas de Unión al Retinol/inmunología , Uveítis Posterior/genética , Uveítis Posterior/inmunología , Proteína AIRE
11.
F1000Res ; 52016.
Artículo en Inglés | MEDLINE | ID: mdl-27303636

RESUMEN

The germinal center response is the delayed but sustained phase of the antibody response that is responsible for producing high-affinity antibodies of the IgG, IgA and/or IgE isotypes. B cells in the germinal center undergo re-iterative cycles of somatic hypermutation of immunoglobulin gene variable regions, clonal expansion, and Darwinian selection for cells expressing higher-affinity antibody variants. Alternatively, selected B cells can terminally differentiate into long-lived plasma cells or into a broad diversity of mutated memory B cells; the former secrete the improved antibodies to fight an infection and to provide continuing protection from re-infection, whereas the latter may jumpstart immune responses to subsequent infections with related but distinct infecting agents. Our understanding of the molecules involved in the germinal center reaction has been informed by studies of human immunodeficiency patients with selective defects in the production of antibodies. Recent studies have begun to reveal how innate immune recognition via Toll-like receptors can enhance the magnitude and selective properties of the germinal center, leading to more effective control of infection by a subset of viruses. Just as early insights into the nature of the germinal center found application in the development of the highly successful conjugate vaccines, more recent insights may find application in the current efforts to develop new generations of vaccines, including vaccines that can induce broadly protective neutralizing antibodies against influenza virus or HIV-1.

12.
Proc Natl Acad Sci U S A ; 111(31): E3224-33, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-25053813

RESUMEN

Recent studies have demonstrated important roles of nucleic acid-sensing Toll-like receptors (TLRs) in promoting protective antibody responses against several viruses. To dissect how recognition of nucleic acids by TLRs enhances germinal center (GC) responses, mice selectively deleted for myeloid differentiation primary-response protein 88 (MyD88) in B cells or dendritic cells (DCs) were immunized with a haptenated protein antigen bound to a TLR9 ligand. TLR9 signaling in DCs led to greater numbers of follicular helper T (TFH) cells and GC B cells, and accelerated production of broad-affinity antihapten IgG. In addition to modulating GC selection by increasing inducible costimulator (ICOS) expression on TFH cells and reducing the number of follicular regulatory T cells, MyD88-dependent signaling in B cells enhanced GC output by augmenting a class switch to IgG2a, affinity maturation, and the memory antibody response. Thus, attachment of a TLR9 ligand to an oligovalent antigen acted on DCs and B cells to coordinate changes in the T-cell compartment and also promoted B cell-intrinsic effects that ultimately programmed a more potent GC response.


Asunto(s)
Formación de Anticuerpos/inmunología , Centro Germinal/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 9/metabolismo , Animales , Afinidad de Anticuerpos/efectos de los fármacos , Formación de Anticuerpos/efectos de los fármacos , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Pollos , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Factores de Transcripción Forkhead/metabolismo , Haptenos/farmacología , Memoria Inmunológica/efectos de los fármacos , Ligandos , Activación de Linfocitos/efectos de los fármacos , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/metabolismo , Nitrofenoles/farmacología , Fenotipo , gammaglobulinas/farmacología
13.
J Exp Med ; 211(5): 887-907, 2014 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-24752299

RESUMEN

Low-grade systemic inflammation is often associated with metabolic syndrome, which plays a critical role in the development of the obesity-associated inflammatory diseases, including insulin resistance and atherosclerosis. Here, we investigate how Toll-like receptor-MyD88 signaling in myeloid and endothelial cells coordinately participates in the initiation and progression of high fat diet-induced systemic inflammation and metabolic inflammatory diseases. MyD88 deficiency in myeloid cells inhibits macrophage recruitment to adipose tissue and their switch to an M1-like phenotype. This is accompanied by substantially reduced diet-induced systemic inflammation, insulin resistance, and atherosclerosis. MyD88 deficiency in endothelial cells results in a moderate reduction in diet-induced adipose macrophage infiltration and M1 polarization, selective insulin sensitivity in adipose tissue, and amelioration of spontaneous atherosclerosis. Both in vivo and ex vivo studies suggest that MyD88-dependent GM-CSF production from the endothelial cells might play a critical role in the initiation of obesity-associated inflammation and development of atherosclerosis by priming the monocytes in the adipose and arterial tissues to differentiate into M1-like inflammatory macrophages. Collectively, these results implicate a critical MyD88-dependent interplay between myeloid and endothelial cells in the initiation and progression of obesity-associated inflammatory diseases.


Asunto(s)
Células Endoteliales/metabolismo , Inflamación/metabolismo , Inflamación/fisiopatología , Insulina/metabolismo , Células Mieloides/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Obesidad/complicaciones , Análisis de Varianza , Animales , Antígeno CD11b/metabolismo , Citometría de Flujo , Inmunohistoquímica , Inflamación/etiología , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
J Immunol ; 192(3): 875-85, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24379120

RESUMEN

The intracellular tyrosine kinase Lyn mediates inhibitory receptor function in B cells and myeloid cells, and Lyn(-/-) mice spontaneously develop an autoimmune and inflammatory disease that closely resembles human systemic lupus erythematosus. TLR-signaling pathways have been implicated in the production of anti-nuclear Abs in systemic lupus erythematosus and mouse models of it. We used a conditional allele of Myd88 to determine whether the autoimmunity of Lyn(-/-) mice is dependent on TLR/MyD88 signaling in B cells and/or in dendritic cells (DCs). The production of IgG anti-nuclear Abs, as well as the deposition of these Abs in the glomeruli of the kidneys, leading to glomerulonephritis in Lyn(-/-) mice, were completely abolished by selective deletion of Myd88 in B cells, and autoantibody production and glomerulonephritis were delayed or decreased by deletion of Myd88 in DCs. The reduced autoantibody production in mice lacking MyD88 in B cells or DCs was accompanied by a dramatic decrease in the spontaneous germinal center (GC) response, suggesting that autoantibodies in Lyn(-/-) mice may depend on GC responses. Consistent with this view, IgG anti-nuclear Abs were absent if T cells were deleted (TCRß(-/-) TCRδ(-/-) mice) or if T cells were unable to contribute to GC responses as the result of mutation of the adaptor molecule SAP. Thus, the autoimmunity of Lyn(-/-) mice was dependent on T cells and on TLR/MyD88 signaling in B cells and in DCs, supporting a model in which DC hyperactivity combines with defects in tolerance in B cells to lead to a T cell-dependent systemic autoimmunity in Lyn(-/-) mice.


Asunto(s)
Anticuerpos Antinucleares/biosíntesis , Linfocitos B/inmunología , Células Dendríticas/inmunología , Centro Germinal/inmunología , Inmunoglobulina G/biosíntesis , Nefritis Lúpica/inmunología , Factor 88 de Diferenciación Mieloide/fisiología , Familia-src Quinasas/deficiencia , Animales , Anticuerpos Antinucleares/genética , Anticuerpos Antinucleares/inmunología , Complejo Antígeno-Anticuerpo/análisis , Modelos Animales de Enfermedad , Eliminación de Gen , Humanos , Inmunoglobulina G/genética , Inmunoglobulina G/inmunología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Lupus Eritematoso Sistémico , Nefritis Lúpica/patología , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Receptores de Antígenos de Linfocitos T gamma-delta/deficiencia , Autotolerancia/inmunología , Transducción de Señal/inmunología , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria , Organismos Libres de Patógenos Específicos , Receptores Toll-Like/inmunología
15.
J Immunol ; 192(3): 919-28, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24376269

RESUMEN

The Lyn tyrosine kinase regulates inhibitory signaling in B and myeloid cells: loss of Lyn results in a lupus-like autoimmune disease with hyperactive B cells and myeloproliferation. We have characterized the relative contribution of Lyn-regulated signaling pathways in B cells specifically to the development of autoimmunity by crossing the novel lyn(flox/flox) animals with mice carrying the Cre recombinase under the control of the Cd79a promoter, resulting in deletion of Lyn in B cells. The specific deletion of Lyn in B cells is sufficient for the development of immune complex-mediated glomerulonephritis. The B cell-specific Lyn-deficient mice have no defects in early bone marrow B cell development but have reduced numbers of mature B cells with poor germinal centers, as well as increased numbers of plasma and B1a cells, similar to the lyn(-/-) animals. Within 8 mo of life, B cell-specific Lyn mutant mice develop high titers of IgG anti-Smith Ag ribonucleoprotein and anti-dsDNA autoantibodies, which deposit in their kidneys, resulting in glomerulonephritis. B cell-specific Lyn mutant mice also develop myeloproliferation, similar to the lyn(-/-) animals. The additional deletion of MyD88 in B cells, achieved by crossing lyn(flox/flox)Cd79a-cre mice with myd88(flox/flox) animals, reversed the autoimmune phenotype observed in B cell-specific Lyn-deficient mice by blocking production of class-switched pathogenic IgG autoantibodies. Our results demonstrate that B cell-intrinsic Lyn-dependent signaling pathways regulate B cell homeostasis and activation, which in concert with B cell-specific MyD88 signaling pathways can drive the development of autoimmune disease.


Asunto(s)
Autoinmunidad/inmunología , Linfocitos B/enzimología , Familia-src Quinasas/deficiencia , Animales , Anticuerpos Antinucleares/biosíntesis , Anticuerpos Antinucleares/genética , Anticuerpos Antinucleares/inmunología , Especificidad de Anticuerpos , Linfocitos B/inmunología , Señalización del Calcio/inmunología , Recuento de Células , Modelos Animales de Enfermedad , Centro Germinal/inmunología , Centro Germinal/patología , Homeostasis , Enfermedades del Complejo Inmune/inmunología , Enfermedades del Complejo Inmune/patología , Cambio de Clase de Inmunoglobulina , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/inmunología , Inmunoglobulina M/biosíntesis , Inmunoglobulina M/inmunología , Riñón/inmunología , Riñón/patología , Nefritis Lúpica/enzimología , Nefritis Lúpica/etiología , Nefritis Lúpica/inmunología , Activación de Linfocitos , Linfopoyesis/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/inmunología , Trastornos Mieloproliferativos/enzimología , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/inmunología , Especificidad de Órganos , Células Plasmáticas/inmunología , Bazo/inmunología , Bazo/patología , Subgrupos de Linfocitos T/inmunología , Familia-src Quinasas/genética , Familia-src Quinasas/inmunología
16.
Sci Signal ; 6(297): ra91, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24129701

RESUMEN

Signaling downstream of the B cell antigen receptor (BCR) is tightly regulated to enable cells to gauge the strength and duration of antigen-receptor interactions and to respond appropriately. We investigated whether metabolism of the second messenger diacylglycerol (DAG) by members of the family of DAG kinases (DGKs) played a role in modulating the magnitude of signaling by DAG downstream of the BCR. In the absence of DGKζ, the threshold for BCR signaling, measured as activation of the Ras-extracellular signal-regulated kinase (ERK) pathway, was markedly reduced in mature follicular B cells, which resulted in enhanced responses to antigen in vitro and in vivo. Inhibition of DAG signaling by DGKζ limited the number of antibody-secreting cells that were generated early in response to T cell-independent type 2 antigens, as well as to T cell-dependent antigens. Furthermore, the effect of loss of DGKζ closely resembled the effect of increasing the affinity of the BCR for antigen during the T cell-dependent antibody response. These results suggest that the magnitude of DAG signaling is important for translating the affinity of the BCR for antigen into the amount of antibody produced during the early stages of an immune response.


Asunto(s)
Formación de Anticuerpos/inmunología , Diacilglicerol Quinasa/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Receptores de Antígenos de Linfocitos B/inmunología , Traslado Adoptivo , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/trasplante , Western Blotting , Proliferación Celular , Diacilglicerol Quinasa/genética , Diacilglicerol Quinasa/metabolismo , Diglicéridos/inmunología , Diglicéridos/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Citometría de Flujo , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Muramidasa/genética , Muramidasa/inmunología , Muramidasa/metabolismo , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Proteína bcl-X/inmunología , Proteína bcl-X/metabolismo
17.
Proc Natl Acad Sci U S A ; 110(35): E3311-20, 2013 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-23940344

RESUMEN

Deletion of lyn, a Src-family tyrosine kinase expressed by B, myeloid, and dendritic cells (DCs), triggers lupus-like disease in mice, characterized by autoantibody production and renal immune complex deposition leading to chronic glomerulonephritis. B cells from these mice are hyperactive to antigen-receptor stimulation owing to a loss of inhibitory signaling mediated by Lyn kinase. The hyperactive B-cell responses are thought to underlie the development of autoimmunity in this model. Lyn-deficient mice also manifest significant myeloexpansion. To test the contribution of different immune cell types to the lupus-like disease in this model, we generated a lyn(flox/flox) transgenic mouse strain. To our surprise, when we crossed these mice to Cd11c-cre animals, generating DC-specific deletion of Lyn, the animals developed spontaneous B- and T-cell activation and subsequent production of autoantibodies and severe nephritis. Remarkably, the DC-specific Lyn-deficient mice also developed severe tissue inflammatory disease, which was not present in the global lyn(-/-) strain. Lyn-deficient DCs were hyperactivated and hyperresponsive to Toll-like receptor agonists and IL-1ß. To test whether dysregulation of these signaling pathways in DCs contributed to the inflammatory/autoimmune phenotype, we crossed the lyn(f/f) Cd11c-cre(+) mice to myd88(f/f) animals, generating double-mutant mice lacking both Lyn and the adaptor protein myeloid differentiation factor 88 (MyD88) in DCs, specifically. Deletion of MyD88 in DCs alone completely reversed the inflammatory autoimmunity in the DC-specific Lyn-mutant mice. Thus, we demonstrate that hyperactivation of MyD88-dependent signaling in DCs is sufficient to drive pathogenesis of lupus-like disease, illuminating the fact that dysregulation in innate immune cells alone can lead to autoimmunity.


Asunto(s)
Autoinmunidad , Células Dendríticas/metabolismo , Inflamación/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal , Familia-src Quinasas/genética , Animales , Autoanticuerpos/biosíntesis , Células Dendríticas/enzimología , Células Dendríticas/inmunología , Enfermedades Linfáticas/genética , Enfermedades Linfáticas/metabolismo , Activación de Linfocitos , Ratones , Ratones Noqueados , Esplenomegalia/genética , Esplenomegalia/metabolismo
18.
Immunity ; 38(6): 1211-22, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23791643

RESUMEN

The intracellular signaling molecule TRAF6 is critical for Toll-like receptor (TLR)-mediated activation of dendritic cells (DCs). We now report that DC-specific deletion of TRAF6 (TRAF6ΔDC) resulted, unexpectedly, in loss of mucosal tolerance, characterized by spontaneous development of T helper 2 (Th2) cells in the lamina propria and eosinophilic enteritis and fibrosis in the small intestine. Loss of tolerance required the presence of gut commensal microbiota but was independent of DC-expressed MyD88. Further, TRAF6ΔDC mice exhibited decreased regulatory T (Treg) cell numbers in the small intestine and diminished induction of iTreg cells in response to model antigen. Evidence suggested that this defect was associated with diminished DC expression of interleukin-2 (IL-2). Finally, we demonstrate that aberrant Th2 cell-associated responses in TRAF6ΔDC mice could be mitigated via restoration of Treg cell activity. Collectively, our findings reveal a role for TRAF6 in directing DC maintenance of intestinal immune tolerance through balanced induction of Treg versus Th2 cell immunity.


Asunto(s)
Células Dendríticas/inmunología , Enteritis/inmunología , Eosinofilia/inmunología , Eosinófilos/inmunología , Gastritis/inmunología , Intestinos/inmunología , Linfocitos T Reguladores/inmunología , Factor 6 Asociado a Receptor de TNF/metabolismo , Células Th2/inmunología , Animales , Células Cultivadas , Células Dendríticas/microbiología , Enteritis/genética , Eosinofilia/genética , Gastritis/genética , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Tolerancia Inmunológica/genética , Interleucina-2/genética , Interleucina-2/metabolismo , Intestinos/microbiología , Intestinos/patología , Activación de Linfocitos/genética , Metagenoma/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal/genética , Linfocitos T Reguladores/microbiología , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/inmunología , Células Th2/microbiología
19.
Nat Immunol ; 14(2): 136-42, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23263554

RESUMEN

Activation of Toll-like receptors (TLRs) by pathogens triggers cytokine production and T cell activation, immune defense mechanisms that are linked to immunopathology. Here we show that IFN-γ production by CD4(+) T(H)1 cells during mucosal responses to the protozoan parasite Toxoplasma gondii resulted in dysbiosis and the elimination of Paneth cells. Paneth cell death led to loss of antimicrobial peptides and occurred in conjunction with uncontrolled expansion of the Enterobacteriaceae family of Gram-negative bacteria. The expanded intestinal bacteria were required for the parasite-induced intestinal pathology. The investigation of cell type-specific factors regulating T(H)1 polarization during T. gondii infection identified the T cell-intrinsic TLR pathway as a major regulator of IFN-γ production in CD4(+) T cells responsible for Paneth cell death, dysbiosis and intestinal immunopathology.


Asunto(s)
Infecciones por Enterobacteriaceae/patología , Enterobacteriaceae/crecimiento & desarrollo , Células de Paneth/patología , Transducción de Señal/inmunología , Células TH1/patología , Toxoplasma/crecimiento & desarrollo , Toxoplasmosis Animal/patología , Animales , Linfocitos T CD4-Positivos , Muerte Celular , Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/complicaciones , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Regulación de la Expresión Génica , Interacciones Huésped-Parásitos , Interacciones Huésped-Patógeno , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-12/genética , Interleucina-12/inmunología , Activación de Linfocitos , Ratones , Ratones Transgénicos , Células de Paneth/microbiología , Células de Paneth/parasitología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Células TH1/microbiología , Células TH1/parasitología , Toxoplasma/inmunología , Toxoplasmosis Animal/complicaciones , Toxoplasmosis Animal/inmunología , Toxoplasmosis Animal/parasitología , alfa-Defensinas/deficiencia
20.
PLoS One ; 7(10): e48126, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23144737

RESUMEN

Type I interferons (T1IFNs) are among the earliest cytokines produced during infections due to their direct regulation by innate immune signaling pathways. Reports have suggested that T1IFNs are produced during malaria infection, but little is known about the in vivo cellular origins of T1IFNs or their role in protection. We have found that in addition to plasmacytoid dendritic cells, splenic red pulp macrophages (RPMs) can generate significant quantities of T1IFNs in response to P. chabaudi infection in a TLR9-, MYD88-, and IRF7-dependent manner. Furthermore, T1IFNs regulate expression of interferon-stimulated genes redundantly with Interferon-gamma (IFNG), resulting in redundancy for resistance to experimental malaria infection. Despite their role in sensing and promoting immune responses to infection, we observe that RPMs are dispensable for control of parasitemia. Our results reveal that RPMs are early sentinels of malaria infection, but that effector mechanisms previously attributed to RPMs are not essential for control.


Asunto(s)
Interferón Tipo I/inmunología , Macrófagos/inmunología , Malaria/inmunología , Plasmodium chabaudi/inmunología , Bazo/inmunología , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Citometría de Flujo , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/metabolismo , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Macrófagos/metabolismo , Malaria/metabolismo , Malaria/parasitología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Plasmodium chabaudi/fisiología , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Receptor de Interferón alfa y beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/inmunología , Bazo/metabolismo , Factores de Tiempo , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/inmunología , Receptor Toll-Like 9/metabolismo , Transcriptoma/genética , Transcriptoma/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...