Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Immunity ; 56(4): 687-694, 2023 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-37044059

RESUMEN

Type 2 immunity defends against macro-parasites and can cause allergic diseases. Our understanding of the mechanisms governing the initiation of type 2 immunity is limited, whereas we know more about type 1 immune responses. Type 2 immunity can be triggered by a wide array of inducers that do not share common features and via diverse pathways and mechanisms. To address the complexity of the type 2 initiation pathways, we suggest a framework that conceptualizes different modes of induction of type 2 immunity. We discuss categories of type 2 inducers and their immunogenicity, types of tissue perturbations that are caused by these inducers, sensing strategies for the initiation of Th2 immune responses, and categorization of the signals that are produced in response to type 2 challenges. We describe tissue-specific examples of functional disruption that could lead to type 2 inflammation and propose that different sensing strategies that operate at the tissue level converge on the initiation of type 2 immune responses.


Asunto(s)
Hipersensibilidad , Inmunidad , Humanos , Inflamación , Células Th2
2.
Immunity ; 40(1): 78-90, 2014 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-24439266

RESUMEN

Innate immune recognition controls adaptive immune responses through multiple mechanisms. The MyD88 signaling adaptor operates in many cell types downstream of Toll-like receptors (TLRs) and interleukin-1 (IL-1) receptor family members. Cell-type-specific functions of MyD88 signaling remain poorly characterized. Here, we have shown that the T cell-specific ablation of MyD88 in mice impairs not only T helper 17 (Th17) cell responses, but also Th1 cell responses. MyD88 relayed signals of TLR-induced IL-1, which became dispensable for Th1 cell responses in the absence of T regulatory (Treg) cells. Treg cell-specific ablation of MyD88 had no effect, suggesting that IL-1 acts on naive CD4(+) T cells instead of Treg cells themselves. Together, these findings demonstrate that IL-1 renders naive CD4(+) T cells refractory to Treg cell-mediated suppression in order to allow their differentiation into Th1 cells. In addition, IL-1 was also important for the generation of functional CD4(+) memory T cells.


Asunto(s)
Interleucina-1/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Células Th17/inmunología , Inmunidad Adaptativa , Animales , Células Cultivadas , Inmunidad Innata , Memoria Inmunológica , Terapia de Inmunosupresión , Interleucina-18/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor 88 de Diferenciación Mieloide/genética , Especificidad de Órganos , Receptores de Interleucina-1/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología
3.
Immunity ; 39(4): 722-32, 2013 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-24076050

RESUMEN

CD4⁺ T cell differentiation is regulated by specialized antigen-presenting cells. Dendritic cells (DCs) produce cytokines that promote naive CD4⁺ T cell differentiation into T helper 1 (Th1), Th17, and inducible T regulatory (iTreg) cells. However, the initiation of Th2 cell responses remains poorly understood, although it is likely that more than one mechanism might be involved. Here we have defined a specific DC subset that is involved in Th2 cell differentiation in vivo in response to a protease allergen, as well as infection with Nippostrongylus brasiliensis. We have demonstrated that this subset is controlled by the transcription factor interferon regulatory factor 4 (IRF4), which is required for their differentiation and Th2 cell-inducing function. IRF4 is known to control Th2 cell differentiation and Th2 cell-associated suppressing function in Treg cells. Our finding suggests that IRF4 also plays a role in DCs where it controls the initiation of Th2 cell responses.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Celular , Factores Reguladores del Interferón/inmunología , Infecciones por Strongylida/inmunología , Células Th2/inmunología , Alérgenos/inmunología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/parasitología , Diferenciación Celular , Técnicas de Cocultivo , Células Dendríticas/parasitología , Células Dendríticas/patología , Regulación de la Expresión Génica , Factores Reguladores del Interferón/genética , Ratones , Ratones Transgénicos , Nippostrongylus/inmunología , Ovalbúmina/inmunología , Transducción de Señal , Infecciones por Strongylida/parasitología , Infecciones por Strongylida/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/parasitología , Linfocitos T Reguladores/patología , Células TH1/inmunología , Células TH1/parasitología , Células TH1/patología , Células Th2/parasitología , Células Th2/patología
4.
Nat Immunol ; 10(7): 713-20, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19465907

RESUMEN

T helper type 2 (T(H)2)-mediated immune responses are induced after infection with multicellular parasites and can be triggered by a variety of allergens. The mechanisms of induction and the antigen-presenting cells involved in the activation of T(H)2 responses remain poorly defined, and the innate immune sensing pathways activated by parasites and allergens are largely unknown. Basophils are required for the in vivo induction of T(H)2 responses by protease allergens. Here we show that basophils also function as antigen-presenting cells. We show that although dendritic cells were dispensable for allergen-induced activation of T(H)2 responses in vitro and in vivo, antigen presentation by basophils was necessary and sufficient for this. Thus, basophils function as antigen-presenting cells for T(H)2 differentiation in response to protease allergens.


Asunto(s)
Alérgenos/inmunología , Células Presentadoras de Antígenos/inmunología , Basófilos/inmunología , Células Th2/inmunología , Traslado Adoptivo , Animales , Células Presentadoras de Antígenos/citología , Células Presentadoras de Antígenos/metabolismo , Antígenos Helmínticos/inmunología , Basófilos/metabolismo , Basófilos/trasplante , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular/inmunología , Células Cultivadas , Endocitosis/inmunología , Femenino , Citometría de Flujo , Expresión Génica , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Interleucina-4/genética , Interleucina-4/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Proteínas Nucleares/metabolismo , Papaína/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Th2/citología , Transactivadores/genética , Transactivadores/inmunología , Transactivadores/metabolismo
5.
J Immunol ; 193(3): 1013-6, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24973450

RESUMEN

Central memory (CM) CD8(+) T cells "remember" prior encounters because they maintain themselves through cell division in the absence of ongoing challenge (homeostatic self-renewal), as well as reproduce the CM fate while manufacturing effector cells during secondary Ag encounters (rechallenge self-renewal). We tested the consequence of conditional deletion of the bone marrow homing receptor CXCR4 on antiviral T cell responses. CXCR4-deficient CD8(+) T cells have impaired memory cell maintenance due to defective homeostatic proliferation. Upon rechallenge, however, CXCR4-deficient T cells can re-expand and renew the CM pool while producing secondary effector cells. The critical bone marrow-derived signals essential for CD8(+) T cell homeostatic self-renewal appear to be dispensable to yield self-renewing, functionally asymmetric cell fates during rechallenge.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Homeostasis/inmunología , Memoria Inmunológica , Receptores CXCR4/deficiencia , Receptores CXCR4/fisiología , Traslado Adoptivo , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Trasplante de Médula Ósea , Linfocitos T CD8-positivos/citología , Células Clonales , Homeostasis/genética , Humanos , Memoria Inmunológica/genética , Inmunofenotipificación , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores CXCR4/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Células Madre/citología , Células Madre/inmunología , Células Madre/metabolismo
6.
Cell Rep ; 22(4): 860-868, 2018 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-29420173

RESUMEN

Unequal transmission of nutritive signaling during cell division establishes fate disparity between sibling lymphocytes, but how asymmetric signaling becomes organized is not understood. We show that receptor-associated class I phosphatidylinositol 3-kinase (PI3K) signaling activity, indexed by phosphatidylinositol (3,4,5)-trisphosphate (PIP3) staining, is spatially restricted to the microtubule-organizing center and subsequently to one pole of the mitotic spindle in activated T and B lymphocytes. Asymmetric PI3K activity co-localizes with polarization of antigen receptor components implicated in class I PI3K signaling and with facultative glucose transporters whose trafficking is PI3K dependent and whose abundance marks cells destined for differentiation. Perturbation of class I PI3K activity disrupts asymmetry of upstream antigen receptors and downstream glucose transporter traffic. The roles of PI3K signaling in nutrient utilization, proliferation, and gene expression may have converged with the conserved role of PI3K signaling in cellular symmetry breaking to form a logic for regenerative lymphocyte divisions.


Asunto(s)
Linfocitos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Diferenciación Celular , Humanos , Transducción de Señal
7.
Trends Cell Biol ; 27(12): 946-954, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28818395

RESUMEN

Activated lymphocytes perform a clonal balancing act, yielding a daughter cell that differentiates owing to intense PI3K signaling, alongside a self-renewing sibling cell with blunted anabolic signaling. Divergent cellular anabolism versus catabolism is emerging as a feature of several developmental and regenerative paradigms. Metabolism can dictate cell fate, in part, because lineage-specific regulators are embedded in the circuitry of conserved metabolic switches. Unequal transmission of PI3K signaling during regenerative divisions is reminiscent of compartmentalized PI3K activity during directed motility or polarized information flow in non-dividing cells. The diverse roles of PI3K pathways in membrane traffic, cell polarity, metabolism, and gene expression may have converged to instruct sibling cell feast and famine, thereby enabling clonal differentiation alongside self-renewal.


Asunto(s)
Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Polaridad Celular/inmunología , Linfocitos/inmunología , Animales , División Celular/inmunología , Células Clonales , Humanos , Linfocitos/citología , Linfocitos/metabolismo , Fosfatidilinositol 3-Quinasas/inmunología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/inmunología
8.
Immunohorizons ; 1(7): 156-161, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28944344

RESUMEN

Anabolic metabolism in lymphocytes promotes plasmablast and cytotoxic T cell differentiation at the expense of self-renewal. Heightened expression and function of the transcription factor IFN regulatory factor 4 (IRF4) accompany enhanced anabolic induction and full commitment to functional differentiation in B cells and CD8+ T cells. In this study, we used a genetic approach to determine whether IRF4 plays an analogous role in Th1 cell induction. Our findings indicate that IRF4 promotes determined Th1 cell differentiation in tandem with anabolic metabolism of CD4+ T cells. IRF4-deficient CD4+ T cells stimulated in vitro exhibit impaired induction of Th1 gene expression and defective silencing of T cell factor 1 expression. IRF4-deficient CD4+ T cells also undergo a shift toward catabolic metabolism, with reduced mammalian target of rapamycin activation, cell size, and nutrient uptake, as well as increased mitochondrial clearance. These findings suggest that the ability to remodel metabolic states can be an essential gateway for altering cell fate.

9.
J Exp Med ; 214(1): 39-47, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27923906

RESUMEN

Upon infection, an activated CD4+ T cell produces terminally differentiated effector cells and renews itself for continued defense. In this study, we show that differentiation and self-renewal arise as opposing outcomes of sibling CD4+ T cells. After influenza challenge, antigen-specific cells underwent several divisions in draining lymph nodes (LN; DLNs) while maintaining expression of TCF1. After four or five divisions, some cells silenced, whereas some cells maintained TCF1 expression. TCF1-silenced cells were T helper 1-like effectors and concentrated in the lungs. Cells from earliest divisions were memory-like and concentrated in nondraining LN. TCF1-expressing cells from later divisions in the DLN could self-renew, clonally yielding a TCF1-silenced daughter cell as well as a sibling cell maintaining TCF1 expression. Some TCF1-expressing cells in DLNs acquired an alternative, follicular helper-like fate. Modeled differentiation experiments in vitro suggested that unequal PI3K/mechanistic target of rapamycin signaling drives intraclonal cell fate heterogeneity. Asymmetric division enables self-renewal to be coupled to production of differentiated CD4+ effector T cells during clonal selection.


Asunto(s)
División Celular Asimétrica/fisiología , Linfocitos T CD4-Positivos/inmunología , Animales , División Celular , Células Cultivadas , Factor Nuclear 1-alfa del Hepatocito/análisis , Factor Nuclear 1-alfa del Hepatocito/genética , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/fisiología , Serina-Treonina Quinasas TOR/fisiología
10.
J Exp Med ; 213(3): 415-31, 2016 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-26903243

RESUMEN

A common genetic alteration in acute myeloid leukemia is the internal tandem duplication (ITD) in FLT3, the receptor for cytokine FLT3 ligand (FLT3L). Constitutively active FLT3-ITD promotes the expansion of transformed progenitors, but also has pleiotropic effects on hematopoiesis. We analyzed the effect of FLT3-ITD on dendritic cells (DCs), which express FLT3 and can be expanded by FLT3L administration. Pre-leukemic mice with the Flt3(ITD) knock-in allele manifested an expansion of classical DCs (cDCs) and plasmacytoid DCs. The expansion originated in DC progenitors, was cell intrinsic, and was further enhanced in Flt3(ITD/ITD) mice. The mutation caused the down-regulation of Flt3 on the surface of DCs and reduced their responsiveness to Flt3L. Both canonical Batf3-dependent CD8(+) cDCs and noncanonical CD8(+) cDCs were expanded and showed specific alterations in their expression profiles. Flt3(ITD) mice showed enhanced capacity to support T cell proliferation, including a cell-extrinsic expansion of regulatory T (T reg) cells. Accordingly, these mice restricted alloreactive T cell responses during graft-versus-host reaction, but failed to control autoimmunity without T reg cells. Thus, the FLT3-ITD mutation directly affects DC development, indirectly modulating T cell homeostasis and supporting T reg cell expansion. We hypothesize that this effect of FLT3-ITD might subvert immunosurveillance and promote leukemogenesis in a cell-extrinsic manner.


Asunto(s)
Células Dendríticas/inmunología , Duplicación de Gen , Leucemia/genética , Leucemia/inmunología , Proteínas de la Membrana/genética , Mutación/genética , Linfocitos T/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linaje de la Célula , Proliferación Celular , Regulación hacia Abajo , Regulación Leucémica de la Expresión Génica , Homeostasis , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Transducción de Señal
11.
Cell Rep ; 17(12): 3142-3152, 2016 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-28009285

RESUMEN

Regeneration requires related cells to diverge in fate. We show that activated lymphocytes yield sibling cells with unequal elimination of aged mitochondria. Disparate mitochondrial clearance impacts cell fate and reflects larger constellations of opposing metabolic states. Differentiation driven by an anabolic constellation of PI3K/mTOR activation, aerobic glycolysis, inhibited autophagy, mitochondrial stasis, and ROS production is balanced with self-renewal maintained by a catabolic constellation of AMPK activation, mitochondrial elimination, oxidative metabolism, and maintenance of FoxO1 activity. Perturbations up and down the metabolic pathways shift the balance of nutritive constellations and cell fate owing to self-reinforcement and reciprocal inhibition between anabolism and catabolism. Cell fate and metabolic state are linked by transcriptional regulators, such as IRF4 and FoxO1, with dual roles in lineage and metabolic choice. Instructing some cells to utilize nutrients for anabolism and differentiation while other cells catabolically self-digest and self-renew may enable growth and repair in metazoa.


Asunto(s)
Proteína Forkhead Box O1/genética , Factores Reguladores del Interferón/genética , Activación de Linfocitos/genética , Linfocitos/metabolismo , Mitocondrias/metabolismo , Animales , Autofagia/genética , Diferenciación Celular/genética , Proteína Forkhead Box O1/metabolismo , Glucólisis , Hematopoyesis/genética , Factores Reguladores del Interferón/metabolismo , Metabolismo/genética , Ratones , Mitocondrias/genética , Fosfatidilinositol 3-Quinasas/genética , Especies Reactivas de Oxígeno/metabolismo , Regeneración/genética , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética
12.
Cell Rep ; 17(7): 1773-1782, 2016 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-27829149

RESUMEN

Selected CD8+ T cells must divide, produce differentiated effector cells, and self-renew, often repeatedly. We now show that silencing expression of the transcription factor TCF1 marks loss of self-renewal by determined effector cells and that this requires cell division. In acute infections, the first three CD8+ T cell divisions produce daughter cells with unequal proliferative signaling but uniform maintenance of TCF1 expression. The more quiescent initial daughter cells resemble canonical central memory cells. The more proliferative, effector-prone cells from initial divisions can subsequently undergo division-dependent production of a TCF1-negative effector daughter cell along with a self-renewing TCF1-positive daughter cell, the latter also contributing to the memory cell pool upon resolution of infection. Self-renewal in the face of effector cell determination may promote clonal amplification and memory cell formation in acute infections, sustain effector regeneration during persistent subclinical infections, and be rate limiting, but remediable, in chronic active infections and cancer.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Autorrenovación de las Células , Animales , Ciclo Celular , Diferenciación Celular , División Celular , Proliferación Celular , Células Clonales , Silenciador del Gen , Ratones Endogámicos C57BL , Factor 1 de Transcripción de Linfocitos T/metabolismo
13.
Cell Rep ; 13(10): 2203-18, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26628372

RESUMEN

Metazoan sibling cells often diverge in activity and identity, suggesting links between growth signals and cell fate. We show that unequal transduction of nutrient-sensitive PI3K/AKT/mTOR signaling during cell division bifurcates transcriptional networks and fates of kindred cells. A sibling B lymphocyte with stronger signaling, indexed by FoxO1 inactivation and IRF4 induction, undergoes PI3K-driven Pax5 repression and plasma cell determination, while its sibling with weaker PI3K activity renews a memory or germinal center B cell fate. PI3K-driven effector T cell determination silences TCF1 in one sibling cell, while its PI3K-attenuated sibling self-renews in tandem. Prior to bifurcations achieving irreversible plasma or effector cell fate determination, asymmetric signaling during initial divisions specifies a more proliferative, differentiation-prone lymphocyte in tandem with a more quiescent memory cell sibling. By triggering cell division but transmitting unequal intensity between sibling cells, nutrient-sensitive signaling may be a frequent arbiter of cell fate bifurcations during development and repair.


Asunto(s)
Linfocitos T CD8-positivos/citología , Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Células Plasmáticas/citología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/metabolismo , Linaje de la Célula , Citometría de Flujo , Técnicas de Sustitución del Gen , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Células Plasmáticas/metabolismo , Transducción de Señal/fisiología
14.
Immunol Res ; 27(2-3): 219-34, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12857970

RESUMEN

Anti-double-stranded DNA (dsDNA) B cells persist even in nonautoimmune- prone animals. In this review, we summarize data regarding the activation potential of these cells. Provision of cognate CD4 T cell help to anti-dsDNA B cells in nonautoimmune mice not only drives their maturation and entry into the B cell follicle, but also leads to secretion of anti-dsDNA autoantibodies. Intriguingly, if T regulatory cells are provided along with T helper cells, the antibody response of anti-dsDNA B cells is diminished. We have also found that T-independent stimulation with CpG oligodeoxynucleotides leads to the proliferation and enhanced recovery of antidsDNA B cells in vitro. These data suggest that control of anti-dsDNA antibody production may rely on elements from both the innate and adaptive arms of the immune system.


Asunto(s)
Anticuerpos Antinucleares/inmunología , Autoinmunidad , Linfocitos B/inmunología , Activación de Linfocitos/inmunología , Animales , ADN/inmunología , Cooperación Linfocítica/inmunología , Ratones , Linfocitos T/inmunología
15.
Elife ; 3: e01949, 2014 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-24842874

RESUMEN

Innate immune recognition is critical for the induction of adaptive immune responses; however the underlying mechanisms remain incompletely understood. In this study, we demonstrate that T cell-specific deletion of the IL-6 receptor α chain (IL-6Rα) results in impaired Th1 and Th17 T cell responses in vivo, and a defect in Tfh function. Depletion of Tregs in these mice rescued the Th1 but not the Th17 response. Our data suggest that IL-6 signaling in effector T cells is required to overcome Treg-mediated suppression in vivo. We show that IL-6 cooperates with IL-1ß to block the suppressive effect of Tregs on CD4(+) T cells, at least in part by controlling their responsiveness to IL-2. In addition, although IL-6Rα-deficient T cells mount normal primary Th1 responses in the absence of Tregs, they fail to mature into functional memory cells, demonstrating a key role for IL-6 in CD4(+) T cell memory formation.DOI: http://dx.doi.org/10.7554/eLife.01949.001.


Asunto(s)
Inmunidad Adaptativa , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Inmunidad Innata , Memoria Inmunológica , Interleucina-6/metabolismo , Transducción de Señal , Inmunidad Adaptativa/efectos de los fármacos , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Inmunidad Innata/efectos de los fármacos , Inmunización , Memoria Inmunológica/efectos de los fármacos , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacología , Interleucina-6/inmunología , Interleucina-6/farmacología , Subunidad alfa del Receptor de Interleucina-6/deficiencia , Subunidad alfa del Receptor de Interleucina-6/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células TH1/inmunología , Células TH1/metabolismo , Células Th17/inmunología , Células Th17/metabolismo
17.
Nat Commun ; 3: 1120, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23047678

RESUMEN

Commensal bacterial sensing by Toll-like receptors is critical for maintaining intestinal homeostasis, but can lead to colitis in the absence of interleukin-10. Although Toll-like receptors are expressed in multiple cell types in the colon, the cell type(s) responsible for the development of colitis are currently unknown. Here we generated mice that are selectively deficient in MyD88 in various cellular compartments in an interleukin-10(-/-) setting. Although epithelial expression of MyD88 was dispensable, MyD88 expression in the mononuclear phagocyte compartment was required for colitis development. Specifically, phenotypically distinct populations of colonic mononuclear phagocytes expressed high levels of interleukin-1ß, interleukin-23 and interleukin-6, and promoted T-helper 17 responses in the absence of interleukin-10. Thus, gut bacterial sensing through MyD88 in mononuclear phagocytes drives inflammatory bowel disease when unopposed by interleukin-10.


Asunto(s)
Colitis/metabolismo , Colon/metabolismo , Colon/patología , Interleucina-10/deficiencia , Interleucina-10/metabolismo , Factor 88 de Diferenciación Mieloide/metabolismo , Fagocitos/metabolismo , Animales , Colitis/genética , Colitis/patología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Interleucina-10/genética , Interleucina-1beta/metabolismo , Interleucina-23/metabolismo , Interleucina-6/metabolismo , Ratones , Factor 88 de Diferenciación Mieloide/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología
18.
J Autoimmun ; 31(2): 98-109, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18539433

RESUMEN

In Fas/FasL-deficient mice anti-chromatin Ab production is T cell dependent and is not apparent until after 10 weeks of age. Early control of anti-chromatin antibodies may be due to the counterbalancing influence of Treg cells. Here we show that Treg cells block lpr/lpr gld/gld Th cells from providing help to anti-chromatin B cells in an in vivo transfer system. Interestingly, the percentage and absolute numbers of Foxp3+ Treg cells is elevated in BALB/c-lpr/lpr gld/gld mice and increases with age compared to BALB/c mice. The majority of Foxp3 expression is found in the B220- CD4+ T cell population, and Foxp3-expressing cells are localized in the splenic PALS (periarteriolar lymphocyte sheath). Strikingly, although the lack of functional Fas/FasL does not affect the ability of Treg cells to block Th cell proliferation, Treg cells can block the IFN-gamma differentiation of Th cells from BALB/c or young BALB-lpr/lpr gld/gld mice but not of pre-existing Th1 cells from older BALB/c-lpr/lpr gld/gld mice. Thus, we suggest autoantibody production is not caused by the lack of Treg cells but by a defect in activation-induced cell death that leads to the accumulation of T effector cells that are resistant to regulatory T cell activity.


Asunto(s)
Autoanticuerpos/biosíntesis , Linfocitos T CD4-Positivos/inmunología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Linfocitos B/inmunología , Femenino , Factores de Transcripción Forkhead/metabolismo , Interferón gamma/metabolismo , Interleucina-4/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos MRL lpr , Ratones Mutantes , Subgrupos de Linfocitos T/inmunología
19.
J Immunol ; 175(7): 4255-64, 2005 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16177065

RESUMEN

To investigate the mechanism by which T regulatory (Treg) cells may control the early onset of autoimmunity, we have used an adoptive transfer model to track Treg, Th, and anti-chromatin B cell interactions in vivo. We show that anti-chromatin B cells secrete Abs by day 8 in vivo upon provision of undeviated, Th1- or Th2-type CD4+ T cell help, but this secretion is blocked by the coinjection of CD4+ CD25+ Treg cells. Although Treg cells do not interfere with the initial follicular entry or activation of Th or B cells at day 3, ICOS levels on Th cells are decreased. Furthermore, Treg cells must be administered during the initial phases of the Ab response to exert full suppression of autoantibody production. These studies indicate that CD25+ Treg cells act to inhibit the maturation, rather than the initiation, of autoantibody responses.


Asunto(s)
Autoanticuerpos/biosíntesis , Linfocitos T Reguladores/inmunología , Animales , Linfocitos B/inmunología , Proliferación Celular , Células Cultivadas , Cromatina/inmunología , Femenino , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Hemaglutininas/inmunología , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Receptores de Interleucina-2/biosíntesis , Receptores de Interleucina-2/metabolismo
20.
J Immunol ; 175(1): 104-11, 2005 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15972636

RESUMEN

Circulating autoantibodies against dsDNA and chromatin are a characteristic of systemic lupus erythematosus in humans and many mouse models of this disease. B cells expressing these autoantibodies are normally regulated in nonautoimmune-prone mice but are induced to secrete Abs following T cell help. Likewise, anti-chromatin autoantibody production is T cell-dependent in Fas/Fas ligand (FasL)-deficient (lpr/lpr or gld/gld) mice. In this study, we demonstrate that Th2 cells promote anti-chromatin B cell survival and autoantibody production in vivo. FasL influences the ability of Th2 cells to help B cells, as Th2-gld/gld cells support higher titers of anti-chromatin Abs than their FasL-sufficient counterparts and promote anti-chromatin B cell participation in germinal centers. Th1 cells induce anti-chromatin B cell germinal centers regardless of FasL status; however, their ability to stimulate anti-chromatin Ab production positively correlates with their level of IFN-gamma production. This distinction is lost if FasL-deficient T cells are used: Th1-gld/gld cells promote significant titers of anti-chromatin Abs regardless of IFN-gamma production levels. Thus, FasL from effector T cells plays an important role in determining the fate of anti-chromatin B cells.


Asunto(s)
Linfocitos B/inmunología , Lupus Eritematoso Sistémico/inmunología , Glicoproteínas de Membrana/inmunología , Células Th2/inmunología , Traslado Adoptivo , Animales , Anticuerpos Antinucleares/biosíntesis , Cromatina/inmunología , Proteína Ligando Fas , Humanos , Cadenas kappa de Inmunoglobulina/genética , Cadenas kappa de Inmunoglobulina/metabolismo , Técnicas In Vitro , Cooperación Linfocítica , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Transgénicos , Células TH1/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA