Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Nat Immunol ; 25(2): 330-342, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38172260

RESUMEN

Antibody-secreting plasma cells (PCs) are generated in secondary lymphoid organs but are reported to reside in an emerging range of anatomical sites. Analysis of the transcriptome of different tissue-resident (Tr)PC populations revealed that they each have their own transcriptional signature indicative of functional adaptation to the host tissue environment. In contrast to expectation, all TrPCs were extremely long-lived, regardless of their organ of residence, with longevity influenced by intrinsic factors like the immunoglobulin isotype. Analysis at single-cell resolution revealed that the bone marrow is unique in housing a compendium of PCs generated all over the body that retain aspects of the transcriptional program indicative of their tissue of origin. This study reveals that extreme longevity is an intrinsic property of TrPCs whose transcriptome is imprinted by signals received both at the site of induction and within the tissue of residence.


Asunto(s)
Médula Ósea , Células Plasmáticas , Células de la Médula Ósea
2.
Nat Immunol ; 25(3): 496-511, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38356058

RESUMEN

Visceral adipose tissue (VAT) is an energy store and endocrine organ critical for metabolic homeostasis. Regulatory T (Treg) cells restrain inflammation to preserve VAT homeostasis and glucose tolerance. Here, we show that the VAT harbors two distinct Treg cell populations: prototypical serum stimulation 2-positive (ST2+) Treg cells that are enriched in males and a previously uncharacterized population of C-X-C motif chemokine receptor 3-positive (CXCR3+) Treg cells that are enriched in females. We show that the transcription factors GATA-binding protein 3 and peroxisome proliferator-activated receptor-γ, together with the cytokine interleukin-33, promote the differentiation of ST2+ VAT Treg cells but repress CXCR3+ Treg cells. Conversely, the differentiation of CXCR3+ Treg cells is mediated by the cytokine interferon-γ and the transcription factor T-bet, which also antagonize ST2+ Treg cells. Finally, we demonstrate that ST2+ Treg cells preserve glucose homeostasis, whereas CXCR3+ Treg cells restrain inflammation in lean VAT and prevent glucose intolerance under high-fat diet conditions. Overall, this study defines two molecularly and developmentally distinct VAT Treg cell types with unique context- and sex-specific functions.


Asunto(s)
Proteína 1 Similar al Receptor de Interleucina-1 , Linfocitos T Reguladores , Femenino , Masculino , Humanos , Grasa Intraabdominal , Citocinas , Inflamación , Glucosa
3.
Nat Immunol ; 22(4): 434-448, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33649580

RESUMEN

T cells dynamically interact with multiple, distinct cellular subsets to determine effector and memory differentiation. Here, we developed a platform to quantify cell location in three dimensions to determine the spatial requirements that direct T cell fate. After viral infection, we demonstrated that CD8+ effector T cell differentiation is associated with positioning at the lymph node periphery. This was instructed by CXCR3 signaling since, in its absence, T cells are confined to the lymph node center and alternatively differentiate into stem-like memory cell precursors. By mapping the cellular sources of CXCR3 ligands, we demonstrated that CXCL9 and CXCL10 are expressed by spatially distinct dendritic and stromal cell subsets. Unlike effector cells, retention of stem-like memory precursors in the paracortex is associated with CCR7 expression. Finally, we demonstrated that T cell location can be tuned, through deficiency in CXCL10 or type I interferon signaling, to promote effector or stem-like memory fates.


Asunto(s)
Infecciones por Arenaviridae/metabolismo , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Quimiocina CXCL10/metabolismo , Quimiocina CXCL9/metabolismo , Memoria Inmunológica , Ganglios Linfáticos/metabolismo , Células Precursoras de Linfocitos T/metabolismo , Receptores CXCR3/metabolismo , Animales , Infecciones por Arenaviridae/genética , Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/virología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Linaje de la Célula , Células Cultivadas , Quimiocina CXCL10/genética , Quimiocina CXCL9/genética , Quimiotaxis de Leucocito , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno , Interferón Tipo I/metabolismo , Ligandos , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Células Precursoras de Linfocitos T/inmunología , Células Precursoras de Linfocitos T/virología , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Receptores CCR7/metabolismo , Receptores CXCR3/genética , Transducción de Señal , Nicho de Células Madre , Células del Estroma/inmunología , Células del Estroma/metabolismo
4.
Nat Immunol ; 22(7): 851-864, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099918

RESUMEN

Group 2 innate lymphoid cells (ILC2s) are essential to maintain tissue homeostasis. In cancer, ILC2s can harbor both pro-tumorigenic and anti-tumorigenic functions, but we know little about their underlying mechanisms or whether they could be clinically relevant or targeted to improve patient outcomes. Here, we found that high ILC2 infiltration in human melanoma was associated with a good clinical prognosis. ILC2s are critical producers of the cytokine granulocyte-macrophage colony-stimulating factor, which coordinates the recruitment and activation of eosinophils to enhance antitumor responses. Tumor-infiltrating ILC2s expressed programmed cell death protein-1, which limited their intratumoral accumulation, proliferation and antitumor effector functions. This inhibition could be overcome in vivo by combining interleukin-33-driven ILC2 activation with programmed cell death protein-1 blockade to significantly increase antitumor responses. Together, our results identified ILC2s as a critical immune cell type involved in melanoma immunity and revealed a potential synergistic approach to harness ILC2 function for antitumor immunotherapies.


Asunto(s)
Anticuerpos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Inhibidores de Puntos de Control Inmunológico/farmacología , Interleucina-33/farmacología , Linfocitos/efectos de los fármacos , Melanoma Experimental/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Quimiotaxis de Leucocito/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de los fármacos , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Eosinófilos/metabolismo , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Linfocitos/inmunología , Linfocitos/metabolismo , Masculino , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Melanoma Experimental/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo
5.
Immunity ; 57(5): 1037-1055.e6, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38593796

RESUMEN

Memory B cells (MBCs) are key providers of long-lived immunity against infectious disease, yet in chronic viral infection, they do not produce effective protection. How chronic viral infection disrupts MBC development and whether such changes are reversible remain unknown. Through single-cell (sc)ATAC-seq and scRNA-seq during acute versus chronic lymphocytic choriomeningitis viral infection, we identified a memory subset enriched for interferon (IFN)-stimulated genes (ISGs) during chronic infection that was distinct from the T-bet+ subset normally associated with chronic infection. Blockade of IFNAR-1 early in infection transformed the chromatin landscape of chronic MBCs, decreasing accessibility at ISG-inducing transcription factor binding motifs and inducing phenotypic changes in the dominating MBC subset, with a decrease in the ISG subset and an increase in CD11c+CD80+ cells. However, timing was critical, with MBCs resistant to intervention at 4 weeks post-infection. Together, our research identifies a key mechanism to instruct MBC identity during viral infection.


Asunto(s)
Epigénesis Genética , Interferón Tipo I , Coriomeningitis Linfocítica , Virus de la Coriomeningitis Linfocítica , Células B de Memoria , Animales , Interferón Tipo I/metabolismo , Interferón Tipo I/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Ratones , Virus de la Coriomeningitis Linfocítica/inmunología , Células B de Memoria/inmunología , Ratones Endogámicos C57BL , Receptor de Interferón alfa y beta/genética , Memoria Inmunológica/inmunología , Enfermedad Crónica , Subgrupos de Linfocitos B/inmunología , Análisis de la Célula Individual
6.
Immunity ; 55(10): 1843-1855.e6, 2022 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-36108634

RESUMEN

To optimize immunity to pathogens, B lymphocytes generate plasma cells with functionally diverse antibody isotypes. By lineage tracing single cells within differentiating B cell clones, we identified the heritability of discrete fate controlling mechanisms to inform a general mathematical model of B cell fate regulation. Founder cells highly influenced clonal plasma-cell fate, whereas class switch recombination (CSR) was variegated within clones. In turn, these CSR patterns resulted from independent all-or-none expression of both activation-induced cytidine deaminase (AID) and IgH germline transcription (GLT), with the latter being randomly re-expressed after each cell division. A stochastic model premised on these molecular transition rules accurately predicted antibody switching outcomes under varied conditions in vitro and during an immune response in vivo. Thus, the generation of functionally diverse antibody types follows rules of autonomous cellular programming that can be adapted and modeled for the rational control of antibody classes for potential therapeutic benefit.


Asunto(s)
Cambio de Clase de Inmunoglobulina , Recombinación Genética , Linfocitos B , Citidina Desaminasa/genética , Citidina Desaminasa/metabolismo , Cambio de Clase de Inmunoglobulina/genética , Isotipos de Inmunoglobulinas/genética , Isotipos de Inmunoglobulinas/metabolismo
7.
Immunity ; 55(3): 423-441.e9, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35139355

RESUMEN

Cell death plays an important role during pathogen infections. Here, we report that interferon-γ (IFNγ) sensitizes macrophages to Toll-like receptor (TLR)-induced death that requires macrophage-intrinsic death ligands and caspase-8 enzymatic activity, which trigger the mitochondrial apoptotic effectors, BAX and BAK. The pro-apoptotic caspase-8 substrate BID was dispensable for BAX and BAK activation. Instead, caspase-8 reduced pro-survival BCL-2 transcription and increased inducible nitric oxide synthase (iNOS), thus facilitating BAX and BAK signaling. IFNγ-primed, TLR-induced macrophage killing required iNOS, which licensed apoptotic caspase-8 activity and reduced the BAX and BAK inhibitors, A1 and MCL-1. The deletion of iNOS or caspase-8 limited SARS-CoV-2-induced disease in mice, while caspase-8 caused lethality independent of iNOS in a model of hemophagocytic lymphohistiocytosis. These findings reveal that iNOS selectively licenses programmed cell death, which may explain how nitric oxide impacts disease severity in SARS-CoV-2 infection and other iNOS-associated inflammatory conditions.


Asunto(s)
COVID-19/inmunología , Caspasa 8/metabolismo , Interferón gamma/metabolismo , Linfohistiocitosis Hemofagocítica/inmunología , Macrófagos/inmunología , Mitocondrias/metabolismo , SARS-CoV-2/fisiología , Animales , Caspasa 8/genética , Células Cultivadas , Citotoxicidad Inmunológica , Humanos , Interferón gamma/genética , Activación de Macrófagos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo II/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Transducción de Señal , Proteína Destructora del Antagonista Homólogo bcl-2/genética , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
8.
Nat Immunol ; 24(2): 205-206, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36658242
11.
Cell ; 150(6): 1249-63, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22980984

RESUMEN

A defining feature of vertebrate immunity is the acquisition of immunological memory, which confers enhanced protection against pathogens by mechanisms that are incompletely understood. Here, we compared responses by virus-specific naive T cells (T(N)) and central memory T cells (T(CM)) to viral antigen challenge in lymph nodes (LNs). In steady-state LNs, both T cell subsets localized in the deep T cell area and interacted similarly with antigen-presenting dendritic cells. However, upon entry of lymph-borne virus, only T(CM) relocalized rapidly and efficiently toward the outermost LN regions in the medullary, interfollicular, and subcapsular areas where viral infection was initially confined. This rapid peripheralization was coordinated by a cascade of cytokines and chemokines, particularly ligands for T(CM)-expressed CXCR3. Consequently, in vivo recall responses to viral infection by CXCR3-deficient T(CM) were markedly compromised, indicating that early antigen detection afforded by intranodal chemokine guidance of T(CM) is essential for efficient antiviral memory.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Ganglios Linfáticos/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Quimiocina CXCL9/inmunología , Células Dendríticas/inmunología , Interferón gamma/inmunología , Ganglios Linfáticos/citología , Virus de la Coriomeningitis Linfocítica , Ratones , Ratones Endogámicos C57BL , Monocitos/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores CXCR3/inmunología , Células del Estroma/inmunología , Virus de la Estomatitis Vesicular Indiana
12.
Immunol Rev ; 306(1): 76-92, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34882817

RESUMEN

The lymph node plays a critical role in mounting an adaptive immune response to infection, clearance of foreign pathogens, and cancer immunosurveillance. Within this complex structure, intranodal migration is vital for CD8+ T cell activation and differentiation. Combining tissue clearing and volumetric light sheet fluorescent microscopy of intact lymph nodes has allowed us to explore the spatial regulation of T cell fates. This has determined that short-lived effector (TSLEC ) are imprinted in peripheral lymph node interfollicular regions, due to CXCR3 migration. In contrast, stem-like memory cell (TSCM ) differentiation is determined in the T cell paracortex. Here, we detail the inflammatory and chemokine regulators of spatially restricted T cell differentiation, with a focus on how to promote TSCM . We propose a default pathway for TSCM differentiation due to CCR7-directed segregation of precursors away from the inflammatory effector niche. Although volumetric imaging has revealed the consequences of intranodal migration, we still lack knowledge of how this is orchestrated within a complex chemokine environment. Toward this goal, we highlight the potential of combining microfluidic chambers with pre-determined complexity and subcellular resolution microscopy.


Asunto(s)
Linfocitos T CD8-positivos , Memoria Inmunológica , Diferenciación Celular , Quimiocinas/metabolismo , Humanos , Ganglios Linfáticos , Activación de Linfocitos
13.
PLoS Pathog ; 19(9): e1011666, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37733817

RESUMEN

Prior infection can generate protective immunity against subsequent infection, although the efficacy of such immunity can vary considerably. Live-attenuated vaccines (LAVs) are one of the most effective methods for mimicking this natural process, and analysis of their efficacy has proven instrumental in the identification of protective immune mechanisms. Here, we address the question of what makes a LAV efficacious by characterising immune responses to a LAV, termed TAS2010, which is highly protective (80-90%) against lethal murine salmonellosis, in comparison with a moderately protective (40-50%) LAV, BRD509. Mice vaccinated with TAS2010 developed immunity systemically and were protected against gut-associated virulent infection in a CD4+ T cell-dependent manner. TAS2010-vaccinated mice showed increased activation of Th1 responses compared with their BRD509-vaccinated counterparts, leading to increased Th1 memory populations in both lymphoid and non-lymphoid organs. The optimal development of Th1-driven immunity was closely correlated with the activation of CD11b+Ly6GnegLy6Chi inflammatory monocytes (IMs), the activation of which can be modulated proportionally by bacterial load in vivo. Upon vaccination with the LAV, IMs expressed T cell chemoattractant CXCL9 that attracted CD4+ T cells to the foci of infection, where IMs also served as a potent source of antigen presentation and Th1-promoting cytokine IL-12. The expression of MHC-II in IMs was rapidly upregulated following vaccination and then maintained at an elevated level in immune mice, suggesting IMs may have a role in sustained antigen stimulation. Our findings present a longitudinal analysis of CD4+ T cell development post-vaccination with an intracellular bacterial LAV, and highlight the benefit of inflammation in the development of Th1 immunity. Future studies focusing on the induction of IMs may reveal key strategies for improving vaccine-induced T cell immunity.


Asunto(s)
Linfocitos T CD4-Positivos , Infecciones por Salmonella , Ratones , Animales , Monocitos , Vacunas Atenuadas , Inflamación
14.
Nat Immunol ; 14(4): 389-95, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23455676

RESUMEN

NKp46+ innate lymphoid cells (ILCs) serve important roles in regulating the intestinal microbiota and defense against pathogens. Whether NKp46+ ILCs arise directly from lymphoid tissue-inducer (LTi) cells or represent a separate lineage remains controversial. We report here that the transcription factor T-bet (encoded by Tbx21) was essential for the development of NKp46+ ILCs but not of LTi cells or nuocytes. Deficiency in interleukin 22 (IL-22)-producing NKp46+ ILCs resulted in greater susceptibility of Tbx21-/- mice to intestinal infection. Haploinsufficient T-bet expression resulted in lower expression of the signaling molecule Notch, and Notch signaling was necessary for the transition of LTi cells into NKp46+ ILCs. Furthermore, NKp46+ ILCs differentiated solely from the CD4- LTi population, not the CD4+ LTi population. Our results pinpoint the regulation of Notch signaling by T-bet as a distinct molecular pathway that guides the development of NKp46+ ILCs.


Asunto(s)
Antígenos Ly/metabolismo , Inmunidad Innata , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Proteínas de Dominio T Box/metabolismo , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/inmunología , Subgrupos Linfocitarios/citología , Ratones , Ratones Noqueados , Proteínas de Dominio T Box/genética
15.
Immunol Rev ; 300(1): 203-219, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33586207

RESUMEN

The relationship between the extrinsic environment and the internal transcriptional network is circular. Naive T cells first engage with antigen-presenting cells to set transcriptional differentiation networks in motion. In turn, this regulates specific chemokine receptors that direct migration into distinct lymph node niches. Movement into these regions brings newly activated T cells into contact with accessory cells and cytokines that reinforce the differentiation programming to specify T cell function. We and others have observed similarities in the transcriptional networks that specify both CD4+ T follicular helper (TFH ) cells and CD8+ central memory stem-like (TSCM ) cells. Here, we compare and contrast the current knowledge for these shared differentiation programs, compared to their effector counterparts, CD4+ T-helper 1 (TH1 ) and CD8+ short-lived effector (TSLEC ) cells. Understanding the interplay between cellular interactions and transcriptional programming is essential to harness T cell differentiation that is fit for purpose; to stimulate potent T cell effector function for the elimination of chronic infection and cancer; or to amplify the formation of humoral immunity and longevity of cellular memory to prevent infectious diseases.


Asunto(s)
Comunicación Celular , Linfocitos T , Células Presentadoras de Antígenos , Diferenciación Celular , Receptores de Quimiocina , Linfocitos T Colaboradores-Inductores
16.
Immunol Cell Biol ; 101(4): 345-357, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36710659

RESUMEN

The transcription factor Myc is critically important in driving cell proliferation, a function that is frequently dysregulated in cancer. To avoid this dysregulation Myc is tightly controlled by numerous layers of regulation. One such layer is the use of distal regulatory enhancers to drive Myc expression. Here, using chromosome conformation capture to examine B cells of the immune system in the first hours after their activation, we reveal a previously unidentified enhancer of Myc. The interactivity of this enhancer coincides with a dramatic, but discrete, spike in Myc expression 3 h post-activation. However, genetic deletion of this region, has little impact on Myc expression, Myc protein level or in vitro and in vivo cell proliferation. Examination of the enhancer deleted regulatory landscape suggests that enhancer redundancy likely sustains Myc expression. This work highlights not only the importance of temporally examining enhancers, but also the complexity and dynamics of the regulation of critical genes such as Myc.


Asunto(s)
Elementos de Facilitación Genéticos , Genes myc , Elementos de Facilitación Genéticos/genética , Factores de Transcripción/metabolismo , Regulación de la Expresión Génica , Regiones Promotoras Genéticas
17.
Immunol Rev ; 289(1): 101-114, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30977199

RESUMEN

A fundamental question in immunology is how cells decide between distinct T helper, effector or memory differentiation fates. These decisions are paramount to overcome infection and establish long-lasting protection. The impact of cell location for the determination of T-cell fate decisions is an emerging field. This review will discuss our current understanding of the migration path that T cells follow, within draining lymph nodes, to steer differentiation down distinct paths of either effector or memory fates. In particular, the regulation of migration and cellular encounters mediated by the chemokine receptor CXCR3 and its ligands will be discussed. The combination of increased antigen density and unique cellular partners play a central role in facilitating the site-specific differentiation of effector T cells, within the interfollicular regions of draining lymph nodes. Recent advances have applied this knowledge to optimize vaccine design to target antigen to lymph nodes. Increased understanding of the regulation of CXCR3 ligands and how T cells integrate multiple chemokine cues will help further progress in this field and allow additional applications to direct cell differentiation outside the lymph node, to enhance memory residency in peripheral tissues and effector anti-tumor responses.


Asunto(s)
Quimiocinas/metabolismo , Receptores CXCR3/metabolismo , Linfocitos T/inmunología , Animales , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Humanos , Inmunomodulación , Activación de Linfocitos
18.
Immunol Cell Biol ; 100(5): 312-322, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35233830

RESUMEN

The chemokine receptor CXCR3 is expressed on immune cells to co-ordinate lymphocyte activation and migration. CXCR3 binds three chemokine ligands, CXCL9, CXCL10 and CXCL11. These ligands display distinct expression patterns and ligand signaling biases; however, how each ligand functions individually and collaboratively is incompletely understood. CXCL9 and CXCL10 are considered pro-inflammatory chemokines during viral infection, while CXCL11 may induce a tolerizing state. The investigation of the individual role of CXCL11 in vivo has been hampered as C57BL/6 mice carry several mutations that result in a null allele. Here, CRISPR/Cas9 was used to correct these mutations on a C57BL/6 background. It was validated that CXCL11KI mice expressed CXCL11 protein in dendritic cells, spleen and lung. CXCL11KI mice were largely phenotypically indistinguishable from C57BL/6 mice, both at steady-state and during two models of viral infection. While CXCL11 expression did not modify acute antiviral responses, this study provides a new tool to understand the role of CXCL11 in other experimental settings.


Asunto(s)
Quimiocina CXCL10 , Quimiocina CXCL11/metabolismo , Virosis , Animales , Quimiocina CXCL10/genética , Quimiocina CXCL11/genética , Quimiocina CXCL9/genética , Quimiocina CXCL9/metabolismo , Inmunidad , Ligandos , Ratones , Ratones Endogámicos C57BL
19.
Immunity ; 37(6): 1091-103, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23123063

RESUMEN

Differentiation of naive CD4(+) T cells into T helper (Th) cells is a defining event in adaptive immunity. The cytokines and transcription factors that control Th cell differentiation are understood, but it is not known how this process is orchestrated within lymph nodes (LNs). Here we have shown that the CXCR3 chemokine receptor was required for optimal generation of interferon-γ (IFN-γ)-secreting Th1 cells in vivo. By using a CXCR3 ligand reporter mouse, we found that stromal cells predominately expressed the chemokine ligand CXCL9 whereas hematopoietic cells expressed CXCL10 in LNs. Dendritic cell (DC)-derived CXCL10 facilitated T cell-DC interactions in LNs during T cell priming while both chemokines guided intranodal positioning of CD4(+) T cells to interfollicular and medullary zones. Thus, different chemokines acting on the same receptor can function locally to facilitate DC-T cell interactions and globally to influence intranodal positioning, and both functions contribute to Th1 cell differentiation.


Asunto(s)
Diferenciación Celular/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Receptores CXCR3/metabolismo , Células TH1/citología , Células TH1/inmunología , Animales , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Quimiocina CXCL9/genética , Quimiocina CXCL9/inmunología , Quimiocinas CXC/genética , Quimiocinas CXC/inmunología , Citocinas/biosíntesis , Proteínas de Unión al ADN/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Regulación de la Expresión Génica , Interferón gamma/biosíntesis , Ligandos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/metabolismo , Ratones , Ratones Transgénicos , Unión Proteica , Receptores CXCR3/genética
20.
Proc Natl Acad Sci U S A ; 114(20): 5225-5230, 2017 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-28461487

RESUMEN

The lymphoid tissue that drains the upper respiratory tract represents an important induction site for cytotoxic T lymphocyte (CTL) immunity to airborne pathogens and intranasal vaccines. Here, we investigated the role of the nasal-associated lymphoid tissues (NALTs), which are mucosal-associated lymphoid organs embedded in the submucosa of the nasal passage, in the initial priming and recall expansion of CD8+ T cells following an upper respiratory tract infection with a pathogenic influenza virus and immunization with a live attenuated influenza virus vaccine. Whereas NALTs served as the induction site for the recall expansion of memory CD8+ T cells following influenza virus infection or vaccination, they failed to support activation of naïve CD8+ T cells. Strikingly, NALTs, unlike other lymphoid tissues, were not routinely surveyed during the steady state by circulating T cells. The selective recruitment of memory T cells into these lymphoid structures occurred in response to infection-induced elevation of the chemokine CXCL10, which attracted CXCR3+ memory CD8+ T cells. These results have significant implications for intranasal vaccines, which deliver antigen to mucosal-associated lymphoid tissue and aim to elicit protective CTL-mediated immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Inmunidad Mucosa/inmunología , Linfocitos T Citotóxicos/inmunología , Administración Intranasal , Animales , Inmunización , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Ganglios Linfáticos/fisiología , Tejido Linfoide/metabolismo , Ratones , Ratones Endogámicos C57BL , Mucosa Nasal/metabolismo , Mucosa Nasal/fisiología , Infecciones por Orthomyxoviridae/inmunología , Infecciones del Sistema Respiratorio , Vacunación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA