Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(17): 4512-4530.e22, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34343496

RESUMEN

Cytotoxic T lymphocyte (CTL) responses against tumors are maintained by stem-like memory cells that self-renew but also give rise to effector-like cells. The latter gradually lose their anti-tumor activity and acquire an epigenetically fixed, hypofunctional state, leading to tumor tolerance. Here, we show that the conversion of stem-like into effector-like CTLs involves a major chemotactic reprogramming that includes the upregulation of chemokine receptor CXCR6. This receptor positions effector-like CTLs in a discrete perivascular niche of the tumor stroma that is densely occupied by CCR7+ dendritic cells (DCs) expressing the CXCR6 ligand CXCL16. CCR7+ DCs also express and trans-present the survival cytokine interleukin-15 (IL-15). CXCR6 expression and IL-15 trans-presentation are critical for the survival and local expansion of effector-like CTLs in the tumor microenvironment to maximize their anti-tumor activity before progressing to irreversible dysfunction. These observations reveal a cellular and molecular checkpoint that determines the magnitude and outcome of anti-tumor immune responses.


Asunto(s)
Receptores CXCR6/metabolismo , Linfocitos T Citotóxicos/inmunología , Microambiente Tumoral , Animales , Antígeno B7-H1/metabolismo , Comunicación Celular , Movimiento Celular , Proliferación Celular , Supervivencia Celular , Quimiocina CXCL16 , Células Dendríticas/metabolismo , Interleucina-12/metabolismo , Interleucina-15/metabolismo , Ligandos , Ganglios Linfáticos/metabolismo , Melanoma/inmunología , Melanoma/patología , Ratones Endogámicos C57BL
2.
Cell ; 162(6): 1322-37, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26296422

RESUMEN

Host defense against viruses and intracellular parasites depends on effector CD8(+) T cells, whose optimal clonal expansion, differentiation, and memory properties require signals from CD4(+) T cells. Here, we addressed the role of dendritic cell (DC) subsets in initial activation of the two T cell types and their co-operation. Surprisingly, initial priming of CD4(+) and CD8(+) T cells was spatially segregated within the lymph node and occurred on different DCs with temporally distinct patterns of antigen presentation via MHCI versus MHCII molecules. DCs that co-present antigen via both MHC molecules were detected at a later stage; these XCR1(+) DCs are the critical platform involved in CD4(+) T cell augmentation of CD8(+) T cell responses. These findings delineate the complex choreography of cellular interactions underlying effective cell-mediated anti-viral responses, with implications for basic DC subset biology, as well as for translational application to the development of vaccines that evoke optimal T cell immunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Comunicación Celular , Células Dendríticas/inmunología , Virus Vaccinia/fisiología , Vaccinia/inmunología , Animales , Presentación de Antígeno , Antígenos Virales/inmunología , Células Dendríticas/citología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ratones , Receptores de Quimiocina/genética , Bazo/citología , Bazo/inmunología
3.
Cell ; 163(2): 354-66, 2015 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-26451485

RESUMEN

Infections have been proposed as initiating factors for inflammatory disorders; however, identifying associations between defined infectious agents and the initiation of chronic disease has remained elusive. Here, we report that a single acute infection can have dramatic and long-term consequences for tissue-specific immunity. Following clearance of Yersinia pseudotuberculosis, sustained inflammation and associated lymphatic leakage in the mesenteric adipose tissue deviates migratory dendritic cells to the adipose compartment, thereby preventing their accumulation in the mesenteric lymph node. As a consequence, canonical mucosal immune functions, including tolerance and protective immunity, are persistently compromised. Post-resolution of infection, signals derived from the microbiota maintain inflammatory mesentery remodeling and consequently, transient ablation of the microbiota restores mucosal immunity. Our results indicate that persistent disruption of communication between tissues and the immune system following clearance of an acute infection represents an inflection point beyond which tissue homeostasis and immunity is compromised for the long-term. VIDEO ABSTRACT.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades del Sistema Inmune/microbiología , Enfermedades del Sistema Inmune/patología , Enfermedades Linfáticas/patología , Infecciones por Yersinia pseudotuberculosis/inmunología , Yersinia pseudotuberculosis/fisiología , Movimiento Celular , Enfermedad Crónica , Células Dendríticas/patología , Femenino , Humanos , Enfermedades Linfáticas/microbiología , Tejido Linfoide/inmunología , Tejido Linfoide/patología , Masculino , Mesenterio/inmunología , Mesenterio/patología , Organismos Libres de Patógenos Específicos , Infecciones por Yersinia pseudotuberculosis/patología
5.
Immunity ; 50(5): 1188-1201.e6, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31053504

RESUMEN

Lymph nodes (LNs) play critical roles in adaptive immunity by concentrating in one location the antigens, antigen-presenting cells, and antigen-responsive lymphocytes involved in such responses. Recent studies have revealed nonrandom localization of innate and adaptive immune cells within these organs, suggesting that microanatomical positioning optimizes responses involving sparse cooperating cells. Here, we report that the peripheral localization of LN cDC2 dendritic cells specialized for MHC-II antigen presentation is matched by a similarly biased paracortical distribution of CD4+ T cells directed by the chemoattractant receptor Ebi2. In the absence of Ebi2, CD4+ T cells lose their location bias and are delayed in antigen recognition, proliferative expansion, differentiation, direct effector activity, and provision of help for CD8+ T cell-mediated memory responses, limiting host defense and vaccine responses. These findings demonstrate evolutionary selection for distinct niches within the LN that promote cellular responses, emphasizing the critical link between fine-grained tissue organization and host defense.


Asunto(s)
Inmunidad Adaptativa/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Presentación de Antígeno/inmunología , Antígenos/inmunología , Diferenciación Celular/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Acoplados a Proteínas G/genética
6.
J Immunol ; 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38884660

RESUMEN

Conventionally, immune responses are studied in the context of inflamed tissues and their corresponding draining lymph nodes (LNs). However, little is known about the effects of systemic inflammatory signals generated during local inflammation on distal tissues and nondraining LNs. Using a mouse model of cutaneous immunization, we found that systemic inflammatory stimuli triggered a rapid and selective distal response in the small intestine and the mesenteric LN (mesLN). This consisted of increased permeability of intestinal blood vessels and lymphatic drainage of bloodborne solutes into the mesLN, enhanced activation and migration of intestinal dendritic cells, as well as amplified T cell responses in the mesLNs to systemic but not orally derived Ags. Mechanistically, we found that the small intestine endothelial cells preferentially expressed molecules involved in TNF-α signaling and that TNF-α blockade markedly diminished distal intestinal responses to cutaneous immunization. Together, these findings reveal that the intestinal immune system is rapidly and selectively activated in response to inflammatory cues regardless of their origin, thus identifying an additional layer of defense and enhanced surveillance of a key barrier organ at constant risk of pathogen encounter.

7.
Immunol Rev ; 306(1): 93-107, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34845729

RESUMEN

Immune responses must be rapid, tightly orchestrated, and tailored to the encountered stimulus. Lymphatic vessels facilitate this process by continuously collecting immunological information (ie, antigens, immune cells, and soluble mediators) about the current state of peripheral tissues, and transporting these via the lymph across the lymphatic system. Lymph nodes (LNs), which are critical meeting points for innate and adaptive immune cells, are strategically located along the lymphatic network to intercept this information. Within LNs, immune cells are spatially organized, allowing them to efficiently respond to information delivered by the lymph, and to either promote immune homeostasis or mount protective immune responses. These responses involve the activation and functional cooperation of multiple distinct cell types and are tailored to the specific inflammatory conditions. The natural patterns of lymph flow can also generate spatial gradients of antigens and agonists within draining LNs, which can in turn further regulate innate cell function and localization, as well as the downstream generation of adaptive immunity. In this review, we explore how information transmitted by the lymph shapes the spatiotemporal organization of innate and adaptive immune responses in LNs, with particular focus on steady state and Type-I vs. Type-II inflammation.


Asunto(s)
Inmunidad Adaptativa , Células Dendríticas , Antígenos/metabolismo , Movimiento Celular , Humanos , Inflamación , Ganglios Linfáticos
8.
Nat Methods ; 19(5): 613-619, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35545715

RESUMEN

Light-sheet microscopy has emerged as the preferred means for high-throughput volumetric imaging of cleared tissues. However, there is a need for a flexible system that can address imaging applications with varied requirements in terms of resolution, sample size, tissue-clearing protocol, and transparent sample-holder material. Here, we present a 'hybrid' system that combines a unique non-orthogonal dual-objective and conventional (orthogonal) open-top light-sheet (OTLS) architecture for versatile multi-scale volumetric imaging. We demonstrate efficient screening and targeted sub-micrometer imaging of sparse axons within an intact, cleared mouse brain. The same system enables high-throughput automated imaging of multiple specimens, as spotlighted by a quantitative multi-scale analysis of brain metastases. Compared with existing academic and commercial light-sheet microscopy systems, our hybrid OTLS system provides a unique combination of versatility and performance necessary to satisfy the diverse requirements of a growing number of cleared-tissue imaging applications.


Asunto(s)
Microscopía , Animales , Ratones , Microscopía/métodos
9.
Immunity ; 42(1): 172-85, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25607462

RESUMEN

Upon infection, adaptive immune responses play catch-up with rapidly replicating pathogens. While mechanisms for efficient humoral responses to lymph-borne antigens have been characterized, the current paradigm for T cell responses to infections and particulate vaccines involves delayed migration of peripheral antigen-bearing dendritic cells (DCs) to lymph nodes (LNs), where they elicit effector T cell responses. Utilizing whole LN 3D imaging, histo-cytometry, and intravital 2-photon microscopy, we have identified a specialized population of DCs, enriched in the LN-resident CD11b(+) subset, which resides within the lymphatic sinus endothelium and scans lymph with motile dendrites. These DCs capture draining particles and present associated antigens to T lymphocytes, inducing T cell responses much sooner than and independently of migratory DCs. Thus, strategic DC subset positioning in LNs limits a potentially costly delay in generation of T cell responses to lymph-borne antigens, contributing to effective host defense. These findings are also highly relevant to vaccine design.


Asunto(s)
Células Dendríticas/inmunología , Activación de Linfocitos , Pseudomonas aeruginosa/inmunología , Linfocitos T/inmunología , Animales , Presentación de Antígeno , Antígenos Bacterianos/inmunología , Antígeno CD11b/metabolismo , Movimiento Celular , Células Cultivadas , Dendritas/inmunología , Endotelio Linfático/inmunología , Humanos , Imagenología Tridimensional , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos , Ratones Transgénicos , Material Particulado/inmunología , Vacunación
10.
Nature ; 554(7691): 255-259, 2018 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-29364878

RESUMEN

The mammalian gut is colonized by numerous microorganisms collectively termed the microbiota, which have a mutually beneficial relationship with their host. Normally, the gut microbiota matures during ontogeny to a state of balanced commensalism marked by the absence of adverse inflammation. Subsets of innate lymphoid cells (ILCs) and conventional T cells are considered to have redundant functions in containment and clearance of microbial pathogens, but how these two major lymphoid-cell populations each contribute to shaping the mature commensal microbiome and help to maintain tissue homeostasis has not been determined. Here we identify, using advanced multiplex quantitative imaging methods, an extensive and persistent phosphorylated-STAT3 signature in group 3 ILCs and intestinal epithelial cells that is induced by interleukin (IL)-23 and IL-22 in mice that lack CD4+ T cells. By contrast, in immune-competent mice, phosphorylated-STAT3 activation is induced only transiently by microbial colonization at weaning. This early signature is extinguished as CD4+ T cell immunity develops in response to the expanding commensal burden. Physiologically, the persistent IL-22 production from group 3 ILCs that occurs in the absence of adaptive CD4+ T-cell activity results in impaired host lipid metabolism by decreasing lipid transporter expression in the small bowel. These findings provide new insights into how innate and adaptive lymphocytes operate sequentially and in distinct ways during normal development to establish steady-state commensalism and tissue metabolic homeostasis.


Asunto(s)
Inmunidad Adaptativa , Microbioma Gastrointestinal/inmunología , Inmunidad Innata , Intestino Delgado/inmunología , Intestino Delgado/microbiología , Metabolismo de los Lípidos , Linfocitos/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Células Epiteliales/citología , Células Epiteliales/inmunología , Proteínas de Homeodominio/genética , Homeostasis , Inflamación/inmunología , Inflamación/microbiología , Inflamación/patología , Interleucina-23/inmunología , Interleucinas/biosíntesis , Interleucinas/inmunología , Intestino Delgado/metabolismo , Activación de Linfocitos , Masculino , Ratones , Monocitos/metabolismo , Fosforilación , Receptores CCR2/metabolismo , Factor de Transcripción STAT3/metabolismo , Simbiosis , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Destete , Interleucina-22
11.
Nature ; 537(7620): 417-421, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27501248

RESUMEN

Chronic viral infections are characterized by a state of CD8+ T-cell dysfunction that is associated with expression of the programmed cell death 1 (PD-1) inhibitory receptor. A better understanding of the mechanisms that regulate CD8+ T-cell responses during chronic infection is required to improve immunotherapies that restore function in exhausted CD8+ T cells. Here we identify a population of virus-specific CD8+ T cells that proliferate after blockade of the PD-1 inhibitory pathway in mice chronically infected with lymphocytic choriomeningitis virus (LCMV). These LCMV-specific CD8+ T cells expressed the PD-1 inhibitory receptor, but also expressed several costimulatory molecules such as ICOS and CD28. This CD8+ T-cell subset was characterized by a unique gene signature that was related to that of CD4+ T follicular helper (TFH) cells, CD8+ T cell memory precursors and haematopoietic stem cell progenitors, but that was distinct from that of CD4+ TH1 cells and CD8+ terminal effectors. This CD8+ T-cell population was found only in lymphoid tissues and resided predominantly in the T-cell zones along with naive CD8+ T cells. These PD-1+CD8+ T cells resembled stem cells during chronic LCMV infection, undergoing self-renewal and also differentiating into the terminally exhausted CD8+ T cells that were present in both lymphoid and non-lymphoid tissues. The proliferative burst after PD-1 blockade came almost exclusively from this CD8+ T-cell subset. Notably, the transcription factor TCF1 had a cell-intrinsic and essential role in the generation of this CD8+ T-cell subset. These findings provide a better understanding of T-cell exhaustion and have implications in the optimization of PD-1-directed immunotherapy in chronic infections and cancer.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Inmunoterapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Antígenos CD28/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Proliferación Celular/efectos de los fármacos , Autorrenovación de las Células , Femenino , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Proteína Coestimuladora de Linfocitos T Inducibles/metabolismo , Coriomeningitis Linfocítica , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Ratones , Receptor de Muerte Celular Programada 1/metabolismo , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Colaboradores-Inductores/metabolismo
12.
Immunity ; 37(2): 364-76, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22863836

RESUMEN

Flow cytometry allows highly quantitative analysis of complex dissociated populations at the cost of neglecting their tissue localization. In contrast, conventional microscopy methods provide spatial information, but visualization and quantification of cellular subsets defined by complex phenotypic marker combinations is challenging. Here, we describe an analytical microscopy method, "histo-cytometry," for visualizing and quantifying phenotypically complex cell populations directly in tissue sections. This technology is based on multiplexed antibody staining, tiled high-resolution confocal microscopy, voxel gating, volumetric cell rendering, and quantitative analysis. We have tested this technology on various innate and adaptive immune populations in murine lymph nodes (LNs) and were able to identify complex cellular subsets and phenotypes, achieving quantitatively similar results to flow cytometry, while also gathering cellular positional information. Here, we employ histo-cytometry to describe the spatial segregation of resident and migratory dendritic cell subsets into specialized microanatomical domains, suggesting an unexpected LN demarcation into discrete functional compartments.


Asunto(s)
Células Dendríticas/citología , Citometría de Flujo/métodos , Inmunohistoquímica/métodos , Ganglios Linfáticos/citología , Microscopía Confocal/métodos , Animales , Antígenos CD/análisis , Quimera/inmunología , Células Dendríticas/inmunología , Humanos , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente/métodos , Linfocitos T/inmunología , Linfocitos T/metabolismo
13.
J Immunol ; 203(4): 807-812, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31308091

RESUMEN

Growing evidence suggests the outcome of Mycobacterium tuberculosis infection is established rapidly after exposure, but how the current tuberculosis vaccine, bacillus Calmette-Guérin (BCG), impacts early immunity is poorly understood. In this study, we found that murine BCG immunization promotes a dramatic shift in infected cell types. Although alveolar macrophages are the major infected cell for the first 2 weeks in unimmunized animals, BCG promotes the accelerated recruitment and infection of lung-infiltrating phagocytes. Interestingly, this shift is dependent on CD4 T cells, yet does not require intrinsic recognition of Ag presented by infected alveolar macrophages. M. tuberculosis-specific T cells are first activated in lung regions devoid of infected cells, and these events precede vaccine-induced reduction of the bacterial burden, which occurs only after the colocalization of T cells and infected cells. Understanding how BCG alters early immune responses to M. tuberculosis provides new avenues to improve upon the immunity it confers.


Asunto(s)
Vacuna BCG/inmunología , Linfocitos T CD4-Positivos/inmunología , Macrófagos Alveolares/inmunología , Tuberculosis Pulmonar/inmunología , Animales , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos Alveolares/microbiología , Ratones , Ratones Endogámicos C57BL , Tuberculosis Pulmonar/prevención & control
14.
Nature ; 528(7581): 225-30, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26605524

RESUMEN

FOXP3(+) regulatory T cells (Treg cells) prevent autoimmunity by limiting the effector activity of T cells that have escaped thymic negative selection or peripheral inactivation. Despite the information available about molecular factors mediating the suppressive function of Treg cells, the relevant cellular events in intact tissues remain largely unexplored, and whether Treg cells prevent activation of self-specific T cells or primarily limit damage from such cells has not been determined. Here we use multiplex, quantitative imaging in mice to show that, within secondary lymphoid tissues, highly suppressive Treg cells expressing phosphorylated STAT5 exist in discrete clusters with rare IL-2-positive T cells that are activated by self-antigens. This local IL-2 induction of STAT5 phosphorylation in Treg cells is part of a feedback circuit that limits further autoimmune responses. Inducible ablation of T cell receptor expression by Treg cells reduces their regulatory capacity and disrupts their localization in clusters, resulting in uncontrolled effector T cell responses. Our data thus reveal that autoreactive T cells are activated to cytokine production on a regular basis, with physically co-clustering T cell receptor-stimulated Treg cells responding in a negative feedback manner to suppress incipient autoimmunity and maintain immune homeostasis.


Asunto(s)
Homeostasis/inmunología , Linfocitos T Reguladores/inmunología , Animales , Movimiento Celular , Células Dendríticas/citología , Células Dendríticas/inmunología , Femenino , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Fenotipo , Transporte de Proteínas , Factor de Transcripción STAT5/metabolismo
15.
Proc Natl Acad Sci U S A ; 114(35): E7321-E7330, 2017 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-28808033

RESUMEN

Organ homeostasis, cellular differentiation, signal relay, and in situ function all depend on the spatial organization of cells in complex tissues. For this reason, comprehensive, high-resolution mapping of cell positioning, phenotypic identity, and functional state in the context of macroscale tissue structure is critical to a deeper understanding of diverse biological processes. Here we report an easy to use method, clearing-enhanced 3D (Ce3D), which generates excellent tissue transparency for most organs, preserves cellular morphology and protein fluorescence, and is robustly compatible with antibody-based immunolabeling. This enhanced signal quality and capacity for extensive probe multiplexing permits quantitative analysis of distinct, highly intermixed cell populations in intact Ce3D-treated tissues via 3D histo-cytometry. We use this technology to demonstrate large-volume, high-resolution microscopy of diverse cell types in lymphoid and nonlymphoid organs, as well as to perform quantitative analysis of the composition and tissue distribution of multiple cell populations in lymphoid tissues. Combined with histo-cytometry, Ce3D provides a comprehensive strategy for volumetric quantitative imaging and analysis that bridges the gap between conventional section imaging and disassociation-based techniques.


Asunto(s)
Técnicas Histológicas/métodos , Imagenología Tridimensional/métodos , Animales , Femenino , Fluorescencia , Masculino , Ratones , Ratones Transgénicos , Microscopía Confocal/métodos
16.
Biomacromolecules ; 20(2): 854-870, 2019 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-30608149

RESUMEN

Small molecule Toll-like receptor-7 and -8 agonists (TLR-7/8a) can be used as vaccine adjuvants to induce CD8 T cell immunity but require formulations that prevent systemic toxicity and focus adjuvant activity in lymphoid tissues. Here, we covalently attached TLR-7/8a to polymers of varying composition, chain architecture and hydrodynamic behavior (∼300 nm submicrometer particles, ∼10 nm micelles and ∼4 nm flexible random coils) and evaluated how these parameters of polymer-TLR-7/8a conjugates impact adjuvant activity in vivo. Attachment of TLR-7/8a to any of the polymer compositions resulted in a nearly 10-fold reduction in systemic cytokines (toxicity). Moreover, both lymph node cytokine production and the magnitude of CD8 T cells induced against protein antigen increased with increasing polymer-TLR-7/8a hydrodynamic radius, with the submicrometer particle inducing the highest magnitude responses. Notably, CD8 T cell responses induced by polymer-TLR-7/8a were dependent on CCR2+ monocytes and IL-12, whereas responses by a small molecule TLR-7/8a that unexpectedly persisted in vaccine-site draining lymph nodes (T1/2 = 15 h) had less dependence on monocytes and IL-12 but required Type I IFNs. This study shows how modular properties of synthetic adjuvants can be chemically programmed to alter immunity in vivo through distinct immunological mechanisms.


Asunto(s)
Adyuvantes Inmunológicos/química , Linfocitos T CD8-positivos/efectos de los fármacos , Activación de Linfocitos , Micelas , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/farmacología , Animales , Linfocitos T CD8-positivos/inmunología , Línea Celular , Células Cultivadas , Citocinas/metabolismo , Femenino , Hidrodinámica , Ratones , Ratones Endogámicos C57BL , Unión Proteica
17.
J Immunol ; 198(2): 895-907, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27903737

RESUMEN

Allergic asthma develops in the mucosal tissue of small bronchi. At these sites, local cytokine production by Th2/Th17 cells is believed to be critical for the development of tissue eosinophilia/neutrophilia. Using the mouse trachea as a relevant model of human small airways, we performed advanced in vivo dynamic and in situ static imaging to visualize individual cytokine-producing T cells in the airway mucosa and to define their immediate cellular environment. Upon allergen sensitization, newly recruited CD4+ T cells formed discrete Ag-driven clusters with dendritic cells (DCs). Within T cell-DC clusters, a small fraction of CD4+ T cells produced IL-13 or IL-17 following prolonged Ag-specific interactions with DCs. As a result of local Th2 cytokine signaling, eosinophils were recruited into these clusters. Neutrophils also infiltrated these clusters in a T cell-dependent manner, but their mucosal distribution was more diffuse. Our findings reveal the focal nature of allergen-driven responses in the airways and define multiple steps with potential for interference with the progression of asthmatic pathology.


Asunto(s)
Asma/inmunología , Linfocitos T CD4-Positivos/inmunología , Quimiotaxis de Leucocito/inmunología , Citocinas/biosíntesis , Células Dendríticas/inmunología , Traslado Adoptivo , Animales , Asma/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Citocinas/inmunología , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Hipersensibilidad/inmunología , Inmunidad Mucosa/inmunología , Inmunohistoquímica , Masculino , Ratones , Microscopía Confocal , Mucosa Respiratoria/inmunología
18.
PLoS Pathog ; 11(2): e1004637, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25658939

RESUMEN

Malaria infection begins when a female Anopheles mosquito injects Plasmodium sporozoites into the skin of its host during blood feeding. Skin-deposited sporozoites may enter the bloodstream and infect the liver, reside and develop in the skin, or migrate to the draining lymph nodes (DLNs). Importantly, the DLN is where protective CD8(+) T cell responses against malaria liver stages are induced after a dermal route of infection. However, the significance of parasites in the skin and DLN to CD8(+) T cell activation is largely unknown. In this study, we used genetically modified parasites, as well as antibody-mediated immobilization of sporozoites, to determine that active sporozoite migration to the DLNs is required for robust CD8(+) T cell responses. Through dynamic in vivo and static imaging, we show the direct uptake of parasites by lymph-node resident DCs followed by CD8(+) T cell-DC cluster formation, a surrogate for antigen presentation, in the DLNs. A few hours after sporozoite arrival to the DLNs, CD8(+) T cells are primed by resident CD8α(+) DCs with no apparent role for skin-derived DCs. Together, these results establish a critical role for lymph node resident CD8α(+) DCs in CD8(+) T cell priming to sporozoite antigens while emphasizing a requirement for motile sporozoites in the induction of CD8(+) T cell-mediated immunity.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Malaria/inmunología , Esporozoítos/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno/inmunología , Antígenos de Protozoos/inmunología , Separación Celular , Células Dendríticas/inmunología , Citometría de Flujo , Inmunidad Celular/inmunología , Ganglios Linfáticos/parasitología , Ratones , Microscopía Confocal , Plasmodium berghei/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
J Immunol ; 192(4): 1651-1660, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24431231

RESUMEN

Exposure to pathogens in the periphery elicits effector T cell differentiation in local lymph nodes followed by migration of activated T cells to and within the infected site. However, the relationships among pathogen abundance, Ag display on MHC molecules, effector T cell dynamics, and functional responses at the infected sites are incompletely characterized. In this study, we compared CD4(+) T cell effector dynamics and responses during pulmonary mycobacterial infection versus acute influenza infection. Two-photon imaging together with in situ as well as ex vivo analysis of cytokine production revealed that the proportion of migration-arrested, cytokine-producing effector T cells was dramatically higher in the influenza-infected lungs due to substantial differences in Ag abundance in the two infectious states. Despite the marked inflammatory conditions associated with influenza infection, histocytometric analysis showed that cytokine production was focal, with a restriction to areas of significant Ag burden. Optimal effector function is thus constrained by the availability of TCR ligands, pointing to the value of increasing Ag stimulation rather than effector numbers in harnessing CD4(+) T cells for therapeutic purposes in such conditions.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Citocinas/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Tuberculosis Pulmonar/inmunología , Traslado Adoptivo , Animales , Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/trasplante , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Citocinas/biosíntesis , Inflamación/inmunología , Interferón gamma/metabolismo , Hígado/inmunología , Pulmón/inmunología , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Mycobacterium bovis/inmunología , Mycobacterium tuberculosis/inmunología , Orthomyxoviridae/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
20.
J Immunol ; 191(3): 1011-5, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23804712

RESUMEN

Naive CD8 T cells proliferate in response to TCR and CD28 signals, but require IL-12 or type I IFN to survive and develop optimal effector functions. Although murine CTL generated in vitro in response to IL-12 or IFN-α had comparable effector functions, IL-12-stimulated cells were significantly more effective in controlling tumor in an adoptive immunotherapy model. They maintained high numbers and function, whereas IFN-α-stimulated cells declined in number and became exhausted. Consistent with this, IFN-α-stimulated cells in the tumor expressed higher levels of programmed death 1 (PD-1) inhibitory receptor than did IL-12-stimulated cells. When blocking Ab specific for the PD-L1 ligand of PD-1 was administered, the efficacy of IFN-α-stimulated CTL became comparable with that of IL-12-stimulated cells. Thus, IL-12 and IFN-α differentially program CD8 T cells to re-express distinct levels of PD-1 upon re-encountering Ag, resulting in IL-12-stimulated cells being less susceptible to exhaustion in the face of sustained tumor Ag.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interferón Tipo I/metabolismo , Interferón-alfa/metabolismo , Interleucina-12/metabolismo , Receptor de Muerte Celular Programada 1/metabolismo , Animales , Anticuerpos Monoclonales/inmunología , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Proliferación Celular , Activación de Linfocitos/inmunología , Ratones , Neoplasias/inmunología , Receptor de Muerte Celular Programada 1/biosíntesis , Transducción de Señal/inmunología , Linfocitos T Citotóxicos/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA