Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
J Exp Med ; 221(4)2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38417020

RESUMO

Anti-PD-1 therapy targets intratumoral CD8+ T cells to promote clinical responses in cancer patients. Recent evidence suggests an additional activity in the periphery, but the underlying mechanism is unclear. Here, we show that anti-PD-1 mAb enhances CD8+ T cell responses in tumor-draining lymph nodes by stimulating cytokine production in follicular helper T cells (Tfh). In two different models, anti-PD-1 mAb increased the activation and proliferation of tumor-specific T cells in lymph nodes. Surprisingly, anti-PD-1 mAb did not primarily target CD8+ T cells but instead stimulated IL-4 production by Tfh cells, the major population bound by anti-PD-1 mAb. Blocking IL-4 or inhibiting the Tfh master transcription factor BCL6 abrogated anti-PD-1 mAb activity in lymph nodes while injection of IL-4 complexes was sufficient to recapitulate anti-PD-1 mAb activity. A similar mechanism was observed in a vaccine model. Finally, nivolumab also boosted human Tfh cells in humanized mice. We propose that Tfh cells and IL-4 play a key role in the peripheral activity of anti-PD-1 mAb.


Assuntos
Neoplasias , Células T Auxiliares Foliculares , Humanos , Camundongos , Animais , Linfócitos T Auxiliares-Indutores , Interleucina-4/metabolismo , Linfonodos , Neoplasias/patologia , Linfócitos T CD8-Positivos
2.
Nat Cancer ; 4(7): 968-983, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37248395

RESUMO

CD4+ T cells and CD4+ chimeric antigen receptor (CAR) T cells display highly variable antitumor activity in preclinical models and in patients; however, the mechanisms dictating how and when CD4+ T cells promote tumor regression are incompletely understood. With the help of functional intravital imaging, we report that interferon (IFN)-γ production but not perforin-mediated cytotoxicity was the dominant mechanism for tumor elimination by anti-CD19 CD4+ CAR T cells. Mechanistically, mouse or human CD4+ CAR T-cell-derived IFN-γ diffused extensively to act on tumor cells at distance selectively killing tumors sensitive to cytokine-induced apoptosis, including antigen-negative variants. In anti-CD19 CAR T-cell-treated patients exhibiting elevated CAR CD4:CD8 ratios, strong induction of serum IFN-γ was associated with increased survival. We propose that the sensitivity of tumor cells to the pro-apoptotic activity of IFN-γ is a major determinant of CD4+ CAR T-cell efficacy and may be considered to guide the use of CD4+ T cells during immunotherapy.


Assuntos
Neoplasias , Linfócitos T , Humanos , Animais , Camundongos , Receptores de Antígenos de Linfócitos T , Citocinas , Interferon gama , Linfócitos T CD4-Positivos
3.
iScience ; 26(2): 106068, 2023 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-36824271

RESUMO

T cells become activated following one or multiple contacts with antigen-presenting cells. Calcium influx is a key signaling event elicited during these cellular interactions; however, it is unclear whether T cells recall and integrate calcium signals elicited during temporally separated contacts. To study the integration of calcium signals, we designed a programmable, multiplex illumination strategy for temporally patterned optogenetics (TEMPO). We found that a single round of calcium elevation was insufficient to promote nuclear factor of activated T cells (NFAT) activity and cytokine production in a T cell line. However, robust responses were detected after a second identical stimulation even when signals were separated by several hours. Our results suggest the existence of a biochemical memory of calcium signals in T cells that favors signal integration during temporally separated contacts and promote cytokine production. As illustrated here, TEMPO is a versatile approach for dissecting temporal integration in defined signaling pathways.

4.
EMBO J ; 41(19): e111528, 2022 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-35997165

RESUMO

The regulation of cellular energy metabolism is central to most physiological and pathophysiological processes. However, most current methods have limited ability to functionally probe metabolic pathways in individual cells. Here, we describe SPICE-Met (Single-cell Profiling and Imaging of Cell Energy Metabolism), a method for profiling energy metabolism in single cells using flow cytometry or imaging. We generated a transgenic mouse expressing PercevalHR, a fluorescent reporter for cellular ATP:ADP ratio. Modulation of PercevalHR fluorescence with metabolic inhibitors was used to infer the dependence of energy metabolism on oxidative phosphorylation and glycolysis in defined cell populations identified by flow cytometry. We applied SPICE-Met to analyze T-cell memory development during vaccination. Finally, we used SPICE-Met in combination with real-time imaging to dissect the heterogeneity and plasticity of energy metabolism in single macrophages ex vivo and identify three distinct metabolic patterns. Functional probing of energy metabolism with single-cell resolution should greatly facilitate the study of immunometabolism at a steady state, during disease pathogenesis or in response to therapy.


Assuntos
Metabolismo Energético , Fosforilação Oxidativa , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Metabolismo Energético/fisiologia , Glicólise/fisiologia , Camundongos , Camundongos Transgênicos
5.
Proc Natl Acad Sci U S A ; 119(29): e2203855119, 2022 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-35858359

RESUMO

Neutrophils form cellular clusters or swarms in response to injury or pathogen intrusion. Yet, intracellular signaling events favoring this coordinated response remain to be fully characterized. Here, we show that calcium signals play a critical role during mouse neutrophil clustering around particles of zymosan, a structural fungal component. Pioneer neutrophils recognizing zymosan or live Candida albicans displayed elevated calcium levels. Subsequently, a transient wave of calcium signals in neighboring cells was observed followed by the attraction of neutrophils that exhibited more persistent calcium signals as they reached zymosan particles. Calcium signals promoted LTB4 production while the blocking of extracellular calcium entry or LTB4 signaling abrogated cluster formation. Finally, using optogenetics to manipulate calcium influx in primary neutrophils, we show that calcium signals could initiate recruitment of neighboring neutrophils in an LTB4-dependent manner. Thus, sustained calcium responses at the center of the cluster are necessary and sufficient for the generation of chemoattractive gradients that attract neutrophils in a self-reinforcing process.


Assuntos
Sinalização do Cálcio , Cálcio , Leucotrieno B4 , Neutrófilos , Animais , Cálcio/metabolismo , Candida albicans/imunologia , Leucotrieno B4/genética , Leucotrieno B4/fisiologia , Camundongos , Neutrófilos/imunologia , Zimosan/imunologia
6.
EMBO J ; 40(15): e107176, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34124789

RESUMO

Dendritic cell (DC) activation by viral RNA sensors such as TLR3 and MDA-5 is critical for initiating antiviral immunity. Optimal DC activation is promoted by type I interferon (IFN) signaling which is believed to occur in either autocrine or paracrine fashion. Here, we show that neither autocrine nor paracrine type I IFN signaling can fully account for DC activation by poly(I:C) in vitro and in vivo. By controlling the density of type I IFN-producing cells in vivo, we establish that instead a quorum of type I IFN-producing cells is required for optimal DC activation and that this process proceeds at the level of an entire lymph node. This collective behavior, governed by type I IFN diffusion, is favored by the requirement for prolonged cytokine exposure to achieve DC activation. Furthermore, collective DC activation was found essential for the development of innate and adaptive immunity in lymph nodes. Our results establish how collective rather than cell-autonomous processes can govern the initiation of immune responses.


Assuntos
Células Dendríticas/fisiologia , Interferon Tipo I/metabolismo , Linfonodos/citologia , Percepção de Quorum/fisiologia , Animais , Linfócitos T CD8-Positivos/fisiologia , Contagem de Células , Células Dendríticas/efeitos dos fármacos , Imunidade Inata/imunologia , Inflamação/patologia , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/imunologia , Interferon Tipo I/farmacologia , Linfonodos/imunologia , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Camundongos Transgênicos , Poli I-C/farmacologia
7.
EMBO J ; 40(11): e106658, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33855732

RESUMO

Cytotoxic T cells (CTLs) can eliminate tumor cells through the delivery of lethal hits, but the actual efficiency of this process in the tumor microenvironment is unclear. Here, we visualized the capacity of single CTLs to attack tumor cells in vitro and in vivo using genetically encoded reporters that monitor cell damage and apoptosis. Using two distinct malignant B-cell lines, we found that the majority of cytotoxic hits delivered by CTLs in vitro were sublethal despite proper immunological synapse formation, and associated with reversible calcium elevation and membrane damage in the targets. Through intravital imaging in the bone marrow, we established that the majority of CTL interactions with lymphoma B cells were either unproductive or sublethal. Functional heterogeneity of CTLs contributed to diverse outcomes during CTL-tumor contacts in vivo. In the therapeutic settings of anti-CD19 CAR T cells, the majority of CAR T cell-tumor interactions were also not associated with lethal hit delivery. Thus, differences in CTL lytic potential together with tumor cell resistance to cytotoxic hits represent two important bottlenecks for anti-tumor responses in vivo.


Assuntos
Imunoterapia Adotiva , Linfoma/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos CD19/imunologia , Apoptose , Linfócitos B/imunologia , Cálcio/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Sinapses Imunológicas/imunologia , Linfoma/terapia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos Quiméricos/imunologia
8.
Sci Immunol ; 6(57)2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33771887

RESUMO

Chimeric antigen receptor (CAR) T cell therapy relies on the activity of a large pool of tumor-targeting cytotoxic effectors. Whether CAR T cells act autonomously or require interactions with the tumor microenvironment (TME) remains incompletely understood. Here, we report an essential cross-talk between CAR T cell subsets and the TME for tumor control in an immunocompetent mouse B cell lymphoma model of anti-CD19 CAR T cell therapy. Using single-cell RNA sequencing, we revealed substantial modification of the TME during CAR T cell therapy. Interferon-γ (IFN-γ) produced by CAR T cells not only enhanced endogenous T and natural killer cell activity but was also essential for sustaining CAR T cell cytotoxicity, as revealed by intravital imaging. CAR T cell-derived IFN-γ facilitated host interleukin-12 production that supported host immune and CAR T cell responses. Compared with CD8+ CAR T cells, CD4+ CAR T cells were more efficient at host immune activation but less capable of direct tumor killing. In summary, CAR T cells do not act independently in vivo but rely instead on cytokine-mediated cross-talk with the TME for optimal activity. Invigorating CAR T cell interplay with the host represents an attractive strategy to prevent relapses after therapy.


Assuntos
Comunicação Celular/imunologia , Testes Imunológicos de Citotoxicidade , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos Quiméricos/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Microambiente Tumoral/imunologia , Animais , Antígenos CD19/imunologia , Antígenos de Neoplasias/imunologia , Comunicação Celular/genética , Linhagem Celular Tumoral , Biologia Computacional/métodos , Citocinas/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Humanos , Imunoterapia Adotiva , Interferon gama/biossíntese , Ativação Linfocitária/imunologia , Linfoma de Células B/genética , Linfoma de Células B/imunologia , Linfoma de Células B/patologia , Linfoma de Células B/terapia , Camundongos , Neoplasias/imunologia , Neoplasias/terapia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos Quiméricos/genética
9.
Sci Adv ; 7(8)2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33608271

RESUMO

Anti-CD20 antibody (mAb) represents an effective strategy for the treatment of B cell malignancies, possibly involving complement activity, antibody-dependent cellular cytotoxicity and phagocytosis (ADP). While ADP by Kupffer cells deplete circulating tumors, mechanisms targeting non-circulating tumors remain unclear. Using intravital imaging in a model of B cell lymphoma, we establish here the dominance and limitations of ADP in the bone marrow (BM). We found that tumor cells were stably residing in the BM with little evidence for recirculation. To elucidate the mechanism of depletion, we designed a dual fluorescent reporter to visualize phagocytosis and apoptosis. ADP by BM-associated macrophages was the primary mode of tumor elimination but was no longer active after one hour, resulting in partial depletion. Moreover, macrophages were present at low density in tumor-rich regions, targeting only neighboring tumors. Overcoming spatiotemporal bottlenecks in tumor-targeting Ab therapy thus represents a critical path towards the design of optimized therapies.

10.
Nat Cancer ; 1(3): 302-314, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32803171

RESUMO

The cytokine IFN-γ produced by tumor-reactive T cells is a key effector molecule with pleiotropic effects during anti-tumor immune responses. While IFN-γ production is targeted at the immunological synapse, its spatiotemporal activity within the tumor remains elusive. Here, we report that while IFN-γ secretion requires local antigen recognition, IFN-γ diffuses extensively to alter the tumor microenvironment in distant areas. Using intravital imaging and a reporter for STAT1 translocation, we provide evidence that T cells mediate sustained IFN-γ signaling in remote tumor cells. Furthermore, tumor phenotypic alterations required several hours of exposure to IFN-γ, a feature that disfavored local IFN-γ activity over diffusion and bystander activity. Finally, single-cell RNA-seq data from melanoma patients also suggested bystander IFN-γ activity in human tumors. Thus, tumor-reactive T cells act collectively to create large cytokine fields that profoundly modify the tumor microenvironment.


Assuntos
Interferon gama , Microambiente Tumoral , Citocinas , Humanos , Linfócitos T
11.
Nat Commun ; 11(1): 1143, 2020 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-32123168

RESUMO

By offering the possibility to manipulate cellular functions with spatiotemporal control, optogenetics represents an attractive tool for dissecting immune responses. However, applying these approaches to single cells in vivo remains particularly challenging for immune cells that are typically located in scattering tissues. Here, we introduce an improved calcium actuator with sensitivity allowing for two-photon photoactivation. Furthermore, we identify an actuator/reporter combination that permits the simultaneous manipulation and visualization of calcium signals in individual T cells in vivo. With this strategy, we document the consequences of defined patterns of calcium signals on T cell migration, adhesion, and chemokine release. Manipulation of individual immune cells in vivo should open new avenues for establishing the functional contribution of single immune cells engaged in complex reactions.


Assuntos
Sinalização do Cálcio/fisiologia , Optogenética/métodos , Linfócitos T/metabolismo , Animais , Proteínas de Arabidopsis/genética , Linfócitos T CD8-Positivos/metabolismo , Adesão Celular , Movimento Celular , Quimiocinas/metabolismo , Criptocromos/genética , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fótons , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Análise de Célula Única/métodos , Molécula 1 de Interação Estromal/genética , Molécula 1 de Interação Estromal/metabolismo , Linfócitos T/citologia
12.
J Exp Med ; 216(5): 1038-1049, 2019 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-30936262

RESUMO

CAR T cells represent a potentially curative strategy for B cell malignancies. However, the outcome and dynamics of CAR T cell interactions in distinct anatomical sites are poorly understood. Using intravital imaging, we tracked interactions established by anti-CD19 CAR T cells in B cell lymphoma-bearing mice. Circulating targets trapped CAR T cells in the lungs, reducing their access to lymphoid organs. In the bone marrow, tumor apoptosis was largely due to CAR T cells that engaged, killed, and detached from their targets within 25 min. Notably, not all CAR T cell contacts elicited calcium signaling or killing while interacting with tumors, uncovering extensive functional heterogeneity. Mathematical modeling revealed that direct killing was sufficient for tumor regression. Finally, antigen-loss variants emerged in the bone marrow, but not in lymph nodes, where CAR T cell cytotoxic activity was reduced. Our results identify a previously unappreciated level of diversity in the outcomes of CAR T cell interactions in vivo, with important clinical implications.


Assuntos
Imunoterapia Adotiva/métodos , Microscopia Intravital/métodos , Linfoma de Células B/terapia , Receptores de Antígenos Quiméricos/metabolismo , Análise de Célula Única/métodos , Linfócitos T/metabolismo , Animais , Antígenos CD19/metabolismo , Apoptose , Linhagem Celular Tumoral , Pulmão/metabolismo , Linfoma de Células B/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Modelos Teóricos , Recidiva
13.
Immunity ; 49(4): 654-665.e5, 2018 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-30266340

RESUMO

Recruitment of immune cells with antimicrobial activities is essential to fight local infections but has the potential to trigger immunopathology. Whether the immune system has the ability to sense inflammation intensity and self-adjust accordingly to limit tissue damage remains to be fully established. During local infection with an intracellular pathogen, we have shown that nitric oxide (NO) produced by recruited monocyte-derived cells was essential to limit inflammation and cell recruitment. Mechanistically, we have provided evidence that NO dampened monocyte-derived cell cytokine and chemokine production by inhibiting cellular respiration and reducing cellular ATP:ADP ratio. Such metabolic control operated at the tissue level but only when a sufficient number of NO-producing cells reached the site of infection. Thus, NO production and activity act as a quorum sensing mechanism to help terminate the inflammatory response.


Assuntos
Citocinas/imunologia , Inflamação/imunologia , Monócitos/imunologia , Óxido Nítrico/imunologia , Animais , Células Cultivadas , Citocinas/metabolismo , Células HEK293 , Interações Hospedeiro-Parasita/imunologia , Humanos , Inflamação/metabolismo , Inflamação/parasitologia , Leishmania major/imunologia , Leishmania major/fisiologia , Leishmaniose Cutânea/imunologia , Leishmaniose Cutânea/metabolismo , Leishmaniose Cutânea/parasitologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/parasitologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monócitos/metabolismo , Monócitos/parasitologia , Óxido Nítrico/metabolismo , Percepção de Quorum/imunologia
14.
J Exp Med ; 215(5): 1481-1492, 2018 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-29588347

RESUMO

T cells are primed in secondary lymphoid organs by establishing stable interactions with antigen-presenting cells (APCs). However, the cellular mechanisms underlying the termination of T cell priming and the initiation of clonal expansion remain largely unknown. Using intravital imaging, we observed that T cells typically divide without being associated to APCs. Supporting these findings, we demonstrate that recently activated T cells have an intrinsic defect in establishing stable contacts with APCs, a feature that was reflected by a blunted capacity to stop upon T cell receptor (TCR) engagement. T cell unresponsiveness was caused, in part, by a general block in extracellular calcium entry. Forcing TCR signals in activated T cells antagonized cell division, suggesting that T cell hyporesponsiveness acts as a safeguard mechanism against signals detrimental to mitosis. We propose that transient unresponsiveness represents an essential phase of T cell priming that promotes T cell disengagement from APCs and favors effective clonal expansion.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Divisão Celular , Apresentação Cruzada/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Animais , Sinalização do Cálcio , Comunicação Celular , Regulação para Baixo , Linfonodos/citologia , Ativação Linfocitária/imunologia , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo
15.
Nat Med ; 22(1): 64-71, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26692332

RESUMO

The inflammasome is activated in response to a variety of pathogens and has an important role in shaping adaptive immunity, yet the spatiotemporal orchestration of inflammasome activation in vivo and the mechanisms by which it promotes an effective immune response are not fully understood. Using an in vivo reporter to visualize inflammasome assembly, we establish the distribution, kinetics and propagation of the inflammasome response to a local viral infection. We show that modified vaccinia Ankara virus induces inflammasome activation in subcapsular sinus (SCS) macrophages, which is immediately followed by cell death and release of extracellular ASC specks. This transient inflammasome signaling in the lymph node generates a robust influx of inflammatory cells and mobilizes T cells from the circulation to increase the magnitude of T cell responses. We propose that after infection, SCS macrophages deliver a burst response of inflammasome activity and cell death that translates into the broadening of T cell responses, identifying an important aspect of inflammasome-driven vaccination strategies.


Assuntos
Imunidade Adaptativa/imunologia , Imunidade Inata/imunologia , Inflamassomos/imunologia , Linfonodos/imunologia , Macrófagos/imunologia , Infecções por Poxviridae/imunologia , Linfócitos T/imunologia , Animais , Proteínas Reguladoras de Apoptose/imunologia , Proteínas Adaptadoras de Sinalização CARD , Citometria de Fluxo , Camundongos , Vaccinia virus/imunologia
16.
Proc Natl Acad Sci U S A ; 112(39): 12151-6, 2015 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-26371316

RESUMO

T lymphocytes are highly motile cells that decelerate upon antigen recognition. These cells can either completely stop or maintain a low level of motility, forming contacts referred to as synapses or kinapses, respectively. Whether similar or distinct molecular mechanisms regulate T-cell deceleration during synapses or kinapses is unclear. Here, we used microfabricated channels and intravital imaging to observe and manipulate T-cell kinapses and synapses. We report that high-affinity antigen induced a pronounced deceleration selectively dependent on Ca(2+) signals and actin-related protein 2/3 complex (Arp2/3) activity. In contrast, low-affinity antigens induced a switch of migration mode that promotes T-cell exploratory behavior, characterized by partial deceleration and frequent direction changes. This switch depended on T-cell receptor binding but was largely independent of downstream signaling. We propose that distinct mechanisms of T-cell deceleration can be triggered during antigenic recognition to favor local exploration and signal integration upon suboptimal stimulus and complete arrest on the best antigen-presenting cells.


Assuntos
Antígenos de Diferenciação de Linfócitos T/imunologia , Movimento Celular/imunologia , Microambiente Celular/imunologia , Imunidade Celular/imunologia , Linfócitos T/imunologia , Complexo 2-3 de Proteínas Relacionadas à Actina/imunologia , Análise de Variância , Animais , Cálcio/metabolismo , Citometria de Fluxo , Sinapses Imunológicas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Estatísticas não Paramétricas
17.
J Exp Med ; 210(9): 1839-54, 2013 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-23918956

RESUMO

The small intestine epithelium (SI-Ep) harbors millions of unconventional (γδ and CD4(-) CD8(-) NK1.1(-) TCRαß) and conventional (CD8αß and CD4) T cells, designated intraepithelial lymphocytes (IELs). Here, we identified the circulating pool of SI-Ep-tropic T cells and studied their capacity to colonize the SI-Ep under steady-state conditions in SPF mice. Developmentally regulated levels of α4ß7 endowed recent thymic emigrants (RTEs) of unconventional types with higher SI-Ep tropism than their conventional homologues. SI-Ep-tropic RTEs, which in all lineages emerged naive, homed to the SI-Ep, but this environment was inadequate to stimulate them to cycle. In contrast, conventional and, unexpectedly, unconventional T cells, particularly Vγ7(+) (hallmark of γδ IELs), previously stimulated to cycle in the gut-associated lymphoid tissue (GALT), proliferated in the SI-Ep. Cycling unconventional SI-Ep immigrants divided far more efficiently than their conventional homologues, thereby becoming predominant. This difference impacted on acquisition of high Granzyme B content, which required extensive proliferation. In conclusion, SI-Ep-tropic T cells follow a thymus-SI-Ep or a GALT-SI-Ep pathway, the latter generating highly competitive immigrants that are the sole precursors of cytotoxic IELs. These events occur continuously as part of the normal IEL dynamics.


Assuntos
Linhagem da Célula/imunologia , Movimento Celular/imunologia , Epitélio/imunologia , Intestino Delgado/citologia , Linfócitos T/citologia , Linfócitos T/imunologia , Animais , Divisão Celular , Proliferação de Células , Células Epiteliais/citologia , Células Epiteliais/imunologia , Granzimas/metabolismo , Integrinas/metabolismo , Tecido Linfoide/citologia , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Receptores CCR/metabolismo , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia , Ducto Torácico/citologia , Timócitos/citologia , Timócitos/metabolismo , Timo/citologia , Timo/crescimento & desenvolvimento
18.
J Immunol ; 191(4): 1578-85, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23836060

RESUMO

Effector T cell responses rely on a phenotypically and functionally heterogeneous population of cells. Whether this diversity is programmed before clonal expansion or in later phases as a result of stochastic events or asymmetric cell division is not fully understood. In this study, we first took advantage of a sensitive in vitro assay to analyze the composition of single CD8(+) T cell progenies. Heterogeneity was predominantly observed between progenies of distinct clones, but could also be detected within individual progenies. Furthermore, by physically isolating daughter cells of the first T cell division, we showed that differences in paired daughter cell progenies contributed to intraclonal diversification. Finally, we developed an in vivo limiting dilution assay to compare individual T cell progenies following immunization. We provided evidence for simultaneous intraclonal and interclonal diversification in vivo. Our results support the idea that T cell diversification is a continuous process, initiated before clonal expansion and amplified during the first and subsequent cell divisions.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linhagem da Célula , Imunofenotipagem/métodos , Subpopulações de Linfócitos T/imunologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/citologia , Divisão Celular , Seleção Clonal Mediada por Antígeno/imunologia , Células Clonais/citologia , Células Dendríticas/imunologia , Epitopos de Linfócito T/imunologia , Citometria de Fluxo/métodos , Subunidade alfa de Receptor de Interleucina-2/análise , Selectina L/análise , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Fragmentos de Peptídeos/imunologia , Subpopulações de Linfócitos T/citologia
19.
Blood ; 121(10): 1749-59, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-23287858

RESUMO

Transcription factors orchestrate T-lineage differentiation in the thymus. One critical checkpoint involves Notch1 signaling that instructs T-cell commitment at the expense of the B-lineage program. While GATA-3 is required for T-cell specification, its mechanism of action is poorly understood. We show that GATA-3 works in concert with Notch1 to commit thymic progenitors to the T-cell lineage via 2 distinct pathways. First, GATA-3 orchestrates a transcriptional "repertoire" that is required for thymocyte maturation up to and beyond the pro-T-cell stage. Second, GATA-3 critically suppresses a latent B-cell potential in pro­T cells. As such, GATA-3 is essential to sealing in Notch-induced T-cell fate in early thymocyte precursors by promoting T-cell identity through the repression of alternative developmental options.


Assuntos
Linfócitos B/citologia , Diferenciação Celular/imunologia , Linhagem da Célula/imunologia , Fator de Transcrição GATA3/fisiologia , Transdução de Sinais/imunologia , Linfócitos T/citologia , Timo/citologia , Animais , Linfócitos B/imunologia , Células Cultivadas , Feminino , Citometria de Fluxo , Masculino , Camundongos , Camundongos Knockout , Receptor Notch1/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Linfócitos T/imunologia , Timo/embriologia , Timo/imunologia , Fatores de Transcrição/metabolismo
20.
J Immunol ; 189(12): 5493-7, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23183896

RESUMO

Monoclonal antibodies represent a promising approach to fight a variety of tumors, but their mode of action remains to be fully understood. NK cells can recognize Ab-coated targets, as well as stress ligands, on tumor cells. In this study, we investigated how NK cells integrate both kinds of activating signals. NK cell-mediated killing was maximal with the combined recognition of NKG2D ligands and Ab; surprisingly, only NKG2D engagement substantially enhanced degranulation. Conversely, Ab recognition by NK cells uniquely increased contact stability with tumor cells. Furthermore, using intravital imaging of solid tumors, we showed that Ab recognition favored prolonged interactions between NK cells and targets. Altogether, our results demonstrate that NK cell-mediated killing can be differentially regulated at the level of degranulation and contact stability by distinct activating receptors. Thus, complementary signals mediated by recognition of stress ligands and tumor-specific Abs may contribute to the efficacy of NK cells during mAb therapy.


Assuntos
Anticorpos Antineoplásicos/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Comunicação Celular/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/fisiologia , Regulação para Cima/imunologia , Animais , Degranulação Celular/imunologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Técnicas de Introdução de Genes , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/patologia , Linfoma/imunologia , Linfoma/metabolismo , Linfoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Receptores de IgG/metabolismo , Timoma/imunologia , Timoma/metabolismo , Timoma/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...