Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Nat Immunol ; 25(8): 1367-1382, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38992254

ABSTRACT

Upregulation of diverse self-antigens that constitute components of the inflammatory response overlaps spatially and temporally with the emergence of pathogen-derived foreign antigens. Therefore, discrimination between these inflammation-associated self-antigens and pathogen-derived molecules represents a unique challenge for the adaptive immune system. Here, we demonstrate that CD8+ T cell tolerance to T cell-derived inflammation-associated self-antigens is efficiently induced in the thymus and supported by redundancy in cell types expressing these molecules. In addition to thymic epithelial cells, this included thymic eosinophils and innate-like T cells, a population that expressed molecules characteristic for all major activated T cell subsets. We show that direct T cell-to-T cell antigen presentation by minute numbers of innate-like T cells was sufficient to eliminate autoreactive CD8+ thymocytes. Tolerance to such effector molecules was of critical importance, as its breach caused by decreased thymic abundance of a single model inflammation-associated self-antigen resulted in autoimmune elimination of an entire class of effector T cells.


Subject(s)
Antigen Presentation , Autoantigens , CD8-Positive T-Lymphocytes , Inflammation , Thymocytes , Thymus Gland , Animals , Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Mice , Thymus Gland/immunology , Inflammation/immunology , Antigen Presentation/immunology , Thymocytes/immunology , Thymocytes/metabolism , Mice, Inbred C57BL , Immunity, Innate , Autoimmunity/immunology , Immune Tolerance/immunology , Mice, Transgenic , Mice, Knockout , Lymphocyte Activation/immunology , Eosinophils/immunology
2.
Nat Immunol ; 24(6): 979-990, 2023 06.
Article in English | MEDLINE | ID: mdl-37188942

ABSTRACT

Antiviral CD8+ T cell immunity depends on the integration of various contextual cues, but how antigen-presenting cells (APCs) consolidate these signals for decoding by T cells remains unclear. Here, we describe gradual interferon-α/interferon-ß (IFNα/ß)-induced transcriptional adaptations that endow APCs with the capacity to rapidly activate the transcriptional regulators p65, IRF1 and FOS after CD4+ T cell-mediated CD40 stimulation. While these responses operate through broadly used signaling components, they induce a unique set of co-stimulatory molecules and soluble mediators that cannot be elicited by IFNα/ß or CD40 alone. These responses are critical for the acquisition of antiviral CD8+ T cell effector function, and their activity in APCs from individuals infected with severe acute respiratory syndrome coronavirus 2 correlates with milder disease. These observations uncover a sequential integration process whereby APCs rely on CD4+ T cells to select the innate circuits that guide antiviral CD8+ T cell responses.


Subject(s)
Antiviral Agents , COVID-19 , Humans , Calibration , Antigen-Presenting Cells , CD8-Positive T-Lymphocytes , CD40 Antigens , Interferon-alpha , CD4-Positive T-Lymphocytes
3.
Nat Immunol ; 22(10): 1256-1267, 2021 10.
Article in English | MEDLINE | ID: mdl-34462601

ABSTRACT

Innate lymphoid cells (ILCs) participate in tissue homeostasis, inflammation, and early immunity against infection. It is unclear how ILCs acquire effector function and whether these mechanisms differ between organs. Through multiplexed single-cell mRNA sequencing, we identified cKit+CD127hiTCF-1hi early differentiation stages of T-bet+ ILC1s. These cells were present across different organs and had the potential to mature toward CD127intTCF-1int and CD127-TCF-1- ILC1s. Paralleling a gradual loss of TCF-1, differentiating ILC1s forfeited their expansion potential while increasing expression of effector molecules, reminiscent of T cell differentiation in secondary lymphoid organs. The transcription factor Hobit was induced in TCF-1hi ILC1s and was required for their effector differentiation. These findings reveal sequential mechanisms of ILC1 lineage commitment and effector differentiation that are conserved across tissues. Our analyses suggest that ILC1s emerge as TCF-1hi cells in the periphery and acquire a spectrum of organ-specific effector phenotypes through a uniform Hobit-dependent differentiation pathway driven by local cues.


Subject(s)
Cell Differentiation/immunology , Immunity, Innate/immunology , Lymphocytes/immunology , Transcription Factors/immunology , Animals , Female , Inflammation/immunology , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/immunology , T-Lymphocytes/immunology
4.
Immunity ; 57(1): 124-140.e7, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38157853

ABSTRACT

Natural killer (NK) cells are present in the circulation and can also be found residing in tissues, and these populations exhibit distinct developmental requirements and are thought to differ in terms of ontogeny. Here, we investigate whether circulating conventional NK (cNK) cells can develop into long-lived tissue-resident NK (trNK) cells following acute infections. We found that viral and bacterial infections of the skin triggered the recruitment of cNK cells and their differentiation into Tcf1hiCD69hi trNK cells that share transcriptional similarity with CD56brightTCF1hi NK cells in human tissues. Skin trNK cells arose from interferon (IFN)-γ-producing effector cells and required restricted expression of the transcriptional regulator Blimp1 to optimize Tcf1-dependent trNK cell formation. Upon secondary infection, trNK cells rapidly gained effector function and mediated an accelerated NK cell response. Thus, cNK cells redistribute and permanently position at sites of previous infection via a mechanism promoting tissue residency that is distinct from Hobit-dependent developmental paths of NK cells and ILC1 seeding tissues during ontogeny.


Subject(s)
Coinfection , Humans , Killer Cells, Natural/metabolism , Cell Differentiation
5.
Nat Immunol ; 21(11): 1397-1407, 2020 11.
Article in English | MEDLINE | ID: mdl-32989328

ABSTRACT

Antiviral CD8+ T cell responses are characterized by an initial activation/priming of T lymphocytes followed by a massive proliferation, subset differentiation, population contraction and the development of a stable memory pool. The transcription factor BATF3 has been shown to play a central role in the development of conventional dendritic cells, which in turn are critical for optimal priming of CD8+ T cells. Here we show that BATF3 was expressed transiently within the first days after T cell priming and had long-lasting T cell-intrinsic effects. T cells that lacked Batf3 showed normal expansion and differentiation, yet succumbed to an aggravated contraction and had a diminished memory response. Vice versa, BATF3 overexpression in CD8+ T cells promoted their survival and transition to memory. Mechanistically, BATF3 regulated T cell apoptosis and longevity via the proapoptotic factor BIM. By programing CD8+ T cell survival and memory, BATF3 is a promising molecule to optimize adoptive T cell therapy in patients.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cellular Reprogramming/genetics , Immunologic Memory/genetics , Repressor Proteins/genetics , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Differentiation , Cell Survival/genetics , Gene Expression , Humans , Immunophenotyping , Mice , Mice, Knockout , Mice, Transgenic , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Nat Immunol ; 21(3): 321-330, 2020 03.
Article in English | MEDLINE | ID: mdl-32066949

ABSTRACT

Differentiation of CD4+ T cells into either follicular helper T (TFH) or type 1 helper T (TH1) cells influences the balance between humoral and cellular adaptive immunity, but the mechanisms whereby pathogens elicit distinct effector cells are incompletely understood. Here we analyzed the spatiotemporal dynamics of CD4+ T cells during infection with recombinant vesicular stomatitis virus (VSV), which induces early, potent neutralizing antibodies, or recombinant lymphocytic choriomeningitis virus (LCMV), which induces a vigorous cellular response but inefficient neutralizing antibodies, expressing the same T cell epitope. Early exposure of dendritic cells to type I interferon (IFN), which occurred during infection with VSV, induced production of the cytokine IL-6 and drove TFH cell polarization, whereas late exposure to type I IFN, which occurred during infection with LCMV, did not induce IL-6 and allowed differentiation into TH1 cells. Thus, tight spatiotemporal regulation of type I IFN shapes antiviral CD4+ T cell differentiation and might instruct vaccine design strategies.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interferon Type I/metabolism , Adaptive Immunity , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/classification , Cell Differentiation/immunology , Female , Interleukin-6/biosynthesis , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spatio-Temporal Analysis , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology , Vesicular stomatitis Indiana virus/immunology , Vesicular stomatitis Indiana virus/pathogenicity , Vesicular stomatitis New Jersey virus/immunology , Vesicular stomatitis New Jersey virus/pathogenicity
7.
Immunity ; 56(8): 1778-1793.e10, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37463581

ABSTRACT

Unlike macrophage networks composed of long-lived tissue-resident cells within specific niches, conventional dendritic cells (cDCs) that generate a 3D network in lymph nodes (LNs) are short lived and continuously replaced by DC precursors (preDCs) from the bone marrow (BM). Here, we examined whether specific anatomical niches exist within which preDCs differentiate toward immature cDCs. In situ photoconversion and Prtn3-based fate-tracking revealed that the LN medullary cords are preferential entry sites for preDCs, serving as specific differentiation niches. Repopulation and fate-tracking approaches demonstrated that the cDC1 network unfolded from the medulla along the vascular tree toward the paracortex. During inflammation, collective maturation and migration of resident cDC1s to the paracortex created discontinuity in the medullary cDC1 network and temporarily impaired responsiveness. The decrease in local cDC1 density resulted in higher Flt3L availability in the medullary niche, which accelerated cDC1 development to restore the network. Thus, the spatiotemporal development of the cDC1 network is locally regulated in dedicated LN niches via sensing of cDC1 densities.


Subject(s)
Lymph Nodes , Macrophages , Cell Differentiation , Dendritic Cells
8.
Immunity ; 55(10): 1813-1828.e9, 2022 10 11.
Article in English | MEDLINE | ID: mdl-36002023

ABSTRACT

Lymphatic transport of molecules and migration of myeloid cells to lymph nodes (LNs) continuously inform lymphocytes on changes in drained tissues. Here, using LN transplantation, single-cell RNA-seq, spectral flow cytometry, and a transgenic mouse model for photolabeling, we showed that tissue-derived unconventional T cells (UTCs) migrate via the lymphatic route to locally draining LNs. As each tissue harbored a distinct spectrum of UTCs with locally adapted differentiation states and distinct T cell receptor repertoires, every draining LN was thus populated by a distinctive tissue-determined mix of these lymphocytes. By making use of single UTC lineage-deficient mouse models, we found that UTCs functionally cooperated in interconnected units and generated and shaped characteristic innate and adaptive immune responses that differed between LNs that drained distinct tissues. Lymphatic migration of UTCs is, therefore, a key determinant of site-specific immunity initiated in distinct LNs with potential implications for vaccination strategies and immunotherapeutic approaches.


Subject(s)
Lymph Nodes , T-Lymphocytes , Animals , Disease Models, Animal , Immunity , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell
9.
Immunity ; 55(4): 656-670.e8, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35366396

ABSTRACT

Reinvigoration of exhausted CD8+ T (Tex) cells by checkpoint immunotherapy depends on the activation of precursors of exhausted T (Tpex) cells, but the local anatomical context of their maintenance, differentiation, and interplay with other cells is not well understood. Here, we identified transcriptionally distinct Tpex subpopulations, mapped their differentiation trajectories via transitory cellular states toward Tex cells, and localized these cell states to specific splenic niches. Conventional dendritic cells (cDCs) were critical for successful αPD-L1 therapy and were required to mediate viral control. cDC1s were dispensable for Tpex cell expansion but provided an essential niche to promote Tpex cell maintenance, preventing their overactivation and T-cell-mediated immunopathology. Mechanistically, cDC1s insulated Tpex cells via MHC-I-dependent interactions to prevent their activation within other inflammatory environments that further aggravated their exhaustion. Our findings reveal that cDC1s maintain and safeguard Tpex cells within distinct anatomical niches to balance viral control, exhaustion, and immunopathology.


Subject(s)
CD8-Positive T-Lymphocytes , Dendritic Cells , Cell Differentiation , Immunotherapy , Lymphocyte Count
10.
Cell ; 162(6): 1322-37, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26296422

ABSTRACT

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.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Communication , Dendritic Cells/immunology , Vaccinia virus/physiology , Vaccinia/immunology , Animals , Antigen Presentation , Antigens, Viral/immunology , Dendritic Cells/cytology , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Receptors, Chemokine/genetics , Spleen/cytology , Spleen/immunology
11.
Cell ; 156(3): 456-68, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24485454

ABSTRACT

The phagocytes of the innate immune system, macrophages and neutrophils, contribute to antibacterial defense, but their functional specialization and cooperation is unclear. Here, we report that three distinct phagocyte subsets play highly coordinated roles in bacterial urinary tract infection. Ly6C(-) macrophages acted as tissue-resident sentinels that attracted circulating neutrophils and Ly6C(+) macrophages. Such Ly6C(+) macrophages played a previously undescribed helper role: once recruited to the site of infection, they produced the cytokine TNF, which caused Ly6C(-) macrophages to secrete CXCL2. This chemokine activated matrix metalloproteinase-9 in neutrophils, allowing their entry into the uroepithelium to combat the bacteria. In summary, the sentinel macrophages elicit the powerful antibacterial functions of neutrophils only after confirmation by the helper macrophages, reminiscent of the licensing role of helper T cells in antiviral adaptive immunity. These findings identify helper macrophages and TNF as critical regulators in innate immunity against bacterial infections in epithelia.


Subject(s)
Bacterial Infections/immunology , Macrophages/immunology , Neutrophils/immunology , Urinary Tract Infections/immunology , Animals , Antigens, Ly/metabolism , Chemokine CXCL2/immunology , Female , Immune System Diseases , Kinetics , Leukocyte Disorders , Macrophages/cytology , Matrix Metalloproteinase 9/metabolism , Mice , Neutrophils/cytology , Specific Pathogen-Free Organisms , Tumor Necrosis Factor-alpha/immunology
12.
Nature ; 618(7967): 1033-1040, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37316667

ABSTRACT

Most clinically applied cancer immunotherapies rely on the ability of CD8+ cytolytic T cells to directly recognize and kill tumour cells1-3. These strategies are limited by the emergence of major histocompatibility complex (MHC)-deficient tumour cells and the formation of an immunosuppressive tumour microenvironment4-6. The ability of CD4+ effector cells to contribute to antitumour immunity independently of CD8+ T cells is increasingly recognized, but strategies to unleash their full potential remain to be identified7-10. Here, we describe a mechanism whereby a small number of CD4+ T cells is sufficient to eradicate MHC-deficient tumours that escape direct CD8+ T cell targeting. The CD4+ effector T cells preferentially cluster at tumour invasive margins where they interact with MHC-II+CD11c+ antigen-presenting cells. We show that T helper type 1 cell-directed CD4+ T cells and innate immune stimulation reprogramme the tumour-associated myeloid cell network towards interferon-activated antigen-presenting and iNOS-expressing tumouricidal effector phenotypes. Together, CD4+ T cells and tumouricidal myeloid cells orchestrate the induction of remote inflammatory cell death that indirectly eradicates interferon-unresponsive and MHC-deficient tumours. These results warrant the clinical exploitation of this ability of CD4+ T cells and innate immune stimulators in a strategy to complement the direct cytolytic activity of CD8+ T cells and natural killer cells and advance cancer immunotherapies.


Subject(s)
CD4-Positive T-Lymphocytes , Cell Death , Immunotherapy , Inflammation , Neoplasms , Tumor Microenvironment , Humans , Antigen-Presenting Cells/immunology , CD11c Antigen/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Death/immunology , Histocompatibility Antigens Class II/immunology , Immunity, Innate , Inflammation/immunology , Interferons/immunology , Major Histocompatibility Complex/immunology , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Tumor Microenvironment/immunology , Immunotherapy/methods , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Th1 Cells/cytology , Th1 Cells/immunology
13.
Nat Immunol ; 17(5): 490-4, 2016 May.
Article in English | MEDLINE | ID: mdl-27092806

ABSTRACT

Lymphocytes are essential in innate and adaptive immunity. Recent insights suggest that some innate lymphocytes execute functions with adaptive characteristics, while adaptive lymphocytes can operate in ways reminiscent of innate cells. Rather than partitioning lymphocytes according to the type of effector function they execute, we propose that a relevant discrimination relates to the existence of conventional T cells in a naive state. The naive state can be seen as an actively repressed condition that supports T cell diversity and enables the flexible differentiation of effector cells in a manner that best addresses the antigenic challenge. We discuss these considerations in the context of the relative roles of innate lymphoid cells and antigen-experienced T cells in the immune system.


Subject(s)
Adaptive Immunity/immunology , Immunity, Innate/immunology , Lymphocytes/immunology , Receptors, Antigen/immunology , Animals , Cell Differentiation/immunology , Clone Cells/immunology , Clone Cells/metabolism , Cytokines/immunology , Cytokines/metabolism , Humans , Lymphocytes/cytology , Lymphocytes/metabolism , Models, Immunological , Receptors, Antigen/metabolism
14.
Immunity ; 51(1): 5-7, 2019 07 16.
Article in English | MEDLINE | ID: mdl-31315036

ABSTRACT

The intracellular pathogen L. monocytogenes takes advantage of several myeloid cell populations to establish infection in the spleen. In this issue, Liu et al. (2019) reveal how marginal zone B cells, dendritic cells, and marginal metallophilic macrophages act together with IL-10 to promote L. monocytogenes infection, while simultaneously enabling adaptive CD8+ T cell responses.


Subject(s)
Listeria monocytogenes , Listeriosis , Animals , B-Lymphocytes , Dendritic Cells , Interleukin-10 , Macrophages , Mice
15.
Immunity ; 50(6): 1453-1466.e4, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31053503

ABSTRACT

In lymph nodes, subcapsular sinus macrophages (SSMs) form an immunological barrier that monitors lymph drained from peripheral tissues. Upon infection, SSMs activate B and natural killer T (NKT) cells while secreting inflammatory mediators. Here, we investigated the mechanisms regulating development and homeostasis of SSMs. Embryonic SSMs originated from yolk sac hematopoiesis and were replaced by a postnatal wave of bone marrow (BM)-derived monocytes that proliferated to establish the adult SSM network. The SSM network self-maintained by proliferation with minimal BM contribution. Upon pathogen-induced transient deletion, BM-derived cells contributed to restoring the SSM network. Lymphatic endothelial cells (LECs) were the main source of CSF-1 within the lymph node and conditional deletion of Csf1 in adult LECs decreased the network of SSMs and medullary sinus macrophages (MSMs). Thus, SSMs have a dual hematopoietic origin, and LECs are essential to the niche supporting these macrophages.


Subject(s)
Endothelial Cells/metabolism , Macrophages/metabolism , Animals , Biomarkers , Cell Communication , Cell Differentiation , Gene Expression , Genes, Reporter , Hematopoiesis/genetics , Hematopoiesis/immunology , Homeostasis , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymphatic Vessels , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/cytology , Macrophages/immunology , Mice , Monocytes/cytology , Monocytes/metabolism , Yolk Sac
16.
Immunity ; 51(2): 285-297.e5, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31272808

ABSTRACT

Interactions with the microbiota influence many aspects of immunity, including immune cell development, differentiation, and function. Here, we examined the impact of the microbiota on CD8+ T cell memory. Antigen-activated CD8+ T cells transferred into germ-free mice failed to transition into long-lived memory cells and had transcriptional impairments in core genes associated with oxidative metabolism. The microbiota-derived short-chain fatty acid (SCFA) butyrate promoted cellular metabolism, enhanced memory potential of activated CD8+ T cells, and SCFAs were required for optimal recall responses upon antigen re-encounter. Mechanistic experiments revealed that butyrate uncoupled the tricarboxylic acid cycle from glycolytic input in CD8+ T cells, which allowed preferential fueling of oxidative phosphorylation through sustained glutamine utilization and fatty acid catabolism. Our findings reveal a role for the microbiota in promoting CD8+ T cell long-term survival as memory cells and suggest that microbial metabolites guide the metabolic rewiring of activated CD8+ T cells to enable this transition.


Subject(s)
Butyrates/metabolism , CD8-Positive T-Lymphocytes/immunology , Fatty Acids, Volatile/metabolism , Immunologic Memory , Microbiota/immunology , Adoptive Transfer , Animals , Antigens/immunology , Cell Differentiation , Cells, Cultured , Glycolysis , Humans , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction
17.
Nature ; 609(7926): 354-360, 2022 09.
Article in English | MEDLINE | ID: mdl-35978192

ABSTRACT

CD8+ T cells that respond to chronic viral infections or cancer are characterized by the expression of inhibitory receptors such as programmed cell death protein 1 (PD-1) and by the impaired production of cytokines. This state of restrained functionality-which is referred to as T cell exhaustion1,2-is maintained by precursors of exhausted T (TPEX) cells that express the transcription factor T cell factor 1 (TCF1), self-renew and give rise to TCF1- exhausted effector T cells3-6. Here we show that the long-term proliferative potential, multipotency and repopulation capacity of exhausted T cells during chronic infection are selectively preserved in a small population of transcriptionally distinct CD62L+ TPEX cells. The transcription factor MYB is not only essential for the development of CD62L+ TPEX cells and maintenance of the antiviral CD8+ T cell response, but also induces functional exhaustion and thereby prevents lethal immunopathology. Furthermore, the proliferative burst in response to PD-1 checkpoint inhibition originates exclusively from CD62L+ TPEX cells and depends on MYB. Our findings identify CD62L+ TPEX cells as a stem-like population that is central to the maintenance of long-term antiviral immunity and responsiveness to immunotherapy. Moreover, they show that MYB is a transcriptional orchestrator of two fundamental aspects of exhausted T cell responses: the downregulation of effector function and the long-term preservation of self-renewal capacity.


Subject(s)
CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor , Proto-Oncogene Proteins c-myb , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Cell Self Renewal , Hepatocyte Nuclear Factor 1-alpha/metabolism , Immunotherapy , L-Selectin/metabolism , Precursor Cells, T-Lymphoid/cytology , Precursor Cells, T-Lymphoid/immunology , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Proto-Oncogene Proteins c-myb/metabolism , Viruses/immunology
18.
Annu Rev Cell Dev Biol ; 30: 141-67, 2014.
Article in English | MEDLINE | ID: mdl-25150013

ABSTRACT

Secondary lymphoid tissues are the sites of both innate and adaptive host defense. Aside from the relatively static nonhematopoietic stromal elements and some macrophages and dendritic cells, most of the cells in these tissues are in constant movement, but the organs maintain a defined microanatomy with preferred locations for the bulk of T cells, B cells, and other lymphocytes and subsets of myeloid cells. Here we describe both the cell dynamics and spatial organization of lymph nodes and review how both physical features and molecular cues guide cell movement to optimize host defense. We emphasize the role of locality in improving the efficiency of a system requiring rare cells to find each other and interact productively through membrane-bound or short-range secreted mediators and highlight how changes in steady-state cell positioning during an infectious challenge contribute to rapid generation of productive responses.


Subject(s)
Adaptive Immunity , Immunity, Innate , Lymphoid Tissue/immunology , Animals , Cell Communication , Chemokines/physiology , Chemotaxis, Leukocyte/physiology , Dendritic Cells/immunology , Germinal Center/immunology , Germinal Center/ultrastructure , Humans , Infections/immunology , Inflammation/immunology , Lymph Nodes/immunology , Lymph Nodes/ultrastructure , Lymphocyte Activation , Lymphocyte Subsets/immunology , Lymphoid Tissue/ultrastructure , Macrophages/immunology , Neutrophils/immunology , Organ Specificity , Stromal Cells/immunology , Time Factors , Vertebrates/anatomy & histology , Vertebrates/immunology , Wounds and Injuries/immunology
19.
Cell ; 150(6): 1235-48, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22980983

ABSTRACT

The lymphatic network that transports interstitial fluid and antigens to lymph nodes constitutes a conduit system that can be hijacked by invading pathogens to achieve systemic spread unless dissemination is blocked in the lymph node itself. Here, we show that a network of diverse lymphoid cells (natural killer cells, γδ T cells, natural killer T cells, and innate-like CD8+ T cells) are spatially prepositioned close to lymphatic sinus-lining sentinel macrophages where they can rapidly and efficiently receive inflammasome-generated IL-18 and additional cytokine signals from the pathogen-sensing phagocytes. This leads to rapid IFNγ secretion by the strategically positioned innate lymphocytes, fostering antimicrobial resistance in the macrophage population. Interference with this innate immune response loop allows systemic spread of lymph-borne bacteria. These findings extend our understanding of the functional significance of cellular positioning and local intercellular communication within lymph nodes while emphasizing the role of these organs as highly active locations of innate host defense.


Subject(s)
Bacterial Infections/immunology , Immunity, Innate , Lymph Nodes/cytology , Lymph Nodes/immunology , Virus Diseases/immunology , Animals , Host-Pathogen Interactions , Inflammasomes/metabolism , Interferon-gamma/immunology , Interleukin-18/immunology , Lymph/microbiology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin Diseases, Infectious/immunology , Specific Pathogen-Free Organisms , T-Lymphocytes/immunology
20.
Nat Immunol ; 15(8): 727-37, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24952505

ABSTRACT

Microbes or danger signals trigger inflammasome sensors, which induce polymerization of the adaptor ASC and the assembly of ASC specks. ASC specks recruit and activate caspase-1, which induces maturation of the cytokine interleukin 1ß (IL-1ß) and pyroptotic cell death. Here we found that after pyroptosis, ASC specks accumulated in the extracellular space, where they promoted further maturation of IL-1ß. In addition, phagocytosis of ASC specks by macrophages induced lysosomal damage and nucleation of soluble ASC, as well as activation of IL-1ß in recipient cells. ASC specks appeared in bodily fluids from inflamed tissues, and autoantibodies to ASC specks developed in patients and mice with autoimmune pathologies. Together these findings reveal extracellular functions of ASC specks and a previously unknown form of cell-to-cell communication.


Subject(s)
Apoptosis/immunology , Caspase 1/immunology , Cytoskeletal Proteins/immunology , Inflammation/immunology , Interleukin-1beta/immunology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antibodies/immunology , Apoptosis Regulatory Proteins , Autoantibodies/immunology , Autoimmune Diseases/immunology , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Caspase 1/genetics , Caspase Inhibitors/pharmacology , Cell Communication/immunology , Cytoskeletal Proteins/genetics , Humans , Inflammasomes/immunology , Lysosomes/pathology , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Phagocytosis/immunology , Prions/chemistry , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL