Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Nat Immunol ; 24(9): 1434-1442, 2023 09.
Article in English | MEDLINE | ID: mdl-37500886

ABSTRACT

Cytotoxic T lymphocytes (CTLs) fight intracellular pathogens and cancer by identifying and destroying infected or transformed target cells1. To kill, CTLs form a specialized cytotoxic immune synapse (IS) with a target of interest and then release toxic perforin and granzymes into the interface to elicit programmed cell death2-5. The IS then dissolves, enabling CTLs to search for additional prey and professional phagocytes to clear the corpse6. While the mechanisms governing IS assembly have been studied extensively, far less is known about target cell release. Here, we applied time-lapse imaging to explore the basis for IS dissolution and found that it occurred concomitantly with the cytoskeletal contraction of apoptotic targets. Genetic and pharmacological perturbation of this contraction response indicated that it was both necessary and sufficient for CTL dissociation. We also found that mechanical amplification of apoptotic contractility promoted faster CTL detachment and serial killing. Collectively, these results establish a biophysical basis for IS dissolution and highlight the importance of mechanosensory feedback in the regulation of cell-cell interactions.


Subject(s)
Apoptosis , T-Lymphocytes, Cytotoxic , Apoptosis/genetics , Perforin , Granzymes
2.
Cell ; 165(1): 100-110, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-26924577

ABSTRACT

The immunological synapse formed between a cytotoxic T lymphocyte (CTL) and an infected or transformed target cell is a physically active structure capable of exerting mechanical force. Here, we investigated whether synaptic forces promote the destruction of target cells. CTLs kill by secreting toxic proteases and the pore forming protein perforin into the synapse. Biophysical experiments revealed a striking correlation between the magnitude of force exertion across the synapse and the speed of perforin pore formation on the target cell, implying that force potentiates cytotoxicity by enhancing perforin activity. Consistent with this interpretation, we found that increasing target cell tension augmented pore formation by perforin and killing by CTLs. Our data also indicate that CTLs coordinate perforin release and force exertion in space and time. These results reveal an unappreciated physical dimension to lymphocyte function and demonstrate that cells use mechanical forces to control the activity of outgoing chemical signals.


Subject(s)
Immunological Synapses , T-Lymphocytes, Cytotoxic/physiology , Animals , Biomechanical Phenomena , Cell Degranulation , Cell Line, Tumor , Mice , Perforin/metabolism , Phosphatidylinositol 3-Kinases/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology
3.
Cell ; 164(3): 365-77, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26806130

ABSTRACT

Malignancy can be suppressed by the immune system in a process termed immunosurveillance. However, to what extent immunosurveillance occurs in spontaneous cancers and the composition of participating cell types remains obscure. Here, we show that cell transformation triggers a tissue-resident lymphocyte response in oncogene-induced murine cancer models. Non-circulating cytotoxic lymphocytes, derived from innate, T cell receptor (TCR)αß, and TCRγδ lineages, expand in early tumors. Characterized by high expression of NK1.1, CD49a, and CD103, these cells share a gene-expression signature distinct from those of conventional NK cells, T cells, and invariant NKT cells. Generation of these lymphocytes is dependent on the cytokine IL-15, but not the transcription factor Nfil3 that is required for the differentiation of tumor-infiltrating NK cells, and IL-15 deficiency, but not Nfil3 deficiency, results in accelerated tumor growth. These findings reveal a tumor-elicited immunosurveillance mechanism that engages unconventional type-1-like innate lymphoid cells and type 1 innate-like T cells.


Subject(s)
Lymphocytes/immunology , Mammary Neoplasms, Experimental/immunology , Monitoring, Immunologic , T-Lymphocyte Subsets/immunology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Granzymes/metabolism , Interleukin-15/immunology , Mice , Mice, Inbred C57BL , Receptors, Antigen, T-Cell, alpha-beta/metabolism
4.
Immunity ; 54(5): 1037-1054.e7, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33756102

ABSTRACT

Immune cells identify and destroy tumors by recognizing cellular traits indicative of oncogenic transformation. In this study, we found that myocardin-related transcription factors (MRTFs), which promote migration and metastatic invasion, also sensitize cancer cells to the immune system. Melanoma and breast cancer cells with high MRTF expression were selectively eliminated by cytotoxic lymphocytes in mouse models of metastasis. This immunosurveillance phenotype was further enhanced by treatment with immune checkpoint blockade (ICB) antibodies. We also observed that high MRTF signaling in human melanoma is associated with ICB efficacy in patients. Using biophysical and functional assays, we showed that MRTF overexpression rigidified the filamentous actin cytoskeleton and that this mechanical change rendered mouse and human cancer cells more vulnerable to cytotoxic T lymphocytes and natural killer cells. Collectively, these results suggest that immunosurveillance has a mechanical dimension, which we call mechanosurveillance, that is particularly relevant for the targeting of metastatic disease.


Subject(s)
Lymphocytes/immunology , Neoplasms/immunology , Actin Cytoskeleton/immunology , Actins/immunology , Animals , Cell Communication/immunology , Cell Line , Cell Line, Tumor , Cell Movement/immunology , Female , HEK293 Cells , Humans , Killer Cells, Natural/immunology , MCF-7 Cells , Male , Mice , Mice, Inbred C57BL , Signal Transduction/immunology , Transcription Factors/immunology
5.
Immunity ; 50(6): 1381-1390.e5, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31103381

ABSTRACT

The process of affinity maturation, whereby T and B cells bearing antigen receptors with optimal affinity to the relevant antigen undergo preferential expansion, is a key feature of adaptive immunity. Natural killer (NK) cells are innate lymphocytes capable of "adaptive" responses after cytomegalovirus (CMV) infection. However, whether NK cells are similarly selected on the basis of their avidity for cognate ligand is unknown. Here, we showed that NK cells with the highest avidity for the mouse CMV glycoprotein m157 were preferentially selected to expand and comprise the memory NK cell pool, whereas low-avidity NK cells possessed greater capacity for interferon-γ (IFN-γ) production. Moreover, we provide evidence for avidity selection occurring in human NK cells during human CMV infection. These results delineate how heterogeneity in NK cell avidity diversifies NK cell effector function during antiviral immunity, and how avidity selection might serve to produce the most potent memory NK cells.


Subject(s)
Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , Cytomegalovirus/immunology , Host-Pathogen Interactions/immunology , Killer Cells, Natural/immunology , Animals , Cytomegalovirus Infections/metabolism , Cytotoxicity, Immunologic , Gene Expression Regulation , Herpesviridae Infections/immunology , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Host-Pathogen Interactions/genetics , Humans , Immunologic Memory , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Knockout , Muromegalovirus/immunology , NK Cell Lectin-Like Receptor Subfamily A/genetics , NK Cell Lectin-Like Receptor Subfamily A/metabolism , T-Cell Antigen Receptor Specificity
6.
Immunity ; 46(4): 609-620, 2017 04 18.
Article in English | MEDLINE | ID: mdl-28389069

ABSTRACT

Immune cells communicate by exchanging cytokines to achieve a context-appropriate response, but the distances over which such communication happens are not known. Here, we used theoretical considerations and experimental models of immune responses in vitro and in vivo to quantify the spatial extent of cytokine communications in dense tissues. We established that competition between cytokine diffusion and consumption generated spatial niches of high cytokine concentrations with sharp boundaries. The size of these self-assembled niches scaled with the density of cytokine-consuming cells, a parameter that gets tuned during immune responses. In vivo, we measured interactions on length scales of 80-120 µm, which resulted in a high degree of cell-to-cell variance in cytokine exposure. Such heterogeneous distributions of cytokines were a source of non-genetic cell-to-cell variability that is often overlooked in single-cell studies. Our findings thus provide a basis for understanding variability in the patterning of immune responses by diffusible factors.


Subject(s)
Cell Communication/immunology , Cytokines/immunology , Immune System/immunology , Signal Transduction/immunology , Animals , Cell Line, Tumor , Cells, Cultured , Cytokines/metabolism , Diffusion , Flow Cytometry , Humans , Immune System/cytology , Immune System/metabolism , Immunohistochemistry , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2/pharmacology , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Immunological , STAT5 Transcription Factor/immunology , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
7.
Trends Immunol ; 43(2): 96-105, 2022 02.
Article in English | MEDLINE | ID: mdl-34973924

ABSTRACT

Antitumor immunosurveillance is triggered by immune cell recognition of characteristic biochemical signals on the surfaces of cancer cells. Recent data suggest that the mechanical properties of cancer cells influence the strength of these signals, with physically harder target cells (more rigid) eliciting better, faster, and stronger cytotoxic responses against metastasis. Using analogies to a certain electronic music duo, we argue that the biophysical properties of cancer cells and their environment can adjust the volume and tone of the antitumor immune response. We also consider the potential influence of biomechanics-based immunosurveillance in disease progression and posit that targeting the biophysical properties of cancer cells in concert with their biochemical features could increase the efficacy of immunotherapy.


Subject(s)
Antineoplastic Agents , Neoplasms , Biophysics , Humans , Immunotherapy , Monitoring, Immunologic , Neoplasms/immunology
8.
Nature ; 568(7750): 112-116, 2019 04.
Article in English | MEDLINE | ID: mdl-30918399

ABSTRACT

Chimeric antigen receptors (CARs) are synthetic antigen receptors that reprogram T cell specificity, function and persistence1. Patient-derived CAR T cells have demonstrated remarkable efficacy against a range of B-cell malignancies1-3, and the results of early clinical trials suggest activity in multiple myeloma4. Despite high complete response rates, relapses occur in a large fraction of patients; some of these are antigen-negative and others are antigen-low1,2,4-9. Unlike the mechanisms that result in complete and permanent antigen loss6,8,9, those that lead to escape of antigen-low tumours remain unclear. Here, using mouse models of leukaemia, we show that CARs provoke reversible antigen loss through trogocytosis, an active process in which the target antigen is transferred to T cells, thereby decreasing target density on tumour cells and abating T cell activity by promoting fratricide T cell killing and T cell exhaustion. These mechanisms affect both CD28- and 4-1BB-based CARs, albeit differentially, depending on antigen density. These dynamic features can be offset by cooperative killing and combinatorial targeting to augment tumour responses to immunotherapy.


Subject(s)
Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Leukemia/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Escape/immunology , 4-1BB Ligand/immunology , Animals , CD28 Antigens/immunology , Cytotoxicity, Immunologic , Female , Immunotherapy, Adoptive , Leukemia/pathology , Male , Mice , Mice, Inbred NOD , Neoplasm Recurrence, Local/immunology , T-Lymphocytes/cytology
9.
Nat Immunol ; 12(7): 647-54, 2011 May 22.
Article in English | MEDLINE | ID: mdl-21602810

ABSTRACT

Polarization of the T cell microtubule-organizing center (MTOC) toward the antigen-presenting cell (APC) is driven by the accumulation of diacylglycerol (DAG) at the immunological synapse (IS). The mechanisms that couple DAG to the MTOC are not known. By single-cell photoactivation of the T cell antigen receptor (TCR), we found that three distinct isoforms of protein kinase C (PKC) were recruited by DAG to the IS in two steps. PKC-ɛ and PKC-η accumulated first in a broad region of membrane, whereas PKC-θ arrived later in a smaller zone. Functional experiments indicated that PKC-θ was required for MTOC reorientation and that PKC-ɛ and PKC-η operated redundantly to promote the recruitment of PKC-θ and subsequent polarization responses. Our results establish a previously uncharacterized role for PKC proteins in T cell polarity.


Subject(s)
Cell Polarity/immunology , Cytoskeleton/enzymology , Isoenzymes/immunology , Protein Kinase C-epsilon/immunology , Protein Kinase C/immunology , T-Lymphocytes/enzymology , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/immunology , Cytoskeleton/immunology , Diglycerides/immunology , Immunological Synapses/enzymology , Immunological Synapses/immunology , Mice , Mice, Transgenic , Protein Kinase C-theta , Receptors, Antigen, T-Cell/immunology , Single-Cell Analysis , T-Lymphocytes/cytology , T-Lymphocytes/immunology
10.
Proc Natl Acad Sci U S A ; 117(8): 4310-4319, 2020 02 25.
Article in English | MEDLINE | ID: mdl-32041868

ABSTRACT

Immunological synapse formation between cytotoxic T lymphocytes (CTLs) and the target cells they aim to destroy is accompanied by reorientation of the CTL centrosome to a position beneath the synaptic membrane. Centrosome polarization is thought to enhance the potency and specificity of killing by driving lytic granule fusion at the synapse and thereby the release of perforin and granzymes toward the target cell. To test this model, we employed a genetic strategy to delete centrioles, the core structural components of the centrosome. Centriole deletion altered microtubule architecture as expected but surprisingly had no effect on lytic granule polarization and directional secretion. Nevertheless, CTLs lacking centrioles did display substantially reduced killing potential, which was associated with defects in both lytic granule biogenesis and synaptic actin remodeling. These results reveal an unexpected role for the intact centrosome in controlling the capacity but not the specificity of cytotoxic killing.


Subject(s)
Centrioles/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Centrosome/immunology , Cytotoxicity Tests, Immunologic , Mice, Inbred C57BL , Microtubules/genetics , Microtubules/immunology , Species Specificity
11.
Proc Natl Acad Sci U S A ; 116(40): 19835-19840, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31527238

ABSTRACT

Cells have the remarkable ability to sense the mechanical stiffness of their surroundings. This has been studied extensively in the context of cells interacting with planar surfaces, a conceptually elegant model that also has application in biomaterial design. However, physiological interfaces are spatially complex, exhibiting topographical features that are described over multiple scales. This report explores mechanosensing of microstructured elastomer surfaces by CD4+ T cells, key mediators of the adaptive immune response. We show that T cells form complex interactions with elastomer micropillar arrays, extending processes into spaces between structures and forming local areas of contraction and expansion dictated by the layout of microtubules within this interface. Conversely, cytoskeletal reorganization and intracellular signaling are sensitive to the pillar dimensions and flexibility. Unexpectedly, these measures show different responses to substrate rigidity, suggesting competing processes in overall T cell mechanosensing. The results of this study demonstrate that T cells sense the local rigidity of their environment, leading to strategies for biomaterial design.


Subject(s)
Adaptive Immunity , Biocompatible Materials/chemistry , CD4-Positive T-Lymphocytes/immunology , Immunological Synapses/immunology , Lymphocyte Activation , Actins/metabolism , Animals , CD28 Antigens/immunology , CD3 Complex/immunology , Cell Communication , Cell Proliferation , Cytoskeleton/metabolism , Immune System , Mechanotransduction, Cellular , Mice , Mice, Inbred C57BL , Microtubules/metabolism , Signal Transduction , Spleen/metabolism , Surface Properties
12.
Nat Immunol ; 10(6): 627-35, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19430478

ABSTRACT

The reorientation of the T cell microtubule-organizing center (MTOC) toward the antigen-presenting cell enables the directional secretion of cytokines and lytic factors. By single-cell photoactivation of the T cell antigen receptor, we show that MTOC polarization is driven by localized accumulation of diacylglycerol (DAG). MTOC reorientation was closely preceded first by production of DAG and then by recruitment of the microtubule motor protein dynein. Blocking DAG production or disrupting the localization of DAG impaired MTOC recruitment. Localized DAG accumulation was also required for cytotoxic T cell-mediated killing. Furthermore, photoactivation of DAG itself was sufficient to induce transient polarization. Our data identify a DAG-dependent pathway that signals through dynein to control microtubule polarity in T cells.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , Cell Polarity , Diglycerides/metabolism , Microtubule-Organizing Center/metabolism , T-Lymphocytes, Helper-Inducer/cytology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Dyneins/metabolism , Mice , Phospholipase C gamma/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes, Helper-Inducer/metabolism
13.
Nat Immunol ; 9(10): 1105-11, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18800163

ABSTRACT

T cells use secreted soluble factors for highly specific intercellular communication and targeted cell killing. This specificity is achieved first through T cell receptor-mediated recognition of complexes of peptide and major histocompatibility complex displayed by appropriate antigen-presenting cells and then by the directed secretion of cytokines and lytic factors into the immunological synapse between the T cell and antigen-presenting cell. Studies have begun to probe the molecular basis for this synaptic secretion and have also shown that T cells release chemokines and certain inflammatory factors through a multidirectional pathway directed away from the synapse. Thus, the mode of secretion seems to be tailored to the intended function of the secreted molecule.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Cell Communication/immunology , Inflammation Mediators/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Cell Polarity , Humans , Inflammation Mediators/immunology , Lymphocyte Activation/physiology , Microtubules/immunology , Microtubules/metabolism
14.
Traffic ; 18(8): 491-504, 2017 08.
Article in English | MEDLINE | ID: mdl-28477369

ABSTRACT

T Lymphocyte recognition of antigens leads to the formation of a highly organized structure termed immune synapse (IS) by analogy with the neuronals synapse. Sorting nexin 27 (SNX27) controls the endosomal traffic of PSD95, Dlg1, ZO-1 (PDZ) domain-interacting proteins, and its alteration is associated with impaired synaptic function and neurological diseases. In T-lymphocytes, SNX27-positive vesicles polarize to the IS, the identity of SNX27 interactors in these conditions nonetheless remains unknown. Here we used proteomics to analyze the SNX27 interactome purified from IS-forming T cells, and confirmed the conserved nature of the SNX27/WASH/retromer association in hematopoietic cells. Furthermore, our comparative interactome analysis of SNX27 wild-type and a mutant-deficient for PDZ cargo recognition identified the epithelial cell-cell junction protein zona occludens-2 (ZO-2) as an IS component. Biochemistry and microscopy approaches in T cells confirmed SNX27/ZO-2 PDZ-dependent interaction, and demonstrated its role controlling the dynamic localization of ZO-2 at the IS. This study broadens our knowledge of SNX27 function in T lymphocytes, and suggests that pathways that delimit polarized structures in nervous and epithelial systems also participate in IS regulation.


Subject(s)
Immunological Synapses/metabolism , Protein Interaction Maps , Sorting Nexins/metabolism , T-Lymphocytes/metabolism , Zonula Occludens-2 Protein/metabolism , Cell Line, Tumor , Humans , Protein Transport
15.
Nature ; 491(7425): 554-9, 2012 Nov 22.
Article in English | MEDLINE | ID: mdl-23135404

ABSTRACT

Regulatory T (T(reg)) cells, characterized by expression of the transcription factor forkhead box P3 (Foxp3), maintain immune homeostasis by suppressing self-destructive immune responses. Foxp3 operates as a late-acting differentiation factor controlling T(reg) cell homeostasis and function, whereas the early T(reg)-cell-lineage commitment is regulated by the Akt kinase and the forkhead box O (Foxo) family of transcription factors. However, whether Foxo proteins act beyond the T(reg)-cell-commitment stage to control T(reg) cell homeostasis and function remains largely unexplored. Here we show that Foxo1 is a pivotal regulator of T(reg )cell function. T(reg) cells express high amounts of Foxo1 and display reduced T-cell-receptor-induced Akt activation, Foxo1 phosphorylation and Foxo1 nuclear exclusion. Mice with T(reg)-cell-specific deletion of Foxo1 develop a fatal inflammatory disorder similar in severity to that seen in Foxp3-deficient mice, but without the loss of T(reg) cells. Genome-wide analysis of Foxo1 binding sites reveals ~300 Foxo1-bound target genes, including the pro-inflammatory cytokine Ifng, that do not seem to be directly regulated by Foxp3. These findings show that the evolutionarily ancient Akt-Foxo1 signalling module controls a novel genetic program indispensable for T(reg) cell function.


Subject(s)
Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription, Genetic , Animals , Binding Sites , Cell Nucleus/metabolism , Cell Nucleus/pathology , Female , Forkhead Box Protein O1 , Forkhead Box Protein O3 , Gene Expression Regulation/genetics , Genome/genetics , Immune Tolerance/genetics , Immune Tolerance/immunology , Interferon-gamma/deficiency , Interferon-gamma/genetics , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/pathology
16.
Proc Natl Acad Sci U S A ; 111(6): 2241-6, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24469820

ABSTRACT

Mechanical forces have key roles in regulating activation of T cells and coordination of the adaptive immune response. A recent example is the ability of T cells to sense the rigidity of an underlying substrate through the T-cell receptor (TCR) coreceptor CD3 and CD28, a costimulation signal essential for cell activation. In this report, we show that these two receptor systems provide complementary functions in regulating the cellular forces needed to test the mechanical properties of the extracellular environment. Traction force microscopy was carried out on primary human cells interacting with micrometer-scale elastomer pillar arrays presenting activation antibodies to CD3 and/or CD28. T cells generated traction forces of 100 pN on arrays with both antibodies. By providing one antibody or the other in solution instead of on the pillars, we show that force generation is associated with CD3 and the TCR complex. Engagement of CD28 increases traction forces associated with CD3 through the signaling pathway involving PI3K, rather than providing additional coupling between the cell and surface. Force generation is concentrated to the cell periphery and associated with molecular complexes containing phosphorylated Pyk2, suggesting that T cells use processes that share features with integrin signaling in force generation. Finally, the ability of T cells to apply forces through the TCR itself, rather than the CD3 coreceptor, was tested. Mouse cells expressing the 5C.C7 TCR exerted traction forces on pillars presenting peptide-loaded MHCs that were similar to those with α-CD3, suggesting that forces are applied to antigen-presenting cells during activation.


Subject(s)
CD28 Antigens/physiology , CD3 Complex/physiology , T-Lymphocytes/immunology , CD28 Antigens/immunology , CD3 Complex/immunology , Cells, Cultured , Humans
17.
Immunol Rev ; 251(1): 143-53, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23278746

ABSTRACT

Cell surface receptors bearing immunotyrosine-based inhibitory motifs (ITIMs) maintain natural killer (NK) cell tolerance to normal host tissues. These receptors are difficult to analyze mechanistically because they block activating responses in a rapid and comprehensive manner. The advent of high-resolution single cell imaging techniques has enabled investigators to explore the cell biological basis of the inhibitory response. Recent studies using these approaches indicate that ITIM-containing receptors function at least in part by structurally undermining the immunological synapse between the NK cell and its target. In this review, we discuss these new advances and how they might relate to what is known about the biochemistry of inhibitory signaling in NK cells and other cell types.


Subject(s)
Immunological Synapses/immunology , Killer Cells, Natural/immunology , Receptors, Cell Surface/immunology , Animals , Feedback, Physiological , Humans , Immune Tolerance , Signal Transduction/immunology , Single-Cell Analysis/methods
18.
Immunol Rev ; 256(1): 95-106, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24117815

ABSTRACT

In T lymphocytes, polarization of the microtubule-organizing center (MTOC) to the immunological synapse enables the directional secretion of cytokines, cytolytic factors, and other soluble molecules toward the antigen-presenting cell. This is likely to be crucial for maintaining the specificity of T-cell effector responses. Here, we review recent advances in our understanding of MTOC reorientation in T cells, focusing first on the importance of diacylglycerol and protein kinase C isozymes and then on the molecular motor proteins that function downstream to drive MTOC movement.


Subject(s)
Lipids/physiology , Microtubule-Organizing Center/physiology , Second Messenger Systems/physiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Diglycerides/metabolism , Dyneins/metabolism , Humans , Isoenzymes , Myosin Type II/metabolism , Protein Kinase C/metabolism
19.
Cell Mol Life Sci ; 72(3): 537-556, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25355055

ABSTRACT

Transient,specialized cell-cell interactions play a central role in leukocyte function by enabling specific intercellular communication in the context of a highly dynamic systems level response. The dramatic structural changes required for the formation of these contacts are driven by rapid and precise cytoskeletal remodeling events. In recent years, the immunological synapse that forms between a T lymphocyte and its antigen-presenting target cell has emerged as an important model system for understanding immune cell interactions. In this review, we discuss how regulators of the cortical actin cytoskeleton control synaptic architecture and in this way specify T cell function.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Communication/immunology , Immunological Synapses/immunology , Models, Immunological , Signal Transduction/immunology , T-Lymphocytes/cytology , Actins/metabolism , Humans , Phosphatidylinositol 3-Kinases/metabolism , T-Lymphocytes/immunology , rho GTP-Binding Proteins/metabolism
20.
Proc Natl Acad Sci U S A ; 110(29): 11976-81, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23818610

ABSTRACT

Centrosome reorientation to the immunological synapse maintains the specificity of T-cell effector function by facilitating the directional release of cytokines and cytolytic factors toward the antigen-presenting cell. This polarization response is driven by the localized accumulation of diacylglycerol, which recruits multiple protein kinase (PK)C isozymes to the synaptic membrane. Here, we used T-cell receptor (TCR) photoactivation and imaging methodology to demonstrate that PKCs control centrosome dynamics through the reciprocal localization of two motor complexes, dynein and nonmuscle myosin (NM)II. Dynein accumulated in the region of TCR stimulation, whereas NMII clustered in the back of the cell, behind the polarizing centrosome. PKC activity, which shaped both dynein and NMII accumulation within this framework, controlled NMII localization directly by phosphorylating inhibitory sites within the myosin regulatory light chain, thereby suppressing NMII clustering in the region of TCR stimulation. Concurrently, phosphorylation of distinct sites within myosin regulatory light chain by Rho kinase drove NMII clustering in areas behind the centrosome. These results reveal a role for NMII in T-cell polarity and demonstrate how it is regulated by upstream signals.


Subject(s)
Centrosome/physiology , Diglycerides/metabolism , Dyneins/metabolism , Immunological Synapses/immunology , Myosin Type II/metabolism , T-Lymphocytes/physiology , Animals , Immunoblotting , Immunohistochemistry , Mice , Mice, Transgenic , Phosphorylation , Protein Kinase C/metabolism , RNA, Small Interfering/genetics , Receptors, Antigen, T-Cell/metabolism , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL