Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
Add more filters










Publication year range
1.
Sci Signal ; 17(827): eade3643, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38470955

ABSTRACT

Activation of the endoplasmic reticulum (ER)-resident adaptor protein STING, a component of a cytosolic DNA-sensing pathway, induces the transcription of genes encoding type I interferons (IFNs) and other proinflammatory factors. Because STING is activated at the Golgi apparatus, control of the localization and activation of STING is important in stimulating antiviral and antitumor immune responses. Through a genome-wide CRISPR interference screen, we found that STING activation required the Golgi-resident protein ACBD3, which promotes the generation of phosphatidylinositol 4-phosphate (PI4P) at the trans-Golgi network, as well as other PI4P-associated proteins. Appropriate localization and activation of STING at the Golgi apparatus required ACBD3 and the PI4P-generating kinase PI4KB. In contrast, STING activation was enhanced when the lipid-shuttling protein OSBP, which removes PI4P from the Golgi apparatus, was inhibited by the US Food and Drug Administration-approved antifungal itraconazole. The increase in the abundance of STING-activating phospholipids at the trans-Golgi network resulted in the increased production of IFN-ß and other cytokines in THP-1 cells. Furthermore, a mutant STING that could not bind to PI4P failed to traffic from the ER to the Golgi apparatus in response to a STING agonist, whereas forced relocalization of STING to PI4P-enriched areas elicited STING activation in the absence of stimulation with a STING agonist. Thus, PI4P is critical for STING activation, and manipulating PI4P abundance may therapeutically modulate STING-dependent immune responses.


Subject(s)
Golgi Apparatus , Phospholipids , Phospholipids/metabolism , Golgi Apparatus/metabolism , Adaptor Proteins, Signal Transducing/metabolism
2.
Nat Rev Immunol ; 23(2): 90-105, 2023 02.
Article in English | MEDLINE | ID: mdl-35637393

ABSTRACT

Great strides have been made in recent years towards understanding the roles of natural killer (NK) cells in immunity to tumours and viruses. NK cells are cytotoxic innate lymphoid cells that produce inflammatory cytokines and chemokines. By lysing transformed or infected cells, they limit tumour growth and viral infections. Whereas T cells recognize peptides presented by MHC molecules, NK cells display receptors that recognize stress-induced autologous proteins on cancer cells. At the same time, their functional activity is inhibited by MHC molecules displayed on such cells. The enormous potential of NK cells for immunotherapy for cancer is illustrated by their broad recognition of stressed cells regardless of neoantigen presentation, and enhanced activity against tumours that have lost expression of MHC class I owing to acquired resistance mechanisms. As a result, many efforts are under way to mobilize endogenous NK cells with therapeutics, or to provide populations of ex vivo-expanded NK cells as a cellular therapy, in some cases by equipping the NK cells with chimeric antigen receptors. Here we consider the key features that underlie why NK cells are emerging as important new additions to the cancer therapeutic arsenal.


Subject(s)
Immunity, Innate , Neoplasms , Humans , Killer Cells, Natural , Neoplasms/therapy , T-Lymphocytes , Immunotherapy
3.
ACS Synth Biol ; 11(10): 3426-3439, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36169352

ABSTRACT

Natural killer (NK) cells are a major subset of innate immune cells that are essential for host defense against pathogens and cancer. Two main classes of inhibitory NK receptors (NKR), KIR and CD94/NKG2A, play a key role in suppressing NK activity upon engagement with tumor cells or virus-infected cells, limiting their antitumor and antiviral activities. Here, we find that single-chain NKR antagonists linked to a VHH that binds the cell surface phosphatase CD45 potentiate NK and T activities to a greater extent than NKR blocking antibodies alone in vitro. We also uncovered crosstalk between NKG2A and Ly49 that collectively inhibit NK cell activation, such that CD45-NKG2A and CD45-Ly49 bispecific molecules show synergistic effects in their ability to enhance NK cell activation. The basis of the activity enhancement by CD45 ligation may reflect greater antagonism of inhibitory signaling from engagement of MHC I on target cells, combined with other mechanisms, including avidity effects, tonic signaling, antagonism of weak inhibition from engagement of MHC I on non-target cells, and possible CD45 segregation within the NK cell-target cell synapse. These results uncover a strategy for enhancing the activity of NK and T cells that may improve cancer immunotherapies.


Subject(s)
NK Cell Lectin-Like Receptor Subfamily C , Receptors, Immunologic , Receptors, Natural Killer Cell , Antibodies, Blocking , Receptors, Immunologic/metabolism , Antigens, CD/metabolism , Antiviral Agents
4.
Elife ; 112022 05 26.
Article in English | MEDLINE | ID: mdl-35617021

ABSTRACT

Mitotically stable random monoallelic gene expression (RME) is documented for a small percentage of autosomal genes. We developed an in vivo genetic model to study the role of enhancers in RME using high-resolution single-cell analysis of natural killer (NK) cell receptor gene expression and enhancer deletions in the mouse germline. Enhancers of the RME NK receptor genes were accessible and enriched in H3K27ac on silent and active alleles alike in cells sorted according to allelic expression status, suggesting enhancer activation and gene expression status can be decoupled. In genes with multiple enhancers, enhancer deletion reduced gene expression frequency, in one instance converting the universally expressed gene encoding NKG2D into an RME gene, recapitulating all aspects of natural RME including mitotic stability of both the active and silent states. The results support the binary model of enhancer action, and suggest that RME is a consequence of general properties of gene regulation by enhancers rather than an RME-specific epigenetic program. Therefore, many and perhaps all genes may be subject to some degree of RME. Surprisingly, this was borne out by analysis of several genes that define different major hematopoietic lineages, that were previously thought to be universally expressed within those lineages: the genes encoding NKG2D, CD45, CD8α, and Thy-1. We propose that intrinsically probabilistic gene allele regulation is a general property of enhancer-controlled gene expression, with previously documented RME representing an extreme on a broad continuum.


Subject(s)
NK Cell Lectin-Like Receptor Subfamily K , Regulatory Sequences, Nucleic Acid , Alleles , Animals , Chromosomes , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Mice
5.
Proc Natl Acad Sci U S A ; 119(22): e2200568119, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35588144

ABSTRACT

Cyclic dinucleotides (CDN) and Toll-like receptor (TLR) ligands mobilize antitumor responses by natural killer (NK) cells and T cells, potentially serving as complementary therapies to immune checkpoint therapy. In the clinic thus far, however, CDN therapy targeting stimulator of interferon genes (STING) protein has yielded mixed results, perhaps because it initiates responses potently but does not provide signals to sustain activation and proliferation of activated cytotoxic lymphocytes. To improve efficacy, we combined CDN with a half life-extended interleukin-2 (IL-2) superkine, H9-MSA (mouse serum albumin). CDN/H9-MSA therapy induced dramatic long-term remissions of the most difficult to treat major histocompatibility complex class I (MHC I)­deficient and MHC I+ tumor transplant models. H9-MSA combined with CpG oligonucleotide also induced potent responses. Mechanistically, tumor elimination required CD8 T cells and not NK cells in the case of MHC I+ tumors and NK cells but not CD8 T cells in the case of MHC-deficient tumors. Furthermore, combination therapy resulted in more prolonged and more intense NK cell activation, cytotoxicity, and expression of cytotoxic effector molecules in comparison with monotherapy. Remarkably, in a primary autochthonous sarcoma model that is refractory to PD-1 checkpoint therapy, the combination of CDN/H9-MSA with checkpoint therapy yielded long-term remissions in the majority of the animals, mediated by T cells and NK cells. This combination therapy has the potential to activate responses in tumors resistant to current therapies and prevent MHC I loss accompanying acquired resistance of tumors to checkpoint therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Histocompatibility Antigens Class I , Immunotherapy , Interleukin-2 , Membrane Proteins , Neoplasms , Nucleotides, Cyclic , Oligodeoxyribonucleotides , Serum Albumin , Animals , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/genetics , Humans , Immunotherapy/methods , Interleukin-2/immunology , Killer Cells, Natural/immunology , Membrane Proteins/agonists , Mice , Neoplasms/genetics , Neoplasms/therapy , Nucleotides, Cyclic/therapeutic use , Oligodeoxyribonucleotides/therapeutic use , Serum Albumin/therapeutic use
6.
Dev Cell ; 56(19): 2712-2721.e4, 2021 10 11.
Article in English | MEDLINE | ID: mdl-34496290

ABSTRACT

Cancer patients often die from symptoms that manifest at a distance from any tumor. Mechanisms underlying these systemic physiological perturbations, called paraneoplastic syndromes, may benefit from investigation in non-mammalian systems. Using a non-metastatic Drosophila adult model, we find that malignant-tumor-produced cytokines drive widespread host activation of JAK-STAT signaling and cause premature lethality. STAT activity is particularly high in cells of the blood-brain barrier (BBB), where it induces aberrant BBB permeability. Remarkably, inhibiting STAT in the BBB not only rescues barrier function but also extends the lifespan of tumor-bearing hosts. We identify BBB damage in other pathological conditions that cause elevated inflammatory signaling, including obesity and infection, where BBB permeability also regulates host survival. IL-6-dependent BBB dysfunction is further seen in a mouse tumor model, and it again promotes host morbidity. Therefore, BBB alterations constitute a conserved lethal tumor-host interaction that also underlies other physiological morbidities.


Subject(s)
Blood-Brain Barrier/physiology , Paraneoplastic Syndromes/physiopathology , Animals , Biological Transport , Blood-Brain Barrier/metabolism , Cells, Cultured , Cytokines , Disease Models, Animal , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Endothelial Cells/metabolism , Interleukin-6/immunology , Mice , Mice, Inbred C57BL , Neoplasms/pathology , Permeability , STAT Transcription Factors/metabolism , Signal Transduction/physiology
7.
Cancer Cell ; 39(6): 725-729, 2021 06 14.
Article in English | MEDLINE | ID: mdl-34129817

ABSTRACT

The tumor immune microenvironment (TIME) is a complex ecosystem that contains adaptive and innate immune cells that have tumor-promoting and anti-tumor effects. There is still much to learn about the diversity, plasticity, and functions of innate immune cells in the TIME and their roles in determining the response to immunotherapies. Experts discuss recent advances in our understanding of their biology in cancer as well as outstanding questions and potential therapeutic avenues.


Subject(s)
Immunity, Innate , Immunotherapy/methods , Lymphocytes/immunology , Neoplasms/therapy , Tumor Microenvironment/immunology , Cancer Vaccines/pharmacology , Dendritic Cells/immunology , Humans , Killer Cells, Natural/immunology , Lymphocytes/pathology , Mast Cells/immunology , Mast Cells/pathology , Myeloid-Derived Suppressor Cells/immunology , Neoplasms/immunology , Neoplasms/pathology , Neutrophils/immunology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/pathology
8.
Sci Adv ; 6(50)2020 Dec.
Article in English | MEDLINE | ID: mdl-33298452

ABSTRACT

Thermography detects surface temperature and subsurface thermal activity of an object based on the Stefan-Boltzmann law. Impacts of the technology would be more far-reaching with finer thermal sensitivity, called noise-equivalent differential temperature (NEDT). Existing efforts to advance NEDT are all focused on improving registration of radiation signals with better cameras, driving the number close to the end of the roadmap at 20 to 40 mK. In this work, we take a distinct approach of sensitizing surface radiation against minute temperature variation of the object. The emissivity of the thermal imaging sensitizer (TIS) rises abruptly at a preprogrammed temperature, driven by a metal-insulator transition in cooperation with photonic resonance in the structure. The NEDT is refined by over 15 times with the TIS to achieve single-digit millikelvin resolution near room temperature, empowering ambient thermography for a broad range of applications such as in operando electronics analysis and early cancer screening.

9.
Science ; 368(6494): 943-944, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32467380
10.
Nature ; 579(7800): E12, 2020 03.
Article in English | MEDLINE | ID: mdl-32144410

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

11.
Sci Immunol ; 5(45)2020 03 20.
Article in English | MEDLINE | ID: mdl-32198222

ABSTRACT

Several immunotherapy approaches that mobilize CD8+ T cell responses stimulate tumor rejection, and some, such as checkpoint blockade, have been approved for several cancer indications and show impressive increases in patient survival. However, tumors may evade CD8+ T cell recognition via loss of MHC molecules or because they contain few or no neoantigens. Therefore, approaches are needed to combat CD8+ T cell-resistant cancers. STING-activating cyclic dinucleotides (CDNs) are a new class of immune-stimulating agents that elicit impressive CD8+ T cell-mediated tumor rejection in preclinical tumor models and are now being tested in clinical trials. Here, we demonstrate powerful CDN-induced, natural killer (NK) cell-mediated tumor rejection in numerous tumor models, independent of CD8+ T cells. CDNs enhanced NK cell activation, cytotoxicity, and antitumor effects in part by inducing type I interferon (IFN). IFN acted in part directly on NK cells in vivo and in part indirectly via the induction of IL-15 and IL-15 receptors, which were important for CDN-induced NK activation and tumor control. After in vivo administration of CDNs, dendritic cells (DCs) up-regulated IL-15Rα in an IFN-dependent manner. Mice lacking the type I IFN receptor specifically on DCs had reduced NK cell activation and tumor control. Therapeutics that activate NK cells, such as CDNs, checkpoint inhibitors, NK cell engagers, and cytokines, may represent next-generation approaches to cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , Membrane Proteins/agonists , Neoplasms/drug therapy , Neoplasms/immunology , Animals , CRISPR-Cas Systems/drug effects , CRISPR-Cas Systems/immunology , Interferon Type I/pharmacology , Killer Cells, Natural/drug effects , Membrane Proteins/immunology , Mice , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/pathology , Tumor Cells, Cultured
12.
Nature ; 573(7774): 434-438, 2019 09.
Article in English | MEDLINE | ID: mdl-31511694

ABSTRACT

The accumulation of DNA in the cytosol serves as a key immunostimulatory signal associated with infections, cancer and genomic damage1,2. Cytosolic DNA triggers immune responses by activating the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway3. The binding of DNA to cGAS activates its enzymatic activity, leading to the synthesis of a second messenger, cyclic guanosine monophosphate-adenosine monophosphate (2'3'-cGAMP)4-7. This cyclic dinucleotide (CDN) activates STING8, which in turn activates the transcription factors interferon regulatory factor 3 (IRF3) and nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), promoting the transcription of genes encoding type I interferons and other cytokines and mediators that stimulate a broader immune response. Exogenous 2'3'-cGAMP produced by malignant cells9 and other CDNs, including those produced by bacteria10-12 and synthetic CDNs used in cancer immunotherapy13,14, must traverse the cell membrane to activate STING in target cells. How these charged CDNs pass through the lipid bilayer is unknown. Here we used a genome-wide CRISPR-interference screen to identify the reduced folate carrier SLC19A1, a folate-organic phosphate antiporter, as the major transporter of CDNs. Depleting SLC19A1 in human cells inhibits CDN uptake and functional responses, and overexpressing SLC19A1 increases both uptake and functional responses. In human cell lines and primary cells ex vivo, CDN uptake is inhibited by folates as well as two medications approved for treatment of inflammatory diseases, sulfasalazine and the antifolate methotrexate. The identification of SLC19A1 as the major transporter of CDNs into cells has implications for the immunotherapeutic treatment of cancer13, host responsiveness to CDN-producing pathogenic microorganisms11 and-potentially-for some inflammatory diseases.


Subject(s)
DNA/metabolism , Nucleotides, Cyclic/metabolism , Reduced Folate Carrier Protein/metabolism , Animals , Cytosol , DNA/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interferon Regulatory Factor-3/metabolism , Nucleotides, Cyclic/immunology , Nucleotidyltransferases/metabolism , Reduced Folate Carrier Protein/immunology
13.
JCI Insight ; 52019 06 11.
Article in English | MEDLINE | ID: mdl-31184599

ABSTRACT

Cellular senescence is a tumor suppressive mechanism that can paradoxically contribute to aging pathologies. Despite evidence of immune clearance in mouse models, it is not known how senescent cells (SnCs) persist and accumulate with age or in tumors in individuals. Here, we identify cooperative mechanisms that orchestrate the immunoevasion and persistence of normal and cancer human SnCs through extracellular targeting of natural killer receptor signaling. Damaged SnCs avoid immune recognition through MMPs-dependent shedding of NKG2D-ligands reinforced via paracrine suppression of NKG2D receptor-mediated immunosurveillance. These coordinated immunoediting processes are evident in residual, drug-resistant tumors from cohorts of >700 prostate and breast cancer patients treated with senescence-inducing genotoxic chemotherapies. Unlike in mice, these reversible senescence-subversion mechanisms are independent of p53/p16 and exacerbated in oncogenic RAS-induced senescence. Critically, the p16INK4A tumor suppressor can disengage the senescence growth arrest from the damage-associated immune senescence program, which is manifest in benign nevi lesions where indolent SnCs accumulate over time and preserve a non-pro-inflammatory tissue microenvironment maintaining NKG2D-mediated immunosurveillance. Our study shows how subpopulations of SnCs elude immunosurveillance, and reveals secretome-targeted therapeutic strategies to selectively eliminate -and restore the clearance of- the detrimental SnCs that actively persist after chemotherapy and accumulate at sites of aging pathologies.


Subject(s)
Aging/immunology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cellular Senescence/immunology , Drug Resistance, Neoplasm/immunology , Prostatic Neoplasms/drug therapy , Tumor Escape/immunology , Aging/pathology , Animals , Antineoplastic Agents/therapeutic use , Biopsy , Breast/pathology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA Damage/drug effects , Datasets as Topic , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunologic Surveillance/drug effects , Immunologic Surveillance/immunology , Male , Metalloendopeptidases/metabolism , Mice , NK Cell Lectin-Like Receptor Subfamily K/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily K/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Prostate/pathology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Tissue Array Analysis , Tumor Escape/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
14.
Immunity ; 49(4): 754-763.e4, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30332631

ABSTRACT

Detection of cytosolic DNA by the enzyme cGAS triggers the production of cGAMP, a second messenger that binds and activates the adaptor protein STING, which leads to interferon (IFN) production. Here, we found that in vivo natural killer (NK) cell killing of tumor cells, but not of normal cells, depends on STING expression in non-tumor cells. Experiments using transplantable tumor models in STING- and cGAS-deficient mice revealed that cGAS expression by tumor cells was critical for tumor rejection by NK cells. In contrast, cGAS expression by host cells was dispensable, suggesting that tumor-derived cGAMP is transferred to non-tumor cells, where it activates STING. cGAMP administration triggered STING activation and IFN-ß production in myeloid cells and B cells but not NK cells. Our results reveal that the anti-tumor response of NK cells critically depends on the cytosolic DNA sensing pathway, similar to its role in defense against pathogens, and identify tumor-derived cGAMP as a major determinant of tumor immunogenicity with implications for cancer immunotherapy.


Subject(s)
Interferons/immunology , Killer Cells, Natural/immunology , Membrane Proteins/immunology , Neoplasms/immunology , Nucleotides, Cyclic/immunology , Animals , Cell Line , Cell Line, Tumor , Gene Expression Regulation/immunology , Humans , Interferons/metabolism , Killer Cells, Natural/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/genetics , Neoplasms/metabolism , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/immunology , Nucleotidyltransferases/metabolism , Signal Transduction/immunology
15.
J Clin Invest ; 128(10): 4654-4668, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30198904

ABSTRACT

Checkpoint blockade immunotherapy targeting the PD-1/PD-L1 inhibitory axis has produced remarkable results in the treatment of several types of cancer. Whereas cytotoxic T cells are known to provide important antitumor effects during checkpoint blockade, certain cancers with low MHC expression are responsive to therapy, suggesting that other immune cell types may also play a role. Here, we employed several mouse models of cancer to investigate the effect of PD-1/PD-L1 blockade on NK cells, a population of cytotoxic innate lymphocytes that also mediate antitumor immunity. We discovered that PD-1 and PD-L1 blockade elicited a strong NK cell response that was indispensable for the full therapeutic effect of immunotherapy. PD-1 was expressed on NK cells within transplantable, spontaneous, and genetically induced mouse tumor models, and PD-L1 expression in cancer cells resulted in reduced NK cell responses and generation of more aggressive tumors in vivo. PD-1 expression was more abundant on NK cells with an activated and more responsive phenotype and did not mark NK cells with an exhausted phenotype. These results demonstrate the importance of the PD-1/PD-L1 axis in inhibiting NK cell responses in vivo and reveal that NK cells, in addition to T cells, mediate the effect of PD-1/PD-L1 blockade immunotherapy.


Subject(s)
B7-H1 Antigen/immunology , Immunotherapy , Killer Cells, Natural/immunology , Neoplasms, Experimental/therapy , Programmed Cell Death 1 Receptor/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/genetics , Humans , K562 Cells , Killer Cells, Natural/pathology , Mice , Mice, Knockout , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics
16.
Elife ; 72018 05 14.
Article in English | MEDLINE | ID: mdl-29757143

ABSTRACT

NKG2D is an important immunoreceptor expressed on the surface of NK cells and some T cells. NKG2D recognizes a set of ligands typically expressed on infected or transformed cells, but recent studies have also documented NKG2D ligands on subsets of host non-tumor cells in tumor-bearing animals and humans. Here we show that in transplanted tumors and genetically engineered mouse cancer models, tumor-associated macrophages are induced to express the NKG2D ligand RAE-1δ. We find that a soluble factor produced by tumor cells is responsible for macrophage RAE-1δ induction, and we identify tumor-derived colony-stimulating factor-1 (CSF-1) as necessary and sufficient for macrophage RAE-1δ induction in vitro and in vivo. Furthermore, we show that induction of RAE-1δ on macrophages by CSF-1 requires PI3K p110α kinase signaling. Thus, production of CSF-1 by tumor cells leading to activation of PI3K p110α represents a novel cellular and molecular pathway mediating NKG2D ligand expression on tumor-associated macrophages.


Subject(s)
Macrophage Colony-Stimulating Factor/metabolism , Macrophages/immunology , Membrane Proteins/metabolism , Sarcoma/pathology , Animals , Cells, Cultured , Disease Models, Animal , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Neoplasm Transplantation , Signal Transduction
17.
Elife ; 62017 12 12.
Article in English | MEDLINE | ID: mdl-29231815

ABSTRACT

Natural Killer (NK) cells confer protection from tumors and infections by releasing cytotoxic granules and pro-inflammatory cytokines upon recognition of diseased cells. The responsiveness of NK cells to acute stimulation is dynamically tuned by steady-state receptor-ligand interactions of an NK cell with its cellular environment. Here, we demonstrate that in healthy WT mice the NK activating receptor NKG2D is engaged in vivo by one of its ligands, RAE-1ε, which is expressed constitutively by lymph node endothelial cells and highly induced on tumor-associated endothelium. This interaction causes internalization of NKG2D from the NK cell surface and transmits an NK-intrinsic signal that desensitizes NK cell responses globally to acute stimulation, resulting in impaired NK antitumor responses in vivo.


Subject(s)
Endothelium, Vascular/immunology , Killer Cells, Natural/immunology , Lymph Nodes/immunology , Melanoma, Experimental/immunology , Membrane Proteins/metabolism , Animals , Cells, Cultured , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K/metabolism
18.
Immunol Rev ; 280(1): 93-101, 2017 11.
Article in English | MEDLINE | ID: mdl-29027233

ABSTRACT

Natural killer (NK) cells recognize and kill cancer cells and infected cells by engaging cell surface ligands that are induced preferentially or exclusively on these cells. These ligands are recognized by activating receptors on NK cells, such as NKG2D. In addition to activation by cell surface ligands, the acquisition of optimal effector activity by NK cells is driven in vivo by cytokines and other signals. This review addresses a developing theme in NK cell biology: that NK-activating ligands on cells, and the provision of cytokines and other signals that drive high effector function in NK cells, are driven by abnormalities that arise from transformation or the infected state. The pathways include genomic damage, which causes self DNA to be exposed in the cytosol of affected cells, where it activates the DNA sensor cGAS. The resulting signaling induces NKG2D ligands and also mobilizes NK cell activation. Other key pathways that regulate NKG2D ligands include PI-3 kinase activation, histone acetylation, and the integrated stress response. This review summarizes the roles of these pathways and their relevance in both viral infections and cancer.


Subject(s)
Infections/immunology , Killer Cells, Natural/immunology , Neoplasms/immunology , Animals , Cytotoxicity, Immunologic , DNA Damage , Humans , Immunologic Surveillance , Lymphocyte Activation , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Nucleotidyltransferases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction
19.
Sci Immunol ; 2(7)2017 Jan 13.
Article in English | MEDLINE | ID: mdl-28783669

ABSTRACT

The immune system provides defense against tumors and pathogens. Here, we propose that by elucidating the shared principles of immunity that underlie cancer and infectious disease, oncologists and microbiologists can learn from each other and achieve the deeper mechanistic understanding critical the development of therapeutic approaches.

SELECTION OF CITATIONS
SEARCH DETAIL
...