Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters











Publication year range
1.
Cell Rep ; 42(12): 113506, 2023 12 26.
Article in English | MEDLINE | ID: mdl-38019655

ABSTRACT

Cross-presentation of dead cell-associated antigens by conventional dendritic cells type 1 (cDC1s) is critical for CD8+ T cells response against many tumors and viral infections. It is facilitated by DNGR-1 (CLEC9A), an SYK-coupled cDC1 receptor that detects dead cell debris. Here, we report that DNGR-1 engagement leads to rapid activation of CBL and CBL-B E3 ligases to cause K63-linked ubiquitination of SYK and terminate signaling. Genetic deletion of CBL E3 ligases or charge-conserved mutation of target lysines within SYK abolishes SYK ubiquitination and results in enhanced DNGR-1-dependent antigen cross-presentation. We also find that cDC1 deficient in CBL E3 ligases are more efficient at cross-priming CD8+ T cells to dead cell-associated antigens and promoting host resistance to tumors. Our findings reveal a role for CBL-dependent ubiquitination in limiting cross-presentation of dead cell-associated antigens and highlight an axis of negative regulation of cDC1 activity that could be exploited to increase anti-tumor immunity.


Subject(s)
Cross-Priming , Ubiquitin-Protein Ligases , CD8-Positive T-Lymphocytes , Proto-Oncogene Proteins c-cbl , Ubiquitination , Dendritic Cells , Syk Kinase
2.
Phys Rev Lett ; 130(14): 141901, 2023 Apr 07.
Article in English | MEDLINE | ID: mdl-37084446

ABSTRACT

The high-x data from the ZEUS Collaboration are used to extract parton density distributions of the proton deep in the perturbative regime of QCD. The data primarily constrain the up-quark valence distribution and new results are presented on its x dependence as well as on the momentum carried by the up quark. The results were obtained using Bayesian analysis methods which can serve as a model for future parton density extractions.

3.
Nat Commun ; 14(1): 122, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36653337

ABSTRACT

Large-scale events like the UEFA Euro 2020 football (soccer) championship offer a unique opportunity to quantify the impact of gatherings on the spread of COVID-19, as the number and dates of matches played by participating countries resembles a randomized study. Using Bayesian modeling and the gender imbalance in COVID-19 data, we attribute 840,000 (95% CI: [0.39M, 1.26M]) COVID-19 cases across 12 countries to the championship. The impact depends non-linearly on the initial incidence, the reproduction number R, and the number of matches played. The strongest effects are seen in Scotland and England, where as much as 10,000 primary cases per million inhabitants occur from championship-related gatherings. The average match-induced increase in R was 0.46 [0.18, 0.75] on match days, but important matches caused an increase as large as +3. Altogether, our results provide quantitative insights that help judge and mitigate the impact of large-scale events on pandemic spread.


Subject(s)
COVID-19 , Soccer , Humans , Bayes Theorem , COVID-19/epidemiology , England , Scotland
4.
J Immunother Cancer ; 10(9)2022 09.
Article in English | MEDLINE | ID: mdl-36162919

ABSTRACT

Type 1 conventional dendritic cells (cDC1) play a critical role in priming anticancer cytotoxic CD8+ T cells. DNGR-1 (a.k.a. CLEC9A) is a cDC1 receptor that binds to F-actin exposed on necrotic cancer and normal cells. DNGR-1 signaling enhances cross-presentation of dead-cell associated antigens, including tumor antigens. We have recently shown that secreted gelsolin (sGSN), a plasma protein, competes with DNGR-1 for binding to dead cell-exposed F-actin and dampens anticancer immunity. Here, we investigated the effects of loss of sGSN on various anticancer therapies that are thought to induce cell death and provoke an immune response to cancer. We compared WT (wildtype) with Rag1-/- , Batf3-/- , Clec9agfp/gfp , sGsn-/- or sGsn-/- Clec9agfp/gfp mice implanted with transplantable tumor cell lines, including MCA-205 fibrosarcoma, 5555 BrafV600E melanoma and B16-F10 LifeAct (LA)-ovalbumin (OVA)-mCherry melanoma. Tumor-bearing mice were treated with (1) doxorubicin (intratumoral) chemotherapy for MCA-205, (2) BRAF-inhibitor PLX4720 (oral gavage) targeted therapy for 5555 BrafV600E, and (3) X-ray radiotherapy for B16 LA-OVA-mCherry. We confirmed that efficient tumor control following each therapy requires an immunocompetent host as efficacy was markedly reduced in Rag1-/- compared with WT mice. Notably, across all the therapeutic modalities, loss of sGSN significantly enhanced tumor control compared with treated WT controls. This was an on-target effect as mice deficient in both sGSN and DNGR-1 behaved no differently from WT mice following therapy. In sum, we find that mice deficient in sGsn display enhanced DNGR-1-dependent responsiveness to chemotherapy, targeted therapy and radiotherapy. Our findings are consistent with the notion some cancer therapies induce immunogenic cell death (ICD), which mobilizes anticancer T cells. Our results point to cDC1 and DNGR-1 as decoders of ICD and to sGSN as a negative regulator of such decoding, highlighting sGSN as a possible target in cancer treatment. Further prospective studies are warranted to identify patients who may benefit most from inhibition of sGSN function.


Subject(s)
Gelsolin , Melanoma, Experimental , Actins/metabolism , Animals , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes , Doxorubicin/metabolism , Gelsolin/genetics , Gelsolin/metabolism , Homeodomain Proteins , Lectins, C-Type , Mice , Ovalbumin , Proto-Oncogene Proteins B-raf/metabolism , Receptors, Immunologic/metabolism
5.
Cell ; 184(15): 4016-4031.e22, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34081922

ABSTRACT

Cross-presentation of antigens from dead tumor cells by type 1 conventional dendritic cells (cDC1s) is thought to underlie priming of anti-cancer CD8+ T cells. cDC1 express high levels of DNGR-1 (a.k.a. CLEC9A), a receptor that binds to F-actin exposed by dead cell debris and promotes cross-presentation of associated antigens. Here, we show that secreted gelsolin (sGSN), an extracellular protein, decreases DNGR-1 binding to F-actin and cross-presentation of dead cell-associated antigens by cDC1s. Mice deficient in sGsn display increased DNGR-1-dependent resistance to transplantable tumors, especially ones expressing neoantigens associated with the actin cytoskeleton, and exhibit greater responsiveness to cancer immunotherapy. In human cancers, lower levels of intratumoral sGSN transcripts, as well as presence of mutations in proteins associated with the actin cytoskeleton, are associated with signatures of anti-cancer immunity and increased patient survival. Our results reveal a natural barrier to cross-presentation of cancer antigens that dampens anti-tumor CD8+ T cell responses.


Subject(s)
Cross-Priming/immunology , Gelsolin/metabolism , Immunity , Lectins, C-Type/metabolism , Neoplasms/immunology , Receptors, Immunologic/metabolism , Receptors, Mitogen/metabolism , Actins/metabolism , Amino Acid Sequence , Animals , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cross-Priming/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Gelsolin/chemistry , Gelsolin/deficiency , Gene Expression Regulation, Neoplastic/drug effects , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunity/drug effects , Mice, Inbred C57BL , Mutation/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Protein Binding/drug effects , Survival Analysis
7.
Nat Immunol ; 22(2): 140-153, 2021 02.
Article in English | MEDLINE | ID: mdl-33349708

ABSTRACT

Type 1 conventional dendritic (cDC1) cells are necessary for cross-presentation of many viral and tumor antigens to CD8+ T cells. cDC1 cells can be identified in mice and humans by high expression of DNGR-1 (also known as CLEC9A), a receptor that binds dead-cell debris and facilitates XP of corpse-associated antigens. Here, we show that DNGR-1 is a dedicated XP receptor that signals upon ligand engagement to promote phagosomal rupture. This allows escape of phagosomal contents into the cytosol, where they access the endogenous major histocompatibility complex class I antigen processing pathway. The activity of DNGR-1 maps to its signaling domain, which activates SYK and NADPH oxidase to cause phagosomal damage even when spliced into a heterologous receptor and expressed in heterologous cells. Our data reveal the existence of innate immune receptors that couple ligand binding to endocytic vesicle damage to permit MHC class I antigen presentation of exogenous antigens and to regulate adaptive immunity.


Subject(s)
Antigen Presentation , Cross-Priming , Dendritic Cells/metabolism , Lectins, C-Type/metabolism , Phagosomes/metabolism , Receptors, Immunologic/metabolism , Receptors, Mitogen/metabolism , T-Lymphocytes/metabolism , Animals , Cell Death , Coculture Techniques , Dendritic Cells/immunology , HEK293 Cells , Histocompatibility Antigens Class I/metabolism , Humans , Lectins, C-Type/genetics , Ligands , Mice , NADPH Oxidases/metabolism , Phagosomes/genetics , Phagosomes/immunology , Phosphorylation , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Receptors, Immunologic/genetics , Receptors, Mitogen/genetics , Signal Transduction , Syk Kinase/metabolism , T-Lymphocytes/immunology
8.
Am J Physiol Cell Physiol ; 319(2): C300-C312, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32520607

ABSTRACT

Skeletal muscle is a target of contraction-induced loading (CiL), leading to protein unfolding or cellular perturbations, respectively. While cytoskeletal desmin is responsible for ongoing structural stabilization, in the immediate response to CiL, alpha-crystallin B (CRYAB) is phosphorylated at serine 59 (pCRYABS59) by P38, acutely protecting the cytoskeleton. To reveal adaptation and deadaptation of these myofibrillar subsystems to CiL, we examined CRYAB, P38, and desmin regulation following resistance exercise at diverse time points of a chronic training period. Mechanosensitive JNK phosphorylation (pJNKT183/Y185) was determined to indicate the presence of mechanical components in CiL. Within 6 wk, subjects performed 13 resistance exercise bouts at the 8-12 repetition maximum, followed by 10 days detraining and a final 14th bout. Biopsies were taken at baseline and after the 1st, 3rd, 7th, 10th, 13th, and 14th bout. To assess whether potential desensitization to CiL can be mitigated, one group trained with progressive and a second with constant loading. As no group differences were found, all subjects were combined for statistics. Total and phosphorylated P38 was not regulated over the time course. pCRYABS59 and pJNKT183/Y185 strongly increased following the unaccustomed first bout. This exercise-induced pCRYABS59/pJNKT183/Y185 increase disappeared with the 10th until 13th bout. As response to the detraining period, the 14th bout led to a renewed increase in pCRYABS59. Desmin content followed pCRYABS59 inversely, i.e., was up- when pCRYABS59 was downregulated and vice versa. In conclusion, the pCRYABS59 response indicates increase and decrease in resistance to CiL, in which a reinforced desmin network could play an essential role by structurally stabilizing the cells.


Subject(s)
Adaptation, Physiological/genetics , Desmin/genetics , Muscle, Skeletal/metabolism , alpha-Crystallin B Chain/genetics , Adult , Cytoskeleton/genetics , Cytoskeleton/metabolism , Desmin/metabolism , Gene Expression Regulation/genetics , Humans , Male , Muscle Contraction/physiology , Muscle, Skeletal/physiology , Phosphorylation/genetics , Resistance Training/adverse effects , Young Adult , alpha-Crystallin B Chain/metabolism
9.
Cancer Cell Int ; 19: 346, 2019.
Article in English | MEDLINE | ID: mdl-31889898

ABSTRACT

BACKGROUND: Treatments that generate T cell-mediated immunity to a patient's unique neoantigens are the current holy grail of cancer immunotherapy. In particular, treatments that do not require cumbersome and individualized ex vivo processing or manufacturing processes are especially sought after. Here we report that AGI-134, a glycolipid-like small molecule, can be used for coating tumor cells with the xenoantigen Galα1-3Galß1-4GlcNAc (α-Gal) in situ leading to opsonization with pre-existing natural anti-α-Gal antibodies (in short anti-Gal), which triggers immune cascades resulting in T cell mediated anti-tumor immunity. METHODS: Various immunological effects of coating tumor cells with α-Gal via AGI-134 in vitro were measured by flow cytometry: (1) opsonization with anti-Gal and complement, (2) antibody-dependent cell-mediated cytotoxicity (ADCC) by NK cells, and (3) phagocytosis and antigen cross-presentation by antigen presenting cells (APCs). A viability kit was used to test AGI-134 mediated complement dependent cytotoxicity (CDC) in cancer cells. The anti-tumoral activity of AGI-134 alone or in combination with an anti-programmed death-1 (anti-PD-1) antibody was tested in melanoma models in anti-Gal expressing galactosyltransferase knockout (α1,3GT-/-) mice. CDC and phagocytosis data were analyzed by one-way ANOVA, ADCC results by paired t-test, distal tumor growth by Mantel-Cox test, C5a data by Mann-Whitney test, and single tumor regression by repeated measures analysis. RESULTS: In vitro, α-Gal labelling of tumor cells via AGI-134 incorporation into the cell membrane leads to anti-Gal binding and complement activation. Through the effects of complement and ADCC, tumor cells are lysed and tumor antigen uptake by APCs increased. Antigen associated with lysed cells is cross-presented by CD8α+ dendritic cells leading to activation of antigen-specific CD8+ T cells. In B16-F10 or JB/RH melanoma models in α1,3GT-/- mice, intratumoral AGI-134 administration leads to primary tumor regression and has a robust abscopal effect, i.e., it protects from the development of distal, uninjected lesions. Combinations of AGI-134 and anti-PD-1 antibody shows a synergistic benefit in protection from secondary tumor growth. CONCLUSIONS: We have identified AGI-134 as an immunotherapeutic drug candidate, which could be an excellent combination partner for anti-PD-1 therapy, by facilitating tumor antigen processing and increasing the repertoire of tumor-specific T cells prior to anti-PD-1 treatment.

10.
Sci Immunol ; 3(30)2018 12 07.
Article in English | MEDLINE | ID: mdl-30530727

ABSTRACT

Ectopic expression of transcription factors has been used to reprogram differentiated somatic cells toward pluripotency or to directly reprogram them to other somatic cell lineages. This concept has been explored in the context of regenerative medicine. Here, we set out to generate dendritic cells (DCs) capable of presenting antigens from mouse and human fibroblasts. By screening combinations of 18 transcription factors that are expressed in DCs, we have identified PU.1, IRF8, and BATF3 transcription factors as being sufficient to reprogram both mouse and human fibroblasts to induced DCs (iDCs). iDCs acquire a conventional DC type 1-like transcriptional program, with features of interferon-induced maturation. iDCs secrete inflammatory cytokines and have the ability to engulf, process, and present antigens to T cells. Furthermore, we demonstrate that murine iDCs generated here were able to cross-present antigens to CD8+ T cells. Our reprogramming system should facilitate better understanding of DC specification programs and serve as a platform for the development of patient-specific DCs for immunotherapy.


Subject(s)
Antigen-Presenting Cells/immunology , Fibroblasts/immunology , Animals , Cells, Cultured , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
11.
Science ; 362(6412): 351-356, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30337411

ABSTRACT

Host injury triggers feedback mechanisms that limit tissue damage. Conventional type 1 dendritic cells (cDC1s) express dendritic cell natural killer lectin group receptor-1 (DNGR-1), encoded by the gene Clec9a, which senses tissue damage and favors cross-presentation of dead-cell material to CD8+ T cells. Here we find that DNGR-1 additionally reduces host-damaging inflammatory responses induced by sterile and infectious tissue injury in mice. DNGR-1 deficiency leads to exacerbated caerulein-induced necrotizing pancreatitis and increased pathology during systemic Candida albicans infection without affecting fungal burden. This effect is B and T cell-independent and attributable to increased neutrophilia in DNGR-1-deficient settings. Mechanistically, DNGR-1 engagement activates SHP-1 and inhibits MIP-2 (encoded by Cxcl2) production by cDC1s during Candida infection. This consequently restrains neutrophil recruitment and promotes disease tolerance. Thus, DNGR-1-mediated sensing of injury by cDC1s serves as a rheostat for the control of tissue damage, innate immunity, and immunopathology.


Subject(s)
Candida albicans/immunology , Candidiasis/pathology , Dendritic Cells/immunology , Lectins, C-Type/physiology , Neutrophil Infiltration/immunology , Pancreas/pathology , Pancreatitis, Acute Necrotizing/pathology , Receptors, Immunologic/physiology , Animals , Lectins, C-Type/genetics , Mice , Mice, Mutant Strains , Necrosis , Neutrophil Infiltration/genetics , Pancreas/immunology , Pancreas/microbiology , Pancreatitis, Acute Necrotizing/microbiology , Receptors, Immunologic/genetics
12.
Cell Rep ; 24(2): 419-428, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29996102

ABSTRACT

Conventional type 1 DCs (cDC1s) excel at cross-presentation of dead cell-associated antigens partly because they express DNGR-1, a receptor that recognizes exposed actin filaments on dead cells. In vitro polymerized F-actin can be used as a synthetic ligand for DNGR-1. However, cellular F-actin is decorated with actin-binding proteins, which could affect DNGR-1 recognition. Here, we demonstrate that myosin II, an F-actin-associated motor protein, greatly potentiates the binding of DNGR-1 to F-actin. Latex beads coated with F-actin and myosin II are taken up by DNGR-1+ cDC1s, and antigen associated with those beads is efficiently cross-presented to CD8+ T cells. Myosin II-deficient necrotic cells are impaired in their ability to stimulate DNGR-1 or to serve as substrates for cDC1 cross-presentation to CD8+ T cells. These results provide insights into the nature of the DNGR-1 ligand and have implications for understanding immune responses to cell-associated antigens and for vaccine design.


Subject(s)
Actins/metabolism , Antigens/metabolism , Cross-Priming/immunology , Lectins, C-Type/metabolism , Myosin Type II/metabolism , Receptors, Immunologic/metabolism , Animals , Cell Death , Cell Line , Immunization , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Myosin Heavy Chains/metabolism , Phagocytosis , Protein Binding , T-Lymphocytes, Cytotoxic/immunology
13.
EMBO J ; 35(22): 2484-2497, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27753620

ABSTRACT

DNGR-1 is receptor expressed by certain dendritic cell (DC) subsets and by DC precursors in mouse. It possesses a C-type lectin-like domain (CTLD) followed by a poorly characterized neck region coupled to a transmembrane region and short intracellular tail. The CTLD of DNGR-1 binds F-actin exposed by dead cell corpses and causes the receptor to signal and potentiate cross-presentation of dead cell-associated antigens by DCs. Here, we describe a conformational change that occurs in the neck region of DNGR-1 in a pH- and ionic strength-dependent manner and that controls cross-presentation of dead cell-associated antigens. We identify residues in the neck region that, when mutated, lock DNGR-1 in one of the two conformational states to potentiate cross-presentation. In contrast, we show that chimeric proteins in which the neck region of DNGR-1 is replaced by that of unrelated C-type lectin receptors fail to promote cross-presentation. Our results suggest that the neck region of DNGR-1 is an integral receptor component that senses receptor progression through the endocytic pathway and has evolved to maximize extraction of antigens from cell corpses, coupling DNGR-1 function to its cellular localization.


Subject(s)
Dendritic Cells/metabolism , Hydrogen-Ion Concentration , Lectins, C-Type/chemistry , Lectins, C-Type/metabolism , Osmolar Concentration , Protein Conformation/drug effects , Receptors, Immunologic/chemistry , Receptors, Immunologic/metabolism , Allosteric Regulation , Animals , DNA Mutational Analysis , Lectins, C-Type/genetics , Mice , Receptors, Immunologic/genetics
14.
Science ; 350(6258): 328-34, 2015 Oct 16.
Article in English | MEDLINE | ID: mdl-26405229

ABSTRACT

Dying cells initiate adaptive immunity by providing both antigens and inflammatory stimuli for dendritic cells, which in turn activate CD8(+) T cells through a process called antigen cross-priming. To define how different forms of programmed cell death influence immunity, we established models of necroptosis and apoptosis, in which dying cells are generated by receptor-interacting protein kinase-3 and caspase-8 dimerization, respectively. We found that the release of inflammatory mediators, such as damage-associated molecular patterns, by dying cells was not sufficient for CD8(+) T cell cross-priming. Instead, robust cross-priming required receptor-interacting protein kinase-1 (RIPK1) signaling and nuclear factor κB (NF-κB)-induced transcription within dying cells. Decoupling NF-κB signaling from necroptosis or inflammatory apoptosis reduced priming efficiency and tumor immunity. Our results reveal that coordinated inflammatory and cell death signaling pathways within dying cells orchestrate adaptive immunity.


Subject(s)
Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , NF-kappa B/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Caspase 8/metabolism , Cell Survival , Cross-Priming , Dendritic Cells/immunology , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction
15.
Immunity ; 42(5): 839-849, 2015 05 19.
Article in English | MEDLINE | ID: mdl-25979418

ABSTRACT

DNGR-1 is a C-type lectin receptor that binds F-actin exposed by dying cells and facilitates cross-presentation of dead cell-associated antigens by dendritic cells. Here we present the structure of DNGR-1 bound to F-actin at 7.7 Å resolution. Unusually for F-actin binding proteins, the DNGR-1 ligand binding domain contacts three actin subunits helically arranged in the actin filament, bridging over two protofilaments, as well as two neighboring actin subunits along one protofilament. Mutation of residues predicted to mediate ligand binding led to loss of DNGR-1-dependent cross-presentation of dead cell-associated antigens, formally demonstrating that the latter depends on F-actin recognition. Notably, DNGR-1 has relatively modest affinity for F-actin but multivalent interactions allow a marked increase in binding strength. Our findings shed light on modes of actin binding by cellular proteins and reveal how extracellular detection of cytoskeletal components by dedicated receptors allows immune monitoring of loss of cellular integrity.


Subject(s)
Actins/chemistry , Cross-Priming , Dendritic Cells/immunology , Lectins, C-Type/chemistry , Models, Molecular , Receptors, Immunologic/chemistry , Actins/metabolism , Animals , Cells, Cultured , Humans , Mice , Mutation , Protein Binding
16.
J Clin Invest ; 122(5): 1615-27, 2012 May.
Article in English | MEDLINE | ID: mdl-22505458

ABSTRACT

DNGR-1 (CLEC9A) is a receptor for necrotic cells required by DCs to cross-prime CTLs against dead cell antigens in mice. It is currently unknown how DNGR-1 couples dead cell recognition to cross-priming. Here we found that DNGR-1 did not mediate DC activation by dead cells but rather diverted necrotic cell cargo into a recycling endosomal compartment, favoring cross-presentation to CD8(+) T cells. DNGR-1 regulated cross-priming in non-infectious settings such as immunization with antigen-bearing dead cells, as well as in highly immunogenic situations such as infection with herpes simplex virus type 1. Together, these results suggest that DNGR-1 is a dedicated receptor for cross-presentation of cell-associated antigens. Our work thus underscores the importance of cross-priming in immunity and indicates that antigenicity and adjuvanticity can be decoded by distinct innate immune receptors. The identification of specialized receptors that regulate antigenicity of virus-infected cells reveals determinants of antiviral immunity that might underlie the human response to infection and vaccination.


Subject(s)
Antigens, Surface/immunology , Cross-Priming , Endocytosis , Herpes Simplex/immunology , Lectins, C-Type/physiology , Necrosis/metabolism , Receptors, Immunologic/physiology , Alphavirus Infections/immunology , Alphavirus Infections/pathology , Animals , Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/physiology , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/pathology , Herpes Simplex/pathology , Humans , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Lung/immunology , Lung/pathology , Lung/virology , Mice , Mice, Inbred C57BL , Myeloid Cells/immunology , Myeloid Cells/physiology , Necrosis/virology , Ovalbumin/immunology , Ovalbumin/metabolism , Protein Transport/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Toll-Like Receptor 3/metabolism
17.
Immunity ; 36(4): 635-45, 2012 Apr 20.
Article in English | MEDLINE | ID: mdl-22483800

ABSTRACT

Sterile inflammation can be initiated by innate immune recognition of markers of tissue injury termed damage-associated molecular patterns (DAMPs). DAMP recognition by dendritic cells (DCs) has also been postulated to lead to T cell responses to foreign antigens in tumors or allografts. Many DAMPs represent intracellular contents that are released upon cell damage, notably after necrosis. In this regard, we have previously described DNGR-1 (CLEC9A) as a DC-restricted receptor specific for an unidentified DAMP that is exposed by necrotic cells and is necessary for efficient priming of cytotoxic T cells against dead cell-associated antigens. Here, we have shown that the DNGR-1 ligand is preserved from yeast to man and corresponds to the F-actin component of the cellular cytoskeleton. The identification of F-actin as a DNGR-1 ligand suggests that cytoskeletal exposure is a universal sign of cell damage that can be targeted by the innate immune system to initiate immunity.


Subject(s)
Actins/metabolism , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Necrosis/metabolism , Receptors, Mitogen/immunology , Receptors, Mitogen/metabolism , Saccharomyces cerevisiae Proteins/genetics , Actin Cytoskeleton/metabolism , Actins/genetics , Dendritic Cells/metabolism , HeLa Cells , Humans , Immunity, Innate , Necrosis/immunology , RNA Interference , RNA, Small Interfering , Saccharomyces cerevisiae/genetics , T-Lymphocytes, Cytotoxic/immunology
18.
Mol Nutr Food Res ; 54 Suppl 2: S141-50, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20540146

ABSTRACT

Advanced glycation endproducts (AGEs) accumulate on protein deposits including the beta-amyloid plaques in Alzheimer's disease. AGEs interact with the "receptor for advanced glycation endproducts", and transmit their signals using intracellular reactive oxygen species as second messengers. Ultimately, AGEs induce the expression of a variety of pro-inflammatory markers including the tumor necrosis factor (TNF-alpha) and inducible nitric oxide (NO) synthase. Antioxidants that act intracellularly, including polyphenols, have been shown to scavenge these "signaling" reactive oxygen species, and thus perform in an anti-inflammatory capacity. This study tested the pure compounds apigenin and diosmetin as well as extracts from silymarin, uva ursi (bearberry) and green olive leaf for their ability to attenuate AGE-induced NO and TNF-alpha production. All five tested samples inhibited BSA-AGE-induced NO production in a dose-dependent manner. Apigenin and diosmetin were most potent, and exhibited EC(50) values approximately 10 microM. In contrast, TNF-alpha expression was only reduced by apigenin, diosmetin and silymarin; not by the bearberry and green olive leaf extracts. In addition, the silymarin and bearberry extracts caused significant cell death at concentrations >or=10 microg/mL and >or=50 microg/mL, respectively. In conclusion, we suggest that plant-derived polyphenols might offer therapeutic opportunities to delay the progression of AGE-mediated and receptor for advanced glycation endproducts-mediated neuro-inflammatory diseases including Alzheimer's disease.


Subject(s)
Antioxidants/pharmacology , Flavonoids/pharmacology , Glycation End Products, Advanced/pharmacology , Nitric Oxide/metabolism , Phenols/pharmacology , Plant Extracts/pharmacology , Serum Albumin, Bovine/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Alzheimer Disease/prevention & control , Animals , Apigenin/pharmacology , Apigenin/toxicity , Arctostaphylos/chemistry , Cell Line , Cognition Disorders/prevention & control , Flavonoids/toxicity , Glycation End Products, Advanced/metabolism , Inhibitory Concentration 50 , Mice , Microglia/drug effects , Microglia/metabolism , Olea/chemistry , Osmolar Concentration , Phenols/toxicity , Plant Extracts/toxicity , Plant Leaves/chemistry , Polyphenols , Receptor for Advanced Glycation End Products , Receptors, Immunologic/metabolism , Serum Albumin, Bovine/metabolism , Signal Transduction/drug effects , Silymarin/analysis
19.
Cell Host Microbe ; 7(5): 354-61, 2010 May 20.
Article in English | MEDLINE | ID: mdl-20478537

ABSTRACT

Cytosolic viral RNA recognition by the helicases RIG-I and MDA5 is considered the major pathway for IFN-alpha/beta induction in response to RNA viruses. However, other cytoplasmic RNA sensors, including the double-stranded RNA-binding protein kinase R (PKR), have been implicated in IFN-alpha/beta production, although their relative contribution and mechanism have been unclear. Using cells expressing nonfunctional PKR or reduced levels of kinase, we show that PKR is required for production of IFN-alpha/beta proteins in response to a subset of RNA viruses including encephalomyocarditis, Theiler's murine encephalomyelitis, and Semliki Forest virus, but not influenza or Sendai virus. Surprisingly, although IFN-alpha/beta mRNA induction is largely normal in PKR-deficient cells, much of that mRNA lacks the poly(A) tail, indicating that its integrity is compromised. Our results suggest that PKR plays a nonredundant role in IFN-alpha/beta production in response to some but not all viruses, in part by regulating IFN-alpha/beta mRNA stability.


Subject(s)
Encephalomyocarditis virus/immunology , Interferon-alpha/biosynthesis , Interferon-beta/biosynthesis , RNA, Messenger/metabolism , Semliki forest virus/immunology , Theilovirus/immunology , eIF-2 Kinase/immunology , Animals , Mice , Mice, Knockout , Orthomyxoviridae/immunology , Sendai virus/immunology , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL