Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Cell ; 187(6): 1363-1373.e12, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38366591

ABSTRACT

In response to the 2022 outbreak of mpox driven by unprecedented human-to-human monkeypox virus (MPXV) transmission, we designed BNT166, aiming to create a highly immunogenic, safe, accessible, and scalable next-generation vaccine against MPXV and related orthopoxviruses. To address the multiple viral forms and increase the breadth of immune response, two candidate multivalent mRNA vaccines were evaluated pre-clinically: a quadrivalent vaccine (BNT166a; encoding the MPXV antigens A35, B6, M1, H3) and a trivalent vaccine (BNT166c; without H3). Both candidates induced robust T cell responses and IgG antibodies in mice, including neutralizing antibodies to both MPXV and vaccinia virus. In challenge studies, BNT166a and BNT166c provided complete protection from vaccinia, clade I, and clade IIb MPXV. Furthermore, immunization with BNT166a was 100% effective at preventing death and at suppressing lesions in a lethal clade I MPXV challenge in cynomolgus macaques. These findings support the clinical evaluation of BNT166, now underway (NCT05988203).


Subject(s)
Monkeypox virus , Mpox (monkeypox) , Smallpox Vaccine , Animals , Humans , Mice , Macaca fascicularis , Monkeypox virus/genetics , Mpox (monkeypox)/immunology , Mpox (monkeypox)/prevention & control , Vaccines, Combined , Vaccinia virus/genetics
2.
Cell ; 150(3): 606-19, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22819539

ABSTRACT

Systemic infections with Gram-negative bacteria are characterized by high mortality rates due to the "sepsis syndrome," a widespread and uncontrolled inflammatory response. Though it is well recognized that the immune response during Gram-negative bacterial infection is initiated after the recognition of endotoxin by Toll-like receptor 4, the molecular mechanisms underlying the detrimental inflammatory response during Gram-negative bacteremia remain poorly defined. Here, we identify a TRIF pathway that licenses NLRP3 inflammasome activation by all Gram-negative bacteria. By engaging TRIF, Gram-negative bacteria activate caspase-11. TRIF activates caspase-11 via type I IFN signaling, an event that is both necessary and sufficient for caspase-11 induction and autoactivation. Caspase-11 subsequently synergizes with the assembled NLRP3 inflammasome to regulate caspase-1 activation and leads to caspase-1-independent cell death. These events occur specifically during infection with Gram-negative, but not Gram-positive, bacteria. The identification of TRIF as a regulator of caspase-11 underscores the importance of TLRs as master regulators of inflammasomes during Gram-negative bacterial infection.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Caspases/metabolism , Citrobacter rodentium/metabolism , Enterohemorrhagic Escherichia coli/metabolism , Inflammasomes/metabolism , Interferons/metabolism , Animals , Carrier Proteins/metabolism , Caspases, Initiator , Citrobacter rodentium/immunology , Enterohemorrhagic Escherichia coli/immunology , Gram-Negative Bacteria/immunology , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/immunology , Gram-Positive Bacteria/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction
3.
Nat Immunol ; 14(6): 543-53, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23644505

ABSTRACT

Phagocytosis is a fundamental cellular process that is pivotal for immunity as it coordinates microbial killing, innate immune activation and antigen presentation. An essential step in this process is phagosome acidification, which regulates many functions of these organelles that allow phagosomes to participate in processes that are essential to both innate and adaptive immunity. Here we report that acidification of phagosomes containing Gram-positive bacteria is regulated by the NLRP3 inflammasome and caspase-1. Active caspase-1 accumulates on phagosomes and acts locally to control the pH by modulating buffering by the NADPH oxidase NOX2. These data provide insight into a mechanism by which innate immune signals can modify cellular defenses and establish a new function for the NLRP3 inflammasome and caspase-1 in host defense.


Subject(s)
Carrier Proteins/immunology , Caspase 1/immunology , Inflammasomes/immunology , Membrane Glycoproteins/immunology , NADPH Oxidases/immunology , Phagosomes/immunology , Animals , Carrier Proteins/metabolism , Caspase 1/metabolism , Cells, Cultured , Enzyme Activation/immunology , Flow Cytometry , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Hydrogen-Ion Concentration , Immunoblotting , Inflammasomes/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Membrane Glycoproteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , Microscopy, Electron , NADPH Oxidase 2 , NADPH Oxidases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Phagocytosis/immunology , Phagosomes/metabolism , Phagosomes/microbiology , Phagosomes/ultrastructure , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Staphylococcus aureus/immunology , Staphylococcus aureus/physiology
4.
Nat Immunol ; 14(8): 812-20, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23812099

ABSTRACT

Particulate ligands, including cholesterol crystals and amyloid fibrils, induce production of interleukin 1ß (IL-1ß) dependent on the cytoplasmic sensor NLRP3 in atherosclerosis, Alzheimer's disease and diabetes. Soluble endogenous ligands, including oxidized low-density lipoprotein (LDL), amyloid-ß and amylin peptides, accumulate in such diseases. Here we identify an endocytic pathway mediated by the pattern-recognition receptor CD36 that coordinated the intracellular conversion of those soluble ligands into crystals or fibrils, which resulted in lysosomal disruption and activation of the NLRP3 inflammasome. Consequently, macrophages that lacked CD36 failed to elicit IL-1ß production in response to those ligands, and targeting CD36 in atherosclerotic mice resulted in lower serum concentrations of IL-1ß and accumulation of cholesterol crystals in plaques. Collectively, our findings highlight the importance of CD36 in the accrual and nucleation of NLRP3 ligands from within the macrophage and position CD36 as a central regulator of inflammasome activation in sterile inflammation.


Subject(s)
Alzheimer Disease/immunology , Atherosclerosis/immunology , CD36 Antigens/immunology , Carrier Proteins/immunology , Diabetes Mellitus, Type 2/immunology , Inflammation/immunology , Animals , CD36 Antigens/genetics , Carrier Proteins/genetics , Inflammasomes/immunology , Interleukin-1beta/immunology , Lipoproteins, LDL/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Fluorescence , NLR Family, Pyrin Domain-Containing 3 Protein , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction
5.
Nat Immunol ; 13(2): 136-43, 2012 Jan 08.
Article in English | MEDLINE | ID: mdl-22231519

ABSTRACT

Atherosclerotic plaque formation is fueled by the persistence of lipid-laden macrophages in the artery wall. The mechanisms by which these cells become trapped, thereby establishing chronic inflammation, remain unknown. Here we found that netrin-1, a neuroimmune guidance cue, was secreted by macrophages in human and mouse atheroma, where it inactivated the migration of macrophages toward chemokines linked to their egress from plaques. Acting via its receptor, UNC5b, netrin-1 inhibited the migration of macrophages directed by the chemokines CCL2 and CCL19, activation of the actin-remodeling GTPase Rac1 and actin polymerization. Targeted deletion of netrin-1 in macrophages resulted in much less atherosclerosis in mice deficient in the receptor for low-density lipoprotein and promoted the emigration of macrophages from plaques. Thus, netrin-1 promoted atherosclerosis by retaining macrophages in the artery wall. Our results establish a causative role for negative regulators of leukocyte migration in chronic inflammation.


Subject(s)
Atherosclerosis/immunology , Cell Movement/immunology , Macrophages/immunology , Nerve Growth Factors/metabolism , Plaque, Atherosclerotic/immunology , Tumor Suppressor Proteins/metabolism , Actins/metabolism , Animals , Cells, Cultured , Chemokine CCL19/metabolism , Chemokine CCL2/metabolism , Chimera/metabolism , Gene Deletion , Humans , Mice , Nerve Growth Factors/genetics , Netrin Receptors , Netrin-1 , Neuropeptides/metabolism , Polymerization , Receptors, Cell Surface/metabolism , Tumor Suppressor Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein/metabolism
6.
Immunity ; 40(6): 896-909, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24882217

ABSTRACT

Animal host defense against infection requires the expression of defense genes at the right place and the right time. Understanding such tight control of host defense requires the elucidation of the transcription factors involved. By using an unbiased approach in the model Caenorhabditis elegans, we discovered that HLH-30 (known as TFEB in mammals) is a key transcription factor for host defense. HLH-30 was activated shortly after Staphylococcus aureus infection, and drove the expression of close to 80% of the host response, including antimicrobial and autophagy genes that were essential for host tolerance of infection. TFEB was also rapidly activated in murine macrophages upon S. aureus infection and was required for proper transcriptional induction of several proinflammatory cytokines and chemokines. Thus, our data suggest that TFEB is a previously unappreciated, evolutionarily ancient transcription factor in the host response to infection.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/immunology , Basic Helix-Loop-Helix Transcription Factors/immunology , Caenorhabditis elegans Proteins/immunology , Caenorhabditis elegans/immunology , Caenorhabditis elegans/microbiology , Staphylococcal Infections/immunology , Animals , Autophagy/genetics , Autophagy/immunology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Caenorhabditis elegans Proteins/genetics , Enterococcus faecalis/immunology , Immunity, Innate , Macrophages/immunology , Mice , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , RNA Interference , RNA, Small Interfering , Salmonella Infections/immunology , Salmonella enterica/immunology , Signal Transduction/immunology , Staphylococcus aureus/immunology , Transcriptional Activation/genetics , Transcriptional Activation/immunology
8.
Nat Immunol ; 11(2): 155-61, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20037584

ABSTRACT

In atherosclerosis and Alzheimer's disease, deposition of the altered self components oxidized low-density lipoprotein (LDL) and amyloid-beta triggers a protracted sterile inflammatory response. Although chronic stimulation of the innate immune system is believed to underlie the pathology of these diseases, the molecular mechanisms of activation remain unclear. Here we show that oxidized LDL and amyloid-beta trigger inflammatory signaling through a heterodimer of Toll-like receptors 4 and 6. Assembly of this newly identified heterodimer is regulated by signals from the scavenger receptor CD36, a common receptor for these disparate ligands. Our results identify CD36-TLR4-TLR6 activation as a common molecular mechanism by which atherogenic lipids and amyloid-beta stimulate sterile inflammation and suggest a new model of TLR heterodimerization triggered by coreceptor signaling events.


Subject(s)
CD36 Antigens/immunology , Inflammation/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 6/immunology , Amyloid beta-Peptides/immunology , Animals , Atherosclerosis/immunology , Atherosclerosis/metabolism , Blotting, Western , CD36 Antigens/metabolism , Cell Line , Chemokines/biosynthesis , Chemokines/immunology , Gene Expression , Humans , Immunoprecipitation , Inflammation/metabolism , Lipoproteins, LDL/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Microglia/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 6/metabolism
9.
J Immunol ; 205(7): 1810-1818, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32859730

ABSTRACT

Systemic lupus erythematosus (SLE) is defined by loss of B cell tolerance, resulting in production of autoantibodies against nucleic acids and other cellular Ags. Aberrant activation of TLRs by self-derived RNA and DNA is strongly associated with SLE in patients and in mouse models, but the mechanism by which TLR signaling to self-ligands is regulated remains poorly understood. In this study, we show that αv integrin plays a critical role in regulating B cell TLR signaling to self-antigens in mice. We show that deletion of αv from B cells accelerates autoantibody production and autoimmune kidney disease in the Tlr7.1 transgenic mouse model of SLE. Increased autoimmunity was associated with specific expansion of transitional B cells, extrafollicular IgG2c-producing plasma cells, and activation of CD4 and CD8 T cells. Our data show that αv-mediated regulation of TLR signaling in B cells is critical for preventing autoimmunity and indicate that loss of αv promotes escape from tolerance. Thus, we identify a new regulatory pathway in autoimmunity and elucidate upstream signals that adjust B cell activation to prevent development of autoimmunity in a mouse model.


Subject(s)
B-Lymphocytes/physiology , Integrin alphaV/metabolism , Lupus Erythematosus, Systemic/immunology , Membrane Glycoproteins/metabolism , Toll-Like Receptor 7/metabolism , Animals , Autoantibodies/metabolism , Autoimmunity , Cells, Cultured , Disease Models, Animal , Humans , Immunoglobulin G/metabolism , Immunomodulation , Integrin alphaV/genetics , Lymphocyte Activation , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Toll-Like Receptor 7/genetics
10.
Immunity ; 36(5): 695-6, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22633454

ABSTRACT

In this issue of Immunity, Irving et al. (2012) show that protein kinase R (PKR) regulates the cytoskeleton via an interaction with gelsolin. This alternative role for PKR prevents penetration of virions into the cell.

11.
Immunity ; 35(4): 536-49, 2011 Oct 28.
Article in English | MEDLINE | ID: mdl-22018470

ABSTRACT

Although infections with virulent pathogens often induce a strong inflammatory reaction, what drives the increased immune response to pathogens compared to nonpathogenic microbes is poorly understood. One possibility is that the immune system senses the level of threat from a microorganism and augments the response accordingly. Here, focusing on cytotoxic necrotizing factor 1 (CNF1), an Escherichia coli-derived effector molecule, we showed the host indirectly sensed the pathogen by monitoring for the effector that modified RhoGTPases. CNF1 modified Rac2, which then interacted with the innate immune adaptors IMD and Rip1-Rip2 in flies and mammalian cells, respectively, to drive an immune response. This response was protective and increased the ability of the host to restrict pathogen growth, thus defining a mechanism of effector-triggered immunity that contributes to how metazoans defend against microbes with pathogenic potential.


Subject(s)
Signal Transduction , rac GTP-Binding Proteins/immunology , Adaptor Proteins, Signal Transducing/metabolism , Enzyme Activation , HEK293 Cells , Humans , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , rac GTP-Binding Proteins/metabolism , RAC2 GTP-Binding Protein
12.
13.
Immunol Rev ; 264(1): 363-81, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25703572

ABSTRACT

The road to a more efficacious vaccine that could be a truly transformative tool for decreasing tuberculosis morbidity and mortality, along with Mycobacterium tuberculosis transmission, is quite daunting. Despite this, there are reasons for optimism. Abetted by better conceptual clarity, clear acknowledgment of the degree of our current immunobiological ignorance, the availability of powerful new tools for dissecting the immunopathogenesis of human tuberculosis, the generation of more creative diversity in tuberculosis vaccine concepts, the development of better fit-for-purpose animal models, and the potential of more pragmatic approaches to the clinical testing of vaccine candidates, the field has promise for delivering novel tools for dealing with this worldwide scourge of poverty.


Subject(s)
Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/immunology , Tuberculosis/prevention & control , Animals , Disease Models, Animal , Humans , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/adverse effects , Vaccines, Subunit
14.
J Immunol ; 197(5): 1968-78, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27481847

ABSTRACT

Activation of TGF-ß by dendritic cells (DCs) expressing αvß8 integrin is essential for the generation of intestinal regulatory T cells (Tregs) that in turn promote tolerance to intestinal Ags. We have recently shown that αvß8 integrin is preferentially expressed by CD103(+) DCs and confers their ability to activate TGF-ß and generate Tregs. However, how these DCs become specialized for this vital function is unknown. In this study, we show that ß8 expression is controlled by a combination of factors that include DC lineage and signals derived from the tissue microenvironment and microbiota. Specifically, our data demonstrate that TGF-ß itself, along with retinoic acid and TLR signaling, drives expression of αvß8 in DCs. However, these signals only result in high levels of ß8 expression in cells of the cDC1 lineage, CD8α(+), or CD103(+)CD11b(-) DCs, and this is associated with epigenetic changes in the Itgb8 locus. Together, these data provide a key illustrative example of how microenvironmental factors and cell lineage drive the generation of regulatory αvß8-expressing DCs specialized for activation of TGF-ß to facilitate Treg generation.


Subject(s)
Cell Lineage , Cellular Microenvironment , Dendritic Cells/immunology , Integrin beta Chains/metabolism , Intestines/cytology , Transforming Growth Factor beta/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Cell Differentiation , Dendritic Cells/physiology , Integrin alpha Chains/genetics , Integrin alpha Chains/immunology , Integrin beta Chains/genetics , Integrin beta Chains/immunology , Intestines/immunology , Mice , T-Lymphocytes, Regulatory/physiology , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Tretinoin/metabolism
15.
J Infect Dis ; 210(11): 1844-54, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-24842831

ABSTRACT

Autophagy has been postulated to play role in mammalian host defense against fungal pathogens, although the molecular details remain unclear. Here, we show that primary macrophages deficient in the autophagic factor LC3 demonstrate diminished fungicidal activity but increased cytokine production in response to Candida albicans stimulation. LC3 recruitment to fungal phagosomes requires activation of the fungal pattern receptor dectin-1. LC3 recruitment to the phagosome also requires Syk signaling but is independent of all activity by Toll-like receptors and does not require the presence of the adaptor protein Card9. We further demonstrate that reactive oxygen species generation by NADPH oxidase is required for LC3 recruitment to the fungal phagosome. These observations directly link LC3 to the inflammatory pathway against C. albicans in macrophages.


Subject(s)
Fungi/immunology , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Macrophages/metabolism , Macrophages/microbiology , Microtubule-Associated Proteins/metabolism , Phagosomes/metabolism , Animals , CARD Signaling Adaptor Proteins/metabolism , Candida albicans/immunology , Cell Line , Interleukin-1beta/biosynthesis , Interleukin-6/biosynthesis , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/immunology , Mice , Microtubule-Associated Proteins/genetics , Models, Biological , NADPH Oxidases/metabolism , Phagosomes/immunology , Phagosomes/microbiology , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Proteoglycans , Reactive Oxygen Species/metabolism , Signal Transduction , Syk Kinase , Tumor Necrosis Factor-alpha/biosynthesis , beta-Glucans/metabolism
16.
J Biol Chem ; 288(22): 16043-54, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23609446

ABSTRACT

Elimination of fungal pathogens by phagocytes requires phagosome maturation, a process that involves the recruitment and fusion of intracellular proteins. The role of Dectin-1, a ß-1,3-glucan receptor, critical for fungal recognition and triggering of Th17 responses, to phagosomal maturation has not been defined. We show that GFP-Dectin-1 translocates to the fungal phagosome, but its signal decays after 2 h. Inhibition of acidification results in retention of GFP-Dectin-1 to phagosome membranes highlighting the requirement for an acidic pH. Following ß-1,3-glucan recognition, GFP-Dectin-1 undergoes tyrosine phosphorylation by Src kinases with subsequent Syk activation. Our results demonstrate that Syk is activated independently of intraphagosomal pH. Inhibition of Src or Syk results in prolonged retention of GFP-Dectin-1 to the phagosome signifying a link between Syk and intraphagosomal pH. ß-1,3-glucan phagosomes expressing a signaling incompetent Dectin-1 failed to mature as demonstrated by prolonged Dectin-1 retention, presence of Rab5B, failure to acquire LAMP-1 and inability to acidify. Phagosomes containing Candida albicans also require Dectin-1-dependent Syk activation for phagosomal maturation. Taken together, these results support a model where Dectin-1 not only controls internalization of ß-1,3-glucan containing cargo and triggers proinflammatory cytokines, but also acts as a master regulator for subsequent phagolysosomal maturation through Syk activation.


Subject(s)
Candida albicans/metabolism , Lectins, C-Type/metabolism , Phagosomes/metabolism , beta-Glucans/metabolism , Animals , Cell Line , Cytokines/genetics , Cytokines/metabolism , Enzyme Activation/genetics , Inflammation Mediators/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lectins, C-Type/genetics , Mice , Phagosomes/genetics , Phagosomes/microbiology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Syk Kinase , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
17.
PLoS Pathog ; 8(7): e1002798, 2012.
Article in English | MEDLINE | ID: mdl-22792069

ABSTRACT

Understanding host defense against microbes is key to developing new and more effective therapies for infection and inflammatory disease. However, how animals integrate multiple environmental signals and discriminate between different pathogens to mount specific and tailored responses remains poorly understood. Using the genetically tractable model host Caenorhabditis elegans and pathogenic bacterium Staphylococcus aureus, we describe an important role for hypoxia-inducible factor (HIF) in defining the specificity of the host response in the intestine. We demonstrate that loss of egl-9, a negative regulator of HIF, confers HIF-dependent enhanced susceptibility to S. aureus while increasing resistance to Pseudomonas aeruginosa. In our attempt to understand how HIF could have these apparently dichotomous roles in host defense, we find that distinct pathways separately regulate two opposing functions of HIF: the canonical pathway is important for blocking expression of a set of HIF-induced defense genes, whereas a less well understood noncanonical pathway appears to be important for allowing the expression of another distinct set of HIF-repressed defense genes. Thus, HIF can function either as a gene-specific inducer or repressor of host defense, providing a molecular mechanism by which HIF can have apparently opposing roles in defense and inflammation. Together, our observations show that HIF can set the balance between alternative pathogen-specific host responses, potentially acting as an evolutionarily conserved specificity switch in the host innate immune response.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/immunology , Pseudomonas aeruginosa/immunology , Staphylococcus aureus/immunology , Transcription Factors/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Cullin Proteins/genetics , Cullin Proteins/metabolism , Host Specificity , Immunity, Innate , Intestinal Mucosa/immunology , Intestinal Mucosa/parasitology , RNA Interference , RNA, Small Interfering , Transcription Factors/genetics
18.
Sci Immunol ; 9(91): eabq6541, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38181093

ABSTRACT

Pore-forming toxins (PFTs) are the largest class of bacterial toxins and contribute to virulence by triggering host cell death. Vertebrates also express endogenous pore-forming proteins that induce cell death as part of host defense. To mitigate damage and promote survival, cells mobilize membrane repair mechanisms to neutralize and counteract pores, but how these pathways are activated is poorly understood. Here, we use a transposon-based gene activation screen to discover pathways that counteract the cytotoxicity of the archetypal PFT Staphylococcus aureus α-toxin. We identify the endolysosomal protein LITAF as a mediator of cellular resistance to PFT-induced cell death that is active against both bacterial toxins and the endogenous pore, gasdermin D, a terminal effector of pyroptosis. Activation of the ubiquitin ligase NEDD4 by potassium efflux mobilizes LITAF to recruit the endosomal sorting complexes required for transport (ESCRT) machinery to repair damaged membrane. Cells lacking LITAF, or carrying naturally occurring disease-associated mutations of LITAF, are highly susceptible to pore-induced death. Notably, LITAF-mediated repair occurs at endosomal membranes, resulting in expulsion of damaged membranes as exosomes, rather than through direct excision of pores from the surface plasma membrane. These results identify LITAF as a key effector that links sensing of cellular damage to repair.


Subject(s)
Bacterial Toxins , Pyroptosis , Animals , Cell Death , Cell Membrane , Endosomes
19.
J Biol Chem ; 287(19): 16029-36, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22433861

ABSTRACT

Immunosuppression via cell-cell contact with apoptotic cells is a well studied immunological phenomenon. Although the original studies of immune repression used primary cells, which undergo spontaneous cell death or apoptosis in response to irradiation, more recent studies have relied on chemotherapeutic agents to induce apoptosis in cell lines. In this work, we demonstrate that Jurkat cells induced to die with actinomycin D suppressed inflammatory cytokine production by macrophages, whereas cells treated with etoposide did not. This immune repression mediated by actinomycin D-treated cells did not require phagocytosis or cell-cell contact and thus occurs through a different mechanism from that seen with primary apoptotic neutrophils. Moreover, cells induced to die with etoposide and then treated for a short time with actinomycin D also suppressed macrophage responses, indicating that suppression was mediated by actinomycin D independent of the mechanism of cell death. Finally, phagocytosis of actinomycin D-treated cells caused apoptosis in macrophages, and suppression could be blocked by inhibition of caspase activity in the target macrophage. Together, these data indicate that apoptotic cells act as "Trojan horses," delivering actinomycin D to engulfing macrophages. Suppression of cytokine production by macrophages is therefore due to exposure to actinomycin D from apoptotic cells and is not the result of cell-receptor interactions. These data suggest that drug-induced death may not be an appropriate surrogate for the immunosuppressive activity of apoptotic cells. Furthermore, these effects of cytotoxic drugs on infiltrating immune phagocytes may have clinical ramifications for their use as antitumor therapies.


Subject(s)
Apoptosis/immunology , Cytokines/immunology , Inflammation Mediators/immunology , Macrophages/immunology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Communication/immunology , Cell Line , Coculture Techniques , Cytokines/metabolism , Dactinomycin/pharmacology , Etoposide/pharmacology , Flow Cytometry , Humans , Inflammation Mediators/metabolism , Jurkat Cells , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Phagocytosis/immunology , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
20.
Immunol Rev ; 230(1): 9-21, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19594626

ABSTRACT

Innate immunity is the earliest response to invading microbes and acts to contain infection in the first minutes to hours of challenge. Unlike adaptive immunity that relies upon clonal expansion of cells that emerge days after antigenic challenge, the innate immune response is immediate. Soluble mediators, including complement components and the mannose binding lectin (MBL) make an important contribution to innate immune protection and work along with epithelial barriers, cellular defenses such as phagocytosis, and pattern-recognition receptors that trigger pro-inflammatory signaling cascades. These four aspects of the innate immune system act in concert to protect from pathogen invasion. Our work has focused on understanding the protection provided by this complex defense system and, as discussed in this review, the particular contribution of soluble mediators such as MBL and phagocytic cells. Over the past two decades both human epidemiological data and mouse models have indicated that MBL plays a critical role in innate immune protection against a number of pathogens. As demonstrated by our recent in vitro work, we show that MBL and the innate immune signaling triggered by the canonical pattern-recognition receptors (PRRs), the Toll-like receptors (TLRs), are linked by their spatial localization to the phagosome. These observations demonstrated a novel role for MBL as a TLR co-receptor and establishes a new paradigm for the role of opsonins, which we propose to function not only to increase microbial uptake but also to spatially coordinate, amplify, and synchronize innate immune defenses mechanism. In this review we discuss both the attributes of MBL that make it a unique soluble pattern recognition molecule and also highlight its broader role in coordinating innate immune activation.


Subject(s)
Complement Activation/immunology , Immunity, Innate , Mannose-Binding Lectin/metabolism , Nod Signaling Adaptor Proteins/immunology , Toll-Like Receptors/immunology , Animals , Carbohydrates/immunology , Humans , Inflammation/immunology , Inflammation/metabolism , Mannose-Binding Lectin/genetics , Mannose-Binding Lectin/immunology , Mannose-Binding Protein-Associated Serine Proteases/immunology , Mannose-Binding Protein-Associated Serine Proteases/metabolism , Mice , Nod Signaling Adaptor Proteins/metabolism , Phagocytosis/immunology , Signal Transduction/immunology , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL