Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters











Publication year range
1.
Curr Opin Immunol ; 90: 102457, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39232338

ABSTRACT

The innate immune system employs two different strategies to detect pathogens: first, it recognizes microbial components as ligands of pattern recognition receptors (pattern-triggered immunity [PTI]), and second, it detects the activities of pathogen-encoded effectors (effector-triggered immunity [ETI]). Recently, these pathogen-centric concepts were expanded to include sensing of self-derived signals during cellular distress or damage (damage-triggered immunity [DTI]). This extension relied on broadening the PTI model to include damage-associated molecular patterns (DAMPs). However, applying the pattern recognition framework of PTI to DTI overlooks the critical role of sterile activation of ETI pathways. We argue that both PTI and ETI pathways are prone to erroneous detection of self, which is largely attributable to 'friendly fire' rather than protective immune activation. This erroneous activation is inherent to the trade-off between sensitivity and specificity of immune sensing and might be tolerated because its detrimental effects emerge late in life, a phenomenon known as antagonistic pleiotropy.


Subject(s)
Immunity, Innate , Receptors, Pattern Recognition , Humans , Animals , Receptors, Pattern Recognition/metabolism , Receptors, Pattern Recognition/immunology , Alarmins/immunology , Alarmins/metabolism , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism , Signal Transduction/immunology , Host-Pathogen Interactions/immunology
2.
Annu Rev Immunol ; 41: 453-481, 2023 04 26.
Article in English | MEDLINE | ID: mdl-36750319

ABSTRACT

The innate immune system detects pathogens via germline-encoded receptors that bind to conserved pathogen ligands called pathogen-associated molecular patterns (PAMPs). Here we consider an additional strategy of pathogen sensing called effector-triggered immunity (ETI). ETI involves detection of pathogen-encoded virulence factors, also called effectors. Pathogens produce effectors to manipulate hosts to create a replicative niche and/or block host immunity. Unlike PAMPs, effectors are often diverse and rapidly evolving and can thus be unsuitable targets for direct detection by germline-encoded receptors. Effectors are instead often sensed indirectly via detection of their virulence activities. ETI is a viable strategy for pathogen sensing and is used across diverse phyla, including plants, but the molecular mechanisms of ETI are complex compared to simple receptor/ligand-based PAMP detection. Here we survey the mechanisms and functions of ETI, with a particular focus on emerging insights from animal studies. We suggest that many examples of ETI may remain to be discovered, hiding in plain sight throughout immunology.


Subject(s)
Innate Immunity Recognition , Pathogen-Associated Molecular Pattern Molecules , Humans , Animals , Pathogen-Associated Molecular Pattern Molecules/metabolism , Virulence
3.
Front Immunol ; 13: 1074440, 2022.
Article in English | MEDLINE | ID: mdl-36578489

ABSTRACT

Necroptosis is a form of regulated cell death that can occur downstream of several immune pathways. While previous studies have shown that dysregulated necroptosis can lead to strong inflammatory responses, little is known about the identity of the endogenous molecules that trigger these responses. Using a reductionist in vitro model, we found that soluble TNF is strongly released in the context of necroptosis. On the one hand, necroptosis promotes TNF translation by inhibiting negative regulatory mechanisms acting at the post-transcriptional level. On the other hand, necroptosis markedly enhances TNF release by activating ADAM proteases. In studying TNF release at single-cell resolution, we found that TNF release triggered by necroptosis is activated in a switch-like manner that exceeds steady-state TNF processing in magnitude and speed. Although this shedding response precedes massive membrane damage, it is closely associated with lytic cell death. Further, we found that lytic cell death induction using a pore-forming toxin also triggers TNF shedding, indicating that the activation of ADAM proteases is not strictly related to the necroptotic pathway but likely associated with biophysical changes of the cell membrane upon lytic cell death. These results demonstrate that lytic cell death, particularly necroptosis, is a critical trigger for TNF release and thus qualify TNF as a necroptosis-associated alarmin.


Subject(s)
Alarmins , Apoptosis , Humans , Necrosis , Necroptosis , Tumor Necrosis Factor-alpha/metabolism , Peptide Hydrolases
4.
Immunity ; 55(12): 2271-2284.e7, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36384135

ABSTRACT

The NLRP3 inflammasome plays a central role in antimicrobial defense as well as in the context of sterile inflammatory conditions. NLRP3 activity is governed by two independent signals: the first signal primes NLRP3, rendering it responsive to the second signal, which then triggers inflammasome formation. Our understanding of how NLRP3 priming contributes to inflammasome activation remains limited. Here, we show that IKKß, a kinase activated during priming, induces recruitment of NLRP3 to phosphatidylinositol-4-phosphate (PI4P), a phospholipid enriched on the trans-Golgi network. NEK7, a mitotic spindle kinase that had previously been thought to be indispensable for NLRP3 activation, was redundant for inflammasome formation when IKKß recruited NLRP3 to PI4P. Studying iPSC-derived human macrophages revealed that the IKKß-mediated NEK7-independent pathway constitutes the predominant NLRP3 priming mechanism in human myeloid cells. Our results suggest that PI4P binding represents a primed state into which NLRP3 is brought by IKKß activity.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , I-kappa B Kinase , Inflammasomes/metabolism , Mice, Inbred C57BL , NIMA-Related Kinases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Serine-Threonine Kinases/metabolism , trans-Golgi Network/metabolism
5.
Nature ; 600(7887): 138-142, 2021 12.
Article in English | MEDLINE | ID: mdl-34759314

ABSTRACT

Pathogens use virulence factors to inhibit the immune system1. The guard hypothesis2,3 postulates that hosts monitor (or 'guard') critical innate immune pathways such that their disruption by virulence factors provokes a secondary immune response1. Here we describe a 'self-guarded' immune pathway in human monocytes, in which guarding and guarded functions are combined in one protein. We find that this pathway is triggered by ICP0, a key virulence factor of herpes simplex virus type 1, resulting in robust induction of anti-viral type I interferon (IFN). Notably, induction of IFN by ICP0 is independent of canonical immune pathways and the IRF3 and IRF7 transcription factors. A CRISPR screen identified the ICP0 target MORC34 as an essential negative regulator of IFN. Loss of MORC3 recapitulates the IRF3- and IRF7-independent IFN response induced by ICP0. Mechanistically, ICP0 degrades MORC3, which leads to de-repression of a MORC3-regulated DNA element (MRE) adjacent to the IFNB1 locus. The MRE is required in cis for IFNB1 induction by the MORC3 pathway, but is not required for canonical IFN-inducing pathways. As well as repressing the MRE to regulate IFNB1, MORC3 is also a direct restriction factor of HSV-15. Our results thus suggest a model in which the primary anti-viral function of MORC3 is self-guarded by its secondary IFN-repressing function-thus, a virus that degrades MORC3 to avoid its primary anti-viral function will unleash the secondary anti-viral IFN response.


Subject(s)
Adenosine Triphosphatases/immunology , DNA-Binding Proteins/immunology , Models, Immunological , Virulence Factors/immunology , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/metabolism , CRISPR-Cas Systems , Cell Line , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Gene Editing , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/pathogenicity , Humans , Immediate-Early Proteins/immunology , Immunity, Innate , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon Type I/antagonists & inhibitors , Interferon Type I/genetics , Interferon Type I/immunology , Monocytes/immunology , Receptor, Interferon alpha-beta , Repressor Proteins/deficiency , Repressor Proteins/immunology , Repressor Proteins/metabolism , Response Elements/genetics , Ubiquitin-Protein Ligases/immunology
6.
Elife ; 102021 06 21.
Article in English | MEDLINE | ID: mdl-34151776

ABSTRACT

Type I interferons (IFNs) are essential for anti-viral immunity, but often impair protective immune responses during bacterial infections. An important question is how type I IFNs are strongly induced during viral infections, and yet are appropriately restrained during bacterial infections. The Super susceptibility to tuberculosis 1 (Sst1) locus in mice confers resistance to diverse bacterial infections. Here we provide evidence that Sp140 is a gene encoded within the Sst1 locus that represses type I IFN transcription during bacterial infections. We generated Sp140-/- mice and found that they are susceptible to infection by Legionella pneumophila and Mycobacterium tuberculosis. Susceptibility of Sp140-/- mice to bacterial infection was rescued by crosses to mice lacking the type I IFN receptor (Ifnar-/-). Our results implicate Sp140 as an important negative regulator of type I IFNs that is essential for resistance to bacterial infections.


Subject(s)
Bacterial Infections/immunology , Interferon Type I/metabolism , Transcription Factors/metabolism , Alleles , Animals , Female , Gene Expression Regulation/drug effects , Interferon Type I/genetics , Macrophages/physiology , Male , Mice , Mice, Knockout , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Mycobacterium tuberculosis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Specific Pathogen-Free Organisms , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/pharmacology
7.
J Biol Chem ; 295(52): 18065-18075, 2020 12 25.
Article in English | MEDLINE | ID: mdl-33082141

ABSTRACT

TNF is a highly pro-inflammatory cytokine that contributes not only to the regulation of immune responses but also to the development of severe inflammatory diseases. TNF is synthesized as a transmembrane protein, which is further matured via proteolytic cleavage by metalloproteases such as ADAM17, a process known as shedding. At present, TNF is mainly detected by measuring the precursor or the mature cytokine of bulk cell populations by techniques such as ELISA or immunoblotting. However, these methods do not provide information on the exact timing and extent of TNF cleavage at single-cell resolution and they do not allow the live visualization of shedding events. Here, we generated C-tag TNF as a genetically encoded reporter to study TNF shedding at the single-cell level. The functionality of the C-tag TNF reporter is based on the exposure of a cryptic epitope on the C terminus of the transmembrane portion of pro-TNF on cleavage. In both denatured and nondenatured samples, this epitope can be detected by a nanobody in a highly sensitive and specific manner only upon TNF shedding. As such, C-tag TNF can successfully be used for the detection of TNF cleavage in flow cytometry and live-cell imaging applications. We furthermore demonstrate its applicability in a forward genetic screen geared toward the identification of genetic regulators of TNF maturation. In summary, the C-tag TNF reporter can be employed to gain novel insights into the complex regulation of ADAM-dependent TNF shedding.


Subject(s)
ADAM Proteins/metabolism , Genes, Reporter , Image Processing, Computer-Assisted/methods , Molecular Imaging/methods , Protein Kinase C/metabolism , Tumor Necrosis Factor-alpha/metabolism , ADAM Proteins/genetics , HEK293 Cells , Humans , Protein Kinase C/genetics , Proteolysis , Tumor Necrosis Factor-alpha/genetics
8.
Cell ; 179(6): 1264-1275.e13, 2019 11 27.
Article in English | MEDLINE | ID: mdl-31778653

ABSTRACT

TLR8 is among the highest-expressed pattern-recognition receptors in the human myeloid compartment, yet its mode of action is poorly understood. TLR8 engages two distinct ligand binding sites to sense RNA degradation products, although it remains unclear how these ligands are formed in cellulo in the context of complex RNA molecule sensing. Here, we identified the lysosomal endoribonuclease RNase T2 as a non-redundant upstream component of TLR8-dependent RNA recognition. RNase T2 activity is required for rendering complex single-stranded, exogenous RNA molecules detectable for TLR8. This is due to RNase T2's preferential cleavage of single-stranded RNA molecules between purine and uridine residues, which critically contributes to the supply of catabolic uridine and the generation of purine-2',3'-cyclophosphate-terminated oligoribonucleotides. Thus-generated molecules constitute agonistic ligands for the first and second binding pocket of TLR8. Together, these results establish the identity and origin of the RNA-derived molecular pattern sensed by TLR8.


Subject(s)
Endoribonucleases/metabolism , Proteolysis , Toll-Like Receptor 8/metabolism , Amino Acid Motifs , Base Sequence , Cell Line , Endoribonucleases/deficiency , Humans , Models, Molecular , Monocytes/metabolism , Myeloid Cells/metabolism , Nitrogen Isotopes , Oligonucleotides/metabolism , Purines/metabolism , RNA/metabolism , Staphylococcus aureus/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/chemistry , Uridine/metabolism
9.
Proc Natl Acad Sci U S A ; 116(3): 970-975, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30591564

ABSTRACT

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is a critical regulator of cell death and inflammation, but its relevance for human disease pathogenesis remains elusive. Studies of monogenic disorders might provide critical insights into disease mechanisms and therapeutic targeting of RIPK1 for common diseases. Here, we report on eight patients from six unrelated pedigrees with biallelic loss-of-function mutations in RIPK1 presenting with primary immunodeficiency and/or intestinal inflammation. Mutations in RIPK1 were associated with reduced NF-κB activity, defective differentiation of T and B cells, increased inflammasome activity, and impaired response to TNFR1-mediated cell death in intestinal epithelial cells. The characterization of RIPK1-deficient patients highlights the essential role of RIPK1 in controlling human immune and intestinal homeostasis, and might have critical implications for therapies targeting RIPK1.


Subject(s)
Cell Differentiation , Immunity, Mucosal/genetics , Inflammatory Bowel Diseases , Intestinal Mucosa , Receptor-Interacting Protein Serine-Threonine Kinases , Severe Combined Immunodeficiency , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , HCT116 Cells , HEK293 Cells , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Male , Mutation , NF-kappa B/genetics , NF-kappa B/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology
11.
Cell Rep ; 25(9): 2354-2368.e5, 2018 11 27.
Article in English | MEDLINE | ID: mdl-30485805

ABSTRACT

IL-1ß is a cytokine of pivotal importance to the orchestration of inflammatory responses. Synthesized as an inactive pro-cytokine, IL-1ß requires proteolytic maturation to gain biological activity. Here, we identify intrinsic apoptosis as a non-canonical trigger of IL-1ß maturation. Guided by the discovery of the immunomodulatory activity of vioprolides, cyclic peptides isolated from myxobacteria, we observe IL-1ß maturation independent of canonical inflammasome pathways, yet dependent on intrinsic apoptosis. Mechanistically, vioprolides inhibit MCL-1 and BCL2, which in turn triggers BAX/BAK-dependent mitochondrial outer membrane permeabilization (MOMP). Induction of MOMP results in the release of pro-apoptotic factors initiating intrinsic apoptosis, as well as the depletion of IAPs (inhibitors of apoptosis proteins). IAP depletion, in turn, operates upstream of ripoptosome complex formation, subsequently resulting in caspase-8-dependent IL-1ß maturation. These results establish the ripoptosome/caspase-8 complex as a pro-inflammatory checkpoint that senses the perturbation of mitochondrial integrity.


Subject(s)
Apoptosis , Caspase 8/metabolism , Interleukin-1beta/metabolism , Macrophages/cytology , Macrophages/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Caspase 1/metabolism , Cell Survival/drug effects , Enzyme Activation/drug effects , Humans , L-Lactate Dehydrogenase/metabolism , Mice , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Peptides, Cyclic/pharmacology , Permeability , Protein Synthesis Inhibitors/pharmacology
12.
Methods Mol Biol ; 1714: 57-66, 2018.
Article in English | MEDLINE | ID: mdl-29177855

ABSTRACT

Monocytes and macrophages play a pivotal role in the induction and shaping of immune responses. Expressing a broad array of pattern recognition receptors (PRRs), monocytes and macrophages constitute an integral component of the innate branch of the immune system. Traditionally, the majority of innate immune sensing and signaling pathways have been studied in macrophages of the murine system. This is largely due to the fact that genetic loss-of-function studies are amenable in this species. On the other hand, human cell lines of the monocyte-macrophage cell lineage have been widely used to study myeloid cells in vitro. However, commonly utilized models (e.g., THP-1 cells) only mimic a limited spectrum of the immunobiology of primary human myeloid cells. Recently, we have explored the possibility to fill this gap with a human trans-differentiation cell culture system, in which lineage conversion from malignant B-lineage cells to monocytes/macrophages is caused by the inducible nuclear translocation of a C/EBPα transgene, BLaER1 cells. Using this model, we were able to characterize a novel inflammasome signaling entity that could not have been uncovered in the murine system or THP-1 cells. Here, we describe the handling of BLaER1 cells, providing a detailed protocol for their induced trans-differentiation. We also provide data to demonstrate the applicability of the BLaER1 monocyte/macrophage system to study phagocytosis and various PRR cascades in human cells.


Subject(s)
Cell Differentiation , Macrophages/cytology , Models, Biological , Monocytes/cytology , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line , Humans , Immunity, Innate , Macrophages/metabolism , Monocytes/metabolism , Myeloid Cells/cytology , Myeloid Cells/metabolism , Signal Transduction
13.
J Mol Biol ; 430(2): 133-141, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29203171

ABSTRACT

NLRP3 is the most studied inflammasome sensor due to its crucial involvement in sterile and infection-triggered inflammation. Although its molecular mode of activation remains to be defined, it is well established that low intracellular potassium concentrations result in its activation. This functionality allows the classical NLRP3 pathway to serve as a highly sensitive, but non-specific surveillance mechanism responding to any type of perturbation that breaches plasma membrane integrity and the associated potassium gradient across the membrane. Here, we review our current knowledge on potassium efflux-dependent NLRP3 activation, with a special focus on how major cell death programs are rendered pro-inflammatory by secondary NLRP3 activation. Apart from the "alternative inflammasome" as the major exception to the rule, this connection explains the fundamental importance of NLRP3 in cell death-associated inflammation and firmly establishes NLRP3 as a principal surveillance mechanism of cellular integrity.


Subject(s)
Inflammasomes/immunology , Inflammation/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Animals , Cell Death , Humans , Potassium/immunology
14.
Cell ; 171(5): 1110-1124.e18, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29033128

ABSTRACT

Detection of cytosolic DNA constitutes a central event in the context of numerous infectious and sterile inflammatory conditions. Recent studies have uncovered a bipartite mode of cytosolic DNA recognition, in which the cGAS-STING axis triggers antiviral immunity, whereas AIM2 triggers inflammasome activation. Here, we show that AIM2 is dispensable for DNA-mediated inflammasome activation in human myeloid cells. Instead, detection of cytosolic DNA by the cGAS-STING axis induces a cell death program initiating potassium efflux upstream of NLRP3. Forward genetics identified regulators of lysosomal trafficking to modulate this cell death program, and subsequent studies revealed that activated STING traffics to the lysosome, where it triggers membrane permeabilization and thus lysosomal cell death (LCD). Importantly, the cGAS-STING-NLRP3 pathway constitutes the default inflammasome response during viral and bacterial infections in human myeloid cells. We conclude that targeting the cGAS-STING-LCD-NLRP3 pathway will ameliorate pathology in inflammatory conditions that are associated with cytosolic DNA sensing.


Subject(s)
Cell Death , Inflammasomes/metabolism , Monocytes/cytology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , DNA/metabolism , Humans , Membrane Proteins/metabolism , Monocytes/metabolism , Signal Transduction
15.
Nature ; 549(7672): 394-398, 2017 09 21.
Article in English | MEDLINE | ID: mdl-28902841

ABSTRACT

Cytosolic DNA arising from intracellular pathogens triggers a powerful innate immune response. It is sensed by cyclic GMP-AMP synthase (cGAS), which elicits the production of type I interferons by generating the second messenger 2'3'-cyclic-GMP-AMP (cGAMP). Endogenous nuclear or mitochondrial DNA can also be sensed by cGAS under certain conditions, resulting in sterile inflammation. The cGAS dimer binds two DNA ligands shorter than 20 base pairs side-by-side, but 20-base-pair DNA fails to activate cGAS in vivo and is a poor activator in vitro. Here we show that cGAS is activated in a strongly DNA length-dependent manner both in vitro and in human cells. We also show that cGAS dimers form ladder-like networks with DNA, leading to cooperative sensing of DNA length: assembly of the pioneering cGAS dimer between two DNA molecules is ineffective; but, once formed, it prearranges the flanking DNA to promote binding of subsequent cGAS dimers. Remarkably, bacterial and mitochondrial nucleoid proteins HU and mitochondrial transcription factor A (TFAM), as well as high-mobility group box 1 protein (HMGB1), can strongly stimulate long DNA sensing by cGAS. U-turns and bends in DNA induced by these proteins pre-structure DNA to nucleate cGAS dimers. Our results suggest a nucleation-cooperativity-based mechanism for sensitive detection of mitochondrial DNA and pathogen genomes, and identify HMGB/TFAM proteins as DNA-structuring host factors. They provide an explanation for the peculiar cGAS dimer structure and suggest that cGAS preferentially binds incomplete nucleoid-like structures or bent DNA.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/chemistry , DNA/metabolism , HMGB Proteins/metabolism , High Mobility Group Proteins/metabolism , Mitochondrial Proteins/metabolism , Nucleotidyltransferases/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Female , Humans , Mice , Models, Molecular , Nucleic Acid Conformation , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/chemistry , Protein Multimerization
16.
Oncotarget ; 8(63): 106764-106777, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29290987

ABSTRACT

Mitochondria form a highly dynamic network driven by opposing scission and fusion events. DRP1 is an essential modulator of mitochondrial fission and dynamics within mammalian cells. Its fission activity is regulated by posttranslational modifications such as activating phosphorylation at serine 616. DRP1 activity has recently been implicated as being dysregulated in numerous human disorders such as cancer and neurodegenerative diseases. Here we describe the development of a cell-based screening assay to detect DRP1 activation. We utilized this to undertake focused compound library screening and identified potent modulators that affected DRP1 activity including ICG-001, which is described as WNT/ß-catenin signaling inhibitor. Our findings elucidate novel details about ICG-001's mechanism of action (MOA) in mediating anti-proliferative activity. We show ICG-001 both inhibits mitochondrial fission and activates an early endoplasmic reticulum (ER) stress response to induce cell death in susceptible colorectal cancer cell lines.

17.
Curr Opin Immunol ; 44: 7-13, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27842238

ABSTRACT

Classical modes of NLRP3 activation entail a priming step that enables its activation (signal 1) and a potassium efflux-dependent activation signal (signal 2) that triggers pyroptosome formation and pyroptosis, a lytic cell death necessary for IL-1ß release. Opposing to that, human monocytes engage an alternative NLRP3 inflammasome pathway in response to LPS that proceeds in the absence of signal 2 activation and enables IL-1ß secretion without pyroptosis. This specifically relies on Caspase-8 to propagate signaling to NLRP3, leading to inflammasome activation in absence of pyroptosome formation. Here, we summarize the current knowledge about alternative inflammasome activation, discuss potential extensions of this signaling entity beyond LPS-dependent activation, speculate about its role in tissue homeostasis and sterile inflammation and highlight the implications of pyroptosis-independent IL-1ß secretion.


Subject(s)
Inflammasomes/metabolism , Inflammation/immunology , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Caspase 8/metabolism , Homeostasis , Humans , Lipopolysaccharides/immunology , Potassium-Hydrogen Antiporters/metabolism , Pyroptosis , Signal Transduction
18.
EMBO J ; 35(20): 2167-2169, 2016 10 17.
Article in English | MEDLINE | ID: mdl-27572465
19.
Immunity ; 44(4): 833-46, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27037191

ABSTRACT

Interleukin-1ß (IL-1ß) is a cytokine whose bioactivity is controlled by activation of the inflammasome. However, in response to lipopolysaccharide, human monocytes secrete IL-1ß independently of classical inflammasome stimuli. Here, we report that this constituted a species-specific response that is not observed in the murine system. Indeed, in human monocytes, lipopolysaccharide triggered an "alternative inflammasome" that relied on NLRP3-ASC-caspase-1 signaling, yet was devoid of any classical inflammasome characteristics including pyroptosome formation, pyroptosis induction, and K(+) efflux dependency. Genetic dissection of the underlying signaling pathway in a monocyte transdifferentiation system revealed that alternative inflammasome activation was propagated by TLR4-TRIF-RIPK1-FADD-CASP8 signaling upstream of NLRP3. Importantly, involvement of this signaling cascade was limited to alternative inflammasome activation and did not extend to classical NLRP3 activation. Because alternative inflammasome activation embraces both sensitivity and promiscuity of TLR4, we propose a pivotal role for this signaling cascade in TLR4-driven, IL-1ß-mediated immune responses and immunopathology in humans.


Subject(s)
Carrier Proteins/immunology , Inflammasomes/immunology , Interleukin-1beta/immunology , Monocytes/immunology , Toll-Like Receptor 4/immunology , Animals , Caspase 1/immunology , Cell Line , Cell Transdifferentiation/immunology , Humans , Interleukin-1beta/metabolism , Lipopolysaccharides , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Potassium/metabolism , Potassium Channels/immunology , Pyroptosis/immunology , Signal Transduction/immunology
20.
Methods Mol Biol ; 1338: 261-72, 2016.
Article in English | MEDLINE | ID: mdl-26443227

ABSTRACT

Recent developments in the field of designer nucleases allow the efficient and specific manipulation of genomic architectures in eukaryotic cell lines. To this end, it has become possible to introduce DNA double strand breaks (DSBs) at user-defined genomic loci. If located in critical coding regions of genes, thus induced DSBs can lead to insertions or deletions (indels) that result in frameshift mutations and thereby the knockout of the target gene. In this chapter, we describe a step-by-step workflow for establishing knockout cell clones of the difficult-to-transfect suspension cell line THP1. The here described protocol encompasses electroporation, cell cloning, and a deep sequencing-based genotyping step that allows the in-parallel analysis of 96 cell clones per gene of interest. Furthermore, we describe the use of the analysis tool OutKnocker that allows rapid identification of cell clones with all-allelic frameshift mutations.


Subject(s)
CRISPR-Cas Systems/genetics , DNA Breaks, Double-Stranded , Endonucleases/genetics , Gene Knockout Techniques/methods , Cell Line , Frameshift Mutation , Genome , Genotype , High-Throughput Nucleotide Sequencing/methods , Humans , INDEL Mutation
SELECTION OF CITATIONS
SEARCH DETAIL