Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 24(4): 595-603, 2023 04.
Article in English | MEDLINE | ID: mdl-36941400

ABSTRACT

Upon detecting pathogens or cell stress, several NOD-like receptors (NLRs) form inflammasome complexes with the adapter ASC and caspase-1, inducing gasdermin D (GSDMD)-dependent cell death and maturation and release of IL-1ß and IL-18. The triggers and activation mechanisms of several inflammasome-forming sensors are not well understood. Here we show that mitochondrial damage activates the NLRP10 inflammasome, leading to ASC speck formation and caspase-1-dependent cytokine release. While the AIM2 inflammasome can also sense mitochondrial demise by detecting mitochondrial DNA (mtDNA) in the cytosol, NLRP10 monitors mitochondrial integrity in an mtDNA-independent manner, suggesting the recognition of distinct molecular entities displayed by the damaged organelles. NLRP10 is highly expressed in differentiated human keratinocytes, in which it can also assemble an inflammasome. Our study shows that this inflammasome surveils mitochondrial integrity. These findings might also lead to a better understanding of mitochondria-linked inflammatory diseases.


Subject(s)
Cytokines , Inflammasomes , Humans , Inflammasomes/metabolism , Caspase 1/metabolism , Cytokines/metabolism , Cell Death , DNA, Mitochondrial/genetics , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism
2.
EMBO J ; 41(23): e111289, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36221902

ABSTRACT

The NOD1/2-RIPK2 is a key cytosolic signaling complex that activates NF-κB pro-inflammatory response against invading pathogens. However, uncontrolled NF-κB signaling can cause tissue damage leading to chronic diseases. The mechanisms by which the NODs-RIPK2-NF-κB innate immune axis is activated and resolved remain poorly understood. Here, we demonstrate that bacterial infection induces the formation of endogenous RIPK2 oligomers (RIPosomes) that are self-assembling entities that coat the bacteria to induce NF-κB response. Next, we show that autophagy proteins IRGM and p62/SQSTM1 physically interact with NOD1/2, RIPK2 and RIPosomes to promote their selective autophagy and limit NF-κB activation. IRGM suppresses RIPK2-dependent pro-inflammatory programs induced by Shigella and Salmonella. Consistently, the therapeutic inhibition of RIPK2 ameliorates Shigella infection- and DSS-induced gut inflammation in Irgm1 KO mice. This study identifies a unique mechanism where the innate immune proteins and autophagy machinery are recruited together to the bacteria for defense as well as for maintaining immune homeostasis.


Subject(s)
Bacterial Infections , NF-kappa B , Mice , Animals , NF-kappa B/metabolism , Mice, Inbred NOD , Autophagy , Immunity, Innate , Homeostasis
3.
EMBO Rep ; 25(7): 2914-2949, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38783164

ABSTRACT

Neutrophil extracellular traps (NETs) are a key antimicrobial feature of cellular innate immunity mediated by polymorphonuclear neutrophils (PMNs). NETs counteract microbes but are also linked to inflammation in atherosclerosis, arthritis, or psoriasis by unknown mechanisms. Here, we report that NET-associated RNA (naRNA) stimulates further NET formation in naive PMNs via a unique TLR8-NLRP3 inflammasome-dependent pathway. Keratinocytes respond to naRNA with expression of psoriasis-related genes (e.g., IL17, IL36) via atypical NOD2-RIPK signaling. In vivo, naRNA drives temporary skin inflammation, which is drastically ameliorated by genetic ablation of RNA sensing. Unexpectedly, the naRNA-LL37 'composite damage-associated molecular pattern (DAMP)' is pre-stored in resting neutrophil granules, defining sterile NETs as inflammatory webs that amplify neutrophil activation. However, the activity of the naRNA-LL37 DAMP is transient and hence supposedly self-limiting under physiological conditions. Collectively, upon dysregulated NET release like in psoriasis, naRNA sensing may represent both a potential cause of disease and a new intervention target.


Subject(s)
Alarmins , Cathelicidins , Extracellular Traps , Inflammation , Neutrophils , Extracellular Traps/metabolism , Neutrophils/metabolism , Neutrophils/immunology , Inflammation/metabolism , Inflammation/genetics , Animals , Humans , Mice , Alarmins/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Keratinocytes/metabolism , RNA/genetics , RNA/metabolism , Psoriasis/genetics , Psoriasis/metabolism , Psoriasis/pathology , Signal Transduction , Neutrophil Activation/genetics , Immunity, Innate/genetics
4.
EMBO J ; 40(13): e106272, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33942347

ABSTRACT

Cellular stress has been associated with inflammation, yet precise underlying mechanisms remain elusive. In this study, various unrelated stress inducers were employed to screen for sensors linking altered cellular homeostasis and inflammation. We identified the intracellular pattern recognition receptors NOD1/2, which sense bacterial peptidoglycans, as general stress sensors detecting perturbations of cellular homeostasis. NOD1/2 activation upon such perturbations required generation of the endogenous metabolite sphingosine-1-phosphate (S1P). Unlike peptidoglycan sensing via the leucine-rich repeats domain, cytosolic S1P directly bound to the nucleotide binding domains of NOD1/2, triggering NF-κB activation and inflammatory responses. In sum, we unveiled a hitherto unknown role of NOD1/2 in surveillance of cellular homeostasis through sensing of the cytosolic metabolite S1P. We propose S1P, an endogenous metabolite, as a novel NOD1/2 activator and NOD1/2 as molecular hubs integrating bacterial and metabolic cues.


Subject(s)
Inflammation/metabolism , Lysophospholipids/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Sphingosine/analogs & derivatives , Animals , Cell Line , Cell Line, Tumor , Female , HEK293 Cells , HeLa Cells , Humans , Mice , NF-kappa B/metabolism , Peptidoglycan/metabolism , Signal Transduction/physiology , Sphingosine/metabolism , THP-1 Cells
5.
J Cell Sci ; 134(12)2021 06 15.
Article in English | MEDLINE | ID: mdl-34152391

ABSTRACT

The receptor interacting serine/threonine kinase 2 (RIPK2) is essential for signal transduction induced by the pattern recognition receptors NOD1 and NOD2 (referred to collectively as NOD1/2). Upon NOD1/2 activation, RIPK2 forms complexes in the cytoplasm of human cells. Here, we identified the molecular composition of these complexes. Infection with Shigella flexneri to activate NOD1-RIPK2 revealed that RIPK2 formed dynamic interactions with several cellular proteins, including A20 (also known as TNFAIP3), erlin-1, erlin-2 and 14-3-3. Whereas interaction of RIPK2 with 14-3-3 proteins was strongly reduced upon infection with Shigella, erlin-1 and erlin-2 (erlin-1/2) specifically bound to RIPK2 complexes. The interaction of these proteins with RIPK2 was validated using protein binding assays and immunofluorescence staining. Beside bacterial activation of NOD1/2, depletion of the E3 ubiquitin ligase XIAP and treatment with RIPK2 inhibitors also led to the formation of RIPK2 cytosolic complexes. Although erlin-1/2 were recruited to RIPK2 complexes following XIAP inhibition, these proteins did not associate with RIPK2 structures induced by RIPK2 inhibitors. While the specific recruitment of erlin-1/2 to RIPK2 suggests a role in innate immune signaling, the biological response regulated by the erlin-1/2-RIPK2 association remains to be determined.


Subject(s)
Nod2 Signaling Adaptor Protein , Receptor-Interacting Protein Serine-Threonine Kinase 2 , 14-3-3 Proteins , Cytosol/metabolism , Humans , Nod1 Signaling Adaptor Protein , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , Protein Binding , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Signal Transduction
6.
Nat Immunol ; 12(2): 121-8, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21245903

ABSTRACT

The last 10 years have witnessed the identification of a new class of intracellular pattern-recognition molecules--the nucleotide-binding domain and leucine-rich repeat-containing family (NLR). Members of this family garnered interest as pattern-recognition receptors able to trigger inflammatory responses against pathogens. Many studies support a pathogen-recognition function for human NLR proteins and shed light on their role in the broader control of adaptive immunity and various disease states. Other evidence suggests that NLRs function in processes unrelated to pathogen detection. Here we discuss recent advances in our understanding of the biology of the human NLR proteins and their non-pathogen-recognition function in tissue homeostasis, apoptosis, graft-versus-host disease and early development.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Carrier Proteins/immunology , Graft vs Host Disease/immunology , Th1 Cells/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Apoptosis Regulatory Proteins , Autoimmune Diseases/genetics , Carrier Proteins/genetics , Embryonic Development/immunology , Genetic Predisposition to Disease , Humans , Lymphocyte Activation , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Ovulation/immunology , Polymorphism, Genetic , Signal Transduction/immunology , Transcriptional Activation/immunology , Transplantation Immunology
7.
Nat Immunol ; 12(9): 817-26, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21852785

ABSTRACT

In plants and animals, the NLR family of receptors perceives non-self and modified-self molecules inside host cells and mediates innate immune responses to microbial pathogens. Despite their similar biological functions and protein architecture, animal NLRs are normally activated by conserved microbe- or damage-associated molecular patterns, whereas plant NLRs typically detect strain-specific pathogen effectors. Plant NLRs recognize either the effector structure or effector-mediated modifications of host proteins. The latter indirect mechanism for the perception of non-self, as well as the within-species diversification of plant NLRs, maximize the capacity to recognize non-self through the use of a finite number of innate immunoreceptors. We discuss recent insights into NLR activation, signal initiation through the homotypic association of N-terminal domains and subcellular receptor dynamics in plants and compare those with NLR functions in animals.


Subject(s)
Immunity, Innate , Plant Immunity , Plants/immunology , Receptors, G-Protein-Coupled/immunology , Receptors, Pattern Recognition/immunology , Signal Transduction/immunology , Animals , Bacteria/pathogenicity , Biological Evolution , Cell Death , Cytokines/immunology , Cytokines/metabolism , Fungi/pathogenicity , Gene Expression Regulation, Plant , Genetic Heterogeneity , Host-Parasite Interactions , Mice , Plant Growth Regulators/immunology , Plant Growth Regulators/metabolism , Plant Proteins/immunology , Plants/microbiology , Plants/parasitology , Protein Multimerization , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/metabolism
8.
Int J Mol Sci ; 24(10)2023 May 11.
Article in English | MEDLINE | ID: mdl-37239938

ABSTRACT

Obesity and its associated metabolic morbidities have been and still are on the rise, posing a major challenge to health care systems worldwide. It has become evident over the last decades that a low-grade inflammatory response, primarily proceeding from the adipose tissue (AT), essentially contributes to adiposity-associated comorbidities, most prominently insulin resistance (IR), atherosclerosis and liver diseases. In mouse models, the release of pro-inflammatory cytokines such as TNF-alpha (TNF-α) and interleukin (IL)-1ß and the imprinting of immune cells to a pro-inflammatory phenotype in AT play an important role. However, the underlying genetic and molecular determinants are not yet understood in detail. Recent evidence demonstrates that nucleotide-binding and oligomerization domain (NOD)-like receptor (NLR) family proteins, a group of cytosolic pattern recognition receptors (PRR), contribute to the development and control of obesity and obesity-associated inflammatory responses. In this article, we review the current state of research on the role of NLR proteins in obesity and discuss the possible mechanisms leading to and the outcomes of NLR activation in the obesity-associated morbidities IR, type 2 diabetes mellitus (T2DM), atherosclerosis and non-alcoholic fatty liver disease (NAFLD) and discuss emerging ideas about possibilities for NLR-based therapeutic interventions of metabolic diseases.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Animals , Mice , Tumor Necrosis Factor-alpha/metabolism , Diabetes Mellitus, Type 2/genetics , Carrier Proteins , Insulin Resistance/genetics , NLR Proteins/metabolism , Obesity/metabolism , Morbidity , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism
9.
Int J Mol Sci ; 24(8)2023 Apr 13.
Article in English | MEDLINE | ID: mdl-37108368

ABSTRACT

Aggressive tumors evade cytotoxic T lymphocytes by suppressing MHC class-I (MHC-I) expression that also compromises tumor responsiveness to immunotherapy. MHC-I defects strongly correlate to defective expression of NLRC5, the transcriptional activator of MHC-I and antigen processing genes. In poorly immunogenic B16 melanoma cells, restoring NLRC5 expression induces MHC-I and elicits antitumor immunity, raising the possibility of using NLRC5 for tumor immunotherapy. As the clinical application of NLRC5 is constrained by its large size, we examined whether a smaller NLRC5-CIITA fusion protein, dubbed NLRC5-superactivator (NLRC5-SA) as it retains the ability to induce MHC-I, could be used for tumor growth control. We show that stable NLRC5-SA expression in mouse and human cancer cells upregulates MHC-I expression. B16 melanoma and EL4 lymphoma tumors expressing NLRC5-SA are controlled as efficiently as those expressing full-length NLRC5 (NLRC5-FL). Comparison of MHC-I-associated peptides (MAPs) eluted from EL4 cells expressing NLRC5-FL or NLRC5-SA and analyzed by mass spectrometry revealed that both NLRC5 constructs expanded the MAP repertoire, which showed considerable overlap but also included a substantial proportion of distinct peptides. Thus, we propose that NLRC5-SA, with its ability to increase tumor immunogenicity and promote tumor growth control, could overcome the limitations of NLRC5-FL for translational immunotherapy applications.


Subject(s)
Gene Expression Regulation , Melanoma, Experimental , Humans , Animals , Mice , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Genes, MHC Class I , Histocompatibility Antigens Class I , Antigen Presentation , Intracellular Signaling Peptides and Proteins/genetics
10.
Trends Immunol ; 40(10): 939-951, 2019 10.
Article in English | MEDLINE | ID: mdl-31500957

ABSTRACT

The mammalian innate immune system deals with invading pathogens and stress by activating pattern-recognition receptors (PRRs) in the host. Initially proposed to be triggered by the discrimination of defined molecular signatures from pathogens rather than from self, it is now clear that PRRs can also be activated by endogenous ligands, bacterial metabolites and, following pathogen-induced alterations of cellular processes, changes in the F-actin cytoskeleton. These processes are collectively referred to as effector-triggered immunity (ETI). Here, we summarize the molecular and conceptual advances in our understanding of cell autonomous innate immune responses against bacterial pathogens, and discuss how classical activation of PRRs and ETI interplay to drive inflammatory responses.


Subject(s)
Bacteria/immunology , Immunity, Innate/immunology , Inflammation/immunology , Receptors, Pattern Recognition/immunology , Animals , Humans , Inflammation/pathology
11.
Gastroenterology ; 159(1): 169-182.e8, 2020 07.
Article in English | MEDLINE | ID: mdl-32169428

ABSTRACT

BACKGROUND & AIMS: Helicobacter pylori induces strong inflammatory responses that are directed at clearing the infection, but if not controlled, these responses can be harmful to the host. We investigated the immune-regulatory effects of the innate immune molecule, nucleotide-binding oligomerization domain-like receptors (NLR) family CARD domain-containing 5 (NLRC5), in patients and mice with Helicobacter infection. METHODS: We obtained gastric biopsies from 30 patients in Australia. We performed studies with mice that lack NLRC5 in the myeloid linage (Nlrc5møKO) and mice without Nlrc5 gene disruption (controls). Some mice were gavaged with H pylori SS1 or Helicobacter felis; 3 months later, stomachs, spleens, and sera were collected, along with macrophages derived from bone marrow. Human and mouse gastric tissues and mouse macrophages were analyzed by histology, immunohistochemistry, immunoblots, and quantitative polymerase chain reaction. THP-1 cells (human macrophages, controls) and NLRC5-/- THP-1 cells (generated by CRISPR-Cas9 gene editing) were incubated with Helicobacter and gene expression and production of cytokines were analyzed. RESULTS: Levels of NLRC5 messenger RNA were significantly increased in gastric tissues from patients with H pylori infection, compared with patients without infection (P < .01), and correlated with gastritis severity (P < .05). H pylori bacteria induced significantly higher levels of chemokine and cytokine production by NLRC5-/- THP-1 macrophages than by control THP-1 cells (P < .05). After 3 months of infection with H felis, Nlrc5mø-KO mice developed gastric hyperplasia (P < .0001), splenomegaly (P < .0001), and increased serum antibody titers (P < .01), whereas control mice did not. Nlrc5mø-KO mice with chronic H felis infection had increased numbers of gastric B-cell follicles expressing CD19 (P < .0001); these follicles had features of mucosa-associated lymphoid tissue lymphoma. We identified B-cell-activating factor as a protein that promoted B-cell hyperproliferation in Nlrc5mø-KO mice. CONCLUSIONS: NLRC5 is a negative regulator of gastric inflammation and mucosal lymphoid formation in response to Helicobacter infection. Aberrant NLRC5 signaling in macrophages can promote B-cell lymphomagenesis during chronic Helicobacter infection.


Subject(s)
Helicobacter Infections/complications , Intracellular Signaling Peptides and Proteins/metabolism , Lymphoma, B-Cell, Marginal Zone/immunology , Stomach Neoplasms/immunology , Animals , B-Lymphocytes/immunology , Biopsy , Cell Proliferation , Disease Models, Animal , Female , Gastric Mucosa/immunology , Gastric Mucosa/microbiology , Gastric Mucosa/pathology , Gene Expression Regulation, Neoplastic/immunology , Gene Knockout Techniques , Helicobacter Infections/immunology , Helicobacter Infections/microbiology , Helicobacter Infections/pathology , Helicobacter felis/immunology , Helicobacter pylori/immunology , Humans , Hyperplasia/immunology , Hyperplasia/microbiology , Immunity, Innate , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Lymphoid Tissue/immunology , Lymphoid Tissue/microbiology , Lymphoid Tissue/pathology , Lymphoma, B-Cell, Marginal Zone/microbiology , Lymphoma, B-Cell, Marginal Zone/pathology , Male , Mice , Mice, Knockout , Signal Transduction/immunology , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , THP-1 Cells
12.
J Immunol ; 202(1): 218-227, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30510071

ABSTRACT

NOD-like receptors (NLR) are critical regulators of innate immune signaling. The NLR family consists of 22 human proteins with a conserved structure containing a central oligomerization NACHT domain, an N-terminal interaction domain, and a variable number of C-terminal leucine-rich repeats. Most NLR proteins function as cytosolic pattern recognition receptors with activation of downstream inflammasome signaling, NF-κB, or MAPK activation. Although NLRP10 is the only NLR protein lacking the leucine rich repeats, it has been implicated in multiple immune pathways, including the regulation of inflammatory responses toward Leishmania major and Shigella flexneri infection. In this study, we identify Abin-1, a negative regulator of NF-κB, as an interaction partner of NLRP10 that binds to the NACHT domain of NLRP10. Using S. flexneri as an infection model in human epithelial cells, our work reveals a novel function of NLRP10 in destabilizing Abin-1, resulting in enhanced proinflammatory signaling. Our data give insight into the molecular mechanism underlying the function of NLRP10 in innate immune responses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , DNA-Binding Proteins/metabolism , Dysentery, Bacillary/immunology , Epithelial Cells/physiology , Inflammation/immunology , Shigella flexneri/physiology , Animals , Apoptosis Regulatory Proteins/genetics , HEK293 Cells , Humans , Immunity, Innate , Inflammasomes/metabolism , Mice , Mice, Knockout , NF-kappa B/metabolism , Protein Binding , Protein Stability , Signal Transduction
13.
Int J Mol Sci ; 22(3)2021 Jan 20.
Article in English | MEDLINE | ID: mdl-33498269

ABSTRACT

Bacterial membrane vesicles (BMVs) are nanoparticles produced by both Gram-negative and Gram-positive bacteria that can function to modulate immunity in the host. Both outer membrane vesicles (OMVs) and membrane vesicles (MVs), which are released by Gram-negative and Gram-positive bacteria, respectively, contain cargo derived from their parent bacterium, including immune stimulating molecules such as proteins, lipids and nucleic acids. Of these, peptidoglycan (PG) and lipopolysaccharide (LPS) are able to activate host innate immune pattern recognition receptors (PRRs), known as NOD-like receptors (NLRs), such as nucleotide-binding oligomerisation domain-containing protein (NOD) 1, NOD2 and NLRP3. NLR activation is a key driver of inflammation in the host, and BMVs derived from both pathogenic and commensal bacteria have been shown to package PG and LPS in order to modulate the host immune response using NLR-dependent mechanisms. Here, we discuss the packaging of immunostimulatory cargo within OMVs and MVs, their detection by NLRs and the cytokines produced by host cells in response to their detection. Additionally, commensal derived BMVs are thought to shape immunity and contribute to homeostasis in the gut, therefore we also highlight the interactions of commensal derived BMVs with NLRs and their roles in limiting inflammatory diseases.


Subject(s)
Bacterial Outer Membrane/immunology , NLR Proteins/metabolism , Nanoparticles/chemistry , Adjuvants, Immunologic/administration & dosage , Animals , Bacterial Outer Membrane/chemistry , Humans , Immunity, Innate , Inflammasomes/immunology , Nanoparticles/metabolism
14.
Int J Mol Sci ; 22(3)2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33525590

ABSTRACT

Type I interferon signaling contributes to the development of innate and adaptive immune responses to either viruses, fungi, or bacteria. However, amplitude and timing of the interferon response is of utmost importance for preventing an underwhelming outcome, or tissue damage. While several pathogens evolved strategies for disturbing the quality of interferon signaling, there is growing evidence that this pathway can be regulated by several members of the Nod-like receptor (NLR) family, although the precise mechanism for most of these remains elusive. NLRs consist of a family of about 20 proteins in mammals, which are capable of sensing microbial products as well as endogenous signals related to tissue injury. Here we provide an overview of our current understanding of the function of those NLRs in type I interferon responses with a focus on viral infections. We discuss how NLR-mediated type I interferon regulation can influence the development of auto-immunity and the immune response to infection.


Subject(s)
Interferon Type I/metabolism , NLR Proteins/metabolism , Virus Diseases/metabolism , Adaptive Immunity , Animals , Humans , Immunity, Innate , Signal Transduction
15.
J Biol Chem ; 293(8): 2701-2710, 2018 02 23.
Article in English | MEDLINE | ID: mdl-29301940

ABSTRACT

Mammalian Nod-like receptor (NLR) proteins contribute to the regulation and induction of innate and adaptive immunity in mammals, although the function of about half of the currently identified NLR proteins remains poorly characterized. Here we analyzed the function of the primate-specific NLRP11 gene product. We show that NLRP11 is highly expressed in immune cells, including myeloid cells, B cells, and some B cell lymphoma lines. Overexpression of NLRP11 in human cells did not trigger key innate immune signaling pathways, including NF-κB and type I interferon responses. NLRP11 harbors a pyrin domain, which is responsible for inflammasome formation in related NLR proteins. However, NLRP11 did not interact with the inflammasome adaptor protein ASC, and it did not trigger caspase-1 activation. By contrast, expression of NLRP11 specifically repressed NF-κB and type I interferon responses, two key innate immune pathways involved in inflammation. This effect was independent of the pyrin domain and ATPase activity of NLRP11. siRNA-mediated knockdown of NLRP11 in human myeloid THP1 cells validated these findings and revealed enhanced lipopolysaccharide and Sendai virus-induced cytokine and interferon responses, respectively, in cells with reduced NLRP11 expression. In summary, our work identifies a novel role of NLRP11 in the regulation of inflammatory responses in human cells.


Subject(s)
B-Lymphocytes/metabolism , Down-Regulation , Gene Expression Regulation , Immunity, Innate , Intracellular Signaling Peptides and Proteins/metabolism , Myeloid Cells/metabolism , NLR Proteins/metabolism , Amino Acid Substitution , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Cell Line, Transformed , Cell Line, Tumor , Down-Regulation/drug effects , Female , Gene Expression Regulation/drug effects , Genes, Reporter/drug effects , Humans , Immunity, Innate/drug effects , Interferon Type I/agonists , Interferon Type I/antagonists & inhibitors , Interferon Type I/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Lipopolysaccharides/toxicity , Male , Mutation , Myeloid Cells/cytology , Myeloid Cells/drug effects , Myeloid Cells/immunology , NF-kappa B/agonists , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , NLR Proteins/genetics , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Organ Specificity , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , RNA Interference , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
16.
J Cell Sci ; 130(3): 648-657, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27980067

ABSTRACT

Nuclear factor (NF)-κB transcription factors play major roles in numerous biological processes including development and immunity. Here, we engineered a novel bi-directional NF-κB-responsive reporter, pSGNluc, in which a high-affinity NF-κB promoter fragment simultaneously drives expression of luciferase and GFP. Treatment with TNFα (also known as TNF) induced a strong, dose-dependent luciferase signal in cell culture. The degree of induction over background was comparable to that of other NF-κB-driven luciferase reporters, but the absolute level of expression was at least 20-fold higher. This extends the sensitivity range of otherwise difficult assays mediated exclusively by endogenously expressed receptors, as we show for Nod1 signaling in HEK293 cells. To measure NF-κB activity in the living organism, we established a transgenic zebrafish line carrying the pSGNluc construct. Live in toto imaging of transgenic embryos revealed the activation patterns of NF-κB signaling during embryonic development and as responses to inflammatory stimuli. Taken together, by integrating qualitative and quantitative NF-κB reporter activity, pSGNluc is a valuable tool for studying NF-κB signaling at high spatiotemporal resolution in cultured cells and living animals that goes beyond the possibilities provided by currently available reporters.


Subject(s)
Cell Culture Techniques/methods , Computer Systems , Genes, Reporter , NF-kappa B/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Base Sequence , Cytokines/metabolism , Embryo, Nonmammalian/metabolism , Embryonic Development/drug effects , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Inflammation/pathology , Luciferases/metabolism , Peptidoglycan/metabolism , Plasmids/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects , Tumor Necrosis Factor-alpha/pharmacology , Ultraviolet Rays , Zebrafish/embryology
17.
Semin Immunol ; 27(2): 75-84, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25911384

ABSTRACT

Recognition of bacterial pathogens by the mammalian host relies on the induction of early innate immune responses initiated by the activation of pattern-recognition receptors (PRRs) upon sensing of their cognate microbe-associated-patterns (MAMPs). Successful pathogens have evolved to intercept PRR activation and signaling at multiple steps. The molecular dissection of the underlying mechanisms revealed many of the basic mechanisms used by the immune system. Here we provide an overview of the different strategies used by bacterial pathogens and commensals to subvert and reprogram PPR-mediated innate immune responses. A particular attention is given to recent discoveries highlighting novel molecular details of the host inflammatory response in mammalian cells and current advances in our understanding of the interaction of commensals with PRR-mediated responses.


Subject(s)
Bacteria/immunology , Bacterial Infections/immunology , Immunity, Innate , Receptors, Pattern Recognition/immunology , Signal Transduction , Symbiosis , Animals , Autophagy , Bacteria/classification , Bacterial Infections/microbiology , Bacterial Physiological Phenomena , Cell Membrane , Humans
18.
EMBO J ; 33(19): 2171-87, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25056906

ABSTRACT

The X-linked inhibitor of apoptosis protein (XIAP) is a potent caspase inhibitor, best known for its anti-apoptotic function in cancer. During apoptosis, XIAP is antagonized by SMAC, which is released from the mitochondria upon caspase-mediated activation of BID. Recent studies suggest that XIAP is involved in immune signaling. Here, we explore XIAP as an important mediator of an immune response against the enteroinvasive bacterium Shigella flexneri, both in vitro and in vivo. Our data demonstrate for the first time that Shigella evades the XIAP-mediated immune response by inducing the BID-dependent release of SMAC from the mitochondria. Unlike apoptotic stimuli, Shigella activates the calpain-dependent cleavage of BID to trigger the release of SMAC, which antagonizes the inflammatory action of XIAP without inducing apoptosis. Our results demonstrate how the cellular death machinery can be subverted by an invasive pathogen to ensure bacterial colonization.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/physiology , Carrier Proteins/metabolism , Dysentery, Bacillary/immunology , Mitochondria/immunology , Mitochondrial Proteins/metabolism , Shigella/immunology , X-Linked Inhibitor of Apoptosis Protein/physiology , Animals , Apoptosis , Apoptosis Regulatory Proteins , Blotting, Western , Caspases/metabolism , Cell Proliferation , Cells, Cultured , Dysentery, Bacillary/microbiology , Dysentery, Bacillary/pathology , Female , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/pathology , Immunoenzyme Techniques , Integrases/metabolism , Male , Membrane Potential, Mitochondrial , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proteins/immunology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Shigella/pathogenicity , Signal Transduction , Spectrometry, Mass, Electrospray Ionization , Tandem Mass Spectrometry
19.
Eur J Immunol ; 46(8): 1959-69, 2016 08.
Article in English | MEDLINE | ID: mdl-27221772

ABSTRACT

The nucleotide binding and oligomerization domain-like receptor (NLR) protein NLRP10 is highly expressed in the epidermis and contributes to cell-autonomous responses against invasive bacteria. To investigate the role of NLRP10 in inflammatory responses of the skin we analyzed the effect of full-body and keratinocyte-specific depletion of NLRP10 in croton oil-induced irritant contact dermatitis (ICD) and 1-fluoro-2,4-dinitrobenzene (DNFB)-induced contact hypersensitivity (CHS) in mice. Nlrp10(-/-) mice were phenotypically normal and skin repair after wounding was not affected by lack of NLRP10. Similarly, we did not detect a contribution of NLRP10 to the ICD response induced by croton oil. In contrast, Nlrp10(-/-) mice showed significantly reduced inflammation in the DNFB-induced CHS response as compared to control animals. Microscopic analysis revealed significantly reduced numbers of CD4(+) and CD8(+) T cells in the infiltrates of animals lacking NLRP10 expression after CHS challenge. Epidermis-specific deletion of Nlrp10 by keratin-14 promotor driven Cre-recombinase was sufficient to account for this phenotype, although lymphocyte recruitment seemed to be unaltered in animals lacking NLRP10 expression in keratinocytes. Taken together, we provide evidence that NLRP10 contributes to T-cell-mediated inflammatory responses in the skin and highlight a physiological role of NLRP10 in epidermal keratinocytes.


Subject(s)
Apoptosis Regulatory Proteins/immunology , CD8-Positive T-Lymphocytes/immunology , Dermatitis, Contact/immunology , Dinitrofluorobenzene/adverse effects , Epidermis/immunology , Adaptor Proteins, Signal Transducing , Animals , Apoptosis Regulatory Proteins/genetics , Dermatitis, Contact/genetics , Dinitrofluorobenzene/administration & dosage , Disease Models, Animal , Epidermis/pathology , Inflammation/metabolism , Keratin-14/genetics , Keratinocytes/immunology , Mice , Mice, Knockout , Wound Healing
20.
Eur J Immunol ; 45(3): 758-72, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25404059

ABSTRACT

The NLR protein, NLRC5 is an important regulator of MHC class I gene expression, however, the role of NLRC5 in other innate immune responses is less well defined. In the present study, we report that NLRC5 binds RIG-I and that this interaction is critical for robust antiviral responses against influenza virus. Overexpression of NLRC5 in the human lung epithelial cell line, A549, and normal human bronchial epithelial cells resulted in impaired replication of influenza virus A/Puerto Rico/8/34 virus (PR8) and enhanced IFN-ß expression. Influenza virus leads to induction of IFN-ß that drives RIG-I and NLRC5 expression in host cells. Our results suggest that NLRC5 extends and stabilizes influenza virus induced RIG-I expression and delays expression of the viral inhibitor protein NS1. We show that NS1 binds to NLRC5 to suppress its function. Interaction domain mapping revealed that NLRC5 interacts with RIG-I via its N-terminal death domain and that NLRC5 enhanced antiviral activity in an leucine-rich repeat domain independent manner. Taken together, our findings identify a novel role for NLRC5 in RIG-I-mediated antiviral host responses against influenza virus infection, distinguished from the role of NLRC5 in MHC class I gene regulation.


Subject(s)
DEAD-box RNA Helicases/immunology , Gene Expression Regulation/immunology , Influenza A virus/immunology , Influenza, Human/immunology , Intracellular Signaling Peptides and Proteins/immunology , Respiratory Mucosa/immunology , DEAD Box Protein 58 , Epithelial Cells/immunology , Epithelial Cells/pathology , Epithelial Cells/virology , HEK293 Cells , Histocompatibility Antigens Class I/immunology , Humans , Influenza, Human/pathology , Protein Structure, Tertiary , Receptors, Immunologic , Respiratory Mucosa/pathology , Respiratory Mucosa/virology
SELECTION OF CITATIONS
SEARCH DETAIL