Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Eur J Immunol ; 52(2): 285-296, 2022 02.
Article in English | MEDLINE | ID: mdl-34694641

ABSTRACT

The upregulation of interferon (IFN)-inducible GTPases in response to pathogenic insults is vital to host defense against many bacterial, fungal, and viral pathogens. Several IFN-inducible GTPases play key roles in mediating inflammasome activation and providing host protection after bacterial or fungal infections, though their role in inflammasome activation after viral infection is less clear. Among the IFN-inducible GTPases, the expression of immunity-related GTPases (IRGs) varies widely across species for unknown reasons. Here, we report that IRGB10, but not IRGM1, IRGM2, or IRGM3, is required for NLRP3 inflammasome activation in response to influenza A virus (IAV) infection in mice. While IRGB10 functions to release inflammasome ligands in the context of bacterial and fungal infections, we found that IRGB10 facilitates endosomal maturation and nuclear translocation of IAV, thereby regulating viral replication. Corresponding with our in vitro results, we found that Irgb10-/- mice were more resistant to IAV-induced mortality than WT mice. The results of our study demonstrate a detrimental role of IRGB10 in host immunity in response to IAV and a novel function of IRGB10, but not IRGMs, in promoting viral translocation into the nucleus.


Subject(s)
GTP Phosphohydrolases/immunology , Inflammasomes/immunology , Influenza A Virus, H1N1 Subtype/physiology , Orthomyxoviridae Infections/immunology , Virus Replication/immunology , Animals , GTP Phosphohydrolases/genetics , Inflammasomes/genetics , Mice , Mice, Knockout , Orthomyxoviridae Infections/genetics , Virus Replication/genetics
2.
Life Sci Alliance ; 5(1)2022 01.
Article in English | MEDLINE | ID: mdl-34764207

ABSTRACT

The integrated stress response (ISR) regulates cellular homeostasis and cell survival following exposure to stressors. Cell death processes such as apoptosis and pyroptosis are known to be modulated by stress responses, but the role of the ISR in necroptosis is poorly understood. Necroptosis is an inflammatory, lytic form of cell death driven by the RIPK3-MLKL signaling axis. Here, we show that macrophages that have induced the ISR are protected from subsequent necroptosis. Consistent with a reduction in necroptosis, phosphorylation of RIPK1, RIPK3, and MLKL is reduced in macrophages pre-treated with ISR-inducing agents that are challenged with necroptosis-inducing triggers. The stress granule component DDX3X, which is involved in ISR-mediated regulation of pyroptosis, is not required for protecting ISR-treated cells from necroptosis. Disruption of stress granule assembly or knockdown of Perk restored necroptosis in pre-stressed cells. Together, these findings identify a critical role for the ISR in limiting necroptosis in macrophages.


Subject(s)
Macrophages/metabolism , Necroptosis , Stress, Physiological , Animals , Cell Survival/genetics , Cell Survival/immunology , DEAD-box RNA Helicases/metabolism , Endoplasmic Reticulum Stress , Fibroblasts , Gene Knockdown Techniques , Macrophages/immunology , Mice , Necroptosis/genetics , Necroptosis/immunology , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Stress Granules/metabolism , Toll-Like Receptors , eIF-2 Kinase
3.
Front Immunol ; 12: 701341, 2021.
Article in English | MEDLINE | ID: mdl-34777335

ABSTRACT

The essential micronutrient Selenium (Se) is co-translationally incorporated as selenocysteine into proteins. Selenoproteins contain one or more selenocysteines and are vital for optimum immunity. Interestingly, many pathogenic bacteria utilize Se for various biological processes suggesting that Se may play a role in bacterial pathogenesis. A previous study had speculated that Francisella tularensis, a facultative intracellular bacterium and the causative agent of tularemia, sequesters Se by upregulating Se-metabolism genes in type II alveolar epithelial cells. Therefore, we investigated the contribution of host vs. pathogen-associated selenoproteins in bacterial disease using F. tularensis as a model organism. We found that F. tularensis was devoid of any Se utilization traits, neither incorporated elemental Se, nor exhibited Se-dependent growth. However, 100% of Se-deficient mice (0.01 ppm Se), which express low levels of selenoproteins, succumbed to F. tularensis-live vaccine strain pulmonary challenge, whereas 50% of mice on Se-supplemented (0.4 ppm Se) and 25% of mice on Se-adequate (0.1 ppm Se) diet succumbed to infection. Median survival time for Se-deficient mice was 8 days post-infection while Se-supplemented and -adequate mice was 11.5 and >14 days post-infection, respectively. Se-deficient macrophages permitted significantly higher intracellular bacterial replication than Se-supplemented macrophages ex vivo, corroborating in vivo observations. Since Francisella replicates in alveolar macrophages during the acute phase of pneumonic infection, we hypothesized that macrophage-specific host selenoproteins may restrict replication and systemic spread of bacteria. F. tularensis infection led to an increased expression of several macrophage selenoproteins, suggesting their key role in limiting bacterial replication. Upon challenge with F. tularensis, mice lacking selenoproteins in macrophages (TrspM) displayed lower survival and increased bacterial burden in the lung and systemic tissues in comparison to WT littermate controls. Furthermore, macrophages from TrspM mice were unable to restrict bacterial replication ex vivo in comparison to macrophages from littermate controls. We herein describe a novel function of host macrophage-specific selenoproteins in restriction of intracellular bacterial replication. These data suggest that host selenoproteins may be considered as novel targets for modulating immune response to control a bacterial infection.


Subject(s)
Francisella tularensis/immunology , Host-Pathogen Interactions/immunology , Macrophages/immunology , Macrophages/metabolism , Selenoproteins/metabolism , Tularemia/etiology , Tularemia/metabolism , Animals , Disease Models, Animal , Disease Susceptibility , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Mice , Pneumonia/immunology , Pneumonia/metabolism , Pneumonia/microbiology , Pneumonia/pathology , Tularemia/mortality , Virulence/genetics , Virulence Factors/genetics
4.
mBio ; 12(3): e0105921, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34154417

ABSTRACT

Burkholderia infections can result in serious diseases with high mortality, such as melioidosis, and they are difficult to treat with antibiotics. Innate immunity is critical for cell-autonomous clearance of intracellular pathogens like Burkholderia by regulating programmed cell death. Inflammasome-dependent inflammatory cytokine release and cell death contribute to host protection against Burkholderia pseudomallei and Burkholderia thailandensis; however, the contribution of apoptosis and necroptosis to protection is not known. Here, we found that bone marrow-derived macrophages (BMDMs) lacking key components of pyroptosis died via apoptosis during infection. BMDMs lacking molecules required for pyroptosis, apoptosis, and necroptosis (PANoptosis), however, were significantly resistant to B. thailandensis-induced cell death until later stages of infection. Consequently, PANoptosis-deficient BMDMs failed to limit B. thailandensis-induced cell-cell fusion, which permits increased intercellular spread and replication compared to wild-type or pyroptosis-deficient BMDMs. Respiratory B. thailandensis infection resulted in higher mortality in PANoptosis-deficient mice than in pyroptosis-deficient mice, indicating that, in the absence of pyroptosis, apoptosis is essential for efficient control of infection in vivo. Together, these findings suggest both pyroptosis and apoptosis are necessary for host-mediated control of Burkholderia infection. IMPORTANCEBurkholderia infections result in a high degree of mortality when left untreated; therefore, understanding the host immune response required to control infection is critical. In this study, we found a hierarchical cell death program utilized by infected cells to disrupt the intracellular niche of Burkholderia thailandensis, which limits bacterial intercellular spread, host cell-cell fusion, and bacterial replication. In macrophages, combined loss of key PANoptosis components results in extensive B. thailandensis infection-induced cell-cell fusion, bacterial replication, and increased cell death at later stages of infection compared with both wild-type (WT) and pyroptosis-deficient cells. During respiratory infection, mortality was increased in PANoptosis-deficient mice compared to pyroptosis-deficient mice, identifying an essential role for multiple cell death pathways in controlling B. thailandensis infection. These findings advance our understanding of the physiological role of programmed cell death in controlling Burkholderia infection.


Subject(s)
Apoptosis/immunology , Burkholderia Infections/immunology , Burkholderia/pathogenicity , Immunity, Innate , Macrophages/microbiology , Macrophages/pathology , Animals , Burkholderia/immunology , Caspases/classification , Caspases/genetics , Caspases/immunology , Female , Male , Mice , Necroptosis/immunology , Pyroptosis/immunology
5.
J Immunol ; 207(1): 115-124, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34145059

ABSTRACT

Cellular stress can induce cytoplasmic ribonucleoprotein complexes called stress granules that allow the cells to survive. Stress granules are also central to cellular responses to infections, in which they can act as platforms for viral sensing or modulate innate immune signaling through pattern recognition receptors. However, the effect of innate immune signaling on stress granules is poorly understood. In this study, we report that prior induction of innate immune signaling through TLRs inhibited stress granule assembly in a TLR ligand dose-dependent manner in murine bone marrow-derived macrophages. Time course analysis suggests that TLR stimulation can reverse stress granule assembly even after it has begun. Additionally, both MYD88- and TRIF-mediated TLR signaling inhibited stress granule assembly in response to endoplasmic reticulum stress in bone marrow-derived macrophages and the chemotherapeutic drug oxaliplatin in murine B16 melanoma cells. This inhibition was not due to a decrease in expression of the critical stress granule proteins G3BP1 and DDX3X and was independent of IRAK1/4, JNK, ERK and P38 kinase activity but dependent on IKK complex kinase activity. Overall, we have identified the TLR-IKK complex signaling axis as a regulator of stress granule assembly-disassembly dynamics, highlighting cross-talk between processes that are critical in health and disease.


Subject(s)
I-kappa B Kinase/immunology , Immunity, Innate/immunology , Stress Granules/immunology , Toll-Like Receptors/immunology , Animals , Cells, Cultured , I-kappa B Kinase/genetics , Mice , Mice, Knockout , Signal Transduction/immunology
6.
Microorganisms ; 9(5)2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33946283

ABSTRACT

Francisella tularensis (Ft) is a Gram-negative, facultative intracellular coccobacillus that is the etiological agent of tularemia. Interestingly, the disease tularemia has variable clinical presentations that are dependent upon the route of infection with Ft. Two of the most likely routes of Ft infection include intranasal and intradermal, which result in pneumonic and ulceroglandular tularemia, respectively. While there are several differences between these two forms of tularemia, the most notable disparity is between mortality rates: the mortality rate following pneumonic tularemia is over ten times that of the ulceroglandular disease. Understanding the differences between intradermal and intranasal Ft infections is important not only for clinical diagnoses and treatment but also for the development of a safe and effective vaccine. However, the immune correlates of protection against Ft, especially within the context of infection by disparate routes, are not yet fully understood. Recent advances in different animal models have revealed new insights in the complex interplay of innate and adaptive immune responses, indicating dissimilar patterns in both responses following infection with Ft via different routes. Further investigation of these differences will be crucial to predicting disease outcomes and inducing protective immunity via vaccination or natural infection.

8.
J Biol Chem ; 296: 100579, 2021.
Article in English | MEDLINE | ID: mdl-33766561

ABSTRACT

Viruses and hosts have coevolved for millions of years, leading to the development of complex host-pathogen interactions. Influenza A virus (IAV) causes severe pulmonary pathology and is a recurrent threat to human health. Innate immune sensing of IAV triggers a complex chain of host responses. IAV has adapted to evade host defense mechanisms, and the host has coevolved to counteract these evasion strategies. However, the molecular mechanisms governing the balance between host defense and viral immune evasion is poorly understood. Here, we show that the host protein DEAD-box helicase 3 X-linked (DDX3X) is critical to orchestrate a multifaceted antiviral innate response during IAV infection, coordinating the activation of the nucleotide-binding oligomerization domain-like receptor with a pyrin domain 3 (NLRP3) inflammasome, assembly of stress granules, and type I interferon (IFN) responses. DDX3X activated the NLRP3 inflammasome in response to WT IAV, which carries the immune evasive nonstructural protein 1 (NS1). However, in the absence of NS1, DDX3X promoted the formation of stress granules that facilitated efficient activation of type I IFN signaling. Moreover, induction of DDX3X-containing stress granules by external stimuli after IAV infection led to increased type I IFN signaling, suggesting that NS1 actively inhibits stress granule-mediated host responses and DDX3X-mediated NLRP3 activation counteracts this action. Furthermore, the loss of DDX3X expression in myeloid cells caused severe pulmonary pathogenesis and morbidity in IAV-infected mice. Together, our findings show that DDX3X orchestrates alternate modes of innate host defense which are critical to fight against NS1-mediated immune evasion strategies during IAV infection.


Subject(s)
DEAD-box RNA Helicases/metabolism , Immunity, Innate , Inflammasomes/metabolism , Influenza A virus/physiology , Interferon Type I/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Influenza A virus/immunology , Mice
9.
Nat Commun ; 12(1): 496, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33479228

ABSTRACT

Chronic inflammation during many diseases is associated with bone loss. While interferons (IFNs) are often inhibitory to osteoclast formation, the complex role that IFN and interferon-stimulated genes (ISGs) play in osteoimmunology during inflammatory diseases is still poorly understood. We show that mice deficient in IFN signaling components including IFN alpha and beta receptor 1 (IFNAR1), interferon regulatory factor 1 (IRF1), IRF9, and STAT1 each have reduced bone density and increased osteoclastogenesis compared to wild type mice. The IFN-inducible guanylate-binding proteins (GBPs) on mouse chromosome 3 (GBP1, GBP2, GBP3, GBP5, GBP7) are required to negatively regulate age-associated bone loss and osteoclastogenesis. Mechanistically, GBP2 and GBP5 both negatively regulate in vitro osteoclast differentiation, and loss of GBP5, but not GBP2, results in greater age-associated bone loss in mice. Moreover, mice deficient in GBP5 or chromosome 3 GBPs have greater LPS-mediated inflammatory bone loss compared to wild type mice. Overall, we find that GBP5 contributes to restricting age-associated and inflammation-induced bone loss by negatively regulating osteoclastogenesis.


Subject(s)
Bone Resorption/metabolism , GTP-Binding Proteins/metabolism , Interferons/metabolism , Osteoclasts/metabolism , Osteogenesis/physiology , Age Factors , Animals , Bone Resorption/genetics , Cell Differentiation/genetics , Cell Fusion , Cells, Cultured , GTP-Binding Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Osteoclasts/cytology , Osteogenesis/genetics , Signal Transduction/genetics
12.
Curr Opin Microbiol ; 59: 42-49, 2021 02.
Article in English | MEDLINE | ID: mdl-32829024

ABSTRACT

The immune system has evolved multiple mechanisms to restrict microbial infections and regulate inflammatory responses. Without appropriate regulation, infection-induced inflammatory pathology can be deadly. The innate immune system recognizes the microbial molecules conserved in many pathogens and engages a rapid response by producing inflammatory mediators and activating programmed cell death pathways, including pyroptosis, apoptosis, and necroptosis. Activation of pattern recognition receptors, in combination with inflammatory cytokine-induced signaling through death domain-containing receptors, initiates a highly interconnected cell death process called PANoptosis (pyroptosis, apoptosis, necroptosis). Broadly speaking, PANoptosis is critical for restricting a wide range of pathogens (including bacteria, viruses, fungi, and parasites), which we describe in this review. We propose that re-examining the role of cell death and inflammatory cytokines through the lens of PANoptosis will advance our understanding of host-pathogen evolution and may reveal new treatment strategies for controlling a wide range of infectious diseases.


Subject(s)
Apoptosis , Cell Death , Host-Pathogen Interactions , Infections , Necroptosis , Pyroptosis , Apoptosis/immunology , Bacterial Physiological Phenomena/immunology , Biological Evolution , Cell Death/physiology , Fungi/physiology , Host-Pathogen Interactions/immunology , Infections/immunology , Infections/microbiology , Infections/virology , Necroptosis/immunology , Pyroptosis/immunology , Virus Physiological Phenomena/immunology
13.
Nature ; 588(7839): 688-692, 2020 12.
Article in English | MEDLINE | ID: mdl-33268895

ABSTRACT

Inflammasomes are important sentinels of innate immune defence that are activated in response to diverse stimuli, including pathogen-associated molecular patterns (PAMPs)1. Activation of the inflammasome provides host defence against aspergillosis2,3, which is a major health concern for patients who are immunocompromised. However, the Aspergillus fumigatus PAMPs that are responsible for inflammasome activation are not known. Here we show that the polysaccharide galactosaminogalactan (GAG) of A. fumigatus is a PAMP that activates the NLRP3 inflammasome. The binding of GAG to ribosomal proteins inhibited cellular translation machinery, and thus activated the NLRP3 inflammasome. The galactosamine moiety bound to ribosomal proteins and blocked cellular translation, which triggered activation of the NLRP3 inflammasome. In mice, a GAG-deficient Aspergillus mutant (Δgt4c) did not elicit protective activation of the inflammasome, and this strain exhibited enhanced virulence. Moreover, administration of GAG protected mice from colitis induced by dextran sulfate sodium in an inflammasome-dependent manner. Thus, ribosomes connect the sensing of this fungal PAMP to the activation of an innate immune response.


Subject(s)
Aspergillosis/prevention & control , Aspergillus fumigatus/metabolism , Inflammasomes/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , Polysaccharides/metabolism , Animals , Aspergillosis/immunology , Aspergillosis/microbiology , Aspergillus fumigatus/immunology , Biofilms , Colitis/chemically induced , Colitis/prevention & control , Dextran Sulfate , Female , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Deletion , Immunity, Innate , Inflammasomes/immunology , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Polysaccharides/immunology , Protein Biosynthesis , Ribosomal Proteins/metabolism , Ribosomes/metabolism
14.
Curr Opin Immunol ; 67: 95-105, 2020 12.
Article in English | MEDLINE | ID: mdl-33242752

ABSTRACT

The innate immune system, the first line of defense against pathogens and host tissue damage, initiates pro-inflammatory responses which, when dysregulated, promote inflammation to drive a broad range of autoimmune diseases. Immunomodulatory therapies have been developed to successfully treat several autoimmune diseases, but still many others lack effective treatments. Here, we explore recent advances in how the innate immune system contributes to autoinflammation, from the innate immune sensors that initiate immune responses to how this system regulates the activation of programmed cell death pathways including pyroptosis, apoptosis, necroptosis, and PANoptosis, which involves machinery from the pyroptotic, apoptotic, and necroptotic pathways. Recent advances in our understanding of innate immunity raise important considerations for developing new inflammatory disease treatments that target innate immune signaling and programmed cell death pathways.


Subject(s)
Autoimmune Diseases/immunology , Immune System/immunology , Immunity, Innate/immunology , Inflammation/immunology , Animals , Humans
15.
Article in English | MEDLINE | ID: mdl-32547960

ABSTRACT

Programmed cell death plays crucial roles in organismal development and host defense. Recent studies have highlighted mechanistic overlaps and extensive, multifaceted crosstalk between pyroptosis, apoptosis, and necroptosis, three programmed cell death pathways traditionally considered autonomous. The growing body of evidence, in conjunction with the identification of molecules controlling the concomitant activation of all three pathways by pathological triggers, has led to the development of the concept of PANoptosis. During PANoptosis, inflammatory cell death occurs through the collective activation of pyroptosis, apoptosis, and necroptosis, which can circumvent pathogen-mediated inhibition of individual death pathways. Many of the molecular details of this emerging pathway are unclear. Here, we describe the activation of PANoptosis by bacterial and viral triggers and report protein interactions that reveal the formation of a PANoptosome complex. Infection of macrophages with influenza A virus, vesicular stomatitis virus, Listeria monocytogenes, or Salmonella enterica serovar Typhimurium resulted in robust cell death and the hallmarks of PANoptosis activation. Combined deletion of the PANoptotic components caspase-1 (CASP1), CASP11, receptor-interacting serine/threonine-protein kinase 3 (RIPK3), and CASP8 largely protected macrophages from cell death induced by these pathogens, while deletion of individual components provided reduced or no protection. Further, molecules from the pyroptotic, apoptotic, and necroptotic cell death pathways interacted to form a single molecular complex that we have termed the PANoptosome. Overall, our study identifies pathogens capable of activating PANoptosis and the formation of a PANoptosome complex.


Subject(s)
Apoptosis , Necroptosis , Pyroptosis , Animals , Caspase 1 , Caspase 8 , Caspases, Initiator , Influenza A virus , Listeria monocytogenes , Macrophages , Mice , Receptor-Interacting Protein Serine-Threonine Kinases , Salmonella typhimurium , Vesicular stomatitis Indiana virus
16.
Cell Res ; 30(4): 315-327, 2020 04.
Article in English | MEDLINE | ID: mdl-32152420

ABSTRACT

Inflammasomes are multi-component signaling complexes critical to the initiation of pyroptotic cell death in response to invading pathogens and cellular damage. A number of innate immune receptors have been reported to serve as inflammasome sensors. Activation of these sensors leads to the proteolytic activation of caspase-1, a proinflammatory caspase responsible for the cleavage of proinflammatory cytokines interleukin-1ß and interleukin-18 and the effector of pyroptotic cell death, gasdermin D. Though crucial to the innate immune response to infection, dysregulation of inflammasome activation can lead to the development of inflammatory diseases, neurodegeneration, and cancer. Therefore, clinical interest in the modulation of inflammasome activation is swiftly growing. As such, it is imperative to develop a mechanistic understanding of the regulation of these complexes. In this review, we divide the regulation of inflammasome activation into three parts. We discuss the transcriptional regulation of inflammasome components and related proteins, the post-translational mechanisms of inflammasome activation, and advances in the understanding of the structural basis of inflammasome activation.


Subject(s)
Immunity, Innate/immunology , Inflammasomes/metabolism , Pyroptosis/immunology , Animals , Caspase 1/metabolism , Gene Expression Regulation , Humans , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphate-Binding Proteins/metabolism , Signal Transduction
17.
PLoS Pathog ; 16(3): e1008364, 2020 03.
Article in English | MEDLINE | ID: mdl-32150572

ABSTRACT

Innate immunity responds to pathogens by producing alarm signals and activating pathways that make host cells inhospitable for pathogen replication. The intracellular bacterium Burkholderia thailandensis invades the cytosol, hijacks host actin, and induces cell fusion to spread to adjacent cells, forming multinucleated giant cells (MNGCs) which promote bacterial replication. We show that type I interferon (IFN) restricts macrophage MNGC formation during B. thailandensis infection. Guanylate-binding proteins (GBPs) expressed downstream of type I IFN were required to restrict MNGC formation through inhibition of bacterial Arp2/3-dependent actin motility during infection. GTPase activity and the CAAX prenylation domain were required for GBP2 recruitment to B. thailandensis, which restricted bacterial actin polymerization required for MNGC formation. Consistent with the effects in in vitro macrophages, Gbp2-/-, Gbp5-/-, GbpChr3-KO mice were more susceptible to intranasal infection with B. thailandensis than wildtype mice. Our findings reveal that IFN and GBPs play a critical role in restricting cell-cell fusion and bacteria-induced pathology during infection.


Subject(s)
Burkholderia Infections/immunology , Burkholderia/immunology , GTP-Binding Proteins/immunology , Giant Cells/immunology , Macrophages/immunology , Nose Diseases/immunology , Protein Prenylation/immunology , Animals , Burkholderia Infections/genetics , Burkholderia Infections/pathology , Cell Fusion , GTP-Binding Proteins/genetics , Giant Cells/microbiology , Giant Cells/pathology , Interferon Type I/genetics , Interferon Type I/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , Nose Diseases/genetics , Nose Diseases/microbiology , Nose Diseases/pathology
18.
Physiology (Bethesda) ; 35(2): 112-124, 2020 03 01.
Article in English | MEDLINE | ID: mdl-32027562

ABSTRACT

The innate immune system recognizes conserved pathogen-associated molecular patterns and produces inflammatory cytokines that direct downstream immune responses. The inappropriate localization of DNA within the cell cytosol or endosomal compartments indicates that a cell may either be infected by a DNA virus or bacterium, or has problems with its own nuclear integrity. This DNA is sensed by certain receptors that mediate cytokine production and, in some cases, initiate an inflammatory and lytic form of cell death called pyroptosis. Dysregulation of these DNA-sensing pathways is thought to contribute to autoimmune diseases and the development of cancer. In this review, we will discuss the DNA sensors Toll-like receptor 9 (TLR9), cyclic GMP-AMP synthase (cGAS), stimulator of interferon genes (STING), absent in melanoma 2 (AIM2), and interferon gamma-inducible 16 (IFI16), their ligands, and their physiological significance. We will also examine the less-well-understood DEAH- and DEAD-box helicases DHX9, DHX36, DDX41, and RNA polymerase III, each of which may play an important role in DNA-mediated innate immunity.


Subject(s)
Cytokines/metabolism , DEAD-box RNA Helicases/metabolism , DNA/immunology , Immunity, Innate/immunology , Neoplasm Proteins/metabolism , Toll-Like Receptor 9/metabolism , Animals , DNA/genetics , DNA/metabolism , Humans , Neoplasm Proteins/genetics , Signal Transduction
19.
Nature ; 573(7775): 590-594, 2019 09.
Article in English | MEDLINE | ID: mdl-31511697

ABSTRACT

The cellular stress response has a vital role in regulating homeostasis by modulating cell survival and death. Stress granules are cytoplasmic compartments that enable cells to survive various stressors. Defects in the assembly and disassembly of stress granules are linked to neurodegenerative diseases, aberrant antiviral responses and cancer1-5. Inflammasomes are multi-protein heteromeric complexes that sense molecular patterns that are associated with damage or intracellular pathogens, and assemble into cytosolic compartments known as ASC specks to facilitate the activation of caspase-1. Activation of inflammasomes induces the secretion of interleukin (IL)-1ß and IL-18 and drives cell fate towards pyroptosis-a form of programmed inflammatory cell death that has major roles in health and disease6-12. Although both stress granules and inflammasomes can be triggered by the sensing of cellular stress, they drive contrasting cell-fate decisions. The crosstalk between stress granules and inflammasomes and how this informs cell fate has not been well-studied. Here we show that the induction of stress granules specifically inhibits NLRP3 inflammasome activation, ASC speck formation and pyroptosis. The stress granule protein DDX3X interacts with NLRP3 to drive inflammasome activation. Assembly of stress granules leads to the sequestration of DDX3X, and thereby the inhibition of NLRP3 inflammasome activation. Stress granules and the NLRP3 inflammasome compete for DDX3X molecules to coordinate the activation of innate responses and subsequent cell-fate decisions under stress conditions. Induction of stress granules or loss of DDX3X in the myeloid compartment leads to a decrease in the production of inflammasome-dependent cytokines in vivo. Our findings suggest that macrophages use the availability of DDX3X to interpret stress signals and choose between pro-survival stress granules and pyroptotic ASC specks. Together, our data demonstrate the role of DDX3X in driving NLRP3 inflammasome and stress granule assembly, and suggest a rheostat-like mechanistic paradigm for regulating live-or-die cell-fate decisions under stress conditions.


Subject(s)
Cell Death/genetics , DEAD-box RNA Helicases/metabolism , Inflammasomes/genetics , Macrophages/cytology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Stress, Physiological/genetics , Animals , Cell Line , Cell Survival/genetics , DEAD-box RNA Helicases/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , HEK293 Cells , Humans , Inflammasomes/immunology , Macrophages/immunology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics
20.
J Exp Med ; 216(7): 1474-1486, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31186281

ABSTRACT

Targeting apoptosis to treat diseases has seen tremendous success over the past decades. More recently, alternative forms of regulated cell death, including pyroptosis and necroptosis, have been described. Understanding the molecular cascades regulating both pyroptosis and necroptosis will yield even more targets to treat diseases. These lytic forms of cell death are distinct from apoptosis due to their characteristic lysis and release of cellular components that promote disease or direct a beneficial immune response. In this review, we focus on how pyroptosis and necroptosis, which release potent immune cytokines such as IL-1 and IL-18, contribute to various diseases. We also consider the important role that the executioners of these cell death pathways, GSDMD and MLKL, play in the progression of inflammatory diseases. Crosstalk between the different cell death pathways likely plays a major role physiologically. New therapeutic strategies targeting these specific molecules hold enormous potential for managing inflammatory diseases.


Subject(s)
Cell Death , Cytokines/metabolism , Animals , Humans , Interleukin-1/metabolism , Necroptosis , Pyroptosis
SELECTION OF CITATIONS
SEARCH DETAIL
...