Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 17(8): 922-9, 2016 08.
Article in English | MEDLINE | ID: mdl-27270400

ABSTRACT

Deficiency in mevalonate kinase (MVK) causes systemic inflammation. However, the molecular mechanisms linking the mevalonate pathway to inflammation remain obscure. Geranylgeranyl pyrophosphate, a non-sterol intermediate of the mevalonate pathway, is the substrate for protein geranylgeranylation, a protein post-translational modification that is catalyzed by protein geranylgeranyl transferase I (GGTase I). Pyrin is an innate immune sensor that forms an active inflammasome in response to bacterial toxins. Mutations in MEFV (encoding human PYRIN) result in autoinflammatory familial Mediterranean fever syndrome. We found that protein geranylgeranylation enabled Toll-like receptor (TLR)-induced activation of phosphatidylinositol-3-OH kinase (PI(3)K) by promoting the interaction between the small GTPase Kras and the PI(3)K catalytic subunit p110δ. Macrophages that were deficient in GGTase I or p110δ exhibited constitutive release of interleukin 1ß that was dependent on MEFV but independent of the NLRP3, AIM2 and NLRC4 inflammasomes. In the absence of protein geranylgeranylation, compromised PI(3)K activity allows an unchecked TLR-induced inflammatory responses and constitutive activation of the Pyrin inflammasome.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Familial Mediterranean Fever/metabolism , Inflammasomes/metabolism , Macrophages/physiology , Mutation/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pyrin/genetics , Alkyl and Aryl Transferases/genetics , Animals , Cells, Cultured , Familial Mediterranean Fever/genetics , Humans , Immunity, Innate , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Polyisoprenyl Phosphates/metabolism , Protein Processing, Post-Translational , Signal Transduction , Toll-Like Receptors/metabolism
2.
Nat Immunol ; 16(3): 229-36, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25689443

ABSTRACT

Alzheimer's disease (AD) is the world's most common dementing illness, affecting over 150 million patients. Classically AD has been viewed as a neurodegenerative disease of the elderly, characterized by the extracellular deposition of misfolded amyloid-ß (Aß) peptide and the intracellular formation of neurofibrillary tangles. Only recently has neuroinflammation emerged as an important component of AD pathology. Experimental, genetic and epidemiological data now indicate a crucial role for activation of the innate immune system as a disease-promoting factor. The sustained formation and deposition of Aß aggregates causes chronic activation of the immune system and disturbance of microglial clearance functions. Here we review advances in the molecular understanding of the inflammatory response in AD that point to novel therapeutic approaches for the treatment of this devastating disease.


Subject(s)
Alzheimer Disease/immunology , Immunity, Innate/immunology , Animals , Humans , Inflammation/immunology
3.
Cell ; 148(6): 1284-92, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22424235

ABSTRACT

Although a great public heath success, vaccines provide suboptimal protection in some patient populations and are not available to protect against many infectious diseases. Insights from innate immunity research have led to a better understanding of how existing vaccines work and have informed vaccine development. New adjuvants and delivery systems are being designed based upon their capacity to stimulate innate immune sensors and target antigens to dendritic cells, the cells responsible for initiating adaptive immune responses. Incorporating these adjuvants and delivery systems in vaccines can beneficially alter the quantitative and qualitative nature of the adaptive immune response, resulting in enhanced protection.


Subject(s)
Immunity, Innate , Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Communicable Disease Control , Communicable Diseases/immunology , Empiricism , Humans , Vaccines/therapeutic use
4.
Nat Immunol ; 14(8): 812-20, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23812099

ABSTRACT

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


Subject(s)
Alzheimer Disease/immunology , Atherosclerosis/immunology , CD36 Antigens/immunology , Carrier Proteins/immunology , Diabetes Mellitus, Type 2/immunology , Inflammation/immunology , Animals , CD36 Antigens/genetics , Carrier Proteins/genetics , Inflammasomes/immunology , Interleukin-1beta/immunology , Lipoproteins, LDL/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Fluorescence , NLR Family, Pyrin Domain-Containing 3 Protein , RNA/chemistry , RNA/genetics , Real-Time Polymerase Chain Reaction
5.
Nature ; 575(7784): 669-673, 2019 11.
Article in English | MEDLINE | ID: mdl-31748742

ABSTRACT

Alzheimer's disease is characterized by the accumulation of amyloid-beta in plaques, aggregation of hyperphosphorylated tau in neurofibrillary tangles and neuroinflammation, together resulting in neurodegeneration and cognitive decline1. The NLRP3 inflammasome assembles inside of microglia on activation, leading to increased cleavage and activity of caspase-1 and downstream interleukin-1ß release2. Although the NLRP3 inflammasome has been shown to be essential for the development and progression of amyloid-beta pathology in mice3, the precise effect on tau pathology remains unknown. Here we show that loss of NLRP3 inflammasome function reduced tau hyperphosphorylation and aggregation by regulating tau kinases and phosphatases. Tau activated the NLRP3 inflammasome and intracerebral injection of fibrillar amyloid-beta-containing brain homogenates induced tau pathology in an NLRP3-dependent manner. These data identify an important role of microglia and NLRP3 inflammasome activation in the pathogenesis of tauopathies and support the amyloid-cascade hypothesis in Alzheimer's disease, demonstrating that neurofibrillary tangles develop downstream of amyloid-beta-induced microglial activation.


Subject(s)
Inflammasomes/metabolism , Microglia/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , tau Proteins/metabolism , Animals , Cyclin-Dependent Kinase 5/metabolism , Gene Expression Regulation/genetics , Humans , Inflammasomes/genetics , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Phosphorylation , Protein Aggregation, Pathological/physiopathology , tau Proteins/genetics
6.
Hepatology ; 78(1): 225-242, 2023 07 01.
Article in English | MEDLINE | ID: mdl-36862512

ABSTRACT

BACKGROUND AIMS: Prolonged systemic inflammation contributes to poor clinical outcomes in severe alcohol-associated hepatitis (AH) even after the cessation of alcohol use. However, mechanisms leading to this persistent inflammation remain to be understood. APPROACH RESULTS: We show that while chronic alcohol induces nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome activation in the liver, alcohol binge results not only in NLRP3 inflammasome activation but also in increased circulating extracellular apoptosis-associated speck-like protein containing a caspase recruitment domain (ex-ASC) specks and hepatic ASC aggregates both in patients with AH and in mouse models of AH. These ex-ASC specks persist in circulation even after the cessation of alcohol use. Administration of alcohol-induced-ex-ASC specks in vivo in alcohol-naive mice results in sustained inflammation in the liver and circulation and causes liver damage. Consistent with the key role of ex-ASC specks in mediating liver injury and inflammation, alcohol binge failed to induce liver damage or IL-1ß release in ASC-deficient mice. Our data show that alcohol induces ex-ASC specks in liver macrophages and hepatocytes, and these ex-ASC specks can trigger IL-1ß release in alcohol-naive monocytes, a process that can be prevented by the NLRP3 inhibitor, MCC950. In vivo administration of MCC950 reduced hepatic and ex-ASC specks, caspase-1 activation, IL-1ß production, and steatohepatitis in a murine model of AH. CONCLUSIONS: Our study demonstrates the central role of NLRP3 and ASC in alcohol-induced liver inflammation and unravels the critical role of ex-ASC specks in the propagation of systemic and liver inflammation in AH. Our data also identify NLRP3 as a potential therapeutic target in AH.


Subject(s)
Hepatitis, Alcoholic , Hepatitis , Animals , Mice , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Hepatitis/etiology , Inflammation , Hepatitis, Alcoholic/etiology , Ethanol/adverse effects , Caspase 1/metabolism , Interleukin-1beta/metabolism , CARD Signaling Adaptor Proteins/metabolism
7.
Immunol Rev ; 293(1): 163-189, 2020 01.
Article in English | MEDLINE | ID: mdl-31642531

ABSTRACT

Plasmodium vivax infection, the predominant cause of malaria in Asia and Latin America, affects ~14 million individuals annually, with considerable adverse effects on wellbeing and socioeconomic development. A clinical hallmark of Plasmodium infection, the paroxysm, is driven by pyrogenic cytokines produced during the immune response. Here, we review studies on the role of specific immune cell types, cognate innate immune receptors, and inflammatory cytokines on parasite control and disease symptoms. This review also summarizes studies on recurrent infections in individuals living in endemic regions as well as asymptomatic infections, a serious barrier to eliminating this disease. We propose potential mechanisms behind these repeated and subclinical infections, such as poor induction of immunological memory cells and inefficient T effector cells. We address the role of antibody-mediated resistance to P. vivax infection and discuss current progress in vaccine development. Finally, we review immunoregulatory mechanisms, such as inhibitory receptors, T regulatory cells, and the anti-inflammatory cytokine, IL-10, that antagonizes both innate and acquired immune responses, interfering with the development of protective immunity and parasite clearance. These studies provide new insights for the clinical management of symptomatic as well as asymptomatic individuals and the development of an efficacious vaccine for vivax malaria.


Subject(s)
Host-Parasite Interactions/immunology , Immunity , Malaria, Vivax/immunology , Malaria, Vivax/parasitology , Plasmodium vivax/physiology , Adaptive Immunity , Antibodies, Protozoan/immunology , Antigens, Protozoan/immunology , Cytokines/metabolism , Disease Susceptibility , Host-Parasite Interactions/genetics , Humans , Immunity, Innate , Inflammation Mediators/metabolism , Malaria Vaccines/immunology , Malaria, Vivax/genetics , Malaria, Vivax/metabolism , Plasmodium vivax/growth & development , Toll-Like Receptors/metabolism
8.
Nature ; 552(7685): 355-361, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29293211

ABSTRACT

The spreading of pathology within and between brain areas is a hallmark of neurodegenerative disorders. In patients with Alzheimer's disease, deposition of amyloid-ß is accompanied by activation of the innate immune system and involves inflammasome-dependent formation of ASC specks in microglia. ASC specks released by microglia bind rapidly to amyloid-ß and increase the formation of amyloid-ß oligomers and aggregates, acting as an inflammation-driven cross-seed for amyloid-ß pathology. Here we show that intrahippocampal injection of ASC specks resulted in spreading of amyloid-ß pathology in transgenic double-mutant APPSwePSEN1dE9 mice. By contrast, homogenates from brains of APPSwePSEN1dE9 mice failed to induce seeding and spreading of amyloid-ß pathology in ASC-deficient APPSwePSEN1dE9 mice. Moreover, co-application of an anti-ASC antibody blocked the increase in amyloid-ß pathology in APPSwePSEN1dE9 mice. These findings support the concept that inflammasome activation is connected to seeding and spreading of amyloid-ß pathology in patients with Alzheimer's disease.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , CARD Signaling Adaptor Proteins/metabolism , Microglia/metabolism , Protein Aggregation, Pathological , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/genetics , Animals , Antibodies/administration & dosage , Antibodies/immunology , Antibodies/pharmacology , CARD Signaling Adaptor Proteins/antagonists & inhibitors , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/immunology , Female , Hippocampus/cytology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Presenilin-1/deficiency , Presenilin-1/genetics , Protein Domains , Spatial Memory/physiology
9.
Nat Immunol ; 11(2): 155-61, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20037584

ABSTRACT

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


Subject(s)
CD36 Antigens/immunology , Inflammation/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Toll-Like Receptor 6/immunology , Amyloid beta-Peptides/immunology , Animals , Atherosclerosis/immunology , Atherosclerosis/metabolism , Blotting, Western , CD36 Antigens/metabolism , Cell Line , Chemokines/biosynthesis , Chemokines/immunology , Gene Expression , Humans , Immunoprecipitation , Inflammation/metabolism , Lipoproteins, LDL/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Microglia/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 6/metabolism
10.
Gastroenterology ; 159(6): 2181-2192.e1, 2020 12.
Article in English | MEDLINE | ID: mdl-32841647

ABSTRACT

BACKGROUND & AIMS: Clostridioides difficile toxin A (TcdA) activates the innate immune response. TcdA co-purifies with DNA. Toll-like receptor 9 (TLR9) recognizes bacterial DNA to initiate inflammation. We investigated whether DNA bound to TcdA activates an inflammatory response in murine models of C difficile infection via activation of TLR9. METHODS: We performed studies with human colonocytes and monocytes and macrophages from wild-type and TLR9 knockout mice incubated with TcdA or its antagonist (ODN TTAGGG) or transduced with vectors encoding TLR9 or small-interfering RNAs. Cytokine production was measured with enzyme-linked immunosorbent assay. We studied a transduction domain of TcdA (TcdA57-80), which was predicted by machine learning to have cell-penetrating activity and confirmed by synchrotron small-angle X-ray scattering. Intestines of CD1 mice, C57BL6J mice, and mice that express a form of TLR9 that is not activated by CpG DNA were injected with TcdA, TLR9 antagonist, or both. Enterotoxicity was estimated based on loop weight to length ratios. A TLR9 antagonist was tested in mice infected with C difficile. We incubated human colon explants with an antagonist of TLR9 and measured TcdA-induced production of cytokines. RESULTS: The TcdA57-80 protein transduction domain had membrane remodeling activity that allowed TcdA to enter endosomes. TcdA-bound DNA entered human colonocytes. TLR9 was required for production of cytokines by cultured cells and in human colon explants incubated with TcdA. TLR9 was required in TcdA-induced mice intestinal secretions and in the survival of mice infected by C difficile. Even in a protease-rich environment, in which only fragments of TcdA exist, the TcdA57-80 domain organized DNA into a geometrically ordered structure that activated TLR9. CONCLUSIONS: TcdA from C difficile can bind and organize bacterial DNA to activate TLR9. TcdA and TcdA fragments remodel membranes, which allows them to access endosomes and present bacterial DNA to and activate TLR9. Rather than inactivating the ability of DNA to bind TLR9, TcdA appears to chaperone and organize DNA into an inflammatory, spatially periodic structure.


Subject(s)
Bacterial Toxins/metabolism , Clostridioides difficile/immunology , Clostridium Infections/immunology , Colitis/immunology , Enterotoxins/metabolism , Toll-Like Receptor 9/metabolism , Animals , Anti-Bacterial Agents/adverse effects , Clostridioides difficile/genetics , Clostridioides difficile/metabolism , Clostridium Infections/chemically induced , Clostridium Infections/microbiology , Colitis/chemically induced , Colitis/microbiology , DNA, Bacterial/metabolism , Disease Models, Animal , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Humans , Immunity, Innate , Mice , Mice, Knockout , Molecular Chaperones/metabolism , Signal Transduction/immunology , Toll-Like Receptor 9/genetics
11.
PLoS Pathog ; 15(3): e1007684, 2019 03.
Article in English | MEDLINE | ID: mdl-30883606

ABSTRACT

Phagocytosis is a complex process that eliminates microbes and is performed by specialised cells such as macrophages. Toll-like receptor 4 (TLR4) is expressed on the surface of macrophages and recognizes Gram-negative bacteria. Moreover, TLR4 has been suggested to play a role in the phagocytosis of Gram-negative bacteria, but the mechanisms remain unclear. Here we have used primary human macrophages and engineered THP-1 monocytes to show that the TLR4 sorting adapter, TRAM, is instrumental for phagocytosis of Escherichia coli as well as Staphylococcus aureus. We find that TRAM forms a complex with Rab11 family interacting protein 2 (FIP2) that is recruited to the phagocytic cups of E. coli. This promotes activation of the actin-regulatory GTPases Rac1 and Cdc42. Our results show that FIP2 guided TRAM recruitment orchestrates actin remodelling and IRF3 activation, two events that are both required for phagocytosis of Gram-negative bacteria.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Carrier Proteins/metabolism , Membrane Proteins/metabolism , Phagocytosis/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Carrier Proteins/physiology , Endocytosis , Endosomes , Escherichia coli/pathogenicity , HEK293 Cells , Humans , Interferon Regulatory Factor-3 , Lipopolysaccharides , Macrophages/immunology , Macrophages/metabolism , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88 , Primary Cell Culture , Protein Transport , Signal Transduction , Staphylococcus aureus/pathogenicity , THP-1 Cells , Toll-Like Receptor 4/metabolism , cdc42 GTP-Binding Protein , rab GTP-Binding Proteins , rac1 GTP-Binding Protein
12.
Proc Natl Acad Sci U S A ; 115(36): 9002-9007, 2018 09 04.
Article in English | MEDLINE | ID: mdl-30127003

ABSTRACT

Alzheimer's disease (AD) is characterized by the progressive destruction and dysfunction of central neurons. AD patients commonly have unprovoked seizures compared with age-matched controls. Amyloid peptide-related inflammation is thought to be an important aspect of AD pathogenesis. We previously reported that NLRP3 inflammasome KO mice, when bred into APPswe/PS1ΔE9 (APP/PS1) mice, are completely protected from amyloid-induced AD-like disease, presumably because they cannot produce mature IL1ß or IL18. To test the role of IL18, we bred IL18KO mice with APP/PS1 mice. Surprisingly, IL18KO/APP/PS1 mice developed a lethal seizure disorder that was completely reversed by the anticonvulsant levetiracetam. IL18-deficient AD mice showed a lower threshold in chemically induced seizures and a selective increase in gene expression related to increased neuronal activity. IL18-deficient AD mice exhibited increased excitatory synaptic proteins, spine density, and basal excitatory synaptic transmission that contributed to seizure activity. This study identifies a role for IL18 in suppressing aberrant neuronal transmission in AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid/metabolism , Inflammasomes/metabolism , Interleukin-18/metabolism , Seizures/metabolism , Synaptic Transmission , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid/genetics , Animals , Inflammasomes/genetics , Interleukin-18/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Levetiracetam , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Piracetam/analogs & derivatives , Piracetam/pharmacology , Seizures/drug therapy , Seizures/genetics , Seizures/pathology
13.
Proc Natl Acad Sci U S A ; 115(28): 7404-7409, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29954866

ABSTRACT

Cerebral malaria (CM) is a serious neurological complication caused by Plasmodium falciparum infection. Currently, the only treatment for CM is the provision of antimalarial drugs; however, such treatment by itself often fails to prevent death or development of neurological sequelae. To identify potential improved treatments for CM, we performed a nonbiased whole-brain transcriptomic time-course analysis of antimalarial drug chemotherapy of murine experimental CM (ECM). Bioinformatics analyses revealed IL33 as a critical regulator of neuroinflammation and cerebral pathology that is down-regulated in the brain during fatal ECM and in the acute period following treatment of ECM. Consistent with this, administration of IL33 alongside antimalarial drugs significantly improved the treatment success of established ECM. Mechanistically, IL33 treatment reduced inflammasome activation and IL1ß production in microglia and intracerebral monocytes in the acute recovery period following treatment of ECM. Moreover, treatment with the NLRP3-inflammasome inhibitor MCC950 alongside antimalarial drugs phenocopied the protective effect of IL33 therapy in improving the recovery from established ECM. We further showed that IL1ß release from macrophages was stimulated by hemozoin and antimalarial drugs and that this was inhibited by MCC950. Our results therefore demonstrate that manipulation of the IL33-NLRP3 axis may be an effective therapy to suppress neuroinflammation and improve the efficacy of antimalarial drug treatment of CM.


Subject(s)
Antimalarials/pharmacology , Brain/parasitology , Drug Delivery Systems/methods , Interleukin-33/metabolism , Malaria, Cerebral/drug therapy , Malaria, Falciparum/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Plasmodium falciparum/metabolism , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Gene Expression Profiling , Hemeproteins/metabolism , Interleukin-1beta/biosynthesis , Interleukin-33/antagonists & inhibitors , Macrophages/metabolism , Macrophages/pathology , Malaria, Cerebral/metabolism , Malaria, Cerebral/pathology , Malaria, Falciparum/metabolism , Malaria, Falciparum/pathology , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Transcriptome/drug effects
14.
Nat Immunol ; 9(8): 857-65, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18604209

ABSTRACT

The fibrillar peptide amyloid-beta (A beta) has a chief function in the pathogenesis of Alzheimer's disease. Interleukin 1 beta (IL-1 beta) is a key cytokine in the inflammatory response to A beta. Insoluble materials such as crystals activate the inflammasome formed by the cytoplasmic receptor NALP3, which results in the release of IL-1 beta. Here we identify the NALP3 inflammasome as a sensor of A beta in a process involving the phagocytosis of A beta and subsequent lysosomal damage and release of cathepsin B. Furthermore, the IL-1 beta pathway was essential for the microglial synthesis of proinflammatory and neurotoxic factors, and the inflammasome, caspase-1 and IL-1 beta were critical for the recruitment of microglia to exogenous A beta in the brain. Our findings suggest that activation of the NALP3 inflammasome is important for inflammation and tissue damage in Alzheimer's disease.


Subject(s)
Alzheimer Disease/immunology , Amyloid beta-Peptides/immunology , Immunity, Innate/immunology , Inflammation/metabolism , Carrier Proteins/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation Mediators/physiology , NLR Family, Pyrin Domain-Containing 3 Protein
15.
Immunity ; 35(2): 194-207, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21820332

ABSTRACT

Although Toll-like receptor 9 (TLR9) has been implicated in cytokine and type I interferon (IFN) production during malaria in humans and mice, the high AT content of the Plasmodium falciparum genome prompted us to examine the possibility that malarial DNA triggered TLR9-independent pathways. Over 6000 ATTTTTAC ("AT-rich") motifs are present in the genome of P. falciparum, which we show here potently induce type I IFNs. Parasite DNA, parasitized erythrocytes and oligonucleotides containing the AT-rich motif induce type I IFNs via a pathway that did not involve the previously described sensors TLR9, DAI, RNA polymerase-III or IFI16/p204. Rather, AT-rich DNA sensing involved an unknown receptor that coupled to the STING, TBK1 and IRF3-IRF7 signaling pathway. Mice lacking IRF3, IRF7, the kinase TBK1 or the type I IFN receptor were resistant to otherwise lethal cerebral malaria. Collectively, these observations implicate AT-rich DNA sensing via STING, TBK1 and IRF3-IRF7 in P. falciparum malaria.


Subject(s)
AT Rich Sequence/genetics , DNA, Protozoan/genetics , Malaria, Falciparum/immunology , Oligonucleotides/genetics , Plasmodium falciparum/physiology , Animals , DNA, Protozoan/metabolism , Gene Expression Profiling , Humans , Immunity, Innate/genetics , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , Malaria, Falciparum/parasitology , Malaria, Falciparum/physiopathology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Oligonucleotides/metabolism , Plasmodium falciparum/pathogenicity , Protein Serine-Threonine Kinases/metabolism , Receptor, Interferon alpha-beta/genetics , Signal Transduction/genetics
16.
J Immunol ; 200(2): 768-774, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29212905

ABSTRACT

Innate immune receptors have a key role in the sensing of malaria and initiating immune responses. As a consequence of infection, systemic inflammation emerges and is directly related to signs and symptoms during acute disease. We have previously reported that plasmodial DNA is the primary driver of systemic inflammation in malaria, both within the phagolysosome and in the cytosol of effector cells. In this article, we demonstrate that Plasmodium falciparum genomic DNA delivered to the cytosol of human monocytes binds and activates cyclic GMP-AMP synthase (cGAS). Activated cGAS synthesizes 2'3'-cGAMP, which we subsequently can detect using liquid chromatography-tandem mass spectrometry. 2'3'-cGAMP acts as a second messenger for STING activation and triggers TBK1/IRF3 activation, resulting in type I IFN production in human cells. This induction of type I IFN was independent of IFI16. Access of DNA to the cytosolic compartment is mediated by hemozoin, because incubation of purified malaria pigment with DNase abrogated IFN-ß induction. Collectively, these observations implicate cGAS as an important cytosolic sensor of P. falciparum genomic DNA and reveal the role of the cGAS/STING pathway in the induction of type I IFN in response to malaria parasites.


Subject(s)
DNA, Protozoan/metabolism , Interferon Type I/metabolism , Nucleotidyltransferases/metabolism , Plasmodium falciparum/genetics , Adolescent , Adult , Cells, Cultured , Erythrocytes/metabolism , Erythrocytes/parasitology , Female , Humans , Interferon Regulatory Factor-3/metabolism , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Male , Membrane Proteins/metabolism , Middle Aged , Nucleotides, Cyclic/metabolism , Phosphorylation , Signal Transduction , Young Adult
17.
J Immunol ; 200(4): 1243-1248, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29330325

ABSTRACT

Malarial infection in naive individuals induces a robust innate immune response. In the recently described model of innate immune memory, an initial stimulus primes the innate immune system to either hyperrespond (termed training) or hyporespond (tolerance) to subsequent immune challenge. Previous work in both mice and humans demonstrated that infection with malaria can both serve as a priming stimulus and promote tolerance to subsequent infection. In this study, we demonstrate that initial stimulation with Plasmodium falciparum-infected RBCs or the malaria crystal hemozoin induced human adherent PBMCs to hyperrespond to subsequent ligation of TLR2. This hyperresponsiveness correlated with increased H3K4me3 at important immunometabolic promoters, and these epigenetic modifications were also seen in Kenyan children naturally infected with malaria. However, the use of epigenetic and metabolic inhibitors indicated that the induction of trained immunity by malaria and its ligands may occur via a previously unrecognized mechanism(s).


Subject(s)
Immunity, Innate/immunology , Malaria, Falciparum/immunology , Child , Child, Preschool , Epigenesis, Genetic/immunology , Female , Gene Expression Regulation/immunology , Histones/biosynthesis , Humans , Infant , Malaria, Falciparum/metabolism , Male
18.
Nature ; 493(7434): 674-8, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23254930

ABSTRACT

Alzheimer's disease is the world's most common dementing illness. Deposition of amyloid-ß peptide drives cerebral neuroinflammation by activating microglia. Indeed, amyloid-ß activation of the NLRP3 inflammasome in microglia is fundamental for interleukin-1ß maturation and subsequent inflammatory events. However, it remains unknown whether NLRP3 activation contributes to Alzheimer's disease in vivo. Here we demonstrate strongly enhanced active caspase-1 expression in human mild cognitive impairment and brains with Alzheimer's disease, suggesting a role for the inflammasome in this neurodegenerative disease. Nlrp3(-/-) or Casp1(-/-) mice carrying mutations associated with familial Alzheimer's disease were largely protected from loss of spatial memory and other sequelae associated with Alzheimer's disease, and demonstrated reduced brain caspase-1 and interleukin-1ß activation as well as enhanced amyloid-ß clearance. Furthermore, NLRP3 inflammasome deficiency skewed microglial cells to an M2 phenotype and resulted in the decreased deposition of amyloid-ß in the APP/PS1 model of Alzheimer's disease. These results show an important role for the NLRP3/caspase-1 axis in the pathogenesis of Alzheimer's disease, and suggest that NLRP3 inflammasome inhibition represents a new therapeutic intervention for the disease.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Carrier Proteins/metabolism , Aged , Aged, 80 and over , Alzheimer Disease/enzymology , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Animals , Behavior, Animal , Brain/enzymology , Carrier Proteins/genetics , Caspase 1/genetics , Caspase 1/metabolism , Cognitive Dysfunction/enzymology , Cognitive Dysfunction/physiopathology , Gene Expression Regulation, Enzymologic , Humans , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Memory , Mice , Mice, Inbred C57BL , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein , Nitric Oxide Synthase Type II/metabolism , Phagocytosis/genetics
19.
J Biol Chem ; 292(14): 5634-5644, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28209713

ABSTRACT

Bacterial sepsis involves a complex interaction between the host immune response and bacterial LPS. LPS binds Toll-like receptor (TLR) 4, which leads to the release of proinflammatory cytokines that are essential for a potent innate immune response against pathogens. The innate immune system is tightly regulated, as excessive inflammation can lead to organ failure and death. MicroRNAs have recently emerged as important regulators of the innate immune system. Here we determined the function of miR-718, which is conserved across mammals and overlaps with the 5' UTR of the interleukin 1 receptor-associated kinase (IRAK1) gene. As IRAK1 is a key component of innate immune signaling pathways that are downstream of most TLRs, we hypothesized that miR-718 helps regulate the innate immune response. Activation of TLR4, but not TLR3, induced the expression of miR-718 in macrophages. miR-718 expression was also induced in the spleens of mice upon LPS injection. miR-718 modulates PI3K/Akt signaling by directly down-regulating phosphatase and tensin homolog (PTEN), thereby promoting phosphorylation of Akt, which leads to a decrease in proinflammatory cytokine production. Phosphorylated Akt induces let-7e expression, which, in turn, down-regulates TLR4 and further diminishes TLR4-mediated proinflammatory signals. Decreased miR-718 expression is associated with bacterial burden during Neisseria gonorrhoeae infection and alters the infection dynamics of N. gonorrhoeae in vitro Furthermore, miR-718 regulates the induction of LPS tolerance in macrophages. We propose a role for miR-718 in controlling TLR4 signaling and inflammatory cytokine signaling through a negative feedback regulation loop involving down-regulation of TLR4, IRAK1, and NF-κB.


Subject(s)
5' Untranslated Regions , Cytokines/metabolism , Macrophages/metabolism , MicroRNAs/metabolism , PTEN Phosphohydrolase/metabolism , Signal Transduction , Animals , Cytokines/genetics , Gonorrhea/genetics , Gonorrhea/metabolism , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Knockout , MicroRNAs/genetics , Neisseria gonorrhoeae/metabolism , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
20.
J Immunol ; 196(4): 1732-40, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26773149

ABSTRACT

Neisseria gonorrhoeae, the causative agent of the sexually transmitted infection gonorrhea, has developed resistance to almost every conventional antibiotic. There is an urgent need to develop novel therapies against gonorrhea. Many pathogens, including N. gonorrhoeae, bind the complement inhibitor factor H (FH) to evade complement-dependent killing. Sialylation of gonococcal lipooligosaccharide, as occurs in vivo, augments binding of human FH through its domains 18-20 (FH18-20). We explored the use of fusing FH18-20 with IgG Fc (FH18-20/Fc) to create a novel anti-infective immunotherapeutic. FH18-20 also binds to select host glycosaminoglycans to limit unwanted complement activation on host cells. To identify mutation(s) in FH18-20 that eliminated complement activation on host cells, yet maintained binding to N. gonorrhoeae, we created four mutations in domains 19 or 20 described in atypical hemolytic uremic syndrome that prevented binding of mutated fH to human erythrocytes. One of the mutant proteins (D to G at position 1119 in domain 19; FHD1119G/Fc) facilitated complement-dependent killing of gonococci similar to unmodified FH18-20/Fc but, unlike FH18-20/Fc, did not lyse human erythrocytes. FHD1119G/Fc bound to all (100%) of 15 sialylated clinical N. gonorrhoeae isolates tested (including three contemporary ceftriaxone-resistant strains), mediated complement-dependent killing of 10 of 15 (67%) strains, and enhanced C3 deposition (≥10-fold above baseline levels) on each of the five isolates not directly killed by complement. FHD1119G/Fc facilitated opsonophagocytic killing of a serum-resistant strain by human polymorphonuclear neutrophils. FHD1119G/Fc administered intravaginally significantly reduced the duration and burden of gonococcal infection in the mouse vaginal colonization model. FHD1119G/Fc represents a novel immunotherapeutic against multidrug-resistant N. gonorrhoeae.


Subject(s)
Complement Factor H/immunology , Gonorrhea/immunology , Immunoglobulin Fc Fragments/immunology , Immunotherapy/methods , Recombinant Fusion Proteins/immunology , Animals , Complement Factor H/pharmacology , Disease Models, Animal , Female , Flow Cytometry , Humans , Immunoglobulin Fc Fragments/pharmacology , Mice , Mice, Inbred BALB C , Neisseria gonorrhoeae/immunology , Recombinant Fusion Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL