Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Trends Immunol ; 40(6): 492-510, 2019 06.
Article in English | MEDLINE | ID: mdl-31053495

ABSTRACT

Long noncoding RNAs (lncRNAs) are key molecules that regulate gene expression in a variety of organisms. LncRNAs can drive different transcriptional and post-transcriptional events that impact cellular functions. Recent studies have identified many lncRNAs associated with immune cell development and activation; however, an understanding of their functional role in host immunity to infection is just emerging. Here, we provide a detailed and updated review of the functional roles of lncRNAs in regulating mammalian immune responses during host-pathogen interactions, because these functions may be either beneficial or detrimental to the host. With increased mechanistic insight into the roles of lncRNAs, it may be possible to design and/or improve lncRNA-based therapies to treat a variety of infectious and inflammatory diseases.


Subject(s)
Gene Expression Regulation , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunomodulation/genetics , RNA, Long Noncoding/genetics , Animals , Disease Resistance/genetics , Disease Resistance/immunology , Genome , Genome-Wide Association Study/methods , Genomics/methods , Humans
2.
Immunol Rev ; 266(1): 109-22, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26085210

ABSTRACT

Pellinos are a family of E3 ubiquitin ligases discovered for their role in catalyzing K63-linked polyubiquitination of Pelle, an interleukin-1 (IL-1) receptor-associated kinase homolog in the Drosophila Toll pathway. Subsequent studies have revealed the central and non-redundant roles of mammalian Pellino-1, Pellino-2, and Pelino-3 in signaling pathways emanating from IL-1 receptors, Toll-like receptors, NOD-like receptors, T- and B-cell receptors. While Pellinos ability to interact with many signaling intermediates suggested their scaffolding roles, recent findings in mice expressing ligase-inactive Pellinos demonstrated the importance of Pellino ubiquitin ligase activity. Cell-specific functions of Pellinos have emerged, e.g. Pellino-1 being a negative regulator in T lymphocytes and a positive regulator in myeloid cells, and details of molecular regulation of receptor signaling by various members of the Pellino family have been revealed. In this review, we summarize current information about Pellino-mediated regulation of signaling by pattern recognition receptors, T-cell and B-cell receptors and tumor necrosis factor receptors, and discuss Pellinos roles in sepsis and infectious diseases, as well as in autoimmune, inflammatory, and allergic disorders. We also provide our perspective on the potential of targeting Pellinos with peptide- or small molecule-based drug compounds as a new therapeutic approach for septic shock and autoimmune pathologies.


Subject(s)
B-Lymphocytes/immunology , Immunotherapy , Nuclear Proteins/metabolism , T-Lymphocytes/immunology , Ubiquitin-Protein Ligases/metabolism , Animals , Humans , Molecular Targeted Therapy , Organ Specificity , Receptors, Pattern Recognition/metabolism , Signal Transduction , Ubiquitination
3.
J Biol Chem ; 292(25): 10685-10695, 2017 06 23.
Article in English | MEDLINE | ID: mdl-28442574

ABSTRACT

Toll-like receptor 2 (TLR2) plays a critical role in host defenses against mycobacterial infections. The R753Q TLR2 polymorphism has been associated with increased incidence of tuberculosis and infections with non-tuberculous mycobacteria in human populations, but the mechanisms by which this polymorphism affects TLR2 signaling are unclear. In this study, we determined the impact of the R753Q TLR2 polymorphism on macrophage sensing of Mycobacterium smegmatis Upon infection with M. smegmatis, macrophages from knock-in mice harboring R753Q TLR2 expressed lower levels of TNF-α, IL-1ß, IL-6, and IL-10 compared with cells from WT mice, but both R753Q TLR2- and WT-derived macrophages exhibited comparable bacterial burdens. The decreased cytokine responses in R753Q TLR2-expressing macrophages were accompanied by impaired phosphorylation of IL-1R-associated kinase 1 (IRAK-1), p38, ERK1/2 MAPKs, and p65 NF-κB, suggesting that the R753Q TLR2 polymorphism alters the functions of the myeloid differentiation primary response protein 88 (MyD88)-IRAK-dependent signaling axis. Supporting this notion, HEK293 cells stably transfected with YFP-tagged R753Q TLR2 displayed reduced recruitment of MyD88 to TLR2, decreased NF-κB activation, and impaired IL-8 expression upon exposure to M. smegmatis Collectively, our results indicate that the R753Q polymorphism alters TLR2 signaling competence, leading to impaired MyD88-TLR2 assembly, reduced phosphorylation of IRAK-1, diminished activation of MAPKs and NF-κB, and deficient induction of cytokines in macrophages infected with M. smegmatis.


Subject(s)
Immunity, Innate , Macrophages/immunology , Mutation, Missense , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium smegmatis/immunology , Myeloid Differentiation Factor 88/immunology , Polymorphism, Genetic , Toll-Like Receptor 2/immunology , Amino Acid Substitution , Animals , Cytokines/genetics , Cytokines/immunology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , HEK293 Cells , Humans , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Mutant Strains , Mycobacterium Infections, Nontuberculous/genetics , Myeloid Differentiation Factor 88/genetics , Toll-Like Receptor 2/genetics
4.
Eur J Immunol ; 47(5): 880-891, 2017 05.
Article in English | MEDLINE | ID: mdl-28295231

ABSTRACT

Interleukin-1 receptor-associated kinase (IRAK) 4 mediates host defense against infections. As an active kinase, IRAK4 elicits full spectra of myeloid differentiation primary response protein (MyD) 88-dependent responses, while kinase-inactive IRAK4 induces a subset of cytokines and negative regulators whose expression is not regulated by mRNA stability. IRAK4 kinase activity is critical for resistance against Streptococcus pneumoniae, but its involvement in autoimmunity is incompletely understood. In this study, we determined the role of IRAK4 kinase activity in murine lupus. Lupus development in BXSB mice expressing the Y chromosome autoimmunity accelerator (Yaa) increased basal and Toll-like receptor (TLR) 4/7-induced phosphorylation of mitogen-activated protein kinases, p65 nuclear factor-κB (NF-κB), enhanced tumor necrosis factor (TNF)-α and C-C motif chemokine ligand (CCL) 5 gene expression in splenic macrophages, but decreased levels of Toll-interacting protein and IRAK-M, without affecting IRAK4 or IRAK1 expression. Mice harboring kinase-inactive IRAK4 on the lupus-prone Yaa background manifested blunted TLR signaling in macrophages and reduced glomerulonephritis, splenomegaly, serum anti-nuclear antibodies, numbers of splenic macrophages, total and TNF-α+ dendritic cells, activated T- and B-lymphocytes, and lower TNF-α expression in macrophages compared with lupus-prone mice with functional IRAK4. Thus, IRAK4 kinase activity contributes to murine lupus and could represent a new therapeutic target.


Subject(s)
Autoimmunity , Cytokines/immunology , Immunologic Deficiency Syndromes , Interleukin-1 Receptor-Associated Kinases/metabolism , Lupus Nephritis/immunology , Animals , Chemokine CCL5/genetics , Interleukin-1 Receptor-Associated Kinases/deficiency , Macrophages/immunology , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Phosphorylation , Primary Immunodeficiency Diseases , Signal Transduction , Spleen/cytology , Spleen/immunology , Toll-Like Receptors/immunology , Tumor Necrosis Factor-alpha/metabolism
5.
J Biol Chem ; 290(31): 19218-32, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26082489

ABSTRACT

Endotoxin tolerance reprograms Toll-like receptor (TLR) 4-mediated macrophage responses by attenuating induction of proinflammatory cytokines while retaining expression of anti-inflammatory and antimicrobial mediators. We previously demonstrated deficient TLR4-induced activation of IL-1 receptor-associated kinase (IRAK) 4, IRAK1, and TANK-binding kinase (TBK) 1 as critical hallmarks of endotoxin tolerance, but mechanisms remain unclear. In this study, we examined the role of the E3 ubiquitin ligase Pellino-1 in endotoxin tolerance and TLR signaling. LPS stimulation increased Pellino-1 mRNA and protein expression in macrophages from mice injected with saline and in medium-pretreated human monocytes, THP-1, and MonoMac-6 cells, whereas endotoxin tolerization abrogated LPS inducibility of Pellino-1. Overexpression of Pellino-1 in 293/TLR2 and 293/TLR4/MD2 cells enhanced TLR2- and TLR4-induced nuclear factor κB (NF-κB) and expression of IL-8 mRNA, whereas Pellino-1 knockdown reduced these responses. Pellino-1 ablation in THP-1 cells impaired induction of myeloid differentiation primary response protein (MyD88), and Toll-IL-1R domain-containing adapter inducing IFN-ß (TRIF)-dependent cytokine genes in response to TLR4 and TLR2 agonists and heat-killed Escherichia coli and Staphylococcus aureus, whereas only weakly affecting phagocytosis of heat-killed bacteria. Co-expressed Pellino-1 potentiated NF-κB activation driven by transfected MyD88, TRIF, IRAK1, TBK1, TGF-ß-activated kinase (TAK) 1, and TNFR-associated factor 6, whereas not affecting p65-induced responses. Mechanistically, Pellino-1 increased LPS-driven K63-linked polyubiquitination of IRAK1, TBK1, TAK1, and phosphorylation of TBK1 and IFN regulatory factor 3. These results reveal a novel mechanism by which endotoxin tolerance re-programs TLR4 signaling via suppression of Pellino-1, a positive regulator of MyD88- and TRIF-dependent signaling that promotes K63-linked polyubiquitination of IRAK1, TBK1, and TAK1.


Subject(s)
Nuclear Proteins/physiology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Ubiquitin-Protein Ligases/physiology , Animals , HEK293 Cells , Humans , Interleukin-1 Receptor-Associated Kinases/metabolism , Lipopolysaccharides/pharmacology , MAP Kinase Kinase Kinases/metabolism , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/metabolism , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6/metabolism , Transcriptional Activation/immunology , Ubiquitination
6.
J Immunol ; 188(9): 4506-15, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22474023

ABSTRACT

Asp(299)Gly (D299G) and, to a lesser extent, Thr(399)Ile (T399I) TLR4 polymorphisms have been associated with gram-negative sepsis and other infectious diseases, but the mechanisms by which they affect TLR4 signaling are unclear. In this study, we determined the impact of the D299G and T399I polymorphisms on TLR4 expression, interactions with myeloid differentiation factor 2 (MD2), LPS binding, and LPS-mediated activation of the MyD88- and Toll/IL-1R resistance domain-containing adapter inducing IFN-ß (TRIF) signaling pathways. Complementation of human embryonic kidney 293/CD14/MD2 transfectants with wild-type (WT) or mutant yellow fluorescent protein-tagged TLR4 variants revealed comparable total TLR4 expression, TLR4-MD2 interactions, and LPS binding. FACS analyses with anti-TLR4 Ab showed only minimal changes in the cell-surface levels of the D299G TLR4. Cells transfected with D299G TLR4 exhibited impaired LPS-induced phosphorylation of p38 and TANK-binding kinase 1, activation of NF-κB and IFN regulatory factor 3, and induction of IL-8 and IFN-ß mRNA, whereas T399I TLR4 did not cause statistically significant inhibition. In contrast to WT TLR4, expression of the D299G mutants in TLR4(-/-) mouse macrophages failed to elicit LPS-mediated induction of TNF-α and IFN-ß mRNA. Coimmunoprecipitation revealed diminished LPS-driven interaction of MyD88 and TRIF with the D299G TLR4 species, in contrast to robust adapter recruitment exhibited by WT TLR4. Thus, the D299G polymorphism compromises recruitment of MyD88 and TRIF to TLR4 without affecting TLR4 expression, TLR4-MD2 interaction, or LPS binding, suggesting that it interferes with TLR4 dimerization and assembly of intracellular docking platforms for adapter recruitment.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Macrophages/immunology , Myeloid Differentiation Factor 88/immunology , Polymorphism, Genetic/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cells, Cultured , HEK293 Cells , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Lipopolysaccharides/pharmacology , Lymphocyte Antigen 96/genetics , Lymphocyte Antigen 96/immunology , Lymphocyte Antigen 96/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/genetics , NF-kappa B/immunology , NF-kappa B/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Phosphorylation/immunology , Protein Kinases/genetics , Protein Kinases/immunology , Protein Kinases/metabolism , Protein Multimerization/genetics , Protein Multimerization/immunology , Signal Transduction/genetics , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism
7.
J Biol Chem ; 287(45): 38327-37, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-22992740

ABSTRACT

The R753Q polymorphism in the Toll-IL-1 receptor domain of Toll-like receptor 2 (TLR2) has been linked to increased incidence of tuberculosis and other infectious diseases, but the mechanisms by which it affects TLR2 functions are unclear. Here, we studied the impact of the R753Q polymorphism on TLR2 expression, hetero-dimerization with TLR6, tyrosine phosphorylation, and recruitment of myeloid differentiation primary response protein (MyD) 88 and MyD88 adapter-like (Mal). Complementation of HEK293 cells with transfected WT or R753Q TLR2 revealed their comparable total levels and only minimal changes in cell surface expression of the mutant species. Notably, even a 100-fold increase in amounts of transfected R753Q TLR2 versus WT variant did not overcome the compromised ability of the mutant TLR2 to activate nuclear factor κB (NF-κB), indicating that a minimal decrease in cell surface levels of the R753Q TLR2 cannot account for the signaling deficiency. Molecular modeling studies suggested that the R753Q mutation changes the electrostatic potential of the DD loop and results in a discrete movement of the residues critical for protein-protein interactions. Confirming these predictions, biochemical assays demonstrated that R753Q TLR2 exhibits deficient agonist-induced tyrosine phosphorylation, hetero-dimerization with TLR6, and recruitment of Mal and MyD88. These proximal signaling deficiencies correlated with impaired capacities of the R753Q TLR2 to mediate p38 phosphorylation, NF-κB activation, and induction of IL-8 mRNA in transfected HEK293 cells challenged with inactivated Mycobacterium tuberculosis or mycobacterial components. Thus, the R753Q polymorphism renders TLR2 signaling-incompetent by impairing its tyrosine phosphorylation, dimerization with TLR6, and recruitment of Mal and MyD88.


Subject(s)
Myeloid Differentiation Factor 88/metabolism , Polymorphism, Genetic , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 6/metabolism , Amino Acid Substitution/genetics , Amino Acid Substitution/immunology , Animals , Cells, Cultured , Gene Expression , HEK293 Cells , Humans , Immunoblotting , Interleukin-8/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Knockout , Microscopy, Confocal , Models, Molecular , Mycobacterium tuberculosis/immunology , Myeloid Differentiation Factor 88/genetics , NF-kappa B/metabolism , Phosphorylation , Protein Multimerization , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 2/chemistry , Toll-Like Receptor 6/chemistry , Toll-Like Receptor 6/genetics , Tyrosine/metabolism
8.
J Virol ; 86(13): 7216-26, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22532679

ABSTRACT

Lymphocytic choriomeningitis virus (LCMV), the prototype arenavirus, and Lassa virus (LASV), the causative agent of Lassa fever (LF), have extensive strain diversity and significant variations in pathogenicity for humans and experimental animals. The WE strain of LCMV (LCMV-WE), but not the Armstrong (Arm) strain, induces a fatal LF-like disease in rhesus macaques. We also demonstrated that LASV infection of human macrophages and endothelial cells resulted in reduced levels of proinflammatory cytokines. Here we have shown that cells infected with LASV or with LCMV-WE suppressed Toll-like receptor 2 (TLR2)-dependent proinflammatory cytokine responses. The persisting isolate LCMV clone 13 (CL13) also failed to stimulate interleukin-6 (IL-6) in macrophages. In contrast, nonpathogenic Mopeia virus, which is a genetic relative of LASV and LCMV-Arm induced robust responses that were TLR2/Mal dependent, required virus replication, and were enhanced by CD14. Superinfection experiments demonstrated that the WE strain of LCMV inhibited the Arm-mediated IL-8 response during the early stage of infection. In cells transfected with the NF-κB-luciferase reporter, infection with LCMV-Arm resulted in the induction of NF-κB, but cells infected with LCMV-WE and CL13 did not. These results suggest that pathogenic arenaviruses suppress NF-κB-mediated proinflammatory cytokine responses in infected cells.


Subject(s)
Cytokines/antagonists & inhibitors , Immune Evasion , Lassa virus/pathogenicity , Lymphocytic choriomeningitis virus/pathogenicity , Myelin Proteins/antagonists & inhibitors , Proteolipids/antagonists & inhibitors , Toll-Like Receptor 2/antagonists & inhibitors , Animals , Cell Line , Humans , Lassa virus/immunology , Lymphocytic choriomeningitis virus/immunology , Membrane Transport Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Proteins/immunology , Myelin and Lymphocyte-Associated Proteolipid Proteins , NF-kappa B/antagonists & inhibitors , NF-kappa B/immunology , Proteolipids/immunology , Toll-Like Receptor 2/immunology
9.
J Biol Chem ; 286(10): 7905-7916, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21220427

ABSTRACT

Endotoxin tolerance reprograms Toll-like receptor 4 responses by impairing LPS-elicited production of pro-inflammatory cytokines without inhibiting expression of anti-inflammatory or anti-microbial mediators. In septic patients, Toll-like receptor tolerance is thought to underlie decreased pro-inflammatory cytokine expression in response to LPS and increased incidence of microbial infections. The impact of endotoxin tolerance on recruitment, post-translational modifications and signalosome assembly of IL-1 receptor-associated kinase (IRAK) 4, IRAK1, TNF receptor-associated factor (TRAF) 6, TGF-ß-activated kinase (TAK) 1, and IκB kinase (IKK) γ is largely unknown. We report that endotoxin tolerization of THP1 cells and human monocytes impairs LPS-mediated receptor recruitment and activation of IRAK4, ablates K63-linked polyubiquitination of IRAK1 and TRAF6, compromises assembly of IRAK1-TRAF6 and IRAK1-IKKγ platforms, and inhibits TAK1 activation. Deficiencies in these signaling events in LPS-tolerant cells coincided with increased expression of A20, an essential deubiquitination enzyme, and sustained A20-IRAK1 associations. Overexpression of A20 inhibited LPS-induced activation of NF-κB and ablated NF-κB reporter activation driven by ectopic expression of MyD88, IRAK1, IRAK2, TRAF6, and TAK1/TAB1, while not affecting the responses induced by IKKß and p65. A20 shRNA knockdown abolished LPS tolerization of THP1 cells, mechanistically linking A20 and endotoxin tolerance. Thus, deficient LPS-induced activation of IRAK4 and TAK1, K63-linked polyubiquitination of IRAK1 and TRAF6, and disrupted IRAK1-TRAF6 and IRAK1-IKKγ assembly associated with increased A20 expression and A20-IRAK1 interactions are new determinants of endotoxin tolerance.


Subject(s)
Drug Resistance/drug effects , Gene Expression Regulation/drug effects , I-kappa B Kinase/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , Lipopolysaccharides/pharmacology , MAP Kinase Kinase Kinases/metabolism , Monocytes/metabolism , Nuclear Proteins/biosynthesis , TNF Receptor-Associated Factor 6/metabolism , Ubiquitination/drug effects , Cell Line , DNA-Binding Proteins , Humans , I-kappa B Kinase/genetics , Interleukin-1 Receptor-Associated Kinases/genetics , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Kinase Kinases/genetics , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Nuclear Proteins/genetics , TNF Receptor-Associated Factor 6/genetics , Tumor Necrosis Factor alpha-Induced Protein 3
10.
J Exp Med ; 218(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33216117

ABSTRACT

Two cosegregating single-nucleotide polymorphisms (SNPs) in human TLR4, an A896G transition at SNP rs4986790 (D299G) and a C1196T transition at SNP rs4986791 (T399I), have been associated with LPS hyporesponsiveness and differential susceptibility to many infectious or inflammatory diseases. However, many studies failed to confirm these associations, and transfection experiments resulted in conflicting conclusions about the impact of these SNPs on TLR4 signaling. Using advanced protein modeling from crystallographic data of human and murine TLR4, we identified homologous substitutions of these SNPs in murine Tlr4, engineered a knock-in strain expressing the D298G and N397I TLR4 SNPs homozygously, and characterized in vivo and in vitro responses to TLR4 ligands and infections in which TLR4 is implicated. Our data provide new insights into cellular and molecular mechanisms by which these SNPs decrease the TLR4 signaling efficiency and offer an experimental approach to confirm or refute human data possibly confounded by variables unrelated to the direct effects of the SNPs on TLR4 functionality.


Subject(s)
Lipopolysaccharides/genetics , Polymorphism, Single Nucleotide/genetics , Toll-Like Receptor 4/genetics , Animals , Disease Models, Animal , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Mice , Signal Transduction/genetics
11.
J Exp Med ; 198(4): 521-31, 2003 Aug 18.
Article in English | MEDLINE | ID: mdl-12925671

ABSTRACT

We identified previously a patient with recurrent bacterial infections who failed to respond to gram-negative LPS in vivo, and whose leukocytes were profoundly hyporesponsive to LPS and IL-1 in vitro. We now demonstrate that this patient also exhibits deficient responses in a skin blister model of aseptic inflammation. A lack of IL-18 responsiveness, coupled with diminished LPS and/or IL-1-induced nuclear factor-kappaB and activator protein-1 translocation, p38 phosphorylation, gene expression, and dysregulated IL-1R-associated kinase (IRAK)-1 activity in vitro support the hypothesis that the defect lies within the signaling pathway common to toll-like receptor 4, IL-1R, and IL-18R. This patient expresses a "compound heterozygous" genotype, with a point mutation (C877T in cDNA) and a two-nucleotide, AC deletion (620-621del in cDNA) encoded by distinct alleles of the IRAK-4 gene (GenBank/EMBL/DDBJ accession nos. AF445802 and AY186092). Both mutations encode proteins with an intact death domain, but a truncated kinase domain, thereby precluding expression of full-length IRAK-4 (i.e., a recessive phenotype). When overexpressed in HEK293T cells, neither truncated form augmented endogenous IRAK-1 kinase activity, and both inhibited endogenous IRAK-1 activity modestly. Thus, IRAK-4 is pivotal in the development of a normal inflammatory response initiated by bacterial or nonbacterial insults.


Subject(s)
Bacterial Infections/metabolism , Interleukin-1/immunology , Lipopolysaccharides/immunology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Point Mutation , Adult , Bacterial Infections/immunology , Cell Line , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Interleukin-1/metabolism , Interleukin-1 Receptor-Associated Kinases , Interleukin-18/immunology , Interleukin-18/metabolism , Leukocytes, Mononuclear/metabolism , Lipopolysaccharides/metabolism , Molecular Sequence Data , NF-kappa B/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Signal Transduction/physiology , Transcription Factor AP-1/metabolism
12.
Methods Mol Biol ; 517: 145-67, 2009.
Article in English | MEDLINE | ID: mdl-19378035

ABSTRACT

Toll-like receptors (TLRs) are principal innate immune sensors critically involved in the recognition of evolutionary conserved microbial and viral structures called "pathogen-associated molecular patterns" (PAMPs). Although recognition patterns of many TLRs have been characterized, molecular mechanisms that initiate TLR signaling are poorly understood. Since posttranslational modifications of many receptor systems are important in initiating signaling, we studied whether tyrosine phosphorylation of TLR4, the principal sensor of Gram-negative bacterial lipopolysaccharide (LPS) plays a role in TLR4 signal-transducing functions. We found that LPS induced TLR4 tyrosine phosphorylation and mutations of tyrosine residues in the Toll-IL-1R signaling domain markedly suppressed TLR4-mediated activation of JNK and p38 MAP kinases and transcription factors NF-kappaB, RANTES, and IFN-beta. This chapter summarizes a combination of methodological approaches that can be used to demonstrate an indispensable role of TLR4 tyrosine phosphorylation in receptor signaling, including transient transfections, site-directed mutagenesis, immunoprecipitation and immunoblot analyses, and analyses of transcription factor activation in reporter assays.


Subject(s)
Phosphotyrosine/metabolism , Toll-Like Receptor 4/metabolism , Blotting, Western , Cell Line , Genes, Reporter/genetics , Humans , Mutagenesis, Site-Directed , Phosphorylation , Phosphotyrosine/analysis , Phosphotyrosine/genetics , Plasmids/genetics , Signal Transduction , Toll-Like Receptor 4/genetics , Transgenes/genetics
13.
Article in English | MEDLINE | ID: mdl-32039056

ABSTRACT

Long non-coding RNAs (lncRNAs) have emerged as key regulators of gene expression in several biological systems. The long intergenic RNA-erythroid pro-survival (lincRNA-EPS) has been shown to play a critical role in restraining inflammatory gene expression. However, the function of lincRNA-EPS during bacterial infections remains unknown. Here, we demonstrate that following infection with the intracellular bacterium Listeria monocytogenes, both mouse macrophages and dendritic cells lacking lincRNA-EPS exhibit an enhanced expression of proinflammatory cytokine genes, as well as an increased expression of the inducible nitric oxide synthase (iNos) and nitric oxide (NO) production. Importantly, we found that lincRNA-EPS-/- mice intraperitoneally infected with L. monocytogenes exhibit lower bacterial burdens in spleen and liver and produce more NO than control mice. Furthermore, lincRNA-EPS-/- mice are less susceptible to a lethal dose of L. monocytogenes than wild type (WT) mice. Collectively these findings show that lincRNA-EPS suppresses host protective NO expression and impairs the host defense against L. monocytogenes infection.


Subject(s)
Cytokines/antagonists & inhibitors , Dendritic Cells/immunology , Down-Regulation , Listeria monocytogenes/immunology , Macrophages/immunology , Nitric Oxide Synthase Type II/antagonists & inhibitors , RNA, Long Noncoding/metabolism , Animals , Bacterial Load , Cells, Cultured , Cytokines/genetics , Disease Models, Animal , Immune Tolerance , Immunity, Innate , Listeriosis/immunology , Listeriosis/microbiology , Listeriosis/pathology , Mice, Inbred C57BL , Mice, Knockout , Models, Theoretical , Nitric Oxide Synthase Type II/genetics , RNA, Long Noncoding/genetics , Survival Analysis
14.
J Leukoc Biol ; 104(4): 811-820, 2018 10.
Article in English | MEDLINE | ID: mdl-29749650

ABSTRACT

IL-1 receptor-associated kinase (IRAK) 4 is a central enzyme of the TLR pathways. This study tested the hypothesis that IRAK4 kinase activity is prerequisite for regulating innate immunity during infections with intracellular bacteria. To this end, we analyzed responses of macrophages obtained from mice expressing wild-type (WT) IRAK4 or its kinase-inactive K213M mutant (IRAK4KI ) upon infection with intracellular bacteria Listeria monocytogenes or Mycobacterium smegmatis. In contrast to robust induction of cytokines by macrophages expressing kinase-sufficient IRAK4, IRAK4KI macrophages expressed decreased TNF-α, IL-6, IL-1ß, and C-C motif chemokine ligand 5 upon infection with L. monocytogenes or M. smegmatis. Bacterial infection of IRAK4KI macrophages led to attenuated activation of IRAK1, MAPKs and NF-κB, impaired induction of inducible NO synthase mRNA and secretion of NO, but resulted in elevated microbial burdens. Compared with WT animals, systemic infection of IRAK4KI mice with M. smegmatis or L. monocytogenes resulted in decreased levels of serum IL-6 and CXCL-1 but increased bacterial burdens in the spleen and liver. Thus, a loss of IRAK4 kinase activity underlies deficient cytokine and microbicidal responses during infection with intracellular bacteria L. monocytogenes or M. smegmatis via impaired activation of IRAK1, MAPKs, and NF-κB but increases bacterial burdens, correlating with decreased induction of NO.


Subject(s)
Immunity, Innate , Immunologic Deficiency Syndromes/immunology , Interleukin-1 Receptor-Associated Kinases/immunology , Listeriosis/immunology , Macrophages/microbiology , Mycobacterium Infections, Nontuberculous/immunology , Animals , Cytokines/metabolism , Female , Immunologic Deficiency Syndromes/genetics , Interleukin-1 Receptor-Associated Kinases/deficiency , Interleukin-1 Receptor-Associated Kinases/genetics , Listeria monocytogenes , Listeriosis/enzymology , Liver/microbiology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation, Missense , Mycobacterium Infections, Nontuberculous/enzymology , Mycobacterium smegmatis , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Primary Immunodeficiency Diseases , Spleen/microbiology
15.
J Endotoxin Res ; 12(3): 133-50, 2006.
Article in English | MEDLINE | ID: mdl-16719986

ABSTRACT

Many host cell types, including endothelial and epithelial cells, neutrophils, monocytes, natural killer cells, dendritic cells and macrophages, initiate the first line of defense against infection by sensing conserved microbial structures through Toll-like receptors (TLRs). Recognition of microbial ligands by TLRs induces their oligomerization and triggers intracellular signaling pathways, leading to production of pro- and anti-inflammatory cytokines. Dysregulation of the fine molecular mechanisms that tightly control TLR signaling may lead to hyperactivation of host cells by microbial products and septic shock. A prior exposure to bacterial products such as lipopolysaccharide (LPS) may result in a transient state of refractoriness to subsequent challenge that has been referred to as 'tolerance'. Tolerance has been postulated as a protective mechanism limiting excessive inflammation and preventing septic shock. However, tolerance may compromise the host's ability to counteract subsequent bacterial challenge since many septic patients exhibit an increased incidence of recurrent bacterial infection and suppressed monocyte responsiveness to LPS, closely resembling the tolerant phenotype. Thus, by studying mechanisms of microbial tolerance, we may gain insights into how normal regulatory mechanisms are dysregulated, leading ultimately to microbial hypo-responsiveness and life-threatening disease. In this review, we present current theories of the molecular mechanisms that underlie induction and maintenance of 'microbial tolerance', and discuss the possible relevance of tolerance to several infectious and non-infectious diseases.


Subject(s)
Bacterial Infections/immunology , Immune Tolerance/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology , Animals , Bacterial Infections/microbiology , Humans , Immune Tolerance/genetics , Immunity, Cellular , Immunity, Innate , Ligands
16.
J Leukoc Biol ; 99(6): 839-50, 2016 06.
Article in English | MEDLINE | ID: mdl-26965636

ABSTRACT

Sensing of microbial pathogens and endogenous "alarmins" by macrophages and dendritic cells is reliant on pattern recognition receptors, including membrane-associated TLRs, cytosolic nucleotide-binding and oligomerization domain leucine-rich repeat-containing receptors, retinoic acid-inducible gene I-like receptors, and absent in melanoma 2-like receptors. Engagement of TLRs elicits signaling pathways that activate inflammatory genes whose expression is regulated by chromatin-modifying complexes and transcription factors. Long noncoding RNAs have emerged as new regulators of inflammatory mediators in the immune system. They are expressed in macrophages, dendritic cells, neutrophils, NK cells, and T- and B-lymphocytes and are involved in immune cell differentiation and activation. Long noncoding RNAs act via repression or activation of transcription factors, modulation of stability of mRNA and microRNA, regulation of ribosome entry and translation of mRNAs, and controlling components of the epigenetic machinery. In this review, we focus on recent advances in deciphering the mechanisms by which long noncoding RNAs regulate TLR-driven responses in macrophages and dendritic cells and discuss the involvement of long noncoding RNAs in endotoxin tolerance, autoimmune, and inflammatory diseases. The dissection of the role of long noncoding RNAs will improve our understanding of the mechanisms of regulation of inflammation and may provide new targets for therapeutic intervention.


Subject(s)
Immunity, Innate , RNA, Long Noncoding/metabolism , Signal Transduction , Toll-Like Receptors/metabolism , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Humans , Inflammation/genetics , Inflammation/pathology
17.
J Leukoc Biol ; 100(5): 927-941, 2016 11.
Article in English | MEDLINE | ID: mdl-27343013

ABSTRACT

TLRs play a critical role in the detection of microbes and endogenous "alarmins" to initiate host defense, yet they can also contribute to the development and progression of inflammatory and autoimmune diseases. To avoid pathogenic inflammation, TLR signaling is subject to multilayer regulatory control mechanisms, including cooperation with coreceptors, post-translational modifications, cleavage, cellular trafficking, and interactions with negative regulators. Nucleic acid-sensing TLRs are particularly interesting in this regard, as they can both recognize host-derived structures and require internalization of their ligand as a result of intracellular sequestration of the nucleic acid-sensing TLRs. This review summarizes the regulatory mechanisms of TLRs, including regulation of their access to ligands, receptor folding, intracellular trafficking, and post-translational modifications, as well as how altered control mechanism could contribute to inflammatory and autoimmune disorders.


Subject(s)
Signal Transduction/immunology , Toll-Like Receptors/immunology , Adaptive Immunity , Alarmins/immunology , Animals , Dimerization , Endosomes/immunology , Glycosylation , Humans , Immunity, Innate , Infections/immunology , Ligands , Nucleic Acids/immunology , Phosphorylation , Protein Binding , Protein Domains , Protein Folding , Protein Processing, Post-Translational , Protein Transport , Structure-Activity Relationship , Toll-Like Receptors/ultrastructure , Ubiquitination
18.
J Innate Immun ; 8(2): 171-84, 2016.
Article in English | MEDLINE | ID: mdl-26457672

ABSTRACT

Endotoxin tolerance protects the host by limiting excessive 'cytokine storm' during sepsis, but compromises the ability to counteract infections in septic shock survivors. It reprograms Toll-like receptor (TLR) 4 responses by attenuating the expression of proinflammatory cytokines without suppressing anti-inflammatory and antimicrobial mediators, but the mechanisms of reprogramming remain unclear. In this study, we demonstrate that the induction of endotoxin tolerance in human monocytes, THP-1 and MonoMac-6 cells inhibited lipopolysaccharide (LPS)-mediated phosphorylation of Lyn, c-Src and their recruitment to TLR4, but increased total protein phosphatase (PP) activity and the expression of protein tyrosine phosphatase (PTP) 1B, PP2A, PTP nonreceptor type (PTPN) 22 and mitogen-activated protein kinase phosphatase (MKP)-1. Chemical PP inhibitors, okadaic acid, dephostatin and cantharidic acid markedly decreased or completely abolished LPS tolerance, indicating the importance of phosphatases in endotoxin tolerization. Overexpression of PTPN22 decreased LPS-mediated nuclear factor (NF)-x03BA;B activation, p38 phosphorylation and CXCL8 gene expression, while PTPN22 ablation upregulated LPS-induced p65 NF-x03BA;B and p38 phosphorylation and the expression of TNF-α and pro-IL-1ß mRNA, indicating PTPN22 as an inhibitor of TLR4 signaling. Thus, LPS tolerance interferes with TLR4 signaling by inhibiting Lyn and c-Src phosphorylation and their recruitment to TLR4, while increasing the phosphatase activity and expression of PP2A, PTPN22, PTP1B and MKP1.


Subject(s)
Gene Expression Regulation, Enzymologic/drug effects , Immune Tolerance/drug effects , Lipopolysaccharides/pharmacology , Monocytes/immunology , Phosphoprotein Phosphatases/immunology , Signal Transduction/drug effects , Toll-Like Receptor 4/immunology , src-Family Kinases/immunology , CSK Tyrosine-Protein Kinase , Cell Line, Tumor , Gene Expression Regulation, Enzymologic/immunology , Humans , Immune Tolerance/genetics , Lipopolysaccharides/immunology , Phosphoprotein Phosphatases/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 4/genetics , src-Family Kinases/genetics
19.
J Endotoxin Res ; 11(6): 333-9, 2005.
Article in English | MEDLINE | ID: mdl-16303088

ABSTRACT

In this overview, we will present current information on known mutations in the TLR4 signaling pathway that have been associated with increased susceptibility to disease. To date, mutations in the extracellular domain of TLR4 itself, IRAK-4, NEMO (IKK gamma), and I kappa B alpha have been identified and profoundly affect the host response to infection.


Subject(s)
Disease Susceptibility , Infections/microbiology , Mutation , Signal Transduction , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Humans , Models, Biological
20.
J Leukoc Biol ; 98(6): 963-74, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26310831

ABSTRACT

Development of endotoxin tolerance in macrophages during sepsis reprograms Toll-like receptor 4 signaling to inhibit proinflammatory cytokines without suppressing anti-inflammatory and antimicrobial mediators and protects the host from excessive inflammation and tissue damage. However, endotoxin tolerance renders septic patients immunocompromised and unable to control secondary infections. Although previous studies have revealed the importance of several negative regulators of Toll-like receptor signaling in endotoxin tolerance, the role of Pellino proteins has not been addressed. The present report shows that the induction of endotoxin tolerance in vivo in mice and in vitro in human monocytes and THP-1 and MonoMac-6 macrophages increases the expression of Pellino-3. Overexpression of Pellino-3 in human embryonic kidney 293/Toll-like receptor 2 or 293/Toll-like receptor 4/myeloid differentiation factor-2 cells inhibited Toll-like receptor 2/4-mediated activation of nuclear factor-κB and induction of CXCL-8 mRNA, and Pellino-3 ablation increased these responses. Pellino-3-deficient THP-1 cells had elevated Toll-like receptor 2/4-driven tumor necrosis factor-α, interleukin-6 mRNA, and Toll-like receptor 4-driven CCL5 gene expression in response to Toll-like receptor agonists and heat-killed Escherichia coli and Staphylococcus aureus, cytokines controlled by the MyD88 and Toll-interleukin-1R domain-containing protein inducing interferon-ß-mediated pathways, respectively. In addition, deficiency in Pellino-3 slightly increased phagocytosis of heat-killed bacteria. Transfected Pellino-3 inhibited nuclear factor-κB activation driven by overexpression of MyD88, TIR domain-containing adapter inducing interferon-ß, interleukin-1R-associated kinase-1, and tumor necrosis factor receptor activator of nuclear factor-κB-binding kinase-1, TGF-ß-activated kinase 1, and tumor necrosis factor receptor-associated factor-6, and inhibited interleukin-1R-associated kinase 1 modifications and tumor necrosis factor receptor activator of nuclear factor-κB-binding kinase 1 phosphorylation. Finally, Pellino-3 ablation in THP-1 decreased the extent of endotoxin tolerization. Thus, Pellino-3 is involved in endotoxin tolerance and functions as a negative regulator of Toll-like receptor 2/4 signaling.


Subject(s)
Immune Tolerance/drug effects , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 4/immunology , Ubiquitin-Protein Ligases/immunology , Animals , Cell Line, Tumor , Cytokines/genetics , Cytokines/immunology , Escherichia coli/immunology , Humans , Immune Tolerance/genetics , Mice , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Staphylococcus aureus/immunology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL