Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
mBio ; 14(5): e0120823, 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37768050

ABSTRACT

IMPORTANCE: Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, impacts millions of individuals worldwide and severely impairs the quality of life for patients. Dysregulation of innate immune signaling pathways reduces barrier function and exacerbates disease progression. Macrophage (Mφ) signaling pathways are potential targets for IBD therapies. While multiple treatments are available for IBD, (i) not all patients respond, (ii) responses may diminish over time, and (iii) treatments often have undesirable side effects. Genetic studies have shown that the inheritance of two co-segregating SNPs expressed in the innate immune receptor, TLR4, is associated with human IBD. Mice expressing homologous SNPs ("TLR4-SNP" mice) exhibited more severe colitis than WT mice in a DSS-induced colonic inflammation/repair model. We identified a critical role for M2a "tissue repair" Mφ in the resolution of colitis. Our findings provide insight into potential development of novel therapies targeting Mφ signaling pathways that aim to alleviate the debilitating symptoms experienced by individuals with IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Humans , Mice , Animals , Toll-Like Receptor 4 , Polymorphism, Single Nucleotide , Quality of Life , Colitis/chemically induced , Macrophages , Inflammatory Bowel Diseases/chemically induced , Mice, Inbred C57BL , Disease Models, Animal
2.
Int J Mol Sci ; 22(4)2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33670600

ABSTRACT

Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammation of the synovial joints. Inflammation, new blood vessel formation (angiogenesis) and bone resorption (osteoclastogenesis) are three key processes involved in the joint damage and deformities of arthritis. Various gut microbiota-derived metabolites are implicated in RA pathogenesis. However, there is barely any information about the impact of two such metabolites, indole-3-aldehyde (IAld) and indole-3-acetic acid (I3AA), on arthritis-related processes. We conducted a comparative analysis of IAld and I3AA using established cell-based models to understand how they might influence RA pathogenesis. Although structurally similar, the bioactivities of these two metabolites were profoundly different. IAld but not I3AA, inhibited the expression of pro-inflammatory cytokines (IL-1ß and IL-6) in RAW 264.7 (RAW) cells stimulated with heat-killed M. tuberculosis sonicate (Mtb) and lipopolysaccharide (LPS). IAld also exhibited pro-angiogenic activity and pro-osteoclastogenic activity. In contrast, I3AA exhibited anti-angiogenic activity on endothelial cell tube formation but had no effect on osteoclastogenesis. Both IAld and I3AA have been proposed as aryl hydrocarbon receptor (AhR) agonists. Use of CH-223191, an inhibitor of the AhR, suppressed the anti-angiogenic activity of I3AA but failed to mitigate the effects of IAld. Further investigation of the anti-inflammatory activities of IAld and I3AA in LPS-treated RAW cells indicated that inhibition of MyD88-dependent activation of NF-κB and MAPK pathways was not likely involved. Our results suggest that the relative bioavailability of these indole derivatives may differentially impact RA progression and possibly other diseases that share similar cellular processes.


Subject(s)
Arthritis, Rheumatoid/immunology , Autoimmune Diseases/immunology , Cytokines/immunology , Indoleacetic Acids/immunology , Indoles/immunology , Microbiota/immunology , Animals , Arthritis, Rheumatoid/metabolism , Autoimmune Diseases/metabolism , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Cytokines/metabolism , Hot Temperature , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/physiology , Humans , Indoleacetic Acids/metabolism , Indoleacetic Acids/pharmacology , Indoles/metabolism , Indoles/pharmacology , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/metabolism , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/immunology , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/immunology , RAW 264.7 Cells
3.
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
4.
Innate Immun ; 27(2): 133-142, 2021 02.
Article in English | MEDLINE | ID: mdl-33241977

ABSTRACT

The capacity for macrophages to polarize into distinct functional activation states (e.g., M1, M2) is critical to tune an inflammatory response to the relevant infection or injury. Alternative or M2 polarization of macrophages is most often achieved in vitro in response to IL-4/IL-13 and results in the transcriptional up-regulation of a constellation of characteristic M2 marker genes. In vivo, additional signals from the inflammatory milieu can further increase or decrease M2 marker expression. Particularly, activation of cAMP-generating G protein-coupled receptors is reported to increase M2 markers, but whether this is strictly dependent upon cAMP production is unclear. We report herein that increased cAMP alone can increase IL-4-dependent M2 marker expression through a PKA/C/EBPß/CREB dependent pathway in murine macrophages.


Subject(s)
Biomarkers/metabolism , Cyclic AMP/metabolism , Macrophages/metabolism , Animals , Cell Differentiation , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation , Interleukin-4/metabolism , Macrophage Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , RAW 264.7 Cells , Signal Transduction , Steroid Isomerases/metabolism , Th2 Cells/immunology
5.
Vaccine ; 38(27): 4298-4308, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32389496

ABSTRACT

Toll-like receptors (TLRs), a family of "pattern recognition receptors," bind microbial and host-derived molecules, leading to intracellular signaling and proinflammatory gene expression. TLR4 is unique in that ligand-mediated activation requires the co-receptor myeloid differentiation 2 (MD2) to initiate two signaling cascades: the MyD88-dependent pathway is initiated at the cell membrane, and elicits rapid MAP kinase and NF-κB activation, while the TIR-domain containing adaptor inducing interferon-ß (TRIF)-dependent pathway is initiated from TLR4-containing endosomes and results in IRF3 activation. Previous studies associated inflammation with the MyD88 pathway and adjuvanticity with the TRIF pathway. Gram-negative lipopolysaccharide (LPS) is a potent TLR4 agonist, and structurally related molecules signal through TLR4 to differing extents. Herein, we compared monophosphoryl lipid A (sMPL) and E6020, two synthetic, non-toxic LPS lipid A analogs used as vaccine adjuvants, for their capacities to activate TLR4-mediated innate immune responses and to enhance antibody production. In mouse macrophages, high dose sMPL activates MyD88-dependent signaling equivalently to E6020, while E6020 exhibits significantly more activation of the TRIF pathway (a "TRIF bias") than sMPL. Eritoran, a TLR4/MD2 antagonist, competitively inhibited sMPL more strongly than E6020. Despite these differences, sMPL and E6020 adjuvants enhanced antibody responses to comparable extents, with balanced immunoglobulin (Ig) isotypes in two immunization models. These data indicate that a TRIF bias is not necessarily predictive of superior adjuvanticity.


Subject(s)
Myeloid Differentiation Factor 88 , Toll-Like Receptor 4 , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Dissociative Disorders , Lipopolysaccharides , Mice , Toll-Like Receptor 4/metabolism , Toll-Like Receptors
6.
Bio Protoc ; 9(18)2019 Sep 20.
Article in English | MEDLINE | ID: mdl-31649967

ABSTRACT

Internalization of cell surface Toll Like Receptor 4 (TLR4) is a convenient read-out to measure LPS dependent activation of the TRIF adaptor pathway. We here provide a protocol to quantify the LPS dependent internalization of TLR4 using thioglycollate-elicited peritoneal macrophages by flow cytometry.

7.
mBio ; 10(3)2019 05 07.
Article in English | MEDLINE | ID: mdl-31064834

ABSTRACT

We previously reported that the Toll-like receptor 4 (TLR4) antagonist Eritoran blocks acute lung injury (ALI) therapeutically in mouse and cotton rat models of influenza. However, secondary (2°) bacterial infection following influenza virus infection is associated with excess morbidity and mortality. Wild-type (WT) mice infected with mouse-adapted influenza A/Puerto Rico/8/34 virus (PR8) and, 7 days later, with Streptococcus pneumoniae serotype 3 (Sp3) exhibited significantly enhanced lung pathology and lethality that was reversed by Eritoran therapy after PR8 infection but before Sp3 infection. Cotton rats infected with nonadapted pH1N1 influenza virus and then superinfected with methicillin-resistant Staphylococcus aureus also exhibited increased lung pathology and serum high-mobility-group box 1 (HMGB1) levels, both of which were blunted by Eritoran therapy. In mice, PR8 infection suppressed Sp3-induced CXCL1 and CXCL2 mRNA, reducing neutrophil infiltration and increasing the bacterial burden, all of which were reversed by Eritoran treatment. While beta interferon (IFN-ß)-deficient (IFN-ß-/-) mice are highly susceptible to PR8, they exhibited delayed death upon Sp3 superinfection, indicating that while IFN-ß was protective against influenza, it negatively impacted the host response to Sp3 IFN-ß-treated WT macrophages selectively suppressed Sp3-induced CXCL1/CXCL2 transcriptionally, as evidenced by reduced recruitment of RNA polymerase II to the CXCL1 promoter. Thus, influenza establishes a "trained" state of immunosuppression toward 2° bacterial infection, in part through the potent induction of IFN-ß and its downstream transcriptional regulation of chemokines, an effect reversed by Eritoran.IMPORTANCE Enhanced susceptibility to 2° bacterial infections following infection with influenza virus is a global health concern that accounts for many hospitalizations and deaths, particularly during pandemics. The complexity of the impaired host immune response during 2° bacterial infection has been widely studied. Both type I IFN and neutrophil dysfunction through decreased chemokine production have been implicated as mechanisms underlying enhanced susceptibility to 2° bacterial infections. Our findings support the conclusion that selective suppression of CXCL1/CXCL2 represents an IFN-ß-mediated "training" of the macrophage transcriptional response to TLR2 agonists and that blocking of TLR4 therapeutically with Eritoran after influenza virus infection reverses this suppression by blunting influenza-induced IFN-ß.


Subject(s)
Coinfection/microbiology , Lung/microbiology , Orthomyxoviridae Infections/microbiology , Superinfection , Acute Lung Injury/microbiology , Acute Lung Injury/virology , Animals , Chemokine CXCL1/genetics , Chemokine CXCL1/immunology , Chemokine CXCL2/genetics , Chemokine CXCL2/immunology , Disaccharides/administration & dosage , Disease Susceptibility , Female , Immunocompromised Host , Influenza A virus , Interferon-beta/immunology , Male , Methicillin-Resistant Staphylococcus aureus , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/complications , Sigmodontinae , Streptococcus pneumoniae/immunology , Sugar Phosphates/administration & dosage , Toll-Like Receptor 4/immunology
8.
Nat Immunol ; 19(12): 1309-1318, 2018 12.
Article in English | MEDLINE | ID: mdl-30397349

ABSTRACT

The unique cell biology of Toll-like receptor 4 (TLR4) allows it to initiate two signal-transduction cascades: a signal dependent on the adaptors TIRAP (Mal) and MyD88 that begins at the cell surface and regulates proinflammatory cytokines, and a signal dependent on the adaptors TRAM and TRIF that begins in the endosomes and drives the production of type I interferons. Negative feedback circuits to limit TLR4 signals from both locations are necessary to balance the inflammatory response. We describe a negative feedback loop driven by autocrine-paracrine prostaglandin E2 (PGE2) and the PGE2 receptor EP4 that restricted TRIF-dependent signals and the induction of interferon-ß through the regulation of TLR4 trafficking. Inhibition of PGE2 production or antagonism of EP4 increased the rate at which TLR4 translocated to endosomes and amplified TRIF-dependent activation of the transcription factor IRF3 and caspase-8. This PGE2-driven mechanism restricted TLR4-TRIF signaling in vitro after infection of macrophages by the Gram-negative pathogens Escherichia coli or Citrobacter rodentium and protected mice against mortality induced by Salmonella enteritidis serovar Typhimurium. Thus, PGE2 restricted TLR4-TRIF signaling specifically in response to lipopolysaccharide.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Dinoprostone/immunology , Immunity, Innate/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Animals , Bacterial Infections/immunology , Feedback, Physiological/physiology , Humans , Lipopolysaccharides/immunology , Lipopolysaccharides/toxicity , Mice , Mice, Inbred C57BL , THP-1 Cells
9.
Infect Immun ; 86(12)2018 12.
Article in English | MEDLINE | ID: mdl-30249748

ABSTRACT

A notable proportion of Salmonella-associated gastroenteritis in the United States is attributed to Salmonella enterica serovar Typhimurium. We have previously shown that live-attenuated S Typhimurium vaccine candidate CVD 1921 (I77 ΔguaBA ΔclpP) was safe and immunogenic in rhesus macaques but was shed for an undesirably long time postimmunization. In mice, occasional mortality postvaccination was also noted (approximately 1 in every 15 mice). Here we describe a further attenuated vaccine candidate strain harboring deletions in two additional genes, htrA and pipA We determined that S Typhimurium requires pipA to elicit fluid accumulation in a rabbit ileal loop model of gastroenteritis, as an S Typhimurium ΔpipA mutant induced significantly less fluid accumulation in rabbit loops than the wild-type strain. New vaccine strain CVD 1926 (I77 ΔguaBA ΔclpP ΔpipA ΔhtrA) was assessed for inflammatory potential in an organoid model of human intestinal mucosa, where it induced less inflammatory cytokine production than organoids exposed to the precursor vaccine, CVD 1921. To assess vaccine safety and efficacy, mice were given three doses of CVD 1926 (109 CFU/dose) by oral gavage, and at 1 or 3 months postimmunization, mice were challenged with 700 or 100 LD50 (50% lethal doses), respectively, of wild-type strain I77. CVD 1926 was well tolerated and exhibited 47% vaccine efficacy following challenge with a high inoculum and 60% efficacy after challenge with a low inoculum of virulent S Typhimurium. CVD 1926 is less reactogenic yet equally as immunogenic and protective as previous iterations in a mouse model.


Subject(s)
Immunogenicity, Vaccine , Inflammation/immunology , Intestinal Mucosa/immunology , Salmonella Infections/prevention & control , Salmonella Vaccines/immunology , Animals , Antibodies, Bacterial/blood , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Cytokines/immunology , Disease Models, Animal , Female , Gene Deletion , Humans , Intestinal Mucosa/microbiology , Mice , Mice, Inbred BALB C , Mutation , Organoids/immunology , Organoids/microbiology , Rabbits , Salmonella Infections/immunology , Salmonella Vaccines/adverse effects , Salmonella typhimurium/immunology , Vaccines, Attenuated/immunology
10.
J Biol Chem ; 292(1): 292-304, 2017 Jan 06.
Article in English | MEDLINE | ID: mdl-27879319

ABSTRACT

The host protein Stimulator of Interferon Genes (STING) has been shown to be essential for recognition of both viral and intracellular bacterial pathogens, but its regulation remains unclear. Previously, we reported that mitochondrial membrane potential regulates STING-dependent IFN-ß induction independently of ATP synthesis. Because mitochondrial membrane potential controls calcium homeostasis, and AMP-activated protein kinase (AMPK) is regulated, in part, by intracellular calcium, we postulated that AMPK participates in STING activation; however, its role has yet to be been defined. Addition of an intracellular calcium chelator or an AMPK inhibitor to either mouse macrophages or mouse embryonic fibroblasts (MEFs) suppressed IFN-ß and TNF-α induction following stimulation with the STING-dependent ligand 5,6-dimethyl xanthnone-4-acetic acid (DMXAA). These pharmacological findings were corroborated by showing that MEFs lacking AMPK activity also failed to up-regulate IFN-ß and TNF-α after treatment with DMXAA or the natural STING ligand cyclic GMP-AMP (cGAMP). As a result, AMPK-deficient MEFs exhibit impaired control of vesicular stomatitis virus (VSV), a virus sensed by STING that can cause an influenza-like illness in humans. This impairment could be overcome by pretreatment of AMPK-deficient MEFs with type I IFN, illustrating that de novo production of IFN-ß in response to VSV plays a key role in antiviral defense during infection. Loss of AMPK also led to dephosphorylation at Ser-555 of the known STING regulator, UNC-51-like kinase 1 (ULK1). However, ULK1-deficient cells responded normally to DMXAA, indicating that AMPK promotes STING-dependent signaling independent of ULK1 in mouse cells.


Subject(s)
AMP-Activated Protein Kinases/physiology , Antiviral Agents , Autophagy-Related Protein-1 Homolog/physiology , Immunity, Innate/immunology , Membrane Proteins/metabolism , Protein Serine-Threonine Kinases/physiology , Vesicular stomatitis Indiana virus/immunology , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/immunology , Embryo, Mammalian/metabolism , Embryo, Mammalian/virology , Fibroblasts/cytology , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/virology , Macrophages, Peritoneal , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Vesicular Stomatitis/immunology , Vesicular Stomatitis/metabolism , Vesicular Stomatitis/virology
11.
PLoS Pathog ; 12(8): e1005803, 2016 08.
Article in English | MEDLINE | ID: mdl-27482714

ABSTRACT

Rickettsial agents are sensed by pattern recognition receptors but lack pathogen-associated molecular patterns commonly observed in facultative intracellular bacteria. Due to these molecular features, the order Rickettsiales can be used to uncover broader principles of bacterial immunity. Here, we used the bacterium Anaplasma phagocytophilum, the agent of human granulocytic anaplasmosis, to reveal a novel microbial surveillance system. Mechanistically, we discovered that upon A. phagocytophilum infection, cytosolic phospholipase A2 cleaves arachidonic acid from phospholipids, which is converted to the eicosanoid prostaglandin E2 (PGE2) via cyclooxygenase 2 (COX2) and the membrane associated prostaglandin E synthase-1 (mPGES-1). PGE2-EP3 receptor signaling leads to activation of the NLRC4 inflammasome and secretion of interleukin (IL)-1ß and IL-18. Importantly, the receptor-interacting serine/threonine-protein kinase 2 (RIPK2) was identified as a major regulator of the immune response against A. phagocytophilum. Accordingly, mice lacking COX2 were more susceptible to A. phagocytophilum, had a defect in IL-18 secretion and exhibited splenomegaly and damage to the splenic architecture. Remarkably, Salmonella-induced NLRC4 inflammasome activation was not affected by either chemical inhibition or genetic ablation of genes associated with PGE2 biosynthesis and signaling. This divergence in immune circuitry was due to reduced levels of the PGE2-EP3 receptor during Salmonella infection when compared to A. phagocytophilum. Collectively, we reveal the existence of a functionally distinct NLRC4 inflammasome illustrated by the rickettsial agent A. phagocytophilum.


Subject(s)
Anaplasma phagocytophilum/immunology , Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/immunology , Dinoprostone/immunology , Ehrlichiosis/immunology , Inflammasomes/immunology , Receptors, Prostaglandin E, EP3 Subtype/immunology , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction
12.
J Interferon Cytokine Res ; 36(7): 454-61, 2016 07.
Article in English | MEDLINE | ID: mdl-27379867

ABSTRACT

Toll-like receptors (TLRs) are major receptors of the host innate immune system that recognize conserved pathogen-associated molecular patterns (PAMPs) of invading microbes. Activation of TLR signaling culminates in the expression of multiple genes in a coordinate and kinetically defined manner. In this review, we summarize the current studies describing the chromatin landscape of TLR-responsive inflammatory genes and how changes to this chromatin landscape govern cell type-specific and temporal gene expression. We further elaborate classical endotoxin tolerance and epigenetic mechanisms controlling tolerance and interferon priming effects on inflammatory promoters.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation , Immunity, Innate/genetics , Inflammation/genetics , Inflammation/immunology , Animals , Chromatin Assembly and Disassembly , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immune Tolerance/genetics , Immune Tolerance/immunology , Inflammation/metabolism , Inflammation/microbiology , Inflammation Mediators/metabolism , Interferons/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Promoter Regions, Genetic , Toll-Like Receptors/metabolism
13.
Innate Immun ; 22(5): 363-72, 2016 07.
Article in English | MEDLINE | ID: mdl-27231145

ABSTRACT

The innate inflammatory response to Francisella tularensis (Ft) in macrophages is significantly governed by the expression of type I interferon (IFN). Previously, the proteolytic processing and maturation of pro-IL-1ß protein was shown to depend upon type I IFN expression. We show in this report that paracrine type I IFN can profoundly enhance innate recognition and TLR-dependent transcriptional responses to Ft infection upstream of its role in inflammasome regulation in both primary human monocyte-derived macrophages and primary murine peritoneal macrophages but not murine bone marrow-derived macrophages. This type I IFN-enhanced response is synergistic with TLR2 transcriptional responses, partially TLR2-independent, but strictly MyD88-dependent.


Subject(s)
Francisella tularensis/immunology , Interferon-beta/metabolism , Macrophages, Peritoneal/physiology , Macrophages/physiology , Myeloid Differentiation Factor 88/metabolism , Animals , Humans , Immunity, Innate , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Organ Specificity , RAW 264.7 Cells , Toll-Like Receptor 2/metabolism , Transcription, Genetic , Tularemia
14.
Infect Immun ; 84(6): 1796-1805, 2016 06.
Article in English | MEDLINE | ID: mdl-27045038

ABSTRACT

Tick saliva contains a number of effector molecules that inhibit host immunity and facilitate pathogen transmission. How tick proteins regulate immune signaling, however, is incompletely understood. Here, we describe that loop 2 of sialostatin L2, an anti-inflammatory tick protein, binds to annexin A2 and impairs the formation of the NLRC4 inflammasome during infection with the rickettsial agent Anaplasma phagocytophilum Macrophages deficient in annexin A2 secreted significantly smaller amounts of interleukin-1ß (IL-1ß) and IL-18 and had a defect in NLRC4 inflammasome oligomerization and caspase-1 activation. Accordingly, Annexin a2-deficient mice were more susceptible to A. phagocytophilum infection and showed splenomegaly, thrombocytopenia, and monocytopenia. Providing translational support to our findings, better binding of annexin A2 to sialostatin L2 in sera from 21 out of 23 infected patients than in sera from control individuals was also demonstrated. Overall, we establish a unique mode of inflammasome evasion by a pathogen, centered on a blood-feeding arthropod.


Subject(s)
Anaplasma phagocytophilum/immunology , Annexin A2/immunology , Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/immunology , Cystatins/immunology , Ehrlichiosis/microbiology , Immune Evasion , Amino Acid Sequence , Anaplasma phagocytophilum/genetics , Animals , Annexin A2/chemistry , Annexin A2/genetics , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Arachnid Vectors/chemistry , Arachnid Vectors/genetics , Arachnid Vectors/immunology , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Caspase 1/genetics , Caspase 1/immunology , Caspases/genetics , Caspases/immunology , Caspases, Initiator , Cystatins/chemistry , Cystatins/genetics , Ehrlichiosis/immunology , Ehrlichiosis/pathology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation , Humans , Inflammasomes/genetics , Inflammasomes/immunology , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Ixodes/chemistry , Ixodes/genetics , Ixodes/immunology , Macrophages/immunology , Macrophages/microbiology , Mice , Models, Molecular , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Signal Transduction
15.
Nat Rev Rheumatol ; 12(4): 198-200, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27006311

ABSTRACT

The selective utilization of IRAK kinases, which are thought to be recruited to MyD88 to form the 'Myddosome', has been shown to differ substantially in mouse and human cells. This finding has important implications for the development of therapeutics for inflammatory and autoimmune disorders associated with Toll-like receptors.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Signal Transduction/immunology , Animals , Humans , Inflammation , Toll-Like Receptors
16.
J Immunol ; 195(5): 2461-71, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26202980

ABSTRACT

Innate immune inflammatory responses are subject to complex layers of negative regulation at intestinal mucosal surfaces. Although the type I IFN system is critical for amplifying antiviral immunity, it has been shown to play a homeostatic role in some models of autoimmune inflammation. Type I IFN is triggered in the gut by select bacterial pathogens, but whether and how the type I IFN might regulate innate immunity in the intestinal environment have not been investigated in the context of Salmonella enterica serovar Typhimurium (ST). ST infection of human or murine macrophages reveals that IFN-ß selectively restricts the transcriptional responses mediated by both the TLRs and the NOD-like receptors. Specifically, IFN-ß potently represses ST-dependent innate induction of IL-1 family cytokines and neutrophil chemokines. This IFN-ß-mediated transcriptional repression was independent of the effects of IFN-ß on ST-induced macrophage cell death, but significantly dependent on IL-10 regulation. We further evaluated ST pathogenesis in vivo following oral inoculation of mice lacking IFN-ß. We show that IFN-ß(-/-) mice exhibit greater resistance to oral ST infection and a slower spread of ST to distal sterile sites. This work provides mechanistic insight into the relationship between ST and type I IFN, and demonstrates an additional mechanism by which IFN-ß may promote spread of enteric pathogens.


Subject(s)
Gene Expression/immunology , Immunity, Innate/immunology , Interferon-beta/immunology , Macrophages/immunology , Salmonella typhimurium/immunology , Animals , Blotting, Western , Cell Line , Cells, Cultured , Chemokines/genetics , Chemokines/immunology , Chemokines/metabolism , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Gene Expression/drug effects , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Ileum/cytology , Immunity, Innate/drug effects , Immunity, Innate/genetics , Interferon-beta/genetics , Interferon-beta/pharmacology , Macrophages/metabolism , Macrophages/microbiology , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Salmonella Infections, Animal/genetics , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Salmonella typhimurium/physiology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
17.
Proc Natl Acad Sci U S A ; 112(27): 8391-6, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26106158

ABSTRACT

Dimerization of Toll-like receptor 4 (TLR4)/myeloid differentiation factor 2 (MD2) heterodimers is critical for both MyD88- and TIR-domain-containing adapter-inducing IFN-ß (TRIF)-mediated signaling pathways. Recently, Zanoni et al. [(2011) Cell 147(4):868-880] reported that cluster of differentiation 14 (CD14) is required for LPS-/Escherichia coli- induced TLR4 internalization into endosomes and activation of TRIF-mediated signaling in macrophages. We confirmed their findings with LPS but report here that CD14 is not required for receptor endocytosis and downstream signaling mediated by TLR4/MD2 agonistic antibody (UT12) and synthetic small-molecule TLR4 ligands (1Z105) in murine macrophages. CD14 deficiency completely ablated the LPS-induced TBK1/IRF3 signaling axis that mediates production of IFN-ß in murine macrophages without affecting MyD88-mediated signaling, including NF-κB, MAPK activation, and TNF-α and IL-6 production. However, neither the MyD88- nor TRIF-signaling pathways and their associated cytokine profiles were altered in the absence of CD14 in UT12- or 1Z105-treated murine macrophages. Eritoran (E5564), a lipid A antagonist that binds the MD2 "pocket," completely blocked LPS- and 1Z105-driven, but not UT12-induced, TLR4 dimerization and endocytosis. Furthermore, TLR4 endocytosis is induced in macrophages tolerized by exposure to either LPS or UT12 and is independent of CD14. These data indicate that TLR4 receptor endocytosis and the TRIF-signaling pathway are dissociable and that TLR4 internalization in macrophages can be induced by UT12, 1Z105, and during endotoxin tolerance in the absence of CD14.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/pharmacology , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Vesicular Transport/genetics , Animals , Blotting, Western , Cells, Cultured , Disaccharides/pharmacology , Dose-Response Relationship, Drug , Endocytosis/drug effects , Flow Cytometry , HEK293 Cells , Humans , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Ligands , Lipopolysaccharide Receptors/genetics , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Sugar Phosphates/pharmacology , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/antagonists & inhibitors
18.
Cytokine ; 74(2): 171-4, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25819430

ABSTRACT

The Toll like receptors (TLRs) and the type I interferons have critical roles to play in innate immunity. In this review we will discuss new developments relating to the important area of TLR/IFN cross regulation.


Subject(s)
Immunity, Innate , Interferon Type I/immunology , Toll-Like Receptors/immunology , Animals , Humans
19.
PLoS Negl Trop Dis ; 9(1): e3394, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25569606

ABSTRACT

Invasive non-typhoidal Salmonella (iNTS) are an important cause of septicemia in children under the age of five years in sub-Saharan Africa. A novel genotype of Salmonella enterica subsp. enterica serovar Typhimurium (multi-locus sequence type [ST] 313) circulating in this geographic region is genetically different to from S. Typhimurium ST19 strains that are common throughout the rest of the world. S. Typhimurium ST313 strains have acquired pseudogenes and genetic deletions and appear to be evolving to become more like the typhoidal serovars S. Typhi and S. Paratyphi A. Epidemiological and clinical data show that S. Typhimurium ST313 strains are clinically associated with invasive systemic disease (bacteremia, septicemia, meningitis) rather than with gastroenteritis. The current work summarizes investigations of the broad hypothesis that S. Typhimurium ST313 isolates from Mali, West Africa, will behave differently from ST19 isolates in various in vitro assays. Here, we show that strains of the ST313 genotype are phagocytosed more efficiently and are highly resistant to killing by macrophage cell lines and primary mouse and human macrophages compared to ST19 strains. S. Typhimurium ST313 strains survived and replicated within different macrophages. Infection of macrophages with S. Typhimurium ST19 strains resulted in increased apoptosis and higher production of proinflammatory cytokines, as measured by gene expression and protein production, compared to S. Typhimurium ST313 strains. This difference in proinflammatory cytokine production and cell death between S. Typhimurium ST19 and ST313 strains could be explained, in part, by an increased production of flagellin by ST19 strains. These observations provide further evidence that S. Typhimurium ST313 strains are phenotypically different to ST19 strains and instead share similar pathogenic characteristics with typhoidal Salmonella serovars.


Subject(s)
Flagellin/metabolism , Inflammation/microbiology , Leukocytes, Mononuclear/microbiology , Macrophages/microbiology , Salmonella typhimurium/pathogenicity , Animals , Cell Line , Female , Flagellin/genetics , Gene Expression Regulation, Bacterial/physiology , Humans , Mice , Mice, Inbred BALB C , Salmonella typhimurium/cytology , Salmonella typhimurium/genetics , Specific Pathogen-Free Organisms
20.
PLoS Pathog ; 9(7): e1003479, 2013.
Article in English | MEDLINE | ID: mdl-23853595

ABSTRACT

The cell surface/endosomal Toll-like Receptors (TLRs) are instrumental in initiating immune responses to both bacteria and viruses. With the exception of TLR2, all TLRs and cytosolic RIG-I-like receptors (RLRs) with known virus-derived ligands induce type I interferons (IFNs) in macrophages or dendritic cells. Herein, we report that prior ligation of TLR2, an event previously shown to induce "homo" or "hetero" tolerance, strongly "primes" macrophages for increased Type I IFN production in response to subsequent TLR/RLR signaling. This occurs by increasing activation of the transcription factor, IFN Regulatory Factor-3 (IRF-3) that, in turn, leads to enhanced induction of IFN-ß, while expression of other pro-inflammatory genes are suppressed (tolerized). In vitro or in vivo "priming" of murine macrophages with TLR2 ligands increase virus-mediated IFN induction and resistance to infection. This priming effect of TLR2 is mediated by the selective upregulation of the K63 ubiquitin ligase, TRAF3. Thus, we provide a mechanistic explanation for the observed antiviral actions of MyD88-dependent TLR2 and further define the role of TRAF3 in viral innate immunity.


Subject(s)
Cellular Reprogramming , Immunity, Innate , Interferon Type I/biosynthesis , Macrophages, Peritoneal/immunology , TNF Receptor-Associated Factor 3/metabolism , Toll-Like Receptor 2/metabolism , Up-Regulation , Animals , Cell Line , Cells, Cultured , Female , Humans , Influenza A virus/immunology , Interferon Type I/genetics , Interferon Type I/metabolism , Ligands , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins/metabolism , Signal Transduction , TNF Receptor-Associated Factor 3/genetics , Toll-Like Receptor 2/genetics , Vaccinia virus/immunology , Vesicular stomatitis Indiana virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...