Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
Cell Rep ; 41(4): 111562, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36288690

ABSTRACT

Infection by the Ebola virus, a member of the Filoviridae family of RNA viruses, leads to acute viral hemorrhagic fever. End-stage Ebola virus disease is characterized by a cytokine storm that causes tissue damage, vascular disintegration, and multi-organ failure. Previous studies showed that a shed form of the viral spike glycoprotein (sGP1,2) drives this hyperinflammatory response by activating Toll-like receptor 4 (TLR4). Here, we find that glycosylation is not required for activation of TLR4 by sGP1,2 and identify the internal fusion loop (IFL) as essential for inflammatory signaling. sGP1,2 competes with lipid antagonists of TLR4, and the IFL interacts directly with TLR4 and co-receptor MD2. Together, these findings indicate that sGP1,2 activates TLR4 analogously to bacterial agonist lipopolysaccharide (LPS) by binding into a hydrophobic pocket in MD2 and promoting the formation of an active heterotetramer. This conclusion is supported by docking studies that predict binding sites for sGP1,2 on TLR4 and MD2.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Humans , Toll-Like Receptor 4/metabolism , Ebolavirus/metabolism , Lipopolysaccharides/metabolism , Glycoproteins
2.
Nat Commun ; 13(1): 5110, 2022 08 30.
Article in English | MEDLINE | ID: mdl-36042238

ABSTRACT

Aedes aegypti has evolved to become an efficient vector for arboviruses but the mechanisms of host-pathogen tolerance are unknown. Immunoreceptor Toll and its ligand Spaetzle have undergone duplication which may allow neofunctionalization and adaptation. Here we present cryo-EM structures and biophysical characterisation of low affinity Toll5A complexes that display transient but specific interactions with Spaetzle1C, forming asymmetric complexes, with only one ligand clearly resolved. Loop structures of Spaetzle1C and Toll5A intercalate, temporarily bridging the receptor C-termini to promote signalling. By contrast unbound receptors form head-to-head homodimers that keep the juxtamembrane regions far apart in an inactive conformation. Interestingly the transcriptional signature of Spaetzle1C differs from other Spaetzle cytokines and controls genes involved in innate immunity, metabolism and tissue regeneration. Taken together our results explain how upregulation of Spaetzle1C in the midgut and Toll5A in the salivary gland shape the concomitant immune response.


Subject(s)
Aedes , Arboviruses , Animals , Immunity, Innate , Ligands , Mosquito Vectors/genetics
4.
Viruses ; 11(5)2019 05 01.
Article in English | MEDLINE | ID: mdl-31052481

ABSTRACT

The host immune response and virus-encoded immune evasion proteins pose constant, mutual selective pressure on each other. Virally encoded immune evasion proteins also indicate which host pathways must be inhibited to allow for viral replication. Here, we show that IIV-6 is capable of inhibiting the two Drosophila NF-κB signaling pathways, Imd and Toll. Antimicrobial peptide (AMP) gene induction downstream of either pathway is suppressed when cells infected with IIV-6 are also stimulated with Toll or Imd ligands. We find that cleavage of both Imd and Relish, as well as Relish nuclear translocation, three key points in Imd signal transduction, occur in IIV-6 infected cells, indicating that the mechanism of viral inhibition is farther downstream, at the level of Relish promoter binding or transcriptional activation. Additionally, flies co-infected with both IIV-6 and the Gram-negative bacterium, Erwinia carotovora carotovora, succumb to infection more rapidly than flies singly infected with either the virus or the bacterium. These findings demonstrate how pre-existing infections can have a dramatic and negative effect on secondary infections, and establish a Drosophila model to study confection susceptibility.


Subject(s)
Drosophila Proteins/immunology , Drosophila melanogaster/immunology , Drosophila melanogaster/virology , Iridovirus/physiology , Toll-Like Receptors/immunology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Host-Pathogen Interactions , Immunity, Innate , Iridovirus/genetics , Toll-Like Receptors/genetics , Transcription Factors/genetics , Transcription Factors/immunology , Virus Replication
5.
Cell Mol Life Sci ; 76(18): 3667-3678, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31062071

ABSTRACT

Cardiolipins (CLs) are tetra-acylated diphosphatidylglycerols found in bacteria, yeast, plants, and animals. In healthy mammals, CLs are unsaturated, whereas saturated CLs are found in blood cells from Barth syndrome patients and in some Gram-positive bacteria. Here, we show that unsaturated but not saturated CLs block LPS-induced NF-κB activation, TNF-α and IP-10 secretion in human and murine macrophages, as well as LPS-induced TNF-α and IL-1ß release in human blood mononuclear cells. Using HEK293 cells transfected with Toll-like receptor 4 (TLR4) and its co-receptor Myeloid Differentiation 2 (MD2), we demonstrate that unsaturated CLs compete with LPS for binding TLR4/MD2 preventing its activation, whereas saturated CLs are TLR4/MD2 agonists. As a consequence, saturated CLs induce a pro-inflammatory response in macrophages characterized by TNF-α and IP-10 secretion, and activate the alternative NLRP3 inflammasome pathway in human blood-derived monocytes. Thus, we identify that double bonds discriminate between anti- and pro-inflammatory properties of tetra-acylated molecules, providing a rationale for the development of TLR4 activators and inhibitors for use as vaccine adjuvants or in the treatment of TLR4-related diseases.


Subject(s)
Cardiolipins/pharmacology , Macrophages/drug effects , Toll-Like Receptor 4/metabolism , Animals , Anti-Infective Agents/chemistry , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Binding, Competitive , Cardiolipins/chemistry , Cardiolipins/metabolism , Cell Survival/drug effects , Chemokine CXCL10/metabolism , HEK293 Cells , Humans , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Lymphocyte Antigen 96/genetics , Lymphocyte Antigen 96/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Monocytes/cytology , Monocytes/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Binding , Signal Transduction/drug effects , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
Mol Ther Nucleic Acids ; 11: 159-169, 2018 Jun 01.
Article in English | MEDLINE | ID: mdl-29858051

ABSTRACT

Therapeutics based on small interfering RNA (siRNA) have promising potential as antiviral and anti-inflammatory agents. To deliver siRNA across cell membranes to reach the RNAi pathway in the cytosol of target cells, non-viral nanoparticulate delivery approaches are explored. Recently, we showed that encapsulation of siRNA in lipid-polymer hybrid nanoparticles (LPNs), based on poly(DL-lactic-co-glycolic acid) (PLGA) and cationic lipid-like materials (lipidoids), remarkably enhances intracellular delivery of siRNA as compared to siRNA delivery with LPNs modified with dioleoyltrimethylammoniumpropane (DOTAP) as the lipid component. However, the potential immune modulation by these cationic lipids remains unexplored. By testing lipidoids and DOTAP for innate immune-receptor-activating properties in vitro, we found that neither lipidoids nor DOTAP activate human Toll-like receptor (TLR) 2, 3, 7, and 9. However, in contrast to DOTAP, lipidoids are strong agonists for TLR4 and activate murine antigen-presenting cells in vitro. This agonistic effect was further confirmed in silico using a prediction model based on crystal structures. Also, lipidoids formulated as lipoplexes or as stable nucleic acid lipid particles, which was the reference formulation for siRNA delivery, proved to activate TLR4. However, by combining lipidoids with PLGA into LPNs, TLR4 activation was abrogated. Thus, lipidoid-mediated TLR4 activation during siRNA delivery may be modulated via optimization of the formulation design.

7.
Immunity ; 47(4): 635-647.e6, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29045898

ABSTRACT

In the Drosophila immune response, bacterial derived diaminopimelic acid-type peptidoglycan binds the receptors PGRP-LC and PGRP-LE, which through interaction with the adaptor protein Imd leads to activation of the NF-κB homolog Relish and robust antimicrobial peptide gene expression. PGRP-LC, PGRP-LE, and Imd each contain a motif with some resemblance to the RIP Homotypic Interaction Motif (RHIM), a domain found in mammalian RIPK proteins forming functional amyloids during necroptosis. Here we found that despite sequence divergence, these Drosophila cryptic RHIMs formed amyloid fibrils in vitro and in cells. Amyloid formation was required for signaling downstream of Imd, and in contrast to the mammalian RHIMs, was not associated with cell death. Furthermore, amyloid formation constituted a regulatable step and could be inhibited by Pirk, an endogenous feedback regulator of this pathway. Thus, diverse sequence motifs are capable of forming amyloidal signaling platforms, and the formation of these platforms may present a regulatory point in multiple biological processes.


Subject(s)
Amyloid/immunology , Carrier Proteins/immunology , Drosophila Proteins/immunology , NF-kappa B/immunology , Receptors, Cell Surface/immunology , Signal Transduction/immunology , Amino Acid Motifs/genetics , Amino Acid Motifs/immunology , Amino Acid Sequence , Amyloid/metabolism , Animals , Binding Sites/genetics , Binding Sites/immunology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Drosophila melanogaster/immunology , Female , Gene Expression/immunology , Male , Microscopy, Confocal , Models, Immunological , Mutation , NF-kappa B/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
8.
J Cell Biol ; 216(5): 1421-1438, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28373203

ABSTRACT

Cell number plasticity is coupled to circuitry in the nervous system, adjusting cell mass to functional requirements. In mammals, this is achieved by neurotrophin (NT) ligands, which promote cell survival via their Trk and p75NTR receptors and cell death via p75NTR and Sortilin. Drosophila NTs (DNTs) bind Toll receptors instead to promote neuronal survival, but whether they can also regulate cell death is unknown. In this study, we show that DNTs and Tolls can switch from promoting cell survival to death in the central nervous system (CNS) via a three-tier mechanism. First, DNT cleavage patterns result in alternative signaling outcomes. Second, different Tolls can preferentially promote cell survival or death. Third, distinct adaptors downstream of Tolls can drive either apoptosis or cell survival. Toll-6 promotes cell survival via MyD88-NF-κB and cell death via Wek-Sarm-JNK. The distribution of adaptors changes in space and time and may segregate to distinct neural circuits. This novel mechanism for CNS cell plasticity may operate in wider contexts.


Subject(s)
Nerve Growth Factors/metabolism , Toll-Like Receptors/metabolism , Animals , Cell Count , Cell Death , Cell Survival , Drosophila , Neuronal Plasticity , Signal Transduction
9.
J Control Release ; 247: 182-193, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28040465

ABSTRACT

Lipopolyamines (LPAs) are cationic lipids; they interact spontaneously with nucleic acids to form lipoplexes used for gene delivery. The main hurdle to using lipoplexes in gene therapy lies in their immunostimulatory properties, so far attributed to the nucleic acid cargo, while cationic lipids were considered as inert to the immune system. Here we demonstrate for the first time that di-C18 LPAs trigger pro-inflammatory responses through Toll-like receptor 2 (TLR2) activation, and this whether they are bound to nucleic acids or not. Molecular docking experiments suggest potential TLR2 binding modes reminiscent of bacterial lipopeptide sensing. The di-C18 LPAs share the ability of burying their lipid chains in the hydrophobic cavity of TLR2 and, in some cases, TLR1, at the vicinity of the dimerization interface; the cationic headgroups form multiple hydrogen bonds, thus crosslinking TLRs into functional complexes. Unravelling the molecular basis of TLR1 and TLR6-driven heterodimerization upon LPA binding underlines the highly collaborative and promiscuous ligand binding mechanism. The prevalence of non-specific main chain-mediated interactions demonstrates that potentially any saturated LPA currently used or proposed as transfection agent is likely to activate TLR2 during transfection. Hence our study emphasizes the urgent need to test the inflammatory properties of transfection agents and proposes the use of docking analysis as a preliminary screening tool for the synthesis of new non-immunostimulatory nanocarriers.


Subject(s)
Inflammation/chemically induced , Lipids/immunology , Polyamines/immunology , Toll-Like Receptor 2/immunology , Cell Line , HEK293 Cells , Humans , Inflammation/immunology , Lipids/adverse effects , Macrophages/drug effects , Macrophages/immunology , Molecular Docking Simulation , Nucleic Acids/administration & dosage , Nucleic Acids/genetics , Polyamines/adverse effects , Transfection , Tumor Necrosis Factor-alpha/immunology
10.
Methods Mol Biol ; 1390: 29-39, 2016.
Article in English | MEDLINE | ID: mdl-26803620

ABSTRACT

Continual advancements in computing power and sophistication, coupled with rapid increases in protein sequence and structural information, have made bioinformatic tools an invaluable resource for the molecular and structural biologist. With the degree of sequence information continuing to expand at an almost exponential rate, it is essential that scientists today have a basic understanding of how to utilise, manipulate and analyse this information for the benefit of their own experiments. In the context of Toll-Interleukin I Receptor domain containing proteins, we describe here a series of the more common and user-friendly bioinformatic tools available as Internet-based resources. These will enable the identification and alignment of protein sequences; the identification of functional motifs; the characterisation of protein secondary structure; the identification of protein structural folds and distantly homologous proteins; and the validation of the structural geometry of modelled protein structures.


Subject(s)
Amino Acid Sequence , Computational Biology/methods , Protein Conformation , Protein Interaction Domains and Motifs , Toll-Like Receptors/chemistry , Animals , Databases, Genetic , Humans , Models, Molecular , Protein Processing, Post-Translational , Reproducibility of Results , Structure-Activity Relationship , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
11.
Cell Mol Life Sci ; 72(20): 3971-82, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25956320

ABSTRACT

DiC14-amidine is a cationic lipid that was originally designed as a lipid nanocarrier for nucleic acid transport, and turned out to be a Toll-like receptor 4 (TLR4) agonist as well. We found that while E. coli lipopolysaccharide (LPS) is a TLR4 agonist in all species, diC14-amidine nanoliposomes are full agonists for human, mouse and cat receptors but weak horse agonists. Taking advantage of this unusual species specificity, we used chimeric constructs based on the human and horse sequences and identified two regions in the human TLR4 that modulate the agonist activity of diC14-amidine. Interestingly, these regions lie outside the known LPS-binding domain. Competition experiments also support our hypothesis that diC14-amidine interacts primarily with TLR4 hydrophobic crevices located at the edges of the TLR4/TLR4* dimerization interface. We have characterized potential binding modes using molecular docking analysis and suggest that diC14-amidine nanoliposomes activate TLR4 by facilitating its dimerization in a process that is myeloid differentiation 2 (MD-2)-dependent and cluster of differentiation 14 (CD14)-independent. Our data suggest that TLR4 may be activated through binding at different anchoring points, expanding the repertoire of TLR4 ligands to non-MD-2-binding lipids.


Subject(s)
Lipopolysaccharides/chemistry , Toll-Like Receptor 4/chemistry , Amino Acid Sequence , Animals , Binding Sites , HEK293 Cells , Horses , Humans , Lipid Metabolism , Lipopolysaccharide Receptors/physiology , Lipopolysaccharides/metabolism , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/metabolism , Lymphocyte Antigen 96/physiology , Mice , Models, Molecular , Molecular Docking Simulation , Recombinant Fusion Proteins , Signal Transduction , Species Specificity , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/physiology
12.
Nat Rev Immunol ; 14(8): 546-58, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25060580

ABSTRACT

Signal transduction by the Toll-like receptors (TLRs) is central to host defence against many pathogenic microorganisms and also underlies a large burden of human disease. Thus, the mechanisms and regulation of signalling by TLRs are of considerable interest. In this Review, we discuss the molecular basis for the recognition of pathogen-associated molecular patterns, the nature of the protein complexes that mediate signalling, and the way in which signals are regulated and integrated at the level of allosteric assembly, post-translational modification and subcellular trafficking of the components of the signalling complexes. These fundamental molecular mechanisms determine whether the signalling output leads to a protective immune response or to serious pathologies such as sepsis. A detailed understanding of these processes at the molecular level provides a rational framework for the development of new drugs that can specifically target pathological rather than protective signalling in inflammatory and autoimmune disease.


Subject(s)
Immunity, Innate , Receptors, Pattern Recognition/immunology , Toll-Like Receptors/immunology , Animals , Drosophila Proteins/immunology , Drosophila melanogaster , Humans , Myeloid Differentiation Factor 88/immunology , Protein Processing, Post-Translational , Protein Transport , Signal Transduction/immunology
13.
PLoS One ; 9(5): e98776, 2014.
Article in English | MEDLINE | ID: mdl-24879320

ABSTRACT

The molecular determinants underpinning how hexaacylated lipid A and tetraacylated precursor lipid IVa activate Toll-like receptor 4 (TLR4) are well understood, but how activation is induced by other lipid A species is less clear. Species specificity studies have clarified how TLR4/MD-2 recognises different lipid A structures, for example tetraacylated lipid IVa requires direct electrostatic interactions for agonism. In this study, we examine how pentaacylated lipopolysaccharide from Rhodobacter sphaeroides (RSLPS) antagonises human TLR4/MD-2 and activates the horse receptor complex using a computational approach and cross-species mutagenesis. At a functional level, we show that RSLPS is a partial agonist at horse TLR4/MD-2 with greater efficacy than lipid IVa. These data suggest the importance of the additional acyl chain in RSLPS signalling. Based on docking analysis, we propose a model for positioning of the RSLPS lipid A moiety (RSLA) within the MD-2 cavity at the TLR4 dimer interface, which allows activity at the horse receptor complex. As for lipid IVa, RSLPS agonism requires species-specific contacts with MD-2 and TLR4, but the R2 chain of RSLA protrudes from the MD-2 pocket to contact the TLR4 dimer in the vicinity of proline 442. Our model explains why RSLPS is only partially dependent on horse TLR4 residue R385, unlike lipid IVa. Mutagenesis of proline 442 into a serine residue, as found in human TLR4, uncovers the importance of this site in RSLPS signalling; horse TLR4 R385G/P442S double mutation completely abolishes RSLPS activity without its counterpart, human TLR4 G384R/S441P, being able to restore it. Our data highlight the importance of subtle changes in ligand positioning, and suggest that TLR4 and MD-2 residues that may not participate directly in ligand binding can determine the signalling outcome of a given ligand. This indicates a cooperative binding mechanism within the receptor complex, which is becoming increasingly important in TLR signalling.


Subject(s)
Lipopolysaccharides/metabolism , Lymphocyte Antigen 96/metabolism , Rhodobacter sphaeroides/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cell Line , HEK293 Cells , Horses , Humans , Ligands , Lipopolysaccharides/genetics , Lymphocyte Antigen 96/genetics , Mutagenesis/genetics , Protein Binding/genetics , Signal Transduction/genetics , Species Specificity , Toll-Like Receptor 4/genetics
14.
Proc Natl Acad Sci U S A ; 110(51): 20461-6, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24282309

ABSTRACT

Drosophila Toll functions in embryonic development and innate immunity and is activated by an endogenous ligand, Spätzle (Spz). The related Toll-like receptors in vertebrates also function in immunity but are activated directly by pathogen-associated molecules such as bacterial endotoxin. Here, we present the crystal structure at 2.35-Å resolution of dimeric Spz bound to a Toll ectodomain encompassing the first 13 leucine-rich repeats. The cystine knot of Spz binds the concave face of the Toll leucine-rich repeat solenoid in an area delineated by N-linked glycans and induces a conformational change. Mutagenesis studies confirm that the interface observed in the crystal structure is relevant for signaling. The asymmetric binding mode of Spz to Toll is similar to that of nerve growth factor (NGF) in complex with the p75 neurotrophin receptor but is distinct from that of microbial ligands bound to the Toll-like receptors. Overall, this study indicates an allosteric signaling mechanism for Toll in which ligand binding to the N terminus induces a conformational change that couples to homodimerization of juxtamembrane structures in the Toll ectodomain C terminus.


Subject(s)
Drosophila Proteins/chemistry , Protein Multimerization/physiology , Toll-Like Receptors/chemistry , Animals , Crystallography, X-Ray , Drosophila Proteins/immunology , Drosophila Proteins/metabolism , Drosophila melanogaster , Endotoxins/chemistry , Endotoxins/immunology , Endotoxins/metabolism , Immunity, Innate/physiology , Nerve Growth Factor/chemistry , Nerve Growth Factor/immunology , Nerve Growth Factor/metabolism , Protein Binding , Protein Structure, Quaternary , Receptor, Nerve Growth Factor/chemistry , Receptor, Nerve Growth Factor/immunology , Receptor, Nerve Growth Factor/metabolism , Repetitive Sequences, Amino Acid , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism
15.
Vet Res ; 44: 50, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23826682

ABSTRACT

TLR2 recognises bacterial lipopeptides and lipoteichoic acid, and forms heterodimers with TLR1 or TLR6. TLR2 is relatively well characterised in mice and humans, with published crystal structures of human TLR2/1/Pam3CSK4 and murine TLR2/6/Pam2CSK4. Equine TLR4 is activated by a different panel of ligands to human and murine TLR4, but less is known about species differences at TLR2. We therefore cloned equine TLR2, TLR1 and TLR6, which showed over 80% sequence identity with these receptors from other mammals, and performed a structure-function analysis. TLR2/1 and TLR2/6 from both horses and humans dose-dependently responded to lipoteichoic acid from Staphylococcus aureus, with no significant species difference in EC50 at either receptor pair. The EC50 of Pam2CSK4 was the same for equine and human TLR2/6, indicating amino acid differences between the two species' TLRs do not significantly affect ligand recognition. Species differences were seen between the responses to Pam2CSK4 and Pam3CSK4 at TLR2/1. Human TLR2/1, as expected, responded to Pam3CSK4 with greater potency and efficacy than Pam2CSK4. At equine TLR2/1, however, Pam3CSK4 was less potent than Pam2CSK4, with both ligands having similar efficacies. Molecular modelling indicates that the majority of non-conserved ligand-interacting residues are at the periphery of the TLR2 binding pocket and in the ligand peptide-interacting regions, which may cause subtle effects on ligand positioning. These results suggest that there are potentially important species differences in recognition of lipopeptides by TLR2/1, which may affect how the horse deals with bacterial infections.


Subject(s)
Horses/genetics , Toll-Like Receptor 1/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 6/genetics , Animals , HEK293 Cells , Horses/metabolism , Humans , Ligands , Lipopeptides/metabolism , Lipopolysaccharides/metabolism , Mice , Molecular Conformation , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Staphylococcus aureus/physiology , Teichoic Acids/metabolism , Toll-Like Receptor 1/chemistry , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/chemistry , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 6/chemistry , Toll-Like Receptor 6/metabolism
16.
J Immunol ; 191(4): 1529-35, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23878318

ABSTRACT

Allergic responses can be triggered by structurally diverse allergens. Most allergens are proteins, yet extensive research has not revealed how they initiate the allergic response and why the myriad of other inhaled proteins do not. Among these allergens, the cat secretoglobulin protein Fel d 1 is a major allergen and is responsible for severe allergic responses. In this study, we show that similar to the mite dust allergen Der p 2, Fel d 1 substantially enhances signaling through the innate receptors TLR4 and TLR2. In contrast to Der p 2, however, Fel d 1 does not act by mimicking the TLR4 coreceptor MD2 and is not able to bind stably to the TLR4/MD2 complex in vitro. Fel d 1 does, however, bind to the TLR4 agonist LPS, suggesting that a lipid transfer mechanism may be involved in the Fel d 1 enhancement of TLR signaling. We also show that the dog allergen Can f 6, a member of a distinct class of lipocalin allergens, has very similar properties to Fel d 1. We propose that Fel d 1 and Can f 6 belong to a group of allergen immunomodulatory proteins that enhance innate immune signaling and promote airway hypersensitivity reactions in diseases such as asthma.


Subject(s)
Allergens/immunology , Cats/immunology , Glycoproteins/immunology , Lipopolysaccharides/immunology , Respiratory Hypersensitivity/immunology , Allergens/chemistry , Animals , Cells, Cultured , Cytokines/biosynthesis , Dogs , Flagellin/immunology , Glycoproteins/chemistry , Glycosylation , Granulocytes/immunology , Granulocytes/metabolism , Humans , Immunity, Innate , Ligands , Lipocalins/immunology , Lipopolysaccharide Receptors/genetics , Lipopolysaccharide Receptors/immunology , Lipopolysaccharides/metabolism , Lymphocyte Antigen 96/genetics , Lymphocyte Antigen 96/immunology , Lymphocyte Antigen 96/metabolism , Macromolecular Substances , Macrophages/immunology , Macrophages/metabolism , Mice , Models, Immunological , Protein Binding , Protein Processing, Post-Translational , Recombinant Fusion Proteins/immunology , Respiratory Hypersensitivity/etiology , Species Specificity , Specific Pathogen-Free Organisms , Structure-Activity Relationship , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism , Transfection
17.
J Appl Crystallogr ; 46(Pt 2): 337-345, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23596340

ABSTRACT

Generating high-quality crystals remains a bottleneck in biological and materials sciences. Here a counter-diffusion method was used to improve the X-ray diffraction quality of the N-terminal domain of Drosophila melanogaster Toll receptor crystals. It was observed that crystallization occurred with a peculiar pattern along the capillary resembling Liesegang bands; this phenomenon is described at both macroscopic and atomic levels. It was found that bands appeared for native protein as well as for co-crystals of magic triangle (I3C)-bound protein even though they crystallize in different space groups. Crystallization occurred with a linear recurrence independent of the precipitant concentration and a protein-specific spacing coefficient. Bandwidth varied along the capillary, oscillating between large precipitation areas and single crystals. The reported data suggest that repetitive patterns can be generated with biological macromolecules in the presence of sodium malonate as a crystallization agent. A comparison with typical Liesegang patterns and the possible mechanism underlying this phenomenon are discussed.

18.
Structure ; 21(1): 143-153, 2013 Jan 08.
Article in English | MEDLINE | ID: mdl-23245851

ABSTRACT

Drosophila melanogaster Toll is the founding member of an important family of pathogen-recognition receptors in humans, the Toll-like receptor (TLR) family. In contrast, the prototypical receptor is a cytokine-like receptor for Spätzle (Spz) protein and plays a dual role in both development and immunity. Here, we present the crystal structure of the N-terminal domain of the receptor that encompasses the first 201 amino acids at 2.4 Å resolution. To our knowledge, the cysteine-rich cap adopts a novel fold unique to Toll-1 orthologs in insects and that is not critical for ligand binding. However, we observed that an antibody directed against the first ten LRRs blocks Spz signaling in a Drosophila cell-based assay. Supplemented by point mutagenesis and deletion analysis, our data suggests that the region up to LRR 14 is involved in Spz binding. Comparison with mammalian TLRs reconciles previous contradictory findings about the mechanism of Toll activation.


Subject(s)
Drosophila Proteins/chemistry , Drosophila melanogaster/metabolism , Toll-Like Receptors/chemistry , Amino Acid Motifs , Amino Acid Sequence , Animals , Antibodies/pharmacology , Cell Line , Crystallography, X-Ray , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Stability , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Sequence Homology, Amino Acid , Signal Transduction , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/metabolism
19.
Trends Biochem Sci ; 37(3): 92-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22196451

ABSTRACT

TLR4 is unique among pathogen-recognition receptors in that it initiates different pathways in different cellular locations. Binding of a bridging factor, Mal, allows recruitment of an adapter protein, MyD88, at the plasma membrane, which leads to the production of proinflammatory cytokines. Upon internalization, TLR4 uses a different bridging factor, TRAM, to activate a MyD88-independent pathway that results in type I interferon expression. Interestingly, both Mal and TRAM are localised initially at the plasma membrane. In this Opinion, I suggest a possible mechanism by which endosomal acidification triggers the differential adaptor usage of TLR4. I discuss the evidence of the pH sensitivity of TLR4 and propose a new dimerisation mode for TLR4 based on the crystal structure of the related receptor TLR3 bound to its ligand, double-stranded RNA.


Subject(s)
Endosomes/metabolism , Inflammation/metabolism , Protein Multimerization , Toll-Like Receptor 4/metabolism , Cytokines/metabolism , Humans , Hydrogen-Ion Concentration , Inflammation/therapy , Membrane Glycoproteins/metabolism , Membrane Microdomains/metabolism , Myeloid Differentiation Factor 88/metabolism , Protein Conformation , Signal Transduction , Toll-Like Receptor 4/chemistry
20.
Trends Immunol ; 32(3): 104-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21269878

ABSTRACT

Signaling by the toll-like receptor (TLR) and interleukin-1 receptor superfamily requires the adapter protein myeloid differentiation primary response protein 88 (MyD88). The recent determination of the structure of the so-called Myddosome provides us with new insights into the structural basis for innate immune signaling. Other information on the biochemistry and genetics of MyD88 and other adapters, such as MyDD adapter-like and TRIF-related adapter molecule, allows us to describe in some detail the signaling process activated by TLRs and provides new insights into the role these important proteins play in innate immunity.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Immunity, Innate , Adaptor Proteins, Signal Transducing/metabolism , Animals , Humans , Protein Multimerization , Signal Transduction , Toll-Like Receptors/chemistry , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...