Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters










Publication year range
2.
Vaccine ; 35(10): 1395-1402, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28190740

ABSTRACT

The bacillus Calmette Guérin (BCG) vaccine, the only licensed vaccine against TB, displays partial and variable efficacy, thus making the exploitation of novel vaccination strategies a major priority. Most of the current vaccines in pre-clinical or clinical development are based on the induction of T cells recognizing protein antigens. However, a large number of T cells specific for mycobacterial lipids are induced during infection, suggesting that lipid-based vaccines might represent an important component of novel sub-unit vaccines. Here, we investigated whether immunization with defined mycobacterial lipid antigens induces protection in guinea pigs challenged with M. tuberculosis. Two purified mycobacterial lipid antigens, the diacylated sulfoglycolipids (Ac2SGL) and the phosphatidyl-myo-inositol dimannosides (PIM2) were formulated in biophysically characterized liposomes made of dimethyl-dioctadecyl-ammonium (DDA) and synthetic trehalose 6,6'-dibehenate (TDB). In three protection trials, a reduction of bacterial load in the spleen of inoculated animals was consistently observed compared to the unvaccinated group. Moreover, a reduction in the number of lesions and severity of pathology was detected in the lungs and spleen of the lipid vaccine group compared to unvaccinated controls. As the degree of protection achieved is similar to that observed using protein antigens in the same guinea pig model, these promising results pave the way to future investigations of lipid antigens as subunit vaccines.


Subject(s)
Antigens, Bacterial/immunology , Glycolipids/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/isolation & purification , Bacterial Load , Disease Models, Animal , Female , Glycolipids/administration & dosage , Glycolipids/isolation & purification , Guinea Pigs , Liposomes/administration & dosage , Lung/microbiology , Lung/pathology , Spleen/microbiology , Spleen/pathology , Treatment Outcome , Tuberculosis/microbiology , Tuberculosis/pathology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/isolation & purification
3.
Cell Chem Biol ; 23(9): 1147-1156, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27662254

ABSTRACT

Complex antigens require processing within antigen-presenting cells (APCs) to form T cell stimulatory complexes with CD1 antigen-presenting molecules. It remains unknown whether lipids with multi-acylated moieties also necessitate digestion by lipases to become capable of binding CD1 molecules and stimulate T cells. Here, we show that the mycobacterial tetra-acylated glycolipid antigens phosphatidyl-myo-inositol mannosides (PIM) are digested to di-acylated forms by pancreatic lipase-related protein 2 (PLRP2) and lysosomal phospholipase A2 (LPLA2) within APCs. Recombinant PLRP2 and LPLA2 removed the sn1- and sn2-bound fatty acids from the PIM glycerol moiety, as revealed by mass spectrometry and nuclear magnetic resonance studies. PLRP2 or LPLA2 gene silencing in APCs abolished PIM presentation to T cells, thus revealing an essential role of both lipases in vivo. These findings show that endosomal lipases participate in lipid antigen presentation by processing lipid antigens and have a role in T cell immunity against mycobacteria.


Subject(s)
Antigens/immunology , Lipase/metabolism , Lipids , Lysosomes/enzymology , Mycobacterium/metabolism , Phospholipases A2/metabolism , T-Lymphocytes/immunology , Acylation , Antigen Presentation/genetics , Antigens/metabolism , Cell Line , Humans , Lipase/genetics , Lymphocyte Activation , Phospholipases A2/genetics , T-Lymphocytes/cytology
4.
Int J Mycobacteriol ; 5(2): 120-7, 2016 06.
Article in English | MEDLINE | ID: mdl-27242221

ABSTRACT

OBJECTIVE/BACKGROUND: The development of new tools capable of targeting Mycobacterium tuberculosis (Mtb)-infected cells have potential applications in diagnosis, treatment, and prevention of tuberculosis. In Mtb-infected cells, CD1b molecules present Mtb lipids to the immune system (Mtb lipid-CD1b complexes). Because of the lack of CD1b polymorphism, specific Mtb lipid-CD1b complexes could be considered as universal Mtb infection markers. 2-Stearoyl-3-hydroxyphthioceranoyl-2'-sulfate-α-α'-d-trehalose (Ac2SGL) is specific for Mtb, and is not present in other mycobacterial species. The CD1b-Ac2SGL complexes are expressed on the surface of human cells infected with Mtb. The aim of this study was to generate ligands capable of binding these CD1b-Ac2SGL complexes. METHODS: A synthetic human scFv phage antibody library was used to select phage-displayed antibody fragments that recognized CD1b-Ac2SGL using CD1b-transfected THP-1 cells loaded with Ac2SGL. RESULTS: One clone, D11-a single, light-variable domain (kappa) antibody (dAbκ11)-showed high relative binding to the Ac2SGL-CD1b complex. CONCLUSION: A ligand recognizing the Ac2SGL-CD1b complex was obtained, which is a potential candidate to be further tested for diagnostic and therapeutic applications.


Subject(s)
Antibodies, Bacterial/immunology , Antigens, CD1/immunology , Glycolipids/immunology , Mycobacterium tuberculosis/immunology , Single-Chain Antibodies/genetics , Tuberculosis/immunology , Antibodies, Bacterial/genetics , Antigens, CD1/genetics , Bacteriophages/genetics , Bacteriophages/metabolism , Gene Expression , Humans , Mycobacterium tuberculosis/genetics , Single-Chain Antibodies/immunology , Tuberculosis/microbiology
5.
J Biol Chem ; 290(44): 26576-86, 2015 Oct 30.
Article in English | MEDLINE | ID: mdl-26391398

ABSTRACT

Specific and coordinated regulation of innate immune receptor-driven signaling networks often determines the net outcome of the immune responses. Here, we investigated the cross-regulation of toll-like receptor (TLR)2 and nucleotide-binding oligomerization domain (NOD)2 pathways mediated by Ac2PIM, a tetra-acylated form of mycobacterial cell wall component and muramyl dipeptide (MDP), a peptidoglycan derivative respectively. While Ac2PIM treatment of macrophages compromised their ability to induce NOD2-dependent immunomodulators like cyclooxygenase (COX)-2, suppressor of cytokine signaling (SOCS)-3, and matrix metalloproteinase (MMP)-9, no change in the NOD2-responsive NO, TNF-α, VEGF-A, and IL-12 levels was observed. Further, genome-wide microRNA expression profiling identified Ac2PIM-responsive miR-150 and miR-143 to target NOD2 signaling adaptors, RIP2 and TAK1, respectively. Interestingly, Ac2PIM was found to activate the SRC-FAK-PYK2-CREB cascade via TLR2 to recruit CBP/P300 at the promoters of miR-150 and miR-143 and epigenetically induce their expression. Loss-of-function studies utilizing specific miRNA inhibitors establish that Ac2PIM, via the miRNAs, abrogate NOD2-induced PI3K-PKCδ-MAPK pathway to suppress ß-catenin-mediated expression of COX-2, SOCS-3, and MMP-9. Our investigation has thus underscored the negative regulatory role of Ac2PIM-TLR2 signaling on NOD2 pathway which could broaden our understanding on vaccine potential or adjuvant utilities of Ac2PIM and/or MDP.


Subject(s)
MAP Kinase Kinase Kinases/metabolism , Macrophages/metabolism , MicroRNAs/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Animals , Cell Line , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/pharmacology , Epigenesis, Genetic , Immunity, Innate , Immunologic Factors/pharmacology , MAP Kinase Kinase Kinases/genetics , Macrophages/cytology , Macrophages/drug effects , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Mitogen-Activated Protein Kinases , Nitric Oxide/metabolism , Nod2 Signaling Adaptor Protein/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Polysaccharides, Bacterial/pharmacology , Protein Binding , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Suppressor of Cytokine Signaling Proteins/pharmacology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism
6.
Glycobiology ; 25(11): 1163-71, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26261090

ABSTRACT

Mycobacterium tuberculosis lipoarabinomannan (LAM) and biosynthetically related lipoglycans and glycans play an important role in host-pathogen interactions. Therefore, the elucidation of the complete biosynthetic pathways of these important molecules is expected to afford novel therapeutic targets. The characterization of biosynthetic enzymes and transporters involved in the formation and localization of these complex macromolecules in the bacterial cell envelope largely relies on genetic manipulation of mycobacteria and subsequent analyses of lipoglycan structural alterations. However, lipoglycans are present in relatively low amounts. Their purification to homogeneity remains tedious and time-consuming. To overcome these issues and to reduce the biomass and time required for lipoglycan purification, we report here the development of a methodology to efficiently purify lipoglycans by sodium deoxycholate-polyacrylamide gel electrophoresis. This faster purification method can be applied on a small amount of mycobacterial cells biomass (10-50 mg), resulting in tens of micrograms of purified lipoglycans. This amount of purified products was found to be sufficient to undertake structural analyses of lipoglycans and glycans carbohydrate domains by a combination of highly sensitive analytical procedures, involving cryoprobe NMR analysis of intact macromolecules and chemical degradations monitored by gas chromatography and capillary electrophoresis. This glycomic approach was successfully applied to the purification and structural characterization of a newly identified polysaccharide, structurally related to LAM, in the model fast-growing species Mycobacterium smegmatis.


Subject(s)
Lipopolysaccharides/chemistry , Mycobacterium tuberculosis/chemistry , Polysaccharides, Bacterial/chemistry , Glycomics/methods , Lipopolysaccharides/metabolism , Mycobacterium tuberculosis/metabolism , Polysaccharides, Bacterial/metabolism
7.
Tuberculosis (Edinb) ; 95(1): 75-85, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25488848

ABSTRACT

Mycobacterium tuberculosis (Mtb) lipids including glycolipids and lipoglycans play a crucial role in the modulation of the host immune response by targeting the innate receptors C-type lectins, TLRs and the CD1 proteins of class 1. Glycolipids have been shown to be biomarkers of M. tuberculosis strains and also of opportunistic mycobacteria called non-tuberculous mycobacteria. Most of the structural and functional work of the Mtb lipids has been done using lipids arising from M. tuberculosis cell growth in vitro. However it is likely that lipid structures can change during infection or among the M. tuberculosis or opportunistic clinical strains. Here we describe a new, rapid and sensitive analysis of lipids directly on whole mycobacteria which can be done in few minutes and on less than 1000 mycobacteria by direct matrix-assisted laser desorption/ionization mass spectrometry using an unusual solvent matrix. By this new methodology, which does not require extraction or purification steps, we are able to discriminate mycobacteria belonging to the Mtb complex as well as opportunistic and non-pathogenic mycobacteria. This method was also found to be successful for identification of an envelope lipid mutant. This work opens a new analytical route for in vivo analysis of mycobacterial lipids.


Subject(s)
Membrane Lipids/isolation & purification , Mycobacterium/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Cell Membrane/metabolism , Mycobacterium/ultrastructure , Phenotype
9.
PLoS One ; 8(10): e75983, 2013.
Article in English | MEDLINE | ID: mdl-24098413

ABSTRACT

House dust mite, Dermatophagoides pteronyssinus (Der p), is one of the major allergens responsible for allergic asthma. However, the putative receptors involved in the signalization of Der p to the innate immune cells are still poorly defined as well as the impact of their activation on the outcome of the allergen-induced cell response. We previously reported that the HDM activation of mouse alveolar macrophages (AM) involves the TLR4/CD14 cell surface receptor complex. Here using a TLR ligand screening essay, we demonstrate that HDM protein extract engages the TLR2, in addition to the TLR4, in engineered TLR-transfected HEK cells but also in the MH-S mouse alveolar macrophage cell line model. Moreover we found that the concomitant recruitment of the MH-S cell's TLR2 and TLR4 receptors by the HDM extract activates the MyD88-dependent signaling pathway and leads to the secretion of the NF-κB regulated pro-inflammatory factors NO and TNF-α. However unlike with the canonical TLR4 ligand (i.e. the bacterial LPS) mobilization of TLR4 by the HDM extract induces a reduced production of the IL-12 pro-inflammatory cytokine and fails to trigger the expression of the T-bet transcription factor. Finally we demonstrated that HDM extract down-regulates LPS induced IL-12 and T-bet expression through a TLR2 dependent mechanism. Therefore, we propose that the simultaneous engagement of the TLR2 and TLR4 receptors by the HDM extract results in a cross regulated original activation pattern of the AM which may contribute to the Th2 polarization of the allergen-induced immune response. The deciphering of these cross-regulation networks is of prime importance to open the way for original therapeutic strategies taking advantage of these receptors and their associated signaling pathways to treat allergic asthma.


Subject(s)
Antigens, Dermatophagoides/immunology , Immunity, Innate , Macrophages, Alveolar/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , HEK293 Cells , Humans , Interleukin-12/biosynthesis , Ligands , Mice , Nitric Oxide/biosynthesis , T-Box Domain Proteins/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Toll-Like Receptor 2/agonists , Toll-Like Receptor 4/agonists , Tumor Necrosis Factor-alpha/biosynthesis
10.
Proc Natl Acad Sci U S A ; 110(22): 8795-800, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23671078

ABSTRACT

Mycobacterium tuberculosis mannose-capped lipoarabinomannan inhibits the release of proinflammatory cytokines by LPS-stimulated human dendritic cells (DCs) via targeting the C-type lectin receptor DC-specific intercellular adhesion molecule 3-grabbing nonintegrin (DC-SIGN). With the aim of mimicking the bioactive supramolecular structure of mannose-capped lipoarabinomannan, we designed and synthesized a set of poly(phosphorhydrazone) dendrimers grafted with mannose units, called mannodendrimers, that differed by size and the number and length of their (α1→2)-oligommanoside caps. A third-generation dendrimer bearing 48 trimannoside caps (3T) and a fourth-generation dendrimer bearing 96 dimannosides (4D) displayed the highest binding avidity for DC-SIGN. Moreover, these dendrimers inhibited proinflammatory cytokines, including TNF-α, production by LPS-stimulated DCs in a DC-SIGN-dependent fashion. Finally, in a model of acute lung inflammation in which mice were exposed to aerosolized LPS, per os administration of 3T mannodendrimer was found to significantly reduce neutrophil influx via targeting the DC-SIGN murine homolog SIGN-related 1. The 3T mannodendrimer therefore represents an innovative fully synthetic compound for the treatment of lung inflammatory diseases.


Subject(s)
Cell Adhesion Molecules/metabolism , Dendrimers/pharmacology , Dendritic Cells/metabolism , Lectins, C-Type/metabolism , Mannosides/pharmacology , Pneumonia/drug therapy , Receptors, Cell Surface/metabolism , Animals , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Dendrimers/chemistry , Flow Cytometry , Humans , Lipopolysaccharides/chemistry , Magnetic Resonance Spectroscopy , Mannosides/chemistry , Mice , Mice, Inbred C57BL , Molecular Structure , Pneumonia/pathology , Protein Binding
11.
Proc Natl Acad Sci U S A ; 110(16): 6560-5, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23550160

ABSTRACT

A posttranslational protein O-mannosylation process resembling that found in fungi and animals has been reported in the major human pathogen Mycobacterium tuberculosis (Mtb) and related actinobacteria. However, the role and incidence of this process, which is essential in eukaryotes, have never been explored in Mtb. We thus analyzed the impact of interrupting O-mannosylation in the nonpathogenic saprophyte Mycobacterium smegmatis and in the human pathogen Mtb by inactivating the respective putative protein mannosyl transferase genes Msmeg_5447 and Rv1002c. Loss of protein O-mannosylation in both mutant strains was unambiguously demonstrated by efficient mass spectrometry-based glycoproteomics analysis. Unexpectedly, although the M. smegmatis phenotype was unaffected by the lack of manno-proteins, the Mtb mutant had severely impacted growth in vitro and in cellulo associated with a strong attenuation of its pathogenicity in immunocompromised mice. These data are unique in providing evidence of the biological significance of protein O-mannosylation in mycobacteria and demonstrate the crucial contribution of this protein posttranslational modification to Mtb virulence in the host.


Subject(s)
Mannose/metabolism , Mannosyltransferases/metabolism , Mycobacterium smegmatis/enzymology , Mycobacterium tuberculosis/enzymology , Mycobacterium tuberculosis/pathogenicity , Protein Processing, Post-Translational/physiology , Animals , Gene Silencing , Mannosyltransferases/genetics , Mass Spectrometry , Mice , Mycobacterium tuberculosis/growth & development , Proteomics/methods , Species Specificity , Virulence
12.
Biochimie ; 95(1): 33-42, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22706280

ABSTRACT

Innate immune system is the first line of host defense against invading microorganisms. It relies on a limited number of germline-encoded pattern recognition receptors that recognize conserved molecular structures of microbes, referred to as pathogen-/microbe-associated molecular patterns (PAMPs/MAMPs). Bacterial cell wall macroamphiphiles, namely Gram-negative bacteria lipopolysaccharide (LPS), Gram-positive bacteria lipoteichoic acid (LTA), lipoproteins and mycobacterial lipoglycans, are important molecules for the physiology of bacteria and evidently meet PAMP/MAMP criteria. They are well suited to innate immune recognition and constitute non-self signatures detected by the innate immune system to signal the presence of an infective agent. They are notably recognized via their lipid anchor by Toll-like receptors (TLRs) 4 or 2. Here, we review our current knowledge of the molecular bases of macroamphiphile recognition by TLRs, with a special emphasis on mycobacterial lipoglycan detection by TLR2.


Subject(s)
Cell Wall , Immunity, Innate , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Bacteria/chemistry , Bacteria/immunology , Cell Wall/chemistry , Cell Wall/immunology , Host-Parasite Interactions/genetics , Host-Parasite Interactions/immunology , Humans , Lipopolysaccharides/chemistry , Lipopolysaccharides/immunology , Teichoic Acids/chemistry , Teichoic Acids/immunology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
13.
Cell Microbiol ; 15(4): 660-74, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23121245

ABSTRACT

Mannose-capped lipoarabinomannan (ManLAM) is considered an important virulence factor of Mycobacterium tuberculosis. However, while mannose caps have been reported to be responsible for various immunosuppressive activities of ManLAM observed in vitro, there is conflicting evidence about their contribution to mycobacterial virulence in vivo. Therefore, we used Mycobacterium bovis BCG and M. tuberculosis mutants that lack the mannose cap of LAM to assess the role of ManLAM in the interaction of mycobacteria with the host cells, to evaluate vaccine-induced protection and to determine its importance in M. tuberculosis virulence. Deletion of the mannose cap did not affect BCG survival and replication in macrophages, although the capless mutant induced a somewhat higher production of TNF. In dendritic cells, the capless mutant was able to induce the upregulation of co-stimulatory molecules and the only difference we detected was the secretion of slightly higher amounts of IL-10 as compared to the wild type strain. In mice, capless BCG survived equally well and induced an immune response similar to the parental strain. Furthermore, the efficacy of vaccination against a M. tuberculosis challenge in low-dose aerosol infection models in mice and guinea pigs was not affected by the absence of the mannose caps in the BCG. Finally, the lack of the mannose cap in M. tuberculosis did not affect its virulence in mice nor its interaction with macrophages in vitro. Thus, these results do not support a major role for the mannose caps of LAM in determining mycobacterial virulence and immunogenicity in vivo in experimental animal models of infection, possibly because of redundancy of function.


Subject(s)
Host-Pathogen Interactions , Lipopolysaccharides/analysis , Mannose/analysis , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology , Animals , Dendritic Cells/immunology , Dendritic Cells/microbiology , Disease Models, Animal , Guinea Pigs , Macrophages/microbiology , Mice , Microbial Viability , Mycobacterium bovis/chemistry , Mycobacterium bovis/genetics , Mycobacterium bovis/growth & development , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Tuberculosis, Pulmonary/microbiology , Virulence Factors/analysis
14.
J Biol Chem ; 287(47): 39933-41, 2012 Nov 16.
Article in English | MEDLINE | ID: mdl-23038254

ABSTRACT

The biosynthesis of the major cell envelope glycoconjugates of Mycobacterium tuberculosis is topologically split across the plasma membrane, yet nothing is known of the transporters required for the translocation of lipid-linked sugar donors and oligosaccharide intermediates from the cytoplasmic to the periplasmic side of the membrane in mycobacteria. One of the mechanisms used by prokaryotes to translocate lipid-linked phosphate sugars across the plasma membrane relies on translocases that share resemblance with small multidrug resistance transporters. The presence of an small multidrug resistance-like gene, Rv3789, located immediately upstream from dprE1/dprE2 responsible for the formation of decaprenyl-monophosphoryl-ß-D-arabinose (DPA) in the genome of M. tuberculosis led us to investigate its potential involvement in the formation of the major arabinosylated glycopolymers, lipoarabinomannan (LAM) and arabinogalactan (AG). Disruption of the ortholog of Rv3789 in Mycobacterium smegmatis resulted in a reduction of the arabinose content of both AG and LAM that accompanied the accumulation of DPA in the mutant cells. Interestingly, AG and LAM synthesis was restored in the mutant not only upon expression of Rv3789 but also upon that of the undecaprenyl phosphate aminoarabinose flippase arnE/F genes from Escherichia coli. A bacterial two-hybrid system further indicated that Rv3789 interacts in vivo with the galactosyltransferase that initiates the elongation of the galactan domain of AG. Biochemical and genetic evidence is thus consistent with Rv3789 belonging to an AG biosynthetic complex, where its role is to reorient DPA to the periplasm, allowing this arabinose donor to then be used in the buildup of the arabinan domains of AG and LAM.


Subject(s)
Bacterial Proteins/metabolism , Drug Resistance, Multiple, Bacterial/physiology , Galactans/metabolism , Lipopolysaccharides/metabolism , Membrane Transport Proteins/metabolism , Mycobacterium tuberculosis/metabolism , Arabinose/genetics , Arabinose/metabolism , Bacterial Proteins/genetics , Galactans/genetics , Genetic Complementation Test , Glycosylation , Lipopolysaccharides/genetics , Membrane Transport Proteins/genetics , Mutation , Mycobacterium smegmatis/genetics , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/genetics
15.
J Biol Chem ; 287(37): 31494-502, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22782895

ABSTRACT

Lipids are important antigens that induce T cell-mediated specific immune responses. They are presented to T lymphocytes by a specific class of MHC-I like proteins, termed CD1. The majority of the described CD1-presented mycobacterial antigens are presented by the CD1b isoform. We previously demonstrated that the stimulation of CD1b-restricted T cells by the hexamannosylated phosphatidyl-myo-inositol (PIM(6)), a family of mycobacterial antigens, requires a prior partial digestion of the antigen oligomannoside moiety by α-mannosidase and that CD1e is an accessory protein absolutely required for the generation of the lipid immunogenic form. Here, we show that CD1e behaves as a lipid transfer protein influencing lipid immunoediting and membrane transfer of PIM lipids. CD1e selectively assists the α-mannosidase-dependent digestion of PIM(6) species according to their degree of acylation. Moreover, CD1e transfers only diacylated PIM from donor to acceptor liposomes and also from membranes to CD1b. This study provides new insight into the molecular mechanisms by which CD1e contributes to lipid immunoediting and CD1-restricted presentation to T cells.


Subject(s)
Antigen Presentation/physiology , Antigens, Bacterial/immunology , Antigens, CD1/immunology , Glycolipids/immunology , Mycobacterium tuberculosis/immunology , T-Lymphocytes/immunology , Antigens, Bacterial/genetics , Antigens, Bacterial/metabolism , Antigens, CD1/genetics , Antigens, CD1/metabolism , Cell Line , Glycolipids/genetics , Glycolipids/metabolism , Humans , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , T-Lymphocytes/metabolism , alpha-Mannosidase/chemistry
16.
PLoS One ; 6(12): e28476, 2011.
Article in English | MEDLINE | ID: mdl-22164297

ABSTRACT

Innate immune recognition is based on the detection, by pattern recognition receptors (PRRs), of molecular structures that are unique to microorganisms. Lipoglycans are macromolecules specific to the cell envelope of mycobacteria and related genera. They have been described to be ligands, as purified molecules, of several PRRs, including the C-type lectins Mannose Receptor and DC-SIGN, as well as TLR2. However, whether they are really sensed by these receptors in the context of a bacterium infection remains unclear. To address this question, we used the model organism Mycobacterium smegmatis to generate mutants altered for the production of lipoglycans. Since their biosynthesis cannot be fully abrogated, we manipulated the biosynthesis pathway of GDP-Mannose to obtain some strains with either augmented (∼1.7 fold) or reduced (∼2 fold) production of lipoglycans. Interestingly, infection experiments demonstrated a direct correlation between the amount of lipoglycans in the bacterial cell envelope on one hand and the magnitude of innate immune signaling in TLR2 reporter cells, monocyte/macrophage THP-1 cell line and human dendritic cells, as revealed by NF-κB activation and IL-8 production, on the other hand. These data establish that lipoglycans are bona fide Microbe-Associated Molecular Patterns contributing to innate immune detection of mycobacteria, via TLR2 among other PRRs.


Subject(s)
Immunity, Innate , Lipopolysaccharides/chemistry , Mycobacterium smegmatis/metabolism , Carbohydrates/chemistry , Cell Adhesion Molecules/metabolism , Cell Line , Dendritic Cells/cytology , Electrophoresis, Polyacrylamide Gel , Guanosine Diphosphate/chemistry , HEK293 Cells , Humans , Interleukin-8/metabolism , Lectins, C-Type/metabolism , Mannose/chemistry , Mannose Receptor , Mannose-Binding Lectins/metabolism , Models, Biological , Mutation , NF-kappa B/metabolism , Plasmids/metabolism , Receptors, Cell Surface/metabolism , Recombinant Proteins/metabolism
17.
PLoS One ; 6(10): e26316, 2011.
Article in English | MEDLINE | ID: mdl-22028855

ABSTRACT

Gram positive bacteria produce cell envelope macroamphiphile glycopolymers, i.e. lipoteichoic acids or lipoglycans, whose functions and biosynthesis are not yet fully understood. We report for the first time a detailed structure of lipoteichoic acid isolated from a Streptomyces species, i.e. Streptomyces hygroscopicus subsp. hygroscopicus NRRL 2387T. Chemical, MS and NMR analyses revealed a polyglycerolphosphate backbone substituted with α-glucosaminyl and α-N-acetyl-glucosaminyl residues but devoid of any amino-acid substituent. This structure is very close, if not identical, to that of the wall teichoic acid of this organism. These data not only contribute to the growing recognition that lipoteichoic acid is a cell envelope component of gram positive Actinobacteria but also strongly support the recently proposed hypothesis of an overlap between the pathways of lipoteichoic acid and wall teichoic acid synthesis in these bacteria. S. hygroscopicus lipoteichoic acid induced signalling by human innate immune receptor TLR2, confirming its role as a microbe-associated molecular pattern. Its activity was partially dependant on TLR1, TLR6 and CD14. Moreover, it stimulated TNF-α and IL-6 production by a human macrophage cell line to an extent similar to that of Staphylococcus aureus lipoteichoic acid. These results provide new clues on lipoteichoic acid structure/function relationships, most particularly on the role of the polyglycerolphosphate backbone substituents.


Subject(s)
Immunologic Factors/chemistry , Immunologic Factors/pharmacology , Lipopolysaccharides/chemistry , Lipopolysaccharides/pharmacology , Models, Molecular , Streptomyces/chemistry , Teichoic Acids/chemistry , Teichoic Acids/pharmacology , Cytokines/biosynthesis , HEK293 Cells , Humans , Immunologic Factors/biosynthesis , Immunologic Factors/isolation & purification , Lipopolysaccharides/biosynthesis , Lipopolysaccharides/isolation & purification , Signal Transduction/drug effects , Streptomyces/metabolism , Structure-Activity Relationship , Teichoic Acids/biosynthesis , Teichoic Acids/isolation & purification , Toll-Like Receptor 2/metabolism
18.
Proc Natl Acad Sci U S A ; 108(43): 17755-60, 2011 Oct 25.
Article in English | MEDLINE | ID: mdl-22006319

ABSTRACT

The mechanisms permitting nonpolymorphic CD1 molecules to present lipid antigens that differ considerably in polar head and aliphatic tails remain elusive. It is also unclear why hydrophobic motifs in the aliphatic tails of some antigens, which presumably embed inside CD1 pockets, contribute to determinants for T-cell recognition. The 1.9-Å crystal structure of an active complex of CD1b and a mycobacterial diacylsulfoglycolipid presented here provides some clues. Upon antigen binding, endogenous spacers of CD1b, which consist of a mixture of diradylglycerols, moved considerably within the lipid-binding groove. Spacer displacement was accompanied by F' pocket closure and an extensive rearrangement of residues exposed to T-cell receptors. Such structural reorganization resulted in reduction of the A' pocket capacity and led to incomplete embedding of the methyl-ramified portion of the phthioceranoyl chain of the antigen, explaining why such hydrophobic motifs are critical for T-cell receptor recognition. Mutagenesis experiments supported the functional importance of the observed structural alterations for T-cell stimulation. Overall, our data delineate a complex molecular mechanism combining spacer repositioning and ligand-induced conformational changes that, together with pocket intricacy, endows CD1b with the required molecular plasticity to present a broad range of structurally diverse antigens.


Subject(s)
Antigens, CD1/chemistry , Glycolipids/chemistry , Models, Molecular , Mycobacterium tuberculosis/chemistry , Protein Conformation , Antigens, CD1/metabolism , Chromatography, Thin Layer , Crystallography, X-Ray , Fourier Analysis , Glycolipids/metabolism , Humans , Mutagenesis , Spectrometry, Mass, Electrospray Ionization
19.
Proc Natl Acad Sci U S A ; 108(34): 14228-33, 2011 Aug 23.
Article in English | MEDLINE | ID: mdl-21844346

ABSTRACT

CD1e is a member of the CD1 family that participates in lipid antigen presentation without interacting with the T-cell receptor. It binds lipids in lysosomes and facilitates processing of complex glycolipids, thus promoting editing of lipid antigens. We find that CD1e may positively or negatively affect lipid presentation by CD1b, CD1c, and CD1d. This effect is caused by the capacity of CD1e to facilitate rapid formation of CD1-lipid complexes, as shown for CD1d, and also to accelerate their turnover. Similar results were obtained with antigen-presenting cells from CD1e transgenic mice in which lipid complexes are assembled more efficiently and show faster turnover than in WT antigen-presenting cells. These effects maximize and temporally narrow CD1-restricted responses, as shown by reactivity to Sphingomonas paucimobilis-derived lipid antigens. CD1e is therefore an important modulator of both group 1 and group 2 CD1-restricted responses influencing the lipid antigen availability as well as the generation and persistence of CD1-lipid complexes.


Subject(s)
Antigens, CD1/immunology , Immunity/immunology , Lipids/immunology , Animals , Antigen Presentation/immunology , Clone Cells , Dendritic Cells/immunology , Glycolipids/immunology , Glycoproteins/immunology , Gram-Negative Bacterial Infections/immunology , Humans , Kinetics , Mice , Mice, Transgenic , Natural Killer T-Cells/immunology , Sphingomonas/immunology
20.
Proc Natl Acad Sci U S A ; 108(32): 13230-5, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21788486

ABSTRACT

CD1e is the only human CD1 protein existing in soluble form in the late endosomes of dendritic cells, where it facilitates the processing of glycolipid antigens that are ultimately recognized by CD1b-restricted T cells. The precise function of CD1e remains undefined, thus impeding efforts to predict the participation of this protein in the presentation of other antigens. To gain insight into its function, we determined the crystal structure of recombinant CD1e expressed in human cells at 2.90-Å resolution. The structure revealed a groove less intricate than in other CD1 proteins, with a significantly wider portal characterized by a 2 Å-larger spacing between the α1 and α2 helices. No electron density corresponding to endogenous ligands was detected within the groove, despite the presence of ligands unequivocally established by native mass spectrometry in recombinant CD1e. Our structural data indicate that the water-exposed CD1e groove could ensure the establishment of loose contacts with lipids. In agreement with this possibility, lipid association and dissociation processes were found to be considerably faster with CD1e than with CD1b. Moreover, CD1e was found to mediate in vitro the transfer of lipids to CD1b and the displacement of lipids from stable CD1b-antigen complexes. Altogether, these data support that CD1e could have evolved to mediate lipid-exchange/editing processes with CD1b and point to a pathway whereby the repertoire of lipid antigens presented by human dendritic cells might be expanded.


Subject(s)
Antigens, CD1/chemistry , Antigens, CD1/metabolism , Lipid Metabolism , Lipids/chemistry , Acylation , Crystallography, X-Ray , Humans , Ligands , Models, Molecular , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...