Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Lipid Res ; 65(7): 100533, 2024 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-38522749

RESUMEN

Mycobacterial plasma membrane, together with the peptidoglycan-arabinogalactan cell wall and waxy outer membrane, creates a robust permeability barrier against xenobiotics. The fact that several antituberculosis drugs target plasma membrane-embedded enzymes underscores the importance of the plasma membrane in bacterial physiology and pathogenesis. Nevertheless, its accurate phospholipid composition remains undefined, with conflicting reports on the abundance of phosphatidylinositol mannosides (PIMs), physiologically important glycolipids evolutionarily conserved among mycobacteria and related bacteria. Some studies indicate cardiolipin, phosphatidylethanolamine, and phosphatidylinositol as dominant structural phospholipids. Conversely, some suggest PIMs dominate the plasma membrane. A striking example of the latter is the use of reverse micelle extraction, showing diacyl phosphatidylinositol dimannoside (Ac2PIM2) as the most abundant phospholipid in a model organism, Mycobacterium smegmatis. Our recent work reveals a rapid response mechanism to membrane-fluidizing stress in mycobacterial plasma membrane: monoacyl phosphatidylinositol dimannoside and hexamannoside (AcPIM2 and AcPIM6) are converted to diacyl forms (Ac2PIM2 and Ac2PIM6). Given the dynamic nature of PIMs, we aimed to resolve the conflicting data in the literature. We show that unstressed M. smegmatis lacks an Ac2PIM2-dominated plasma membrane. Ac2PIM2 accumulation is induced by experimental conditions involving sodium docusate, a component of the reverse micellar solution. Using chemically synthesized PIMs as standards, we accurately quantified phospholipid ratio in M. smegmatis through liquid chromatography-mass spectrometry, revealing that mycobacterial plasma membrane is dominated by cardiolipin, phosphatidylethanolamine, and phosphatidylinositol. PIMs are quantitatively minor but responsive to environmental stresses in M. smegmatis. Our study paves the way for accurate modeling of mycobacterial plasma membrane.

2.
Proc Natl Acad Sci U S A ; 114(10): 2675-2680, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28223515

RESUMEN

The advances in subunit vaccines development have intensified the search for potent adjuvants, particularly adjuvants inducing cell-mediated immune responses. Identification of the C-type lectin Mincle as one of the receptors underlying the remarkable immunogenicity of the mycobacterial cell wall, via recognition of trehalose-6,6'-dimycolate (TDM), has opened avenues for the rational design of such molecules. Using a combination of chemical synthesis, biological evaluation, molecular dynamics simulations, and protein mutagenesis, we gained insight into the molecular bases of glycolipid recognition by Mincle. Unexpectedly, the fine structure of the fatty acids was found to play a key role in the binding of a glycolipid to the carbohydrate recognition domain of the lectin. Glucose and mannose esterified at O-6 by a synthetic α-ramified 32-carbon fatty acid showed agonist activity similar to that of TDM, despite their much simpler structure. Moreover, they were seen to stimulate proinflammatory cytokine production in primary human and murine cells in a Mincle-dependent fashion. Finally, they were found to induce strong Th1 and Th17 immune responses in vivo in immunization experiments in mice and conferred protection in a murine model of Mycobacterium tuberculosis infection. Here we describe the rational development of new molecules with powerful adjuvant properties.


Asunto(s)
Adyuvantes Inmunológicos/química , Lectinas Tipo C/inmunología , Receptores Inmunológicos/inmunología , Tuberculosis/prevención & control , Vacunas de Subunidad/inmunología , Inmunidad Adaptativa/efectos de los fármacos , Adyuvantes Inmunológicos/uso terapéutico , Animales , Pared Celular/efectos de los fármacos , Pared Celular/inmunología , Factores Cordón/química , Factores Cordón/inmunología , Humanos , Lectinas Tipo C/química , Lectinas Tipo C/uso terapéutico , Ratones , Simulación de Dinámica Molecular , Mutagénesis/efectos de los fármacos , Mycobacterium/inmunología , Mycobacterium/patogenicidad , Receptores Inmunológicos/química , Tuberculosis/inmunología , Tuberculosis/microbiología , Vacunas de Subunidad/uso terapéutico
3.
Proc Natl Acad Sci U S A ; 114(42): 11205-11210, 2017 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-28973928

RESUMEN

Mycobacterium tuberculosis is a major human pathogen that is able to survive inside host cells and resist immune clearance. Most particularly, it inhibits several arms of the innate immune response, including phagosome maturation or cytokine production. To better understand the molecular mechanisms by which M. tuberculosis circumvents host immune defenses, we used a transposon mutant library generated in a virulent clinical isolate of M. tuberculosis of the W/Beijing family to infect human macrophages, utilizing a cell line derivative of THP-1 cells expressing a reporter system for activation of the transcription factor NF-κB, a key regulator of innate immunity. We identified several M. tuberculosis mutants inducing a NF-κB activation stronger than that of the wild-type strain. One of these mutants was found to be deficient for the synthesis of cell envelope glycolipids, namely sulfoglycolipids, suggesting that the latter can interfere with innate immune responses. Using natural and synthetic molecular variants, we determined that sulfoglycolipids inhibit NF-κB activation and subsequent cytokine production or costimulatory molecule expression by acting as competitive antagonists of Toll-like receptor 2, thereby inhibiting the recognition of M. tuberculosis by this receptor. Our study reveals that producing glycolipid antagonists of pattern recognition receptors is a strategy used by M. tuberculosis to undermine innate immune defense. Sulfoglycolipids are major and specific lipids of M. tuberculosis, considered for decades as virulence factors of the bacilli. Our study uncovers a mechanism by which they may contribute to M. tuberculosis virulence.


Asunto(s)
Glucolípidos/metabolismo , Inmunidad Innata , Mycobacterium tuberculosis/metabolismo , Receptor Toll-Like 2/antagonistas & inhibidores , Glucolípidos/farmacología , Humanos , Macrófagos/metabolismo , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/inmunología , FN-kappa B/metabolismo
4.
J Immunol ; 195(10): 4595-603, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26466957

RESUMEN

Human T cells are activated by both peptide and nonpeptide Ags produced by Mycobacterium tuberculosis. T cells recognize cell wall lipids bound to CD1 molecules, but effector functions of CD1-reactive T cells have not been systematically assessed in M. tuberculosis-infected humans. It is also not known how these features correlate with T cell responses to secreted protein Ags. We developed a flow cytometric assay to profile CD1-restricted T cells ex vivo and assessed T cell responses to five cell wall lipid Ags in a cross-sectional study of 19 M. tuberculosis-infected and 22 M. tuberculosis-uninfected South African adolescents. We analyzed six T cell functions using a recently developed computational approach for flow cytometry data in high dimensions. We compared these data with T cell responses to five protein Ags in the same cohort. We show that CD1b-restricted T cells producing antimycobacterial cytokines IFN-γ and TNF-α are detectable ex vivo in CD4(+), CD8(+), and CD4(-)CD8(-) T cell subsets. Glucose monomycolate was immunodominant among lipid Ags tested, and polyfunctional CD4 T cells specific for this lipid simultaneously expressed CD40L, IFN-γ, IL-2, and TNF-α. Lipid-reactive CD4(+) T cells were detectable at frequencies of 0.001-0.01%, and this did not differ by M. tuberculosis infection status. Finally, CD4 T cell responses to lipids were poorly correlated with CD4 T cell responses to proteins (Spearman rank correlation -0.01; p = 0.95). These results highlight the functional diversity of CD1-restricted T cells circulating in peripheral blood as well as the complementary nature of T cell responses to mycobacterial lipids and proteins. Our approach enables further population-based studies of lipid-specific T cell responses during natural infection and vaccination.


Asunto(s)
Antígenos CD1/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Lípidos de la Membrana/inmunología , Mycobacterium tuberculosis/inmunología , Tuberculosis Pulmonar/inmunología , Adolescente , Antígenos Bacterianos/inmunología , Ligando de CD40/biosíntesis , Pared Celular/inmunología , Estudios Transversales , Femenino , Citometría de Flujo , Glucolípidos/inmunología , Humanos , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Células K562 , Activación de Linfocitos/inmunología , Masculino , Sudáfrica/epidemiología , Tuberculosis Pulmonar/epidemiología , Tuberculosis Pulmonar/microbiología , Factor de Necrosis Tumoral alfa/biosíntesis
6.
Proc Natl Acad Sci U S A ; 110(22): 8795-800, 2013 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-23671078

RESUMEN

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.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Dendrímeros/farmacología , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Manósidos/farmacología , Neumonía/tratamiento farmacológico , Receptores de Superficie Celular/metabolismo , Animales , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Dendrímeros/química , Citometría de Flujo , Humanos , Lipopolisacáridos/química , Espectroscopía de Resonancia Magnética , Manósidos/química , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Neumonía/patología , Unión Proteica
7.
Proc Natl Acad Sci U S A ; 108(34): 14228-33, 2011 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-21844346

RESUMEN

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.


Asunto(s)
Antígenos CD1/inmunología , Inmunidad/inmunología , Lípidos/inmunología , Animales , Presentación de Antígeno/inmunología , Células Clonales , Células Dendríticas/inmunología , Glucolípidos/inmunología , Glicoproteínas/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Humanos , Cinética , Ratones , Ratones Transgénicos , Células T Asesinas Naturales/inmunología , Sphingomonas/inmunología
8.
Proc Natl Acad Sci U S A ; 108(32): 13230-5, 2011 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-21788486

RESUMEN

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.


Asunto(s)
Antígenos CD1/química , Antígenos CD1/metabolismo , Metabolismo de los Lípidos , Lípidos/química , Acilación , Cristalografía por Rayos X , Humanos , Ligandos , Modelos Moleculares , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
9.
Proc Natl Acad Sci U S A ; 108(43): 17755-60, 2011 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-22006319

RESUMEN

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.


Asunto(s)
Antígenos CD1/química , Glucolípidos/química , Modelos Moleculares , Mycobacterium tuberculosis/química , Conformación Proteica , Antígenos CD1/metabolismo , Cromatografía en Capa Delgada , Cristalografía por Rayos X , Análisis de Fourier , Glucolípidos/metabolismo , Humanos , Mutagénesis , Espectrometría de Masa por Ionización de Electrospray
10.
J Immunol ; 182(11): 7030-7, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19454700

RESUMEN

CD1b-restricted T lymphocytes recognize a large diversity of mycobacterial lipids, which differ in their hydrophilic heads and the structure of their acyl appendages. Both moieties participate in the antigenicity of lipid Ags, but the structural constraints governing binding to CD1b and generation of antigenic CD1b:lipid Ag complexes are still poorly understood. Here, we investigated the structural requirements conferring antigenicity to Mycobacterium tuberculosis sulfoglycolipid Ags using a combination of CD1b:lipid binding and T cell activation assays with both living dendritic cells and plate-bound recombinant soluble CD1b. Comparison of the antigenicity of a panel of synthetic analogs, sharing the same trehalose-sulfate polar head, but differing in the structure of their acyl tails, shows that the number of C-methyl substituents on the fatty acid, the configuration of the chiral centers, and the respective localization of the two different acyl chains on the sugar moiety govern TCR recognition and T lymphocyte activation. These studies have major implications for the design of sulfoglycolipid analogs with potential use as tuberculosis subunit vaccines.


Asunto(s)
Antígenos CD1/metabolismo , Glucolípidos/inmunología , Mycobacterium tuberculosis/inmunología , Linfocitos T/inmunología , Animales , Antígenos CD1/inmunología , Células Dendríticas , Ácidos Grasos/química , Glucolípidos/química , Glucolípidos/metabolismo , Humanos , Activación de Linfocitos , Ratones , Estructura Molecular , Mycobacterium tuberculosis/química , Unión Proteica , Vacunas contra la Tuberculosis
11.
Sci Adv ; 7(42): eabj4565, 2021 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-34652941

RESUMEN

Glycolipids are prominent components of bacterial membranes that play critical roles not only in maintaining the structural integrity of the cell but also in modulating host-pathogen interactions. PatA is an essential acyltransferase involved in the biosynthesis of phosphatidyl-myo-inositol mannosides (PIMs), key structural elements and virulence factors of Mycobacterium tuberculosis. We demonstrate by electron spin resonance spectroscopy and surface plasmon resonance that PatA is an integral membrane acyltransferase tightly anchored to anionic lipid bilayers, using a two-helix structural motif and electrostatic interactions. PatA dictates the acyl chain composition of the glycolipid by using an acyl chain selectivity "ruler." We established this by a combination of structural biology, enzymatic activity, and binding measurements on chemically synthesized nonhydrolyzable acyl­coenzyme A (CoA) derivatives. We propose an interfacial catalytic mechanism that allows PatA to acylate hydrophobic PIMs anchored in the inner membrane of mycobacteria, through the use of water-soluble acyl-CoA donors.

12.
J Exp Med ; 199(5): 649-59, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14981115

RESUMEN

Mycobacterial lipids comprise a heterogeneous group of molecules capable of inducing T cell responses in humans. To identify novel antigenic lipids and increase our understanding of lipid-mediated immune responses, we established a panel of T cell clones with different lipid specificities. Using this approach we characterized a novel lipid antigen belonging to the group of diacylated sulfoglycolipids purified from Mycobacterium tuberculosis. The structure of this sulfoglycolipid was identified as 2-palmitoyl or 2-stearoyl-3-hydroxyphthioceranoyl-2'-sulfate-alpha-alpha'-D-trehalose (Ac2SGL). Its immunogenicity is dependent on the presence of the sulfate group and of the two fatty acids. Ac2SGL is mainly presented by CD1b molecules after internalization in a cellular compartment with low pH. Ac2SGL-specific T cells release interferon gamma, efficiently recognize M. tuberculosis-infected cells, and kill intracellular bacteria. The presence of Ac2SGL-responsive T cells in vivo is strictly dependent on previous contact with M. tuberculosis, but independent from the development of clinically overt disease. These properties identify Ac2SGL as a promising candidate to be tested in novel vaccines against tuberculosis.


Asunto(s)
Antígenos Bacterianos , Linfocitos T CD8-positivos/inmunología , Glucolípidos/inmunología , Mycobacterium tuberculosis/inmunología , Presentación de Antígeno , Antígenos Bacterianos/química , Antígenos CD1/metabolismo , Línea Celular , Glucolípidos/química , Humanos , Activación de Linfocitos , Estructura Molecular , Mycobacterium tuberculosis/patogenicidad
13.
Front Immunol ; 10: 2913, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31921172

RESUMEN

Mycobacterium leprae, the causative agent of leprosy, is unique amongst human pathogens in its capacity to produce the virulence factor phenolic glycolipid (PGL)-I. In addition to mediating bacterial tropism for neurons, PGL-I interacts with Complement Receptor (CR)3 on macrophages (MPs) to promote infection. We demonstrate here that PGL-I binding to CR3 also enhances bacterial invasion of both polymorphonuclear neutrophils (PMNs) and dendritic cells (DCs). Moreover, in all cell types CR3 engagement by PGL-I activates the Syk tyrosine kinase, inducing calcineurin-dependent nuclear translocation of the transcription factor NFATc. This selectively augments the production of IL-2 by DCs, IL-10 by PMNs and IL-1ß by MPs. In intranasally-infected mice PGL-I binding to CR3 heightens mycobacterial phagocytosis by lung PMNs and MPs, and stimulates NFATc-controlled production of Syk-dependent cytokines. Our study thus identifies the CR3-Syk-NFATc axis as a novel signaling pathway activated by PGL-I in innate immune cells, rewiring host cytokine responses to M. leprae.


Asunto(s)
Antígenos Bacterianos/inmunología , Calcineurina/inmunología , Glucolípidos/inmunología , Inmunidad Innata , Lepra/inmunología , Antígeno de Macrófago-1/inmunología , Mycobacterium leprae/inmunología , Factores de Transcripción NFATC/inmunología , Transducción de Señal/inmunología , Quinasa Syk/inmunología , Animales , Calcineurina/genética , Citocinas/genética , Citocinas/inmunología , Células Dendríticas/inmunología , Lepra/genética , Antígeno de Macrófago-1/genética , Masculino , Ratones , Ratones Noqueados , Factores de Transcripción NFATC/genética , Neutrófilos/inmunología , Fagocitosis , Transducción de Señal/genética , Quinasa Syk/genética
14.
Nat Commun ; 10(1): 56, 2019 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-30610190

RESUMEN

CD1 proteins are expressed on dendritic cells, where they display lipid antigens to T-cell receptors (TCRs). Here we describe T-cell autoreactivity towards ubiquitous human membrane phospholipids presented by CD1b. These T-cells discriminate between two major types of lipids, sphingolipids and phospholipids, but were broadly cross-reactive towards diverse phospholipids including phosphatidylcholine, phosphatidylinositol and phosphatidylethanolamine. The crystal structure of a representative TCR bound to CD1b-phosphatidylcholine provides a molecular mechanism for this promiscuous recognition. We observe a lateral escape channel in the TCR, which shunted phospholipid head groups sideways along the CD1b-TCR interface, without contacting the TCR. Instead the TCR recognition site involved the neck region phosphate that is common to all major self-phospholipids but absent in sphingolipids. Whereas prior studies have focused on foreign lipids or rare self-lipids, we define a new molecular mechanism of promiscuous recognition of common self-phospholipids including those that are known targets in human autoimmune disease.


Asunto(s)
Antígenos CD1/química , Fosfolípidos/química , Receptores de Antígenos de Linfocitos T/química , Linfocitos T/fisiología , Presentación de Antígeno , Unión Competitiva , Línea Celular , Membrana Celular/inmunología , Membrana Celular/metabolismo , Cristalografía por Rayos X , Humanos , Modelos Inmunológicos , Simulación del Acoplamiento Molecular
16.
ACS Chem Biol ; 13(1): 131-140, 2018 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-29185694

RESUMEN

Glycolipids play a central role in a variety of important biological processes in all living organisms. PatA is a membrane acyltransferase involved in the biosynthesis of phosphatidyl-myo-inositol mannosides (PIMs), key structural elements, and virulence factors of Mycobacterium tuberculosis. PatA catalyzes the transfer of a palmitoyl moiety from palmitoyl-CoA to the 6-position of the mannose ring linked to the 2-position of inositol in PIM1/PIM2. We report here the crystal structure of PatA in the presence of 6-O-palmitoyl-α-d-mannopyranoside, unraveling the acceptor binding mechanism. The acceptor mannose ring localizes in a cavity at the end of a surface-exposed long groove where the active site is located, whereas the palmitate moiety accommodates into a hydrophobic pocket deeply buried in the α/ß core of the protein. Both fatty acyl chains of the PIM2 acceptor are essential for the reaction to take place, highlighting their critical role in the generation of a competent active site. By the use of combined structural and quantum-mechanics/molecular-mechanics (QM/MM) metadynamics, we unravel the catalytic mechanism of PatA at the atomic-electronic level. Our study provides a detailed structural rationale for a stepwise reaction, with the generation of a tetrahedral transition state for the rate-determining step. Finally, the crystal structure of PatA in the presence of ß-d-mannopyranose and palmitate suggests an inhibitory mechanism for the enzyme, providing exciting possibilities for inhibitor design and the discovery of chemotherapeutic agents against this major human pathogen.


Asunto(s)
Aciltransferasas/química , Aciltransferasas/metabolismo , Mycobacterium smegmatis/enzimología , Aciltransferasas/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Sitios de Unión , Catálisis , Cristalografía por Rayos X , Manósidos/farmacología , Modelos Moleculares , Mycobacterium smegmatis/metabolismo , Conformación Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
17.
Front Immunol ; 9: 2, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29403489

RESUMEN

Phenolic glycolipids (PGLs) are cell wall components of a subset of pathogenic mycobacteria, with immunomodulatory properties. Here, we show that in addition, PGLs exert antibactericidal activity by limiting the production of nitric oxide synthase (iNOS) in mycobacteria-infected macrophages. PGL-mediated downregulation of iNOS was complement receptor 3-dependent and comparably induced by bacterial and purified PGLs. Using Mycobacterium leprae PGL-1 as a model, we found that PGLs dampen the toll-like receptor (TLR)4 signaling pathway, with macrophage exposure to PGLs leading to significant reduction in TIR-domain-containing adapter-inducing interferon-ß (TRIF) protein level. PGL-driven decrease in TRIF operated posttranscriptionally and independently of Src-family tyrosine kinases, lysosomal and proteasomal degradation. It resulted in the defective production of TRIF-dependent IFN-ß and CXCL10 in TLR4-stimulated macrophages, in addition to iNOS. Our results unravel a mechanism by which PGLs hijack both the bactericidal and inflammatory responses of host macrophages. Moreover, they identify TRIF as a critical node in the crosstalk between CR3 and TLR4.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Antígenos Bacterianos/metabolismo , Glucolípidos/metabolismo , Macrófagos/inmunología , Mycobacterium leprae/inmunología , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Receptor Toll-Like 4/metabolismo , Animales , Pared Celular/metabolismo , Células Cultivadas , Quimiocina CXCL10/biosíntesis , Interferón beta/biosíntesis , Lepra/inmunología , Lepra/microbiología , Lepra/patología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
18.
Sci Rep ; 8(1): 16840, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30443026

RESUMEN

Dectin-2 is a C-type lectin involved in the recognition of several pathogens such as Aspergillus fumigatus, Candida albicans, Schistosoma mansonii, and Mycobacterium tuberculosis that triggers Th17 immune responses. Identifying pathogen ligands and understanding the molecular basis of their recognition is one of the current challenges. Purified M. tuberculosis mannose-capped lipoarabinomannan (ManLAM) was shown to induce signaling via Dectin-2, an activity that requires the (α1 → 2)-linked mannosides forming the caps. Here, using isogenic M. tuberculosis mutant strains, we demonstrate that ManLAM is a bona fide and actually the sole ligand mediating bacilli recognition by Dectin-2, although M. tuberculosis produces a variety of cell envelope mannoconjugates, such as phosphatidyl-myo-inositol hexamannosides, lipomannan or manno(lipo)proteins, that bear (α1 → 2)-linked mannosides. In addition, we found that Dectin-2 can recognize lipoglycans from other bacterial species, such as Saccharotrix aerocolonigenes or the human opportunistic pathogen Tsukamurella paurometabola, suggesting that lipoglycans are prototypical Dectin-2 ligands. Finally, from a structure/function relationship perspective, we show, using lipoglycan variants and synthetic mannodendrimers, that dimannoside caps and multivalent interaction are required for ligand binding to and signaling via Dectin-2. Better understanding of the molecular basis of ligand recognition by Dectin-2 will pave the way for the rational design of potent adjuvants targeting this receptor.


Asunto(s)
Lectinas Tipo C/metabolismo , Lipopolisacáridos/metabolismo , Mycobacterium tuberculosis/metabolismo , Animales , Membrana Celular/metabolismo , Humanos , Ligandos , Lipopolisacáridos/química , Ratones Endogámicos C57BL , Transducción de Señal
19.
Cell Chem Biol ; 25(4): 392-402.e14, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29398561

RESUMEN

Mycobacterial cell wall lipids bind the conserved CD1 family of antigen-presenting molecules and activate T cells via their T cell receptors (TCRs). Sulfoglycolipids (SGLs) are uniquely synthesized by Mycobacterium tuberculosis, but tools to study SGL-specific T cells in humans are lacking. We designed a novel hybrid synthesis of a naturally occurring SGL, generated CD1b tetramers loaded with natural or synthetic SGL analogs, and studied the molecular requirements for TCR binding and T cell activation. Two T cell lines derived using natural SGLs are activated by synthetic analogs independently of lipid chain length and hydroxylation, but differentially by saturation status. By contrast, two T cell lines derived using an unsaturated SGL synthetic analog were not activated by the natural antigen. Our data provide a bioequivalence hierarchy of synthetic SGL analogs and SGL-loaded CD1b tetramers. These reagents can now be applied to large-scale translational studies investigating the diagnostic potential of SGL-specific T cell responses or SGL-based vaccines.


Asunto(s)
Antígenos Bacterianos/inmunología , Antígenos CD1/inmunología , Glucolípidos/inmunología , Activación de Linfocitos , Mycobacterium tuberculosis/inmunología , Linfocitos T/inmunología , Tuberculosis/inmunología , Acilación , Antígenos CD1/química , Línea Celular , Glucolípidos/química , Humanos , Modelos Moleculares , Mycobacterium tuberculosis/química , Multimerización de Proteína
20.
Vaccine ; 35(10): 1395-1402, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28190740

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos/inmunología , Glucolípidos/inmunología , Mycobacterium tuberculosis/inmunología , Vacunas contra la Tuberculosis/inmunología , Tuberculosis/prevención & control , Adyuvantes Inmunológicos/administración & dosificación , Animales , Antígenos Bacterianos/administración & dosificación , Antígenos Bacterianos/aislamiento & purificación , Carga Bacteriana , Modelos Animales de Enfermedad , Femenino , Glucolípidos/administración & dosificación , Glucolípidos/aislamiento & purificación , Cobayas , Liposomas/administración & dosificación , Pulmón/microbiología , Pulmón/patología , Bazo/microbiología , Bazo/patología , Resultado del Tratamiento , Tuberculosis/microbiología , Tuberculosis/patología , Vacunas contra la Tuberculosis/administración & dosificación , Vacunas contra la Tuberculosis/aislamiento & purificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA