Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters










Publication year range
1.
Mol Neurobiol ; 61(3): 1580-1592, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37731080

ABSTRACT

Osteoarthritis (OA) is one of the most common joint disorder, with pain accompanied by functional impairment, as the most pronounced clinical symptom. Currently used pharmacotherapy involves symptomatic treatment that do not always provide adequate pain relief. This may be due to concomitance of central sensitization and development of neuropathic features in OA patients. Here we performed studies in the animal model of OA to investigate of the neuropathic component. Intraarticular injection of monoiodoacetate (MIA, 1 mg) was used to induce OA in Wistar male rats. Development of pain phenotype was assessed by behavioral testing (PAM test and von Frey's test), while corresponding changes in dorsal root ganglia (DRGs L3-L5) and spinal cord (SC) gene expression were assessed by means of qRT-PCR technique. We also performed microtomography of OA-affected knee joints to correlate the level of bone degradation with observed behavioral and molecular changes. We observed gradually developing remote allodynia after MIA treatment, indicating the presence of neuropathic component. Our results showed that, among DRGs innervating knee joint, development of central sensitization is most likely due to peripheral input of stimuli through DRG L5. In SC, development of secondary hypersensitivity correlated with increased expression of TAC1 and NPY. Our studies provided molecular records on abnormal activation of pain transmission markers in DRG and SC during development of OA that are responsible for the manifestation of neuropathic features. The obtained results increase insight into molecular changes occurring in the neuronal tissue during OA development and may contribute to readdressing treatment paradigms.


Subject(s)
Neuralgia , Osteoarthritis , Humans , Rats , Animals , Male , Cord Factors/metabolism , Rats, Sprague-Dawley , Disease Models, Animal , Rats, Wistar , Osteoarthritis/complications , Osteoarthritis/diagnostic imaging , Osteoarthritis/metabolism , Neuralgia/metabolism , Spinal Cord/metabolism , Ganglia, Spinal/metabolism
2.
Structure ; 31(9): 1077-1085.e5, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37348496

ABSTRACT

Mincle (macrophage-inducible C-type lectin, CLEC4E) is a C-type lectin immune-stimulatory receptor for cord factor, trehalose dimycolate (TDM), which serves as a potent component of adjuvants. The recognition of glycolipids by Mincle, especially their lipid parts, is poorly understood. Here, we performed nuclear magnetic resonance analysis, revealing that titration of trehalose harboring a linear short acyl chain showed a chemical shift perturbation of hydrophobic residues next to the Ca-binding site. Notably, there were split signals for Tyr201 upon complex formation, indicating two binding modes for the acyl chain. In addition, most Mincle residues close to the Ca-binding site showed no observable signals, suggesting their mobility on an ∼ ms scale even after complex formation. Mutagenesis study supported two putative lipid-binding modes for branched acyl-chain TDM binding. These results provide novel insights into the plastic-binding modes of Mincle toward a wide range of glycol- and glycerol-lipids, important for rational adjuvant development.


Subject(s)
Glycolipids , Lectins, C-Type , Binding Sites , Cord Factors/chemistry , Cord Factors/metabolism , Glycolipids/chemistry , Glycolipids/metabolism , Lectins, C-Type/chemistry , Mutagenesis , Humans
3.
Biometals ; 36(3): 603-615, 2023 06.
Article in English | MEDLINE | ID: mdl-35976499

ABSTRACT

Lactoferrin (LTF), an iron binding protein, is known to exhibit immune modulatory effects on pulmonary pathology during insult-induced models of primary Mycobacterium tuberculosis (Mtb) infection. The effects of LTF correlate with modulation of the immune related development of the pathology, and altering of the histological nature of the physically compact and dense lung granuloma in mice. Specifically, a recombinant human version of LTF limits immediate progression of granulomatous severity following administration of the Mtb cell wall mycolic acid, trehalose 6,6'-dimycolate (TDM), in part through reduced pro-inflammatory responses known to control these events. This current study investigates a limited course of LTF to modulate not only initiation, but also maintenance and resolution of pathology post development of the granulomatous response in mice. Comparison is made to a fusion of LTF with the Fc domain of IgG2 (FcLTF), which is known to extend LTF half-life in circulation. TDM induced granulomas were examined at extended times post insult (day 7 and 14). Both LTF and the novel FcLTF exerted sustained effects on lung granuloma pathology. Reduction of pulmonary pro-inflammatory cytokines TNF-α and IL-1ß occurred, correlating with reduced pathology. Increase in IL-6, known to regulate granuloma maintenance, was also seen with the LTFs. The FcLTF demonstrated greater impact than the recombinant LTF, and was superior in limiting damage to pulmonary tissues while limiting residual inflammatory cytokine production.


Subject(s)
Cord Factors , Granuloma, Respiratory Tract , Lactoferrin , Lung Diseases , Animals , Humans , Mice , Cord Factors/metabolism , Cord Factors/toxicity , Lactoferrin/therapeutic use , Mycobacterium tuberculosis/metabolism , Granuloma, Respiratory Tract/chemically induced , Granuloma, Respiratory Tract/drug therapy , Lung Diseases/chemically induced , Lung Diseases/drug therapy
4.
PLoS Biol ; 19(8): e3001370, 2021 08.
Article in English | MEDLINE | ID: mdl-34383749

ABSTRACT

The mycobacterial membrane protein large 3 (MmpL3) transporter is essential and required for shuttling the lipid trehalose monomycolate (TMM), a precursor of mycolic acid (MA)-containing trehalose dimycolate (TDM) and mycolyl arabinogalactan peptidoglycan (mAGP), in Mycobacterium species, including Mycobacterium tuberculosis and Mycobacterium smegmatis. However, the mechanism that MmpL3 uses to facilitate the transport of fatty acids and lipidic elements to the mycobacterial cell wall remains elusive. Here, we report 7 structures of the M. smegmatis MmpL3 transporter in its unbound state and in complex with trehalose 6-decanoate (T6D) or TMM using single-particle cryo-electron microscopy (cryo-EM) and X-ray crystallography. Combined with calculated results from molecular dynamics (MD) and target MD simulations, we reveal a lipid transport mechanism that involves a coupled movement of the periplasmic domain and transmembrane helices of the MmpL3 transporter that facilitates the shuttling of lipids to the mycobacterial cell wall.


Subject(s)
Bacterial Proteins/metabolism , Cord Factors/metabolism , Lipid Metabolism , Membrane Transport Proteins/metabolism , Mycobacterium smegmatis/metabolism , Bacterial Proteins/ultrastructure , Cryoelectron Microscopy , Decanoates/metabolism , Escherichia coli , Membrane Transport Proteins/ultrastructure , Molecular Dynamics Simulation , Mycobacterium smegmatis/ultrastructure , Trehalose/metabolism
5.
mBio ; 12(1)2021 01 19.
Article in English | MEDLINE | ID: mdl-33468692

ABSTRACT

The mycomembrane layer of the mycobacterial cell envelope is a barrier to environmental, immune, and antibiotic insults. There is considerable evidence of mycomembrane plasticity during infection and in response to host-mimicking stresses. Since mycobacteria are resource and energy limited under these conditions, it is likely that remodeling has distinct requirements from those of the well-characterized biosynthetic program that operates during unrestricted growth. Unexpectedly, we found that mycomembrane remodeling in nutrient-starved, nonreplicating mycobacteria includes synthesis in addition to turnover. Mycomembrane synthesis under these conditions occurs along the cell periphery, in contrast to the polar assembly of actively growing cells, and both liberates and relies on the nonmammalian disaccharide trehalose. In the absence of trehalose recycling, de novo trehalose synthesis fuels mycomembrane remodeling. However, mycobacteria experience ATP depletion, enhanced respiration, and redox stress, hallmarks of futile cycling and the collateral dysfunction elicited by some bactericidal antibiotics. Inefficient energy metabolism compromises the survival of trehalose recycling mutants in macrophages. Our data suggest that trehalose recycling alleviates the energetic burden of mycomembrane remodeling under stress. Cell envelope recycling pathways are emerging targets for sensitizing resource-limited bacterial pathogens to host and antibiotic pressure.IMPORTANCE The glucose-based disaccharide trehalose is a stress protectant and carbon source in many nonmammalian cells. Mycobacteria are relatively unique in that they use trehalose for an additional, extracytoplasmic purpose: to build their outer "myco" membrane. In these organisms, trehalose connects mycomembrane biosynthesis and turnover to central carbon metabolism. Key to this connection is the retrograde transporter LpqY-SugABC. Unexpectedly, we found that nongrowing mycobacteria synthesize mycomembrane under carbon limitation but do not require LpqY-SugABC. In the absence of trehalose recycling, compensatory anabolism allows mycomembrane biosynthesis to continue. However, this workaround comes at a cost, namely, ATP consumption, increased respiration, and oxidative stress. Strikingly, these phenotypes resemble those elicited by futile cycles and some bactericidal antibiotics. We demonstrate that inefficient energy metabolism attenuates trehalose recycling mutant Mycobacterium tuberculosis in macrophages. Energy-expensive macromolecule biosynthesis triggered in the absence of recycling may be a new paradigm for boosting host activity against bacterial pathogens.


Subject(s)
Cell Membrane/metabolism , Cell Wall/metabolism , Energy Metabolism/drug effects , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/metabolism , Trehalose/metabolism , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/biosynthesis , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Membrane/drug effects , Cell Wall/drug effects , Cord Factors/metabolism , Cord Factors/pharmacology , Diarylquinolines/pharmacology , Energy Metabolism/genetics , Galactans/metabolism , Galactans/pharmacology , Gene Expression/drug effects , Glucose/metabolism , Glucose/pharmacology , Maltose/metabolism , Maltose/pharmacology , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/genetics , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Mycolic Acids/metabolism , Mycolic Acids/pharmacology , Rifampin/pharmacology , Trehalose/pharmacology
6.
J Immunol ; 205(6): 1580-1592, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32796022

ABSTRACT

Mycobacteria survive in macrophages despite triggering pattern recognition receptors and T cell-derived IFN-γ production. Mycobacterial cord factor trehalose-6,6-dimycolate (TDM) binds the C-type lectin receptor MINCLE and induces inflammatory gene expression. However, the impact of TDM on IFN-γ-induced macrophage activation is not known. In this study, we have investigated the cross-regulation of the mouse macrophage transcriptome by IFN-γ and by TDM or its synthetic analogue trehalose-6,6-dibehenate (TDB). As expected, IFN-γ induced genes involved in Ag presentation and antimicrobial defense. Transcriptional programs induced by TDM and TDB were highly similar but clearly distinct from the response to IFN-γ. The glycolipids enhanced expression of a subset of IFN-γ-induced genes associated with inflammation. In contrast, TDM/TDB exerted delayed inhibition of IFN-γ-induced genes, including pattern recognition receptors, MHC class II genes, and IFN-γ-induced GTPases, with antimicrobial function. TDM downregulated MHC class II cell surface expression and impaired T cell activation by peptide-pulsed macrophages. Inhibition of the IFN-γ-induced GTPase GBP1 occurred at the level of transcription by a partially MINCLE-dependent mechanism that may target IRF1 activity. Although activation of STAT1 was unaltered, deletion of Socs1 relieved inhibition of GBP1 expression by TDM. Nonnuclear Socs1 was sufficient for inhibition, suggesting a noncanonical, cytoplasmic mechanism. Taken together, unbiased analysis of transcriptional reprogramming revealed a significant degree of negative regulation of IFN-γ-induced Ag presentation and antimicrobial gene expression by the mycobacterial cord factor that may contribute to mycobacterial persistence.


Subject(s)
Cord Factors/metabolism , GTP-Binding Proteins/metabolism , Inflammation/microbiology , Lectins, C-Type/metabolism , Macrophages/physiology , Membrane Proteins/metabolism , Mycobacterium tuberculosis/physiology , Tuberculosis/microbiology , Animals , Antigen Presentation , Cells, Cultured , GTP-Binding Proteins/genetics , Gene Expression Profiling , Humans , Inflammation/immunology , Interferon-gamma/metabolism , Lectins, C-Type/genetics , Macrophage Activation , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Suppressor of Cytokine Signaling 1 Protein/genetics , Suppressor of Cytokine Signaling 1 Protein/metabolism , Tuberculosis/immunology
7.
Eur J Med Chem ; 200: 112390, 2020 Aug 15.
Article in English | MEDLINE | ID: mdl-32447196

ABSTRACT

The mycobacterial membrane protein Large 3 (MmpL3) is an inner membrane protein that transports trehalose-monomycolates, precursors for trehalose-dimycolates and mycolic acids that make up essential components of the mycobacterial outer membrane. Inhibition of MmpL3 weakens the mycobacterial cell wall and ultimately results in cell death in both in vitro and in vivo infection models. This highlights the therapeutic potential of MmpL3 as a drug target. High-throughput whole-cell screening along with whole genome sequencing of resistant mutants has identified numerous classes of compounds that can be classified as MmpL3 inhibitors. In this review, we provide insights into the current development of various MmpL3 inhibitors and discuss the potential challenges in this area.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Cell Wall/drug effects , Antitubercular Agents/therapeutic use , Bacterial Proteins/metabolism , Cord Factors/metabolism , Humans , Membrane Transport Proteins/metabolism , Mycobacterium tuberculosis/drug effects , Mycolic Acids/metabolism
8.
PLoS Pathog ; 16(4): e1008452, 2020 04.
Article in English | MEDLINE | ID: mdl-32255801

ABSTRACT

The Mycobacterium tuberculosis Ser/Thr protein kinases PknA and PknB are essential for growth and have been proposed as possible drug targets. We used a titratable conditional depletion system to investigate the functions of these kinases. Depletion of PknA or PknB or both kinases resulted in growth arrest, shortening of cells, and time-dependent loss of acid-fast staining with a concomitant decrease in mycolate synthesis and accumulation of trehalose monomycolate. Depletion of PknA and/or PknB resulted in markedly increased susceptibility to ß-lactam antibiotics, and to the key tuberculosis drug rifampin. Phosphoproteomic analysis showed extensive changes in protein phosphorylation in response to PknA depletion and comparatively fewer changes with PknB depletion. These results identify candidate substrates of each kinase and suggest specific and coordinate roles for PknA and PknB in regulating multiple essential physiologies. These findings support these kinases as targets for new antituberculosis drugs and provide a valuable resource for targeted investigation of mechanisms by which protein phosphorylation regulates pathways required for growth and virulence in M. tuberculosis.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Protein Serine-Threonine Kinases/metabolism , Bacterial Proteins/genetics , Cord Factors/metabolism , Gene Expression Regulation, Bacterial/drug effects , Humans , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Protein Serine-Threonine Kinases/genetics , Tuberculosis/microbiology
9.
Proteins ; 88(6): 809-815, 2020 06.
Article in English | MEDLINE | ID: mdl-31833106

ABSTRACT

Trehalose monomycolate (TMM) represents an essential element of the mycobacterial envelope. While synthesized in the cytoplasm, TMM is transported across the inner membrane by MmpL3 but, little is known regarding the MmpL3 partners involved in this process. Recently, the TMM transport factor A (TtfA) was found to form a complex with MmpL3 and to participate in TMM transport, although its biological role remains to be established. Herein, we report the crystal structure of the Mycobacterium smegmatis TtfA core domain. The phylogenetic distribution of TtfA homologues in non-mycolate containing bacteria suggests that TtfA may exert additional functions.


Subject(s)
Bacterial Proteins/chemistry , Cell Wall/chemistry , Cord Factors/chemistry , Membrane Transport Proteins/chemistry , Mycobacterium smegmatis/chemistry , Mycobacterium tuberculosis/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Biological Transport , Cell Wall/metabolism , Cloning, Molecular , Cord Factors/metabolism , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Models, Molecular , Mycobacterium smegmatis/classification , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/classification , Mycobacterium tuberculosis/metabolism , Phylogeny , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Folding , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
10.
J Biol Chem ; 294(46): 17512-17523, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31562241

ABSTRACT

Mycobacterium tuberculosis, the causative agent of tuberculosis, remains a major human pathogen, and current treatment options to combat this disease are under threat because of the emergence of multidrug-resistant and extensively drug-resistant tuberculosis. High-throughput whole-cell screening of an extensive compound library has recently identified a piperidinol-containing molecule, PIPD1, as a potent lead compound against M. tuberculosis Herein, we show that PIPD1 and related analogs exert in vitro bactericidal activity against the M. tuberculosis strain mc26230 and also against a panel of multidrug-resistant and extensively drug-resistant clinical isolates of M. tuberculosis, suggesting that PIPD1's mode of action differs from those of most first- and second-line anti-tubercular drugs. Selection and DNA sequencing of PIPD1-resistant mycobacterial mutants revealed the presence of single-nucleotide polymorphisms in mmpL3, encoding an inner membrane-associated mycolic acid flippase in M. tuberculosis Results from functional assays with spheroplasts derived from a M. smegmatis strain lacking the endogenous mmpL3 gene but harboring the M. tuberculosis mmpL3 homolog indicated that PIPD1 inhibits the MmpL3-driven translocation of trehalose monomycolate across the inner membrane without altering the proton motive force. Using a predictive structural model of MmpL3 from M. tuberculosis, docking studies revealed a PIPD1-binding cavity recently found to accommodate different inhibitors in M. smegmatis MmpL3. In conclusion, our findings have uncovered bactericidal activity of a new chemical scaffold. Its anti-tubercular activity is mediated by direct inhibition of the flippase activity of MmpL3 rather than by inhibition of the inner membrane proton motive force, significantly advancing our understanding of MmpL3-targeted inhibition in mycobacteria.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Mycobacterium tuberculosis/drug effects , Mycolic Acids/metabolism , Piperidines/pharmacology , Antitubercular Agents/chemistry , Bacterial Proteins/metabolism , Biological Transport/drug effects , Cord Factors/metabolism , Humans , Membrane Transport Proteins/metabolism , Microbial Sensitivity Tests , Models, Molecular , Mycobacterium tuberculosis/metabolism , Piperidines/chemistry , Tuberculosis/drug therapy , Tuberculosis/microbiology
11.
Article in English | MEDLINE | ID: mdl-31405862

ABSTRACT

The Mycobacterium tuberculosis mycolate flippase MmpL3 has been the proposed target for multiple inhibitors with diverse chemical scaffolds. This diversity in chemical scaffolds has made it difficult to predict compounds that inhibit MmpL3 without whole-genome sequencing of isolated resistant mutants. Here, we describe the identification of four new inhibitors that select for resistance mutations in mmpL3. Using these resistant mutants, we conducted a targeted whole-cell phenotypic screen of 163 novel M. tuberculosis growth inhibitors for differential growth inhibition of wild-type M. tuberculosis compared to the growth of a pool of 24 unique mmpL3 mutants. The screen successfully identified six additional putative MmpL3 inhibitors. The compounds were bactericidal both in vitro and against intracellular M. tuberculosisM. tuberculosis cells treated with these compounds were shown to accumulate trehalose monomycolates, have reduced levels of trehalose dimycolate, and displace an MmpL3-specific probe, supporting MmpL3 as the target. The inhibitors were mycobacterium specific, with several also showing activity against the nontuberculous mycobacterial species M. abscessus Cluster analysis of cross-resistance profiles generated by dose-response experiments for each combination of 13 MmpL3 inhibitors against each of the 24 mmpL3 mutants defined two clades of inhibitors and two clades of mmpL3 mutants. Pairwise combination studies of the inhibitors revealed interactions that were specific to the clades identified in the cross-resistance profiling. Additionally, modeling of resistance-conferring substitutions to the MmpL3 crystal structure revealed clade-specific localization of the residues to specific domains of MmpL3, with the clades showing differential resistance. Several compounds exhibited high solubility and stability in microsomes and low cytotoxicity in macrophages, supporting their further development. The combined study of multiple mutants and novel compounds provides new insights into structure-function interactions of MmpL3 and small-molecule inhibitors.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/genetics , Benzamides/pharmacology , Benzothiazoles/pharmacology , Drug Resistance, Bacterial/drug effects , Membrane Transport Proteins/genetics , Mycobacterium tuberculosis/drug effects , Pyridines/pharmacology , Antitubercular Agents/chemical synthesis , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Benzamides/chemical synthesis , Benzothiazoles/chemical synthesis , Binding Sites , Biological Transport/drug effects , Cord Factors/antagonists & inhibitors , Cord Factors/biosynthesis , Cord Factors/metabolism , Drug Resistance, Bacterial/genetics , Galactans/metabolism , Gene Expression , High-Throughput Screening Assays , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Microbial Sensitivity Tests , Models, Molecular , Mutation , Mycobacterium abscessus/drug effects , Mycobacterium abscessus/genetics , Mycobacterium abscessus/growth & development , Mycobacterium abscessus/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/metabolism , Mycolic Acids/metabolism , Protein Binding , Protein Structure, Secondary , Pyridines/chemical synthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Whole Genome Sequencing
12.
Article in English | MEDLINE | ID: mdl-31332069

ABSTRACT

1H-benzo[d]imidazole derivatives exhibit antitubercular activity in vitro at a nanomolar range of concentrations and are not toxic to human cells, but their mode of action remains unknown. Here, we showed that these compounds are active against intracellular Mycobacterium tuberculosis To identify their target, we selected drug-resistant M. tuberculosis mutants and then used whole-genome sequencing to unravel mutations in the essential mmpL3 gene, which encodes the integral membrane protein that catalyzes the export of trehalose monomycolate, a precursor of the mycobacterial outer membrane component trehalose dimycolate (TDM), as well as mycolic acids bound to arabinogalactan. The drug-resistant phenotype was also observed in the parental strain overexpressing the mmpL3 alleles carrying the mutations identified in the resistors. However, no cross-resistance was observed between 1H-benzo[d]imidazole derivatives and SQ109, another MmpL3 inhibitor, or other first-line antitubercular drugs. Metabolic labeling and quantitative thin-layer chromatography (TLC) analysis of radiolabeled lipids from M. tuberculosis cultures treated with the benzoimidazoles indicated an inhibition of trehalose dimycolate (TDM) synthesis, as well as reduced levels of mycolylated arabinogalactan, in agreement with the inhibition of MmpL3 activity. Overall, this study emphasizes the pronounced activity of 1H-benzo[d]imidazole derivatives in interfering with mycolic acid metabolism and their potential for therapeutic application in the fight against tuberculosis.


Subject(s)
Antitubercular Agents/pharmacology , Bacterial Proteins/genetics , Benzimidazoles/pharmacology , Cord Factors/antagonists & inhibitors , Drug Resistance, Bacterial/drug effects , Membrane Transport Proteins/genetics , Mycobacterium tuberculosis/drug effects , Amino Acid Motifs , Antitubercular Agents/chemical synthesis , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Benzimidazoles/chemical synthesis , Binding Sites , Biological Transport/drug effects , Cloning, Molecular , Cord Factors/biosynthesis , Cord Factors/metabolism , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Galactans/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Microbial Sensitivity Tests , Models, Molecular , Mutation , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/metabolism , Mycolic Acids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Whole Genome Sequencing
13.
Proc Natl Acad Sci U S A ; 116(23): 11241-11246, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31113875

ABSTRACT

The cell envelope of Mycobacterium tuberculosis is notable for the abundance of mycolic acids (MAs), essential to mycobacterial viability, and of other species-specific lipids. The mycobacterial cell envelope is extremely hydrophobic, which contributes to virulence and antibiotic resistance. However, exactly how fatty acids and lipidic elements are transported across the cell envelope for cell-wall biosynthesis is unclear. Mycobacterial membrane protein Large 3 (MmpL3) is essential and required for transport of trehalose monomycolates (TMMs), precursors of MA-containing trehalose dimycolates (TDM) and mycolyl arabinogalactan peptidoglycan, but the exact function of MmpL3 remains elusive. Here, we report a crystal structure of Mycobacterium smegmatis MmpL3 at a resolution of 2.59 Å, revealing a monomeric molecule that is structurally distinct from all known bacterial membrane proteins. A previously unknown MmpL3 ligand, phosphatidylethanolamine (PE), was discovered inside this transporter. We also show, via native mass spectrometry, that MmpL3 specifically binds both TMM and PE, but not TDM, in the micromolar range. These observations provide insight into the function of MmpL3 and suggest a possible role for this protein in shuttling a variety of lipids to strengthen the mycobacterial cell wall.


Subject(s)
Bacterial Proteins/metabolism , Cord Factors/metabolism , Membrane Transport Proteins/metabolism , Phosphatidylethanolamines/metabolism , Biological Transport/physiology , Cell Membrane/metabolism , Cell Wall/metabolism , Mycobacterium smegmatis/metabolism , Mycolic Acids/metabolism
14.
Tuberculosis (Edinb) ; 116S: S118-S122, 2019 05.
Article in English | MEDLINE | ID: mdl-31072690

ABSTRACT

Mycobacterium tuberculosis (MTB) is a pathogen that infects and kills millions yearly. The mycobacterium's cell wall glycolipid trehalose 6,6'-dimycolate (TDM) has been used historically to model MTB induced inflammation and granuloma formation. Alterations to the model can significantly influence the induced pathology. One such method incorporates intraperitoneal pre-exposure, after which the intravenous injection of TDM generates pathological damage effectively mimicking the hypercoagulation, thrombus formation, and tissue remodeling apparent in lungs of infected individuals. The purpose of these experiments is to examine the histological inflammation involved in the TDM mouse model that induces development of the hemorrhagic response. TDM induced lungs of C57BL/6 mice to undergo granulomatous inflammation. Further histological examination of the peak response demonstrated tissue remodeling consistent with hypercoagulation. The observed vascular occlusion indicates that obstruction likely occurs due to subendothelial localized activity leading to restriction of blood vessel lumens. Trichrome staining revealed that associated damage in the hypercoagulation model is consistent with intra endothelial cell accumulation of innate cells, bordered by collagen deposition in the underlying parenchyma. Overall, the hypercoagulation model represents a comparative pathological instrument for understanding mechanisms underlying development of hemorrhage and vascular occlusion seen during MTB infection.


Subject(s)
Cord Factors/metabolism , Endothelium, Vascular/pathology , Granuloma, Respiratory Tract/pathology , Lung/blood supply , Mycobacterium tuberculosis/metabolism , Pneumonia/pathology , Tuberculosis, Pulmonary/pathology , Animals , Blood Coagulation , Disease Models, Animal , Endothelium, Vascular/microbiology , Female , Granuloma, Respiratory Tract/blood , Granuloma, Respiratory Tract/chemically induced , Granuloma, Respiratory Tract/microbiology , Lung/microbiology , Mice, Inbred C57BL , Pneumonia/blood , Pneumonia/chemically induced , Pneumonia/microbiology , Tuberculosis, Pulmonary/blood , Tuberculosis, Pulmonary/chemically induced , Tuberculosis, Pulmonary/microbiology , Vascular Remodeling
15.
Mol Cell Proteomics ; 18(4): 669-685, 2019 04.
Article in English | MEDLINE | ID: mdl-30635358

ABSTRACT

Immune sensing of Mycobacterium tuberculosis relies on recognition by macrophages. Mycobacterial cord factor, trehalose-6,6'-dimycolate (TDM), is the most abundant cell wall glycolipid and binds to the C-type lectin receptor (CLR) MINCLE. To explore the kinase signaling linking the TDM-MINCLE interaction to gene expression, we employed quantitative phosphoproteome analysis. TDM caused upregulation of 6.7% and suppressed 3.8% of the 14,000 phospho-sites identified on 3727 proteins. MINCLE-dependent phosphorylation was observed for canonical players of CLR signaling (e.g. PLCγ, PKCδ), and was enriched for PKCδ and GSK3 kinase motifs. MINCLE-dependent activation of the PI3K-AKT-GSK3 pathway contributed to inflammatory gene expression and required the PI3K regulatory subunit p85α. Unexpectedly, a substantial fraction of TDM-induced phosphorylation was MINCLE-independent, a finding paralleled by transcriptome data. Bioinformatics analysis of both data sets concurred in the requirement for MINCLE for innate immune response pathways and processes. In contrast, MINCLE-independent phosphorylation and transcriptome responses were linked to cell cycle regulation. Collectively, our global analyses show substantial reprogramming of macrophages by TDM and reveal a dichotomy of MINCLE-dependent and -independent signaling linked to distinct biological responses.


Subject(s)
Cord Factors/metabolism , Lectins, C-Type/metabolism , Macrophages/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Proteome/metabolism , Proteomics , Signal Transduction , Animals , Cell Cycle/genetics , Cell Proliferation/genetics , Cell Survival/genetics , Cord Factors/pharmacology , Cytokines/metabolism , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Glycolipids/metabolism , Kinetics , Macrophage Activation/drug effects , Macrophage Activation/genetics , Macrophages/drug effects , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Mycobacterium tuberculosis/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Syk Kinase/metabolism , Transcriptome/genetics , Trehalose/metabolism
16.
Chembiochem ; 20(10): 1282-1291, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30589191

ABSTRACT

Mycobacteria and related organisms in the Corynebacterineae suborder are characterized by a distinctive outer membrane referred to as the mycomembrane. Biosynthesis of the mycomembrane occurs through an essential process called mycoloylation, which involves antigen 85 (Ag85)-catalyzed transfer of mycolic acids from the mycoloyl donor trehalose monomycolate (TMM) to acceptor carbohydrates and, in some organisms, proteins. We recently described an alkyne-modified TMM analogue (O-AlkTMM-C7) which, in conjunction with click chemistry, acted as a chemical reporter for mycoloylation in intact cells and allowed metabolic labeling of mycoloylated components of the mycomembrane. Here, we describe the synthesis and evaluation of a toolbox of TMM-based reporters bearing alkyne, azide, trans-cyclooctene, and fluorescent tags. These compounds gave further insight into the substrate tolerance of mycoloyltransferases (e.g., Ag85s) in a cellular context and they provide significantly expanded experimental versatility by allowing one- or two-step cell labeling, live cell labeling, and rapid cell labeling via tetrazine ligation. Such capabilities will facilitate research on mycomembrane composition, biosynthesis, and dynamics. Moreover, because TMM is exclusively metabolized by Corynebacterineae, the described probes may be valuable for the specific detection and cell-surface engineering of Mycobacterium tuberculosis and related pathogens. We also performed experiments to establish the dependence of probe incorporation on mycoloyltransferase activity, results from which suggested that cellular labeling is a function not only of metabolic incorporation (and likely removal) pathway(s), but also accessibility across the envelope. Thus, whole-cell labeling experiments with TMM reporters should be carefully designed and interpreted when envelope permeability may be compromised. On the other hand, this property of TMM reporters can potentially be exploited as a convenient way to probe changes in envelope integrity and permeability, facilitating drug development studies.


Subject(s)
Cell Membrane/chemistry , Cord Factors/chemistry , Corynebacterium/chemistry , Acyltransferases/metabolism , Alkynes/chemical synthesis , Alkynes/chemistry , Alkynes/metabolism , Azides/chemical synthesis , Azides/chemistry , Azides/metabolism , Bacillus subtilis/chemistry , Cell Engineering/methods , Cell Membrane/metabolism , Click Chemistry , Cord Factors/chemical synthesis , Cord Factors/metabolism , Escherichia coli/chemistry , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Fluorescent Dyes/metabolism , Molecular Structure , Mycobacterium smegmatis/chemistry , Mycobacterium tuberculosis/chemistry
17.
Cell Host Microbe ; 24(4): 514-525.e6, 2018 10 10.
Article in English | MEDLINE | ID: mdl-30308157

ABSTRACT

Mycobacterial infection leads to the formation of characteristic immune aggregates called granulomas, a process accompanied by dramatic remodeling of the host vasculature. As granuloma angiogenesis favors the infecting mycobacteria, it may be actively promoted by bacterial determinants during infection. Using Mycobacterium marinum-infected zebrafish as a model, we identify the enzyme proximal cyclopropane synthase of alpha-mycolates (PcaA) as an important bacterial determinant of granuloma-associated angiogenesis. cis-Cyclopropanation of mycobacterial mycolic acids by pcaA drives the activation of host Vegf signaling within granuloma macrophages. Cyclopropanation of the mycobacterial cell wall glycolipid trehalose dimycolate is both required and sufficient to induce robust host angiogenesis. Inducible genetic inhibition of angiogenesis and Vegf signaling during granuloma formation results in bacterial growth deficits. Together, these data reveal a mechanism by which PcaA-mediated cis-cyclopropanation of mycolic acids promotes bacterial growth and dissemination in vivo by eliciting granuloma vascularization and suggest potential approaches for host-directed therapies.


Subject(s)
Bacterial Proteins/metabolism , Methyltransferases/metabolism , Mycobacterium marinum/enzymology , Neovascularization, Pathologic/microbiology , Receptors, Vascular Endothelial Growth Factor/metabolism , Tuberculoma/microbiology , Angiogenesis Inhibitors/pharmacology , Animals , Bacterial Proteins/genetics , Cord Factors/metabolism , Disease Models, Animal , Humans , Indazoles , Macrophages/immunology , Macrophages/microbiology , Methyltransferases/genetics , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium marinum/genetics , Mycobacterium marinum/pathogenicity , Mycolic Acids/metabolism , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Pyrimidines/pharmacology , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/drug effects , Signal Transduction , Sulfonamides/pharmacology , Tuberculoma/immunology , Tuberculoma/pathology , Zebrafish
18.
Proc Natl Acad Sci U S A ; 115(20): 5271-5276, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29703753

ABSTRACT

Control and manipulation of bacterial populations requires an understanding of the factors that govern growth, division, and antibiotic action. Fluorescent and chemically reactive small molecule probes of cell envelope components can visualize these processes and advance our knowledge of cell envelope biosynthesis (e.g., peptidoglycan production). Still, fundamental gaps remain in our understanding of the spatial and temporal dynamics of cell envelope assembly. Previously described reporters require steps that limit their use to static imaging. Probes that can be used for real-time imaging would advance our understanding of cell envelope construction. To this end, we synthesized a fluorogenic probe that enables continuous live cell imaging in mycobacteria and related genera. This probe reports on the mycolyltransferases that assemble the mycolic acid membrane. This peptidoglycan-anchored bilayer-like assembly functions to protect these cells from antibiotics and host defenses. Our probe, quencher-trehalose-fluorophore (QTF), is an analog of the natural mycolyltransferase substrate. Mycolyltransferases process QTF by diverting their normal transesterification activity to hydrolysis, a process that unleashes fluorescence. QTF enables high contrast continuous imaging and the visualization of mycolyltransferase activity in cells. QTF revealed that mycolyltransferase activity is augmented before cell division and localized to the septa and cell poles, especially at the old pole. This observed localization suggests that mycolyltransferases are components of extracellular cell envelope assemblies, in analogy to the intracellular divisomes and polar elongation complexes. We anticipate QTF can be exploited to detect and monitor mycobacteria in physiologically relevant environments.


Subject(s)
Cell Wall/metabolism , Cord Factors/metabolism , Corynebacterium glutamicum/growth & development , Fluorescent Dyes/chemistry , Image Processing, Computer-Assisted/methods , Mycobacterium tuberculosis/growth & development , Tuberculosis/diagnosis , Bacterial Proteins/metabolism , Cell Division , Fluorescence , Humans , Peptidoglycan/metabolism , Tuberculosis/metabolism , Tuberculosis/microbiology
19.
Microbiol Immunol ; 61(12): 523-530, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28976590

ABSTRACT

Trehalose 6,6'-dimycolate (TDM), or cord factor, is a crucial stimulus of immune responses during Mycobacterium tuberculosis infection. Although TDM has immuno-stimulatory properties, including adjuvant activity and the ability to induce granuloma formation, the mechanisms underlying these remain unknown. We hypothesized that TDM stimulates transendothelial migration of neutrophils, which are the first immune cells to infiltrate the tissue upon infection. In this study, it was shown that TDM enhances N-formylmethionyl-leucyl-phenylalanine (fMLP)-induced chemotaxis and transendothelial movement by prolonging AKT phosphorylation in human neutrophils. TDM induced expression of macrophage-inducible C-type lectin, a receptor for TDM, and induced secretion of pro-inflammatory cytokines and chemokines in differentiated HL-60 cells. In 2- and 3-D neutrophil migration assays, TDM-stimulated neutrophils showed increased fMLP-induced chemotaxis and transendothelial migration. Interestingly, following fMLP stimulation of TDM-activated neutrophils, AKT, a crucial kinase for neutrophil polarization and chemotaxis, showed prolonged phosphorylation at serine 473. Taken together, these data suggest that TDM modulates transendothelial migration of neutrophils upon mycobacterial infection through prolonged AKT phosphorylation. AKT may therefore be a promising therapeutic target for enhancing immune responses to mycobacterial infection.


Subject(s)
Cell Movement , Cord Factors/metabolism , Mycobacterium tuberculosis/metabolism , Neutrophils/cytology , Proto-Oncogene Proteins c-akt/metabolism , Tuberculosis/enzymology , Amino Acid Motifs , HL-60 Cells , Host-Pathogen Interactions , Humans , Mycobacterium tuberculosis/genetics , N-Formylmethionine Leucyl-Phenylalanine/metabolism , Neutrophils/enzymology , Neutrophils/metabolism , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/genetics , Tuberculosis/genetics , Tuberculosis/microbiology , Tuberculosis/physiopathology
20.
Proc Natl Acad Sci U S A ; 114(30): 7993-7998, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28698380

ABSTRACT

The defining feature of the mycobacterial outer membrane (OM) is the presence of mycolic acids (MAs), which, in part, render the bilayer extremely hydrophobic and impermeable to external insults, including many antibiotics. Although the biosynthetic pathway of MAs is well studied, the mechanism(s) by which these lipids are transported across the cell envelope is(are) much less known. Mycobacterial membrane protein Large 3 (MmpL3), an essential inner membrane (IM) protein, is implicated in MA transport, but its exact function has not been elucidated. It is believed to be the cellular target of several antimycobacterial compounds; however, evidence for direct inhibition of MmpL3 activity is also lacking. Here, we establish that MmpL3 is the MA flippase at the IM of mycobacteria and is the molecular target of BM212, a 1,5-diarylpyrrole compound. We develop assays that selectively access mycolates on the surface of Mycobacterium smegmatis spheroplasts, allowing us to monitor flipping of MAs across the IM. Using these assays, we establish the mechanism of action of BM212 as a potent MmpL3 inhibitor, and use it as a molecular probe to demonstrate the requirement for functional MmpL3 in the transport of MAs across the IM. Finally, we show that BM212 binds MmpL3 directly and inhibits its activity. Our work provides fundamental insights into OM biogenesis and MA transport in mycobacteria. Furthermore, our assays serve as an important platform for accelerating the validation of small molecules that target MmpL3, and their development as future antituberculosis drugs.


Subject(s)
Bacterial Proteins/metabolism , Cord Factors/metabolism , Membrane Proteins/metabolism , Mycobacterium smegmatis/enzymology , Mycolic Acids/metabolism , Lipid Metabolism , Piperazines , Pyrroles , Spheroplasts
SELECTION OF CITATIONS
SEARCH DETAIL
...