Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Org Chem ; 83(15): 8054-8080, 2018 08 03.
Article in English | MEDLINE | ID: mdl-29979042

ABSTRACT

The rhodium(II)-catalyzed oxidative cyclization of glycal 3-carbamates with in situ incorporation of an alcohol nucleophile at the anomeric position provides access to a range of 2-amino sugars having 1,2-trans-2,3-cis stereochemistry, a structural motif present in compounds of medicinal and biological significance such as the streptothricin group of antibiotics and the Chitinase inhibitor allosamidin. All of the diastereomeric d-glycal 3-carbamates have been investigated, revealing significant differences in anomeric stereoselectivity depending on substrate stereochemistry and protecting groups. In addition, some substrates were prone to forming C3-oxidized dihydropyranone byproducts under the reaction conditions. Allal- and gulal 3-carbamates provided uniformly high stereo- and chemoselectivity, while for glucal substrates, acyclic, electron-withdrawing protecting groups at the 4 O and 6 O positions were required. Galactal 3-carbamates have been the most challenging substrates; formation of their amidoglycosylation products is most effective with an electron-withdrawing 6 O-Ts substituent and a sterically demanding 4 O-TBS group. These results suggest a mechanism whereby conformational and electronic factors determine the partitioning of an intermediate acyl nitrenoid between alkene addition, leading to amidoglycosylation, and C3-H insertion, providing the dihydropyranone byproduct. Along the amidoglycosylation pathway, high anomeric selectivity results when a glycosyl aziridine intermediate is favored over an aziridine-opened oxocarbenium donor.


Subject(s)
Carbamates/chemistry , Carbamates/chemical synthesis , Imines/chemistry , Rhodium/chemistry , Carbohydrate Conformation , Catalysis , Chemistry Techniques, Synthetic , Cyclization , Glycosylation , Oxidation-Reduction , Stereoisomerism
2.
Cell Rep ; 23(5): 1435-1447, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29719256

ABSTRACT

There are a limited number of adjuvants that elicit effective cell-based immunity required for protection against intracellular bacterial pathogens. Here, we report that STING-activating cyclic dinucleotides (CDNs) formulated in a protein subunit vaccine elicit long-lasting protective immunity to Mycobacterium tuberculosis in the mouse model. Subcutaneous administration of this vaccine provides equivalent protection to that of the live attenuated vaccine strain Bacille Calmette-Guérin (BCG). Protection is STING dependent but type I IFN independent and correlates with an increased frequency of a recently described subset of CXCR3-expressing T cells that localize to the lung parenchyma. Intranasal delivery results in superior protection compared with BCG, significantly boosts BCG-based immunity, and elicits both Th1 and Th17 immune responses, the latter of which correlates with enhanced protection. Thus, a CDN-adjuvanted protein subunit vaccine has the capability of eliciting a multi-faceted immune response that results in protection from infection by an intracellular pathogen.


Subject(s)
Adjuvants, Immunologic/pharmacology , BCG Vaccine/pharmacology , Membrane Proteins/immunology , Mycobacterium tuberculosis/immunology , Th17 Cells/immunology , Tuberculosis, Pulmonary/prevention & control , Animals , BCG Vaccine/immunology , Disease Models, Animal , Immunity, Cellular/drug effects , Mice , Mice, Knockout , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/pathology , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/pathology , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacokinetics
3.
ACS Infect Dis ; 3(8): 564-574, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28537707

ABSTRACT

Host-directed therapeutics have the potential to combat the global tuberculosis pandemic. We previously identified gefitinib, an inhibitor of EGFR, as a potential host-targeted therapeutic effective against Mycobacterium tuberculosis infection of macrophages and mice. Here we examine the functional consequences of gefitinib treatment on M. tuberculosis infected macrophages. Using phosphoproteomic and transcriptional profiling, we identify two mechanisms by which gefitinib influences macrophage responses to infection to affect cytokine responses and limit replication of M. tuberculosis in macrophages. First, we find that gefitinib treatment of M. tuberculosis infected macrophages inhibits STAT3, a transcription factor known to repress effective immune responses to M. tuberculosis in vivo. Second, we find that gefitinib treatment of M. tuberculosis infected macrophages leads to increased expression of genes involved in lysosomal biogenesis and function and an increase of functional lysosomes in gefitinib treated cells. Furthermore, we show that gefitinib treatment increases the targeting of bacteria to lysosomes, providing an explanation for the cell intrinsic effects of gefitinib treatment on M. tuberculosis infection. Our data provide novel insights into the effects of gefitinib on mammalian cells and into the possible roles for EGFR signaling in macrophages.


Subject(s)
Antitubercular Agents/pharmacology , Lysosomes/drug effects , Macrophages/drug effects , Mycobacterium tuberculosis/drug effects , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Cells/microbiology , Cytokines/antagonists & inhibitors , Cytokines/genetics , Cytokines/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gefitinib , Gene Expression Regulation , Lysosomes/microbiology , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/pathogenicity , Organelle Biogenesis , Phosphoproteins/genetics , Phosphoproteins/metabolism , Primary Cell Culture , Proteomics/methods , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction
4.
ACS Infect Dis ; 2(11): 800-806, 2016 11 11.
Article in English | MEDLINE | ID: mdl-27933784

ABSTRACT

Sulfomenaquinone (SMK) is a recently identified metabolite that is unique to the Mycobacterium tuberculosis (M. tuberculosis) complex and is shown to modulate its virulence. Here, we report the identification of the SMK biosynthetic operon that, in addition to a previously identified sulfotransferase stf3, includes a putative cytochrome P450 gene (cyp128) and a gene of unknown function, rv2269c. We demonstrate that cyp128 and stf3 are sufficient for the biosynthesis of SMK from menaquinone and rv2269c exhibits promoter activity in M. tuberculosis. Loss of Stf3 expression, but not that of Cyp128, is correlated with elevated levels of menaquinone-9, an essential component in the electron-transport chain in M. tuberculosis. Finally, we showed in a mouse model of infection that the loss of cyp128 exhibits a hypervirulent phenotype similar to that in previous studies of the stf3 mutant. These findings provide a platform for defining the molecular basis of SMK's role in M. tuberculosis pathogenesis.


Subject(s)
Mycobacterium tuberculosis/metabolism , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/microbiology , Vitamin K 2/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Mice , Mycobacterium tuberculosis/genetics , Operon , Virulence
5.
J Immunol ; 197(4): 1287-97, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27430718

ABSTRACT

The cytokine IFN-γ coordinates macrophage activation and is essential for control of pathogens, including Mycobacterium tuberculosis However, the mechanisms by which IFN-γ controls M. tuberculosis infection are only partially understood. In this study, we show that the transcription factor hypoxia-inducible factor-1α (HIF-1α) is an essential mediator of IFN-γ-dependent control of M. tuberculosis infection both in vitro and in vivo. M. tuberculosis infection of IFN-γ-activated macrophages results in a synergistic increase in HIF-1α protein levels. This increase in HIF-1α levels is functionally important, as macrophages lacking HIF-1α are defective for IFN-γ-dependent control of infection. RNA-sequencing demonstrates that HIF-1α regulates nearly one-half of all IFN-γ-inducible genes during infection of macrophages. In particular, HIF-1α regulates production of important immune effectors, including inflammatory cytokines and chemokines, eicosanoids, and NO. In addition, we find that during infection HIF-1α coordinates a metabolic shift to aerobic glycolysis in IFN-γ-activated macrophages. We find that this enhanced glycolytic flux is crucial for IFN-γ-dependent control of infection in macrophages. Furthermore, we identify a positive feedback loop between HIF-1α and aerobic glycolysis that amplifies macrophage activation. Finally, we demonstrate that HIF-1α is crucial for control of infection in vivo as mice lacking HIF-1α in the myeloid lineage are strikingly susceptible to infection and exhibit defective production of inflammatory cytokines and microbicidal effectors. In conclusion, we have identified HIF-1α as a novel regulator of IFN-γ-dependent immunity that coordinates an immunometabolic program essential for control of M. tuberculosis infection in vitro and in vivo.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Interferon-gamma/immunology , Macrophage Activation/immunology , Tuberculosis/immunology , Animals , Blotting, Western , Chromatography, Liquid , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Feedback, Physiological , Glycolysis/physiology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mycobacterium tuberculosis , Polymerase Chain Reaction , Tandem Mass Spectrometry
6.
PLoS One ; 8(6): e65080, 2013.
Article in English | MEDLINE | ID: mdl-23762287

ABSTRACT

The genome of Mycobacterium tuberculosis (Mtb) encodes nine putative sulfatases, none of which have a known function or substrate. Here, we characterize Mtb's single putative type II sulfatase, Rv3406, as a non-heme iron (II) and α-ketoglutarate-dependent dioxygenase that catalyzes the oxidation and subsequent cleavage of alkyl sulfate esters. Rv3406 was identified based on its homology to the alkyl sulfatase AtsK from Pseudomonas putida. Using an in vitro biochemical assay, we confirmed that Rv3406 is a sulfatase with a preference for alkyl sulfate substrates similar to those processed by AtsK. We determined the crystal structure of the apo Rv3406 sulfatase at 2.5 Å. The active site residues of Rv3406 and AtsK are essentially superimposable, suggesting that the two sulfatases share the same catalytic mechanism. Finally, we generated an Rv3406 mutant (Δrv3406) in Mtb to study the sulfatase's role in sulfate scavenging. The Δrv3406 strain did not replicate in minimal media with 2-ethyl hexyl sulfate as the sole sulfur source, in contrast to wild type Mtb or the complemented strain. We conclude that Rv3406 is an iron and α-ketoglutarate-dependent sulfate ester dioxygenase that has unique substrate specificity that is likely distinct from other Mtb sulfatases.


Subject(s)
Bacterial Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Sulfatases/metabolism , Sulfates/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biocatalysis , Catalytic Domain , Crystallography, X-Ray , Iron/chemistry , Ketoglutaric Acids/chemistry , Models, Molecular , Molecular Sequence Data , Mycobacterium tuberculosis/genetics , Oxidation-Reduction , Protein Conformation , Pseudomonas putida/enzymology , Pseudomonas putida/genetics , Sequence Homology, Amino Acid , Substrate Specificity , Sulfatases/chemistry , Sulfatases/genetics , Sulfates/chemistry
7.
PLoS One ; 7(1): e29266, 2012.
Article in English | MEDLINE | ID: mdl-22279533

ABSTRACT

Research on the human pathogen Mycobacterium tuberculosis (Mtb) would benefit from novel tools for regulated gene expression. Here we describe the characterization and application of a synthetic riboswitch-based system, which comprises a mycobacterial promoter for transcriptional control and a riboswitch for translational control. The system was used to induce and repress heterologous protein overexpression reversibly, to create a conditional gene knockdown, and to control gene expression in a macrophage infection model. Unlike existing systems for controlling gene expression in Mtb, the riboswitch does not require the co-expression of any accessory proteins: all of the regulatory machinery is encoded by a short DNA segment directly upstream of the target gene. The inducible riboswitch platform has the potential to be a powerful general strategy for creating customized gene regulation systems in Mtb.


Subject(s)
Gene Expression Regulation, Bacterial , Mycobacterium tuberculosis/genetics , Riboswitch/genetics , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Catalase/genetics , Catalase/metabolism , Cell Line , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunoblotting , Macrophages/drug effects , Macrophages/microbiology , Mice , Microscopy, Fluorescence , Molecular Sequence Data , Mutagenesis, Site-Directed , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/growth & development , Promoter Regions, Genetic/genetics , Theophylline/pharmacology , Tuberculosis/microbiology , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
8.
Bioorg Med Chem Lett ; 21(17): 4956-9, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21795043

ABSTRACT

CysQ is a 3'-phosphoadenosine-5'-phosphatase that dephosphorylates intermediates from the sulfate assimilation pathway of Mycobacterium tuberculosis (Mtb). Here, we demonstrate that cysQ disruption attenuates Mtb growth in vitro and decreases the biosynthesis of sulfated glycolipids but not major thiols, suggesting that the encoded enzyme specifically regulates mycobacterial sulfation.


Subject(s)
Glycolipids/biosynthesis , Mycobacterium tuberculosis/enzymology , Phosphoric Monoester Hydrolases/metabolism , Sulfates/chemistry , Chromatography, Liquid , Glycolipids/chemistry , Mycobacterium tuberculosis/growth & development
9.
Org Lett ; 11(7): 1527-30, 2009 Apr 02.
Article in English | MEDLINE | ID: mdl-19254041

ABSTRACT

In the Rh(2)(OAc)(4)-catalyzed amidoglycosylation of glucal 3-carbamates, anomeric stereoselectivity and the extent of competing C3-H oxidation depend on the 4O and 6O protecting groups. Acyclic protection permits high alpha-anomer selectivity with further improvement in less polar solvents, while electron-withdrawing protecting groups limit C3-oxidized byproducts. Stereocontrol and bifurcation between alkene insertion and C3-H oxidation reflect an interplay of conformational, stereoelectronic, and inductive factors.


Subject(s)
Calcium Gluconate/chemistry , Carbamates/chemistry , Hexosamines/chemical synthesis , Mannose/chemical synthesis , Oxazolidinones/chemical synthesis , Catalysis , Combinatorial Chemistry Techniques , Glycosylation , Hexosamines/chemistry , Mannose/analogs & derivatives , Molecular Structure , Oxazolidinones/chemistry , Stereoisomerism
10.
ACS Chem Biol ; 3(10): 619-24, 2008 Oct 17.
Article in English | MEDLINE | ID: mdl-18928249

ABSTRACT

Mycobacterium tuberculosis, the causative agent of tuberculosis, produces unique sulfated metabolites associated with virulence. One such metabolite from M. tuberculosis lipid extracts, S881, has been shown to negatively regulate the virulence of M. tuberculosis in mouse infection studies, and its cell-surface localization suggests a role in modulating host-pathogen interactions. However, a detailed structural analysis of S881 has remained elusive. Here we use high-resolution, high-mass-accuracy, and tandem mass spectrometry to characterize the structure of S881. Exact mass measurements showed that S881 is highly unsaturated, tandem mass spectrometry indicated a polyisoprene-derived structure, and characterization of synthetic structural analogs confirmed that S881 is a previously undescribed sulfated derivative of dihydromenaquinone-9, the primary quinol electron carrier in M. tuberculosis. To our knowledge, this is the first example of a sulfated menaquinone produced in any prokaryote. Together with previous studies, these findings suggest that this redox cofactor may play a role in mycobacterial pathogenesis.


Subject(s)
Mycobacterium tuberculosis/metabolism , Sulfuric Acid Esters/chemistry , Vitamin K 2/chemistry , Animals , Genes, Bacterial , Mice , Molecular Structure , Sulfuric Acid Esters/isolation & purification , Sulfuric Acid Esters/metabolism , Tandem Mass Spectrometry , Vitamin K 2/isolation & purification , Vitamin K 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...