Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Chem Commun (Camb) ; 53(53): 7385-7388, 2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28613292

ABSTRACT

Here we report a ratiometric fluorescent probe for chemoselective conjugation to sulfenic acids in living cells. Our approach couples an α-fluoro-substituted dimedone to an aminonaphthalene fluorophore (F-DiNap), which upon sulfenic acid conjugation is locked as the 1,3-diketone, changing the fluorophore excitation. F-DiNap reacts with S-sulfenylated proteins at equivalent rates to current probes, but the α-fluorine substitution blocks side-reactions with biological aldehydes.

2.
ACS Chem Biol ; 11(12): 3374-3382, 2016 12 16.
Article in English | MEDLINE | ID: mdl-27748579

ABSTRACT

Post-translational S-palmitoylation directs the trafficking and membrane localization of hundreds of cellular proteins, often involving a coordinated palmitoylation cycle that requires both protein acyl transferases (PATs) and acyl protein thioesterases (APTs) to actively redistribute S-palmitoylated proteins toward different cellular membrane compartments. This process is necessary for the trafficking and oncogenic signaling of S-palmitoylated Ras isoforms, and potentially many peripheral membrane proteins. The depalmitoylating enzymes APT1 and APT2 are separately conserved in all vertebrates, suggesting unique functional roles for each enzyme. The recent discovery of the APT isoform-selective inhibitors ML348 and ML349 has opened new possibilities to probe the function of each enzyme, yet it remains unclear how each inhibitor achieves orthogonal inhibition. Herein, we report the high-resolution structure of human APT2 in complex with ML349 (1.64 Å), as well as the complementary structure of human APT1 bound to ML348 (1.55 Å). Although the overall peptide backbone structures are nearly identical, each inhibitor adopts a distinct conformation within each active site. In APT1, the trifluoromethyl group of ML348 is positioned above the catalytic triad, but in APT2, the sulfonyl group of ML349 forms hydrogen bonds with active site resident waters to indirectly engage the catalytic triad and oxyanion hole. Reciprocal mutagenesis and activity profiling revealed several differing residues surrounding the active site that serve as critical gatekeepers for isoform accessibility and dynamics. Structural and biochemical analysis suggests the inhibitors occupy a putative acyl-binding region, establishing the mechanism for isoform-specific inhibition, hydrolysis of acyl substrates, and structural orthogonality important for future probe development.


Subject(s)
Enzyme Inhibitors/pharmacology , Thiolester Hydrolases/antagonists & inhibitors , Amino Acid Sequence , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Protein Conformation, alpha-Helical/drug effects , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Thiolester Hydrolases/chemistry , Thiolester Hydrolases/metabolism
3.
Medchemcomm ; 7(1): 11-27, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26877861

ABSTRACT

Aminoglycoside (AG) antibiotics are used to treat many Gram-negative and some Gram-positive infections and, importantly, multidrug-resistant tuberculosis. Among various bacterial species, resistance to AGs arises through a variety of intrinsic and acquired mechanisms. The bacterial cell wall serves as a natural barrier for small molecules such as AGs and may be further fortified via acquired mutations. Efflux pumps work to expel AGs from bacterial cells, and modifications here too may cause further resistance to AGs. Mutations in the ribosomal target of AGs, while rare, also contribute to resistance. Of growing clinical prominence is resistance caused by ribosome methyltransferases. By far the most widespread mechanism of resistance to AGs is the inactivation of these antibiotics by AG-modifying enzymes. We provide here an overview of these mechanisms by which bacteria become resistant to AGs and discuss their prevalence and potential for clinical relevance.

4.
J Am Chem Soc ; 138(6): 1852-9, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26780921

ABSTRACT

Cysteine S-nitrosation and S-sulfination are naturally occurring post-translational modifications (PTMs) on proteins induced by physiological signals and redox stress. Here we demonstrate that sulfinic acids and nitrosothiols react to form a stable thiosulfonate bond, and leverage this reactivity using sulfinate-linked probes to enrich and annotate hundreds of endogenous S-nitrosated proteins. In physiological buffers, sulfinic acids do not react with iodoacetamide or disulfides, enabling selective alkylation of free thiols and site-specific analysis of S-nitrosation. In parallel, S-nitrosothiol-linked probes enable enrichment and detection of endogenous S-sulfinated proteins, confirming that a single sulfinic acid can react with a nitrosothiol to form a thiosulfonate linkage. Using this approach, we find that hydrogen peroxide addition increases S-sulfination of human DJ-1 (PARK7) at Cys106, whereas Cys46 and Cys53 are fully oxidized to sulfonic acids. Comparative gel-based analysis of different mouse tissues reveals distinct profiles for both S-nitrosation and S-sulfination. Quantitative proteomic analysis demonstrates that both S-nitrosation and S-sulfination are widespread, yet exhibit enhanced occupancy on select proteins, including thioredoxin, peroxiredoxins, and other validated redox active proteins. Overall, we present a direct, bidirectional method to profile select redox cysteine modifications based on the unique nucleophilicity of sulfinic acids.


Subject(s)
Cysteine/chemistry , Cross Reactions , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Nitroso Compounds/chemistry , Oncogene Proteins/chemistry , Oxidation-Reduction , Protein Deglycase DJ-1 , Sulfhydryl Compounds/chemistry , Sulfinic Acids/chemistry
5.
Nat Prod Rep ; 32(5): 641-53, 2015 May.
Article in English | MEDLINE | ID: mdl-25622971

ABSTRACT

Nonribosomal peptides (NRPs) account for a large portion of drugs and drug leads currently available in the pharmaceutical industry. They are one of two main families of natural products biosynthesized on megaenzyme assembly-lines composed of multiple modules that are, in general, each comprised of three core domains and on occasion of accompanying auxiliary domains. The core adenylation (A) domains are known to delineate the identity of the specific chemical components to be incorporated into the growing NRPs. Previously believed to be inactive, A domains interrupted by auxiliary enzymes have recently been proven to be active and capable of performing two distinct chemical reactions. This highlight summarizes current knowledge on A domains and presents the various interrupted A domains found in a number of nonribosomal peptide synthetase (NRPS) assembly-lines, their predicted or proven dual functions, and their potential for manipulation and engineering for chemoenzymatic synthesis of new pharmaceutical agents with increased potency.


Subject(s)
Peptide Biosynthesis, Nucleic Acid-Independent , Peptide Synthases/metabolism , Peptides/metabolism , Molecular Structure , Peptides/chemistry
6.
Biochemistry ; 52(30): 5125-32, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23837529

ABSTRACT

The upsurge in drug-resistant tuberculosis (TB) is an emerging global problem. The increased expression of the enhanced intracellular survival (Eis) protein is responsible for the clinical resistance to aminoglycoside (AG) antibiotics of Mycobacterium tuberculosis . Eis from M. tuberculosis (Eis_Mtb) and M. smegmatis (Eis_Msm) function as acetyltransferases capable of acetylating multiple amines of many AGs; however, these Eis homologues differ in AG substrate preference and in the number of acetylated amine groups per AG. The AG binding cavity of Eis_Mtb is divided into two narrow channels, whereas Eis_Msm contains one large cavity. Five bulky residues lining one of the AG binding channels of Eis_Mtb, His119, Ile268, Trp289, Gln291, and Glu401, have significantly smaller counterparts in Eis_Msm, Thr119, Gly266, Ala287, Ala289, and Gly401, respectively. To identify the residue(s) responsible for AG binding in Eis_Mtb and for the functional differences from Eis_Msm, we have generated single, double, triple, quadruple, and quintuple mutants of these residues in Eis_Mtb by mutating them into their Eis_Msm counterparts, and we tested their acetylation activity with three structurally diverse AGs: kanamycin A (KAN), paromomyin (PAR), and apramycin (APR). We show that penultimate C-terminal residue Glu401 plays a critical role in the overall activity of Eis_Mtb. We also demonstrate that the identities of residues Ile268, Trp289, and Gln291 (in Eis_Mtb nomenclature) dictate the differences between the acetylation efficiencies of Eis_Mtb and Eis_Msm for KAN and PAR. Finally, we show that the mutation of Trp289 in Eis_Mtb into Ala plays a role in APR acetylation.


Subject(s)
Acetyltransferases/metabolism , Aminoglycosides/metabolism , Antibiotics, Antitubercular/metabolism , Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Mycobacterium tuberculosis/enzymology , Acetylation , Acetyltransferases/chemistry , Acetyltransferases/genetics , Amino Acid Sequence , Amino Acid Substitution , Aminoglycosides/chemistry , Aminoglycosides/pharmacology , Antibiotics, Antitubercular/chemistry , Antibiotics, Antitubercular/pharmacology , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Drug Resistance, Multiple, Bacterial , Kanamycin/chemistry , Kanamycin/metabolism , Kanamycin/pharmacology , Kinetics , Molecular Conformation , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mycobacterium smegmatis/drug effects , Mycobacterium smegmatis/enzymology , Mycobacterium smegmatis/metabolism , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/metabolism , Nebramycin/analogs & derivatives , Nebramycin/chemistry , Nebramycin/metabolism , Nebramycin/pharmacology , Paromomycin/chemistry , Paromomycin/metabolism , Paromomycin/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Substrate Specificity
7.
Future Med Chem ; 5(11): 1285-309, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23859208

ABSTRACT

Shortly after the discovery of the first antibiotics, bacterial resistance began to emerge. Many mechanisms give rise to resistance; the most prevalent mechanism of resistance to the aminoglycoside (AG) family of antibiotics is the action of aminoglycoside-modifying enzymes (AMEs). Since the identification of these modifying enzymes, many efforts have been put forth to prevent their damaging alterations of AGs. These diverse strategies are discussed within this review, including: creating new AGs that are unaffected by AMEs; developing inhibitors of AMEs to be co-delivered with AGs; or regulating AME expression. Modern high-throughput methods as well as drug combinations and repurposing are highlighted as recent drug-discovery efforts towards fighting the increasing antibiotic resistance crisis.


Subject(s)
Aminoglycosides/pharmacology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Drug Resistance, Bacterial/drug effects , Acetyltransferases/antagonists & inhibitors , Acetyltransferases/metabolism , Aminoglycosides/chemistry , Aminoglycosides/therapeutic use , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Bacterial Proteins/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Kanamycin Kinase/antagonists & inhibitors , Kanamycin Kinase/metabolism , Nucleotidyltransferases/antagonists & inhibitors , Nucleotidyltransferases/metabolism
8.
J Med Chem ; 56(13): 5613-25, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23786357

ABSTRACT

In this study, we describe the synthesis of a full set of homo- and heterodimers of three intact structures of different ribosome-targeting antibiotics: tobramycin, clindamycin, and chloramphenicol. Several aspects of the biological activity of the dimeric structures were evaluated including antimicrobial activity, inhibition of in vitro bacterial protein translation, and the effect of dimerization on the action of several bacterial resistance mechanisms that deactivate tobramycin and chloramphenicol. This study demonstrates that covalently linking two identical or different ribosome-targeting antibiotics may lead to (i) a broader spectrum of antimicrobial activity, (ii) improved inhibition of bacterial translation properties compared to that of the parent antibiotics, and (iii) reduction in the efficacy of some drug-modifying enzymes that confer high levels of resistance to the parent antibiotics from which the dimers were derived.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Drug Resistance, Bacterial/drug effects , Protein Biosynthesis/drug effects , Ribosomes/drug effects , Anti-Bacterial Agents/chemistry , Bacteria/genetics , Bacteria/metabolism , Chloramphenicol/chemical synthesis , Chloramphenicol/chemistry , Chloramphenicol/pharmacology , Clindamycin/chemical synthesis , Clindamycin/chemistry , Clindamycin/pharmacology , Dimerization , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/genetics , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/genetics , Gram-Positive Bacteria/metabolism , Inhibitory Concentration 50 , Microbial Sensitivity Tests , Models, Chemical , Molecular Structure , Ribosomes/genetics , Ribosomes/metabolism , Tobramycin/chemical synthesis , Tobramycin/chemistry , Tobramycin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL