Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 119
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Proc Natl Acad Sci U S A ; 121(22): e2310677121, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38753503

RÉSUMÉ

Seasonal and pandemic-associated influenza strains cause highly contagious viral respiratory infections that can lead to severe illness and excess mortality. Here, we report on the optimization of our small-molecule inhibitor F0045(S) targeting the influenza hemagglutinin (HA) stem with our Sulfur-Fluoride Exchange (SuFEx) click chemistry-based high-throughput medicinal chemistry (HTMC) strategy. A combination of SuFEx- and amide-based lead molecule diversification and structure-guided design led to identification and validation of ultrapotent influenza fusion inhibitors with subnanomolar EC50 cellular antiviral activity against several influenza A group 1 strains. X-ray structures of six of these compounds with HA indicate that the appended moieties occupy additional pockets on the HA surface and increase the binding interaction, where the accumulation of several polar interactions also contributes to the improved affinity. The compounds here represent the most potent HA small-molecule inhibitors to date. Our divergent HTMC platform is therefore a powerful, rapid, and cost-effective approach to develop bioactive chemical probes and drug-like candidates against viral targets.


Sujet(s)
Antiviraux , Glycoprotéine hémagglutinine du virus influenza , Glycoprotéine hémagglutinine du virus influenza/métabolisme , Glycoprotéine hémagglutinine du virus influenza/composition chimique , Humains , Antiviraux/pharmacologie , Antiviraux/composition chimique , Chimie pharmaceutique/méthodes , Tests de criblage à haut débit/méthodes , Grippe humaine/traitement médicamenteux , Grippe humaine/virologie , Cristallographie aux rayons X/méthodes , Chimie click/méthodes , Animaux , Virus de la grippe A/effets des médicaments et des substances chimiques , Cellules rénales canines Madin-Darby , Inhibiteurs des protéines de fusion virale/pharmacologie , Inhibiteurs des protéines de fusion virale/composition chimique , Chiens
2.
Cell Chem Biol ; 31(3): 477-486.e7, 2024 Mar 21.
Article de Anglais | MEDLINE | ID: mdl-38518746

RÉSUMÉ

Of the targets for HIV-1 therapeutics, the capsid core is a relatively unexploited but alluring drug target due to its indispensable roles throughout virus replication. Because of this, we aimed to identify "clickable" covalent modifiers of the HIV-1 capsid protein (CA) for future functionalization. We screened a library of fluorosulfate compounds that can undergo sulfur(VI) fluoride exchange (SuFEx) reactions, and five compounds were identified as hits. These molecules were further characterized for antiviral effects. Several compounds impacted in vitro capsid assembly. One compound, BBS-103, covalently bound CA via a SuFEx reaction to Tyr145 and had antiviral activity in cell-based assays by perturbing virus production, but not uncoating. The covalent binding of compounds that target the HIV-1 capsid could aid in the future design of antiretroviral drugs or chemical probes that will help study aspects of HIV-1 replication.


Sujet(s)
Protéines de capside , VIH-1 (Virus de l'Immunodéficience Humaine de type 1) , Protéines de capside/métabolisme , Capside/composition chimique , Capside/métabolisme , Assemblage viral , Réplication virale , Antiviraux/pharmacologie
3.
Bioorg Med Chem Lett ; 98: 129596, 2024 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-38142914

RÉSUMÉ

To identify new compounds that can effectively inhibit Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), we screened, synthesized, and evaluated a series of novel aryl fluorosulfate derivatives for their in vitro inhibitory activity against Mtb. Compound 21b exhibited an in vitro minimum inhibitory concentration (MIC) of 0.06 µM against Mtb, no cytotoxicity against both HEK293T and HepG2 mammalian cell lines, and had good in vivo mouse plasma exposure and lung concentration with a 20 mg/kg oral dose, which supports advanced development as a new chemical entity for TB treatment.


Sujet(s)
Mycobacterium tuberculosis , Tuberculose , Animaux , Humains , Souris , Antituberculeux , Cellules HEK293 , Mammifères , Tests de sensibilité microbienne , Relation structure-activité , Tuberculose/traitement médicamenteux , Sulfates organiques/composition chimique , Sulfates organiques/pharmacologie
4.
Joule ; 7(1): 95-111, 2023 Jan 18.
Article de Anglais | MEDLINE | ID: mdl-37034575

RÉSUMÉ

High capacity polymer dielectrics that operate with high efficiencies under harsh electrification conditions are essential components for advanced electronics and power systems. It is, however, fundamentally challenging to design polymer dielectrics that can reliably withstand demanding temperatures and electric fields, which necessitate the balance of key electronic, electrical and thermal parameters. Herein, we demonstrate that polysulfates, synthesized by sulfur(VI) fluoride exchange (SuFEx) catalysis, another near-perfect click chemistry reaction, serve as high-performing dielectric polymers that overcome such bottlenecks. Free-standing polysulfate thin films from convenient solution processes exhibit superior insulating properties and dielectric stability at elevated temperatures, which are further enhanced when ultrathin (~5 nm) oxide coatings are deposited by atomic layer deposition. The corresponding electrostatic film capacitors display high breakdown strength (>700 MV m-1) and discharged energy density of 8.64 J cm-3 at 150 °C, outperforming state-of-the-art free-standing capacitor films based on commercial and synthetic dielectric polymers and nanocomposites.

5.
Proc Natl Acad Sci U S A ; 120(11): e2220767120, 2023 03 14.
Article de Anglais | MEDLINE | ID: mdl-36893261

RÉSUMÉ

The recently developed double-click reaction sequence [G. Meng et al., Nature 574, 86-89 (2019)] is expected to vastly expand the number and diversity of synthetically accessible 1,2,3-triazole derivatives. However, it remains elusive how to rapidly navigate the extensive chemical space created by double-click chemistry for bioactive compound discovery. In this study, we selected a particularly challenging drug target, the glucagon-like-peptide-1 receptor (GLP-1R), to benchmark our new platform for the design, synthesis, and screening of double-click triazole libraries. First, we achieved a streamlined synthesis of customized triazole libraries on an unprecedented scale (composed of 38,400 new compounds). By interfacing affinity-selection mass spectrometry and functional assays, we identified a series of positive allosteric modulators (PAMs) with unreported scaffolds that can selectively and robustly enhance the signaling activity of the endogenous GLP-1(9-36) peptide. Intriguingly, we further revealed an unexpected binding mode of new PAMs which likely act as a molecular glue between the receptor and the peptide agonist. We anticipate the merger of double-click library synthesis with the hybrid screening platform allows for efficient and economic discovery of drug candidates or chemical probes for various therapeutic targets.


Sujet(s)
Récepteur du peptide-1 similaire au glucagon , Peptides , Régulation allostérique , Glucagon-like peptide 1/métabolisme , Récepteur du peptide-1 similaire au glucagon/agonistes , Peptides/composition chimique , Triazoles/composition chimique
6.
Article de Anglais | MEDLINE | ID: mdl-38873592

RÉSUMÉ

Sulfur Fluoride Exchange (SuFEx) is a click reaction par excellence that has revolutionized multiple research fields. In this Primer, we delve into the essential elements of SuFEx operation, catalysis, and SuFExable connective hubs. We also explore the cutting-edge applications of SuFEx in drug development, polymer science, and biochemistry. Additionally, we examine the potential limitations and promising prospects for this versatile click reaction.

7.
Chem ; 9(8): 2128-2143, 2023 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-38882554

RÉSUMÉ

Phosphorus Fluoride Exchange (PFEx) represents a cutting-edge advancement in catalytic click-reaction technology. Drawing inspiration from Nature's phosphate connectors, PFEx facilitates the reliable coupling of P(V)-F loaded hubs with aryl alcohols, alkyl alcohols, and amines to produce stable, multidimensional P(V)-O and P(V)-N linked products. The rate of P-F exchange is significantly enhanced by Lewis amine base catalysis, such as 1,5,7-triazabicyclo[4.4.0]dec-5-ene (TBD). PFEx substrates containing multiple P-F bonds are capable of selective, serial exchange reactions via judicious catalyst selection. In fewer than four synthetic steps, controlled projections can be deliberately incorporated along three of the four tetrahedral axes departing from the P(V) central hub, thus taking full advantage of the potential for generating three-dimensional diversity. Furthermore, late-stage functionalization of drugs and drug fragments can be achieved with the polyvalent PFEx hub, hexafluorocyclotriphosphazene (HFP), as has been demonstrated in prior research.

8.
Isr J Chem ; 63(10-11)2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-38737670

RÉSUMÉ

Truncated mucin-type O-glycans, such as Tn-associated antigens, are aberrantly expressed biomarkers of cancer, but remain challenging to target. Reactive antibodies to these antigens either lack high-affinity or are prone to antigen escape. Here, we have developed a robust chemoenzymatic strategy for the global labeling of Tn-associated antigens, i.e. Tn (GalNAcα-O-Ser/Thr), Thomsen-Friedenreich (Galß1-3GalNAcα-O-Ser/Thr, TF) and STF (Neu5Acα2-3Galß1-3GalNAcα-O-Ser/Thr, STF) antigens, in human whole blood with high efficiency and selectivity. This method relies on the use of the O-glycan sialyltransferase ST6GalNAc1 to transfer a sialic acid-functionalized adaptor to the GalNAc residue of these antigens. By tagging, the adaptor functionalized antigens can be easily targeted by customized strategies such as, but not limited to, chimeric antigen receptor T-Cells (CAR-T). We expect this tagging system to find broad applications in cancer diagnostics and targeting in combination with established strategies.

9.
Proc Natl Acad Sci U S A ; 119(37): e2208540119, 2022 09 13.
Article de Anglais | MEDLINE | ID: mdl-36070343

RÉSUMÉ

Diversity Oriented Clicking (DOC) is a discovery method geared toward the rapid synthesis of functional libraries. It combines the best attributes of both classical and modern click chemistries. DOC strategies center upon the chemical diversification of core "SuFExable" hubs-exemplified by 2-Substituted-Alkynyl-1-Sulfonyl Fluorides (SASFs)-enabling the modular assembly of compounds through multiple reaction pathways. We report here a range of stereoselective Michael-type addition pathways from SASF hubs including reactions with secondary amines, carboxylates, 1H-1,2,3-triazole, and halides. These high yielding conjugate addition pathways deliver unprecedented ß-substituted alkenyl sulfonyl fluorides as single isomers with minimal purification, greatly enriching the repertoire of DOC and holding true to the fundamentals of modular click chemistry. Further, we demonstrate the potential for biological function - a key objective of click chemistry - of this family of SASF-derived molecules as covalent inhibitors of human neutrophil elastase.


Sujet(s)
Chimie click , Fluorures , Leukocyte elastase , Protéines sécrétoires inhibitrices de protéinases , Acides sulfiniques , Chimie click/méthodes , Fluorures/synthèse chimique , Fluorures/composition chimique , Fluorures/pharmacologie , Humains , Leukocyte elastase/antagonistes et inhibiteurs , Protéines sécrétoires inhibitrices de protéinases/synthèse chimique , Protéines sécrétoires inhibitrices de protéinases/composition chimique , Protéines sécrétoires inhibitrices de protéinases/pharmacologie , Acides sulfiniques/synthèse chimique , Acides sulfiniques/composition chimique , Acides sulfiniques/pharmacologie
10.
ACS Cent Sci ; 7(11): 1919-1928, 2021 Nov 24.
Article de Anglais | MEDLINE | ID: mdl-34841062

RÉSUMÉ

Sulfur(VI) fluoride exchange (SuFEx) click chemistry has offered a facile and reliable approach to produce polysulfates and polysulfonates. However, the current SuFEx polymerization methods lack precise control of target molecular weight and dispersity. Herein, we report the first chain-growth SuFEx polycondensation process by exploiting the unique reactivity and selectivity of S-F bonds under SuFEx catalysis. Given the higher reactivity of iminosulfur oxydifluoride versus fluorosulfate, the chain-growth SuFEx polycondensation is realized by using an iminosulfur oxydifluoride-containing compound as the reactive chain initiator and deactivated AB-type aryl silyl ether-fluorosulfates bearing an electron-withdrawing group as monomers. When 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU) was utilized as the polymerization catalyst, precise control over the polymer molecular weight and polydispersity was achieved. The resulting polymers possess great thermal stability but are easily degradable under mild acidic and basic conditions.

11.
Nat Chem ; 13(9): 858-867, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34400816

RÉSUMÉ

Sulfur(VI) fluoride exchange (SuFEx) is a category of click chemistry that enables covalent linking of modular units through sulfur(VI) connective hubs. The efficiency of SuFEx and the stability of the resulting bonds have led to polymer chemistry applications. Now, we report the SuFEx click chemistry synthesis of several structurally diverse SOF4-derived copolymers based on the polymerization of bis(iminosulfur oxydifluorides) and bis(aryl silyl ethers). This polymer class presents two key characteristics. First, the [-N=S(=O)F-O-] polymer backbone linkages are themselves SuFExable and undergo precise SuFEx-based post-modification with phenols or amines to yield branched functional polymers. Second, studies of individual polymer chains of several of these new materials indicate helical polymer structures. The robust nature of SuFEx click chemistry offers the potential for post-polymerization modification, enabling the synthesis of materials with control over composition and conformation.

12.
Angew Chem Int Ed Engl ; 60(39): 21195-21199, 2021 09 20.
Article de Anglais | MEDLINE | ID: mdl-34259368

RÉSUMÉ

Fluorosulfuryl isocyanate (FSI, FSO2 NCO) is established as a reliable bis-electrophilic linker for stepwise attachment of an alcohol bearing module to an amine bearing module and thence a new module RO-C(=O)-NH-SO2 -NR'R'' is created. FSI's isocyanate motif fuses directly and quickly with alcohols and phenols, affording fluorosulfuryl carbamates in nearly quantitative yield. A new reagent and process to deliver the FSI-derived fluorosulfuryl carbamate fragment to amines are also developed. The resulting SVI -F motifs from step-1 are remarkably stable, given the great structural complexities in diverse products. In the step-2 reaction with amines, the best yield of the S-N linked products arise with water alone. This "on water" interfacial reactivity phenomenon is crucial, revealing the latent reactivity of SVI -F probe for potential covalent capture of proteins in vivo which is important in today's drug discovery. The scope of the SuFEx chemistry is largely expanded thereby and the facile entry to these phosphate-like connections should prove useful to click chemistry across diverse fields.

13.
Proc Natl Acad Sci U S A ; 118(28)2021 07 13.
Article de Anglais | MEDLINE | ID: mdl-34244433

RÉSUMÉ

Sulfur fluoride exchange (SuFEx), a next generation of click chemistry, opens an avenue for drug discovery. We report here the discovery and structure-activity relationship studies of a series of arylfluorosulfates, synthesized via SuFEx, as antibacterial agents. Arylfluorosulfates 3, 81, and 101 showed potency to overcome multidrug resistance and were not susceptible to the generation of resistance. They exhibited rapid bactericidal potency and selectively killed gram-positive bacterial strains. These compounds also exhibited the ability to disrupt established bacterial biofilm and kill persisters derived from biofilm. Furthermore, arylfluorosulfate 3 had a synergistic effect with streptomycin and gentamicin. In addition, their anti-MRSA potency was evaluated and determined by the Caenorhabditis elegans model.


Sujet(s)
Antibactériens/pharmacologie , Résistance bactérienne aux médicaments , Sulfates/pharmacologie , Animaux , Biofilms/effets des médicaments et des substances chimiques , Biofilms/croissance et développement , Caenorhabditis elegans/microbiologie , Modèles animaux de maladie humaine , Résistance bactérienne aux médicaments/effets des médicaments et des substances chimiques , Cellules HEK293 , Humains , Cinétique , Staphylococcus aureus résistant à la méticilline/effets des médicaments et des substances chimiques , Tests de sensibilité microbienne , Phénotype , Relation structure-activité , Sulfates/composition chimique
14.
ACS Cent Sci ; 7(5): 815-830, 2021 May 26.
Article de Anglais | MEDLINE | ID: mdl-34079898

RÉSUMÉ

Transcriptional coregulators, which mediate chromatin-dependent transcriptional signaling, represent tractable targets to modulate tumorigenic gene expression programs with small molecules. Genetic loss-of-function studies have recently implicated the transcriptional coactivator, ENL, as a selective requirement for the survival of acute leukemia and highlighted an essential role for its chromatin reader YEATS domain. Motivated by these discoveries, we executed a screen of nearly 300,000 small molecules and identified an amido-imidazopyridine inhibitor of the ENL YEATS domain (IC50 = 7 µM). Improvements to the initial screening hit were enabled by adopting and expanding upon a SuFEx-based approach to high-throughput medicinal chemistry, ultimately demonstrating that it is compatible with cell-based drug discovery. Through these efforts, we discovered SR-0813, a potent and selective ENL/AF9 YEATS domain inhibitor (IC50 = 25 nM). Armed with this tool and a first-in-class ENL PROTAC, SR-1114, we detailed the biological response of AML cells to pharmacological ENL disruption for the first time. Most notably, we discovered that ENL YEATS inhibition is sufficient to selectively suppress ENL target genes, including HOXA9/10, MYB, MYC, and a number of other leukemia proto-oncogenes. Cumulatively, our study establishes YEATS domain inhibition as a viable approach to disrupt the pathogenic function of ENL in acute leukemia and provides the first thoroughly characterized chemical probe for the ENL YEATS domain.

15.
J Pharmacol Exp Ther ; 378(3): 315-321, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34145064

RÉSUMÉ

Inhibition of acetylcholinesterase (AChE) by certain organophosphates (OPs) can be life-threatening and requires reactivating antidote accessibility to the peripheral and central nervous systems to reverse symptoms and enhance survival parameters. In considering dosing requirements for oxime antidotes in OP exposures that inactivate AChE, clearance of proton ionizable, zwitterionic antidotes is rapid and proceeds with largely the parent antidotal compound being cleared by renal transporters. Such transporters may also control disposition between target tissues and plasma as well as overall elimination from the body. An ideal small-molecule antidote should access and be retained in primary target tissues-central nervous system (brain), skeletal muscle, and peripheral autonomic sites-for sufficient periods to reactivate AChE and prevent acute toxicity. We show here that we can markedly prolong the antidotal activity of zwitterionic antidotes by inhibiting P-glycoprotein (P-gp) transporters in the brain capillary and renal systems. We employ the P-gp inhibitor tariquidar as a reference compound and show that tissue and plasma levels of RS194B, a hydroxyl-imino acetamido alkylamine reactivator, are elevated and that plasma clearances are reduced. To examine the mechanism, identify the transporter, and establish the actions of a transport inhibitor, we compare the pharmacokinetic parameters in a P-glycoprotein knockout mouse strain and see dramatic enhancements of short-term plasma and tissue levels. Hence, repurposed transport inhibitors that are candidate or Food and Drug Administration-approved drugs, should enhance target tissue concentrations of the zwitterionic antidote through inhibition of both renal elimination and brain capillary extrusion. SIGNIFICANCE STATEMENT: We examine renal and brain capillary transporter inhibition as means for lowering dose and frequency of dosing of a blood-brain barrier permanent reactivating antidote, RS194B, an ionizable zwitterion. Through a small molecule, tariquidar, and gene knockout mice, CNS antidote concentrations are enhanced, and total body clearances are concomitantly diminished. RS194B with repurposed transport inhibitors should enhance reactivation of central and peripheral OP-inhibited acetylcholinesterase. Activities at both disposition sites are a desired features for replacing the antidote, pralidoxime, for acute OP exposure.


Sujet(s)
Acetylcholinesterase , Cinétique , Organophosphates , Composés de pralidoxime
16.
J Neurochem ; 158(6): 1217-1222, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-33638151

RÉSUMÉ

We detail here distinctive departures from lead classical cholinesterase re-activators, the pyridinium aldoximes, to achieve rapid CNS penetration and reactivation of AChE in the CNS (brain and spinal cord). Such reactivation is consistent with these non-canonical re-activators enhancing survival parameters in both mice and macaques following exposure to organophosphates. Thus, the ideal cholinesterase re-activator should show minimal toxicity, limited inhibitory activity in the absence of an organophosphate, and rapid CNS penetration, in addition to its nucleophilic potential at the target, the conjugated AChE active center. These are structural properties directed to reactivity profiles at the conjugated AChE active center, reinforced by the pharmacokinetic and tissue disposition properties of the re-activator leads. In the case of nicotinic acetylcholine receptor (nAChR) agonists and antagonists, with the many existing receptor subtypes in mammals, we prioritize subtype selectivity in their design. In contrast to nicotine and its analogues that react with panoply of AChR subtypes, the substituted di-2-picolyl amine pyrimidines possess distinctive ionization characteristics reflecting in selectivity for the orthosteric site at the α7 subtypes of receptor. Here, entry to the CNS should be prioritized for the therapeutic objectives of the nicotinic agent influencing aberrant CNS activity in development or in the sequence of CNS ageing (longevity) in mammals, along with general peripheral activities controlling inflammation.


Sujet(s)
Acetylcholinesterase/composition chimique , Réactivateurs de la cholinestérase/composition chimique , Conception de médicament , Agonistes nicotiniques/composition chimique , Antagonistes nicotiniques/composition chimique , Récepteurs nicotiniques/composition chimique , Acetylcholinesterase/métabolisme , Animaux , Réactivateurs de la cholinestérase/métabolisme , Humains , Ligands , Agonistes nicotiniques/métabolisme , Antagonistes nicotiniques/métabolisme , Structure secondaire des protéines , Structure tertiaire des protéines , Récepteurs nicotiniques/métabolisme
17.
J Am Chem Soc ; 143(10): 3753-3763, 2021 03 17.
Article de Anglais | MEDLINE | ID: mdl-33630577

RÉSUMÉ

The lack of efficient [18F]fluorination processes and target-specific organofluorine chemotypes remains the major challenge of fluorine-18 positron emission tomography (PET). We report here an ultrafast isotopic exchange method for the radiosynthesis of novel PET agent aryl [18F]fluorosulfate enabled by the emerging sulfur fluoride exchange (SuFEx) click chemistry. The method has been applied to the fully automated 18F-radiolabeling of 25 structurally and functionally diverse aryl fluorosulfates with excellent radiochemical yield (83-100%, median 98%) and high molar activity (280 GBq µmol-1) at room temperature in 30 s. The purification of radiotracers requires no time-consuming HPLC but rather a simple cartridge filtration. We further demonstrate the imaging application of a rationally designed poly(ADP-ribose) polymerase 1 (PARP1)-targeting aryl [18F]fluorosulfate by probing subcutaneous tumors in vivo.


Sujet(s)
Chimie click , Fluorures/composition chimique , Radiopharmaceutiques/synthèse chimique , Composés du soufre/composition chimique , Animaux , Lignée cellulaire tumorale , Produits de contraste/synthèse chimique , Produits de contraste/composition chimique , Produits de contraste/métabolisme , Théorie de la fonctionnelle de la densité , Stabilité de médicament , Fluorures/synthèse chimique , Fluorures/métabolisme , Radio-isotopes du fluor/composition chimique , Humains , Souris , Tumeurs/imagerie diagnostique , Poly(ADP-ribose) polymerases/composition chimique , Poly(ADP-ribose) polymerases/métabolisme , Tomographie par émission de positons , Radiopharmaceutiques/métabolisme , Composés du soufre/synthèse chimique , Composés du soufre/métabolisme , Transplantation hétérologue
18.
Nat Chem ; 12(10): 906-913, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32868886

RÉSUMÉ

Drug candidates that form covalent linkages with their target proteins have been underexplored compared with the conventional counterparts that modulate biological function by reversibly binding to proteins, in part due to concerns about off-target reactivity. However, toxicity linked to off-target reactivity can be minimized by using latent electrophiles that only become activated towards covalent bond formation on binding a specific protein. Here we study sulfuramidimidoyl fluorides, a class of weak electrophiles that undergo sulfur(VI) fluoride exchange chemistry. We show that equilibrium binding of a sulfuramidimidoyl fluoride to a protein can allow nucleophilic attack by a specific amino acid side chain, which leads to conjugate formation. We incubated small molecules, each bearing a sulfuramidimidoyl fluoride electrophile, with human cell lysate, and the protein conjugates formed were identified by affinity chromatography-mass spectrometry. This inverse drug discovery approach identified a compound that covalently binds to and irreversibly inhibits the activity of poly(ADP-ribose) polymerase 1, an important anticancer target in living cells.


Sujet(s)
Découverte de médicament , Fluorures/composition chimique , Thiols/composition chimique , Soufre/composition chimique , Chromatographie d'affinité , Cellules HEK293 , Humains , Spectrométrie de masse , Structure moléculaire , Thiols/synthèse chimique
19.
J Am Chem Soc ; 142(25): 10899-10904, 2020 06 24.
Article de Anglais | MEDLINE | ID: mdl-32479075

RÉSUMÉ

Optimization of small-molecule probes or drugs is a synthetically lengthy, challenging, and resource-intensive process. Lack of automation and reliance on skilled medicinal chemists is cumbersome in both academic and industrial settings. Here, we demonstrate a high-throughput hit-to-lead process based on the biocompatible sulfur(VI) fluoride exchange (SuFEx) click chemistry. A high-throughput screening hit benzyl (cyanomethyl)carbamate (Ki = 8 µM) against a bacterial cysteine protease SpeB was modified with a SuFExable iminosulfur oxydifluoride [RN═S(O)F2] motif, rapidly diversified into 460 analogs in overnight reactions, and the products were directly screened to yield drug-like inhibitors with 480-fold higher potency (Ki = 18 nM). We showed that the improved molecule is active in a bacteria-host coculture. Since this SuFEx linkage reaction succeeds on picomole scale for direct screening, we anticipate our methodology can accelerate the development of robust biological probes and drug candidates.


Sujet(s)
Protéines bactériennes/antagonistes et inhibiteurs , Inhibiteurs de la cystéine protéinase/pharmacologie , Exotoxines/antagonistes et inhibiteurs , Composés du soufre/composition chimique , Protéines bactériennes/composition chimique , Protéines bactériennes/métabolisme , Domaine catalytique , Chimie click , Cristallographie aux rayons X , Inhibiteurs de la cystéine protéinase/métabolisme , Inhibiteurs de la cystéine protéinase/toxicité , Découverte de médicament , Exotoxines/composition chimique , Exotoxines/métabolisme , Tests de criblage à haut débit , Humains , Cellules Jurkat , Microsomes du foie/métabolisme , Étude de validation de principe , Liaison aux protéines
20.
PLoS Negl Trop Dis ; 14(4): e0008224, 2020 04.
Article de Anglais | MEDLINE | ID: mdl-32302296

RÉSUMÉ

Giardiasis and other protozoan infections are major worldwide causes of morbidity and mortality, yet development of new antimicrobial agents with improved efficacy and ability to override increasingly common drug resistance remains a major challenge. Antimicrobial drug development typically proceeds by broad functional screens of large chemical libraries or hypothesis-driven exploration of single microbial targets, but both strategies have challenges that have limited the introduction of new antimicrobials. Here, we describe an alternative drug development strategy that identifies a sufficient but manageable number of promising targets, while reducing the risk of pursuing targets of unproven value. The strategy is based on defining and exploiting the incompletely understood adduction targets of 5-nitroimidazoles, which are proven antimicrobials against a wide range of anaerobic protozoan and bacterial pathogens. Comprehensive adductome analysis by modified click chemistry and multi-dimensional proteomics were applied to the model pathogen Giardia lamblia to identify dozens of adducted protein targets common to both 5'-nitroimidazole-sensitive and -resistant cells. The list was highly enriched for known targets in G. lamblia, including arginine deiminase, α-tubulin, carbamate kinase, and heat shock protein 90, demonstrating the utility of the approach. Importantly, over twenty potential novel drug targets were identified. Inhibitors of two representative new targets, NADP-specific glutamate dehydrogenase and peroxiredoxin, were found to have significant antigiardial activity. Furthermore, all the identified targets remained available in resistant cells, since giardicidal activity of the respective inhibitors was not impacted by resistance to 5'-nitroimidazoles. These results demonstrate that the combined use of click chemistry and proteomics has the potential to reveal alternative drug targets for overcoming antimicrobial drug resistance in protozoan parasites.


Sujet(s)
Antiparasitaires/pharmacologie , Chimie click/méthodes , Découverte de médicament/méthodes , Giardia lamblia/effets des médicaments et des substances chimiques , Indazoles/pharmacologie , Protéines de protozoaire/métabolisme , Animaux , Antiparasitaires/synthèse chimique , Antiparasitaires/usage thérapeutique , Modèles animaux de maladie humaine , Femelle , Giardiase/traitement médicamenteux , Indazoles/synthèse chimique , Indazoles/usage thérapeutique , Intestin grêle/parasitologie , Mâle , Souris de lignée C57BL , Charge parasitaire , Liaison aux protéines , Protéomique/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...