Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
ACS Chem Biol ; 18(2): 296-303, 2023 02 17.
Article in English | MEDLINE | ID: mdl-36602435

ABSTRACT

Lactic acid transport is a key process maintaining glycolytic flux in tumors. Inhibition of this process will result in glycolytic shutdown, impacting on cell growth and survival and thus has been pursued as a therapeutic approach for cancers. Using a cell-based screen in a MCT4-dependent cell line, we identified and optimized compounds for their ability to inhibit the efflux of intracellular lactic acid with good physical and pharmacokinetic properties. To deconvolute the mechanism of lactic acid efflux inhibition, we have developed three assays to measure cellular target engagement. Specifically, we synthesized a biologically active photoaffinity probe (IC50 < 10 nM), and using this probe, we demonstrated selective engagement of MCT4 of our parent molecule through a combination of confocal microscopy and in-cell chemoproteomics. As an orthogonal assay, the cellular thermal shift assay (CETSA) confirmed binding to MCT4 in the cellular system. Comparisons of lactic acid efflux potencies in cells with differential expression of MCT family members further confirmed that the optimized compounds inhibit the efflux of lactic acid through the inhibition of MCT4. Taken together, these data demonstrate the power of orthogonal chemical biology methods to determine cellular target engagement, particularly for proteins not readily amenable to traditional biophysical methods.


Subject(s)
Biology , Lactic Acid , Lactic Acid/metabolism , Biological Transport , Cell Line, Tumor , Cell Proliferation
2.
SLAS Discov ; 26(4): 518-523, 2021 04.
Article in English | MEDLINE | ID: mdl-33615886

ABSTRACT

Mass spectrometry-based proteomics profiling is a discovery tool that enables researchers to understand the mechanisms of action of drug candidates. When applied to proteolysis targeting chimeras (PROTACs) such approaches provide unbiased perspectives of the binding, degradation selectivity, and mechanism related to efficacy and safety. Specifically, global profiling experiments can identify direct degradation events and assess downstream pathway modulation that may result from degradation or off-target inhibition. Targeted proteomics approaches can be used to quantify the levels of relevant E3 ligases and the protein of interest in cell lines and tissues of interest, which can inform the line of sight and provide insights on possible safety liabilities early in the project. Furthermore, proteomics approaches can be applied to understand protein turnover and resynthesis rates and inform on target tractability, as well as pharmacokinetics/pharmacodynamics understanding. In this perspective, we survey the literature around the impact of mass spectrometry-based proteomics in the development of PROTACs and present our envisioned proteomics cascade for supporting targeted protein degradation projects.


Subject(s)
High-Throughput Screening Assays , Molecular Targeted Therapy/methods , Proteasome Endopeptidase Complex/metabolism , Protein Processing, Post-Translational , Small Molecule Libraries/pharmacology , Ubiquitin-Protein Ligases/metabolism , Drug Discovery/methods , Eukaryotic Cells/cytology , Eukaryotic Cells/drug effects , Eukaryotic Cells/metabolism , Humans , Ligands , Mass Spectrometry/methods , Protein Binding , Proteolysis/drug effects , Proteomics/methods , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacokinetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects
3.
Mol Brain ; 13(1): 66, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32366277

ABSTRACT

Alzheimer's disease (AD), the leading cause of dementia, is a chronic neurodegenerative disease. Apolipoprotein E (apoE), which carries lipids in the brain in the form of lipoproteins, plays an undisputed role in AD pathophysiology. A high-throughput phenotypic screen was conducted using a CCF-STTG1 human astrocytoma cell line to identify small molecules that could upregulate apoE secretion. AZ7235, a previously discovered Axl kinase inhibitor, was identified to have robust apoE activity in brain microglia, astrocytes and pericytes. AZ7235 also increased expression of ATP-binding cassette protein A1 (ABCA1), which is involved in the lipidation and secretion of apoE. Moreover, AZ7235 did not exhibit Liver-X-Receptor (LXR) activity and stimulated apoE and ABCA1 expression in the absence of LXR. Target validation studies using AXL-/- CCF-STTG1 cells showed that Axl is required to mediate AZ7235 upregulation of apoE and ABCA1. Intriguingly, apoE expression and secretion was significantly attenuated in AXL-deficient CCF-STTG1 cells and reconstitution of Axl or kinase-dead Axl significantly restored apoE baseline levels, demonstrating that Axl also plays a role in maintaining apoE homeostasis in astrocytes independent of its kinase activity. Lastly, these effects may require human apoE regulatory sequences, as AZ7235 exhibited little stimulatory activity toward apoE and ABCA1 in primary murine glia derived from neonatal human APOE3 targeted-replacement mice. Collectively, we identified a small molecule that exhibits robust apoE and ABCA1 activity independent of the LXR pathway in human cells and elucidated a novel relationship between Axl and apoE homeostasis in human astrocytes.


Subject(s)
Alzheimer Disease/metabolism , Apolipoproteins E/metabolism , Astrocytes/drug effects , Astrocytoma/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , ATP Binding Cassette Transporter 1/metabolism , Animals , Apolipoproteins E/genetics , Astrocytoma/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Gene Knockdown Techniques , High-Throughput Screening Assays , Humans , Mice , Microglia/drug effects , Microglia/metabolism , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Axl Receptor Tyrosine Kinase
4.
Nat Commun ; 7: 13042, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27727204

ABSTRACT

Inhibition of ß-secretase BACE1 is considered one of the most promising approaches for treating Alzheimer's disease. Several structurally distinct BACE1 inhibitors have been withdrawn from development after inducing ocular toxicity in animal models, but the target mediating this toxicity has not been identified. Here we use a clickable photoaffinity probe to identify cathepsin D (CatD) as a principal off-target of BACE1 inhibitors in human cells. We find that several BACE1 inhibitors blocked CatD activity in cells with much greater potency than that displayed in cell-free assays with purified protein. Through a series of exploratory toxicology studies, we show that quantifying CatD target engagement in cells with the probe is predictive of ocular toxicity in vivo. Taken together, our findings designate off-target inhibition of CatD as a principal driver of ocular toxicity for BACE1 inhibitors and more generally underscore the power of chemical proteomics for discerning mechanisms of drug action.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Cathepsin D/metabolism , Enzyme Inhibitors/toxicity , Eye/pathology , Proteomics/methods , Toxicity Tests , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Eye/drug effects , Humans , Inhibitory Concentration 50 , Mass Spectrometry , Mice, Knockout , Molecular Probes/chemical synthesis , Molecular Probes/chemistry , Peptides/metabolism , Protein Binding , Rats, Wistar , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/pathology , Staining and Labeling
5.
ACS Chem Biol ; 10(4): 925-32, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25602368

ABSTRACT

Platelet-activating factor acetylhydrolases (PAFAHs) 1b2 and 1b3 are poorly characterized serine hydrolases that form a complex with a noncatalytic protein (1b1) to regulate brain development, spermatogenesis, and cancer pathogenesis. Determining physiological substrates and biochemical functions for the PAFAH1b complex would benefit from selective chemical probes that can perturb its activity in living systems. Here, we report a class of tetrahydropyridine reversible inhibitors of PAFAH1b2/3 discovered using a fluorescence polarization-activity-based protein profiling (fluopol-ABPP) screen of the NIH 300,000+ compound library. The most potent of these agents, P11, exhibited IC50 values of ∼40 and 900 nM for PAFAH1b2 and 1b3, respectively. We confirm selective inhibition of PAFAH1b2/3 in cancer cells by P11 using an ABPP protocol adapted for in situ analysis of reversible inhibitors and show that this compound impairs tumor cell survival, supporting a role for PAFAH1b2/3 in cancer.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , 1-Alkyl-2-acetylglycerophosphocholine Esterase/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cell Line , Cell Line, Tumor/drug effects , Cell Survival/drug effects , Fluorescence Polarization/methods , Humans , Inhibitory Concentration 50 , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Proteomics/methods , Pyridines/chemistry , Pyridines/pharmacology , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
6.
Nat Chem Biol ; 9(5): 300-6, 2013 May.
Article in English | MEDLINE | ID: mdl-23455543

ABSTRACT

Nicotinamide N-methyltransferase (NNMT) is overexpressed in a variety of human cancers, where it contributes to tumorigenesis by a mechanism that is still poorly understood. Here we show using metabolomics that NNMT impairs the methylation potential of cancer cells by consuming methyl units from S-adenosyl methionine to create the stable metabolic product 1-methylnicotinamide. As a result, NNMT-expressing cancer cells have an altered epigenetic state that includes hypomethylated histones and other cancer-related proteins combined with heightened expression of protumorigenic gene products. Our findings thus point to a direct mechanistic link between the deregulation of a metabolic enzyme and widespread changes in the methylation landscape of cancer cells.


Subject(s)
Epigenesis, Genetic , Models, Genetic , Neoplasms/metabolism , Nicotinamide N-Methyltransferase/metabolism , Epigenesis, Genetic/genetics , Humans , Methionine/chemistry , Methionine/metabolism , Methylation , Neoplasms/genetics , Neoplasms/pathology , Niacinamide/analogs & derivatives , Niacinamide/chemistry , Niacinamide/metabolism
7.
Chembiochem ; 13(14): 2082-93, 2012 Sep 24.
Article in English | MEDLINE | ID: mdl-22907802

ABSTRACT

The serine hydrolases constitute a large class of enzymes that play important roles in physiology. There is great interest in the development of potent and selective pharmacological inhibitors of these proteins. Traditional active-site inhibitors often have limited selectivity within this superfamily and are tedious and expensive to discover. Using the serine hydrolase RBBP9 as a model target, we designed a rapid and relatively inexpensive route to highly selective peptoid-based inhibitors that can be activated by visible light. This technology provides rapid access to photo-activated tool compounds capable of selectively blocking the function of particular serine hydrolases.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Light , Neoplasm Proteins/antagonists & inhibitors , Catalytic Domain , Cell Cycle Proteins/metabolism , Enzyme Inhibitors/metabolism , Eosine Yellowish-(YS)/chemistry , Fluorescein/chemistry , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Magnetics , Neoplasm Proteins/metabolism , Peptoids/chemical synthesis , Peptoids/chemistry , Protein Binding , Proteome/metabolism , Ruthenium/chemistry
8.
J Am Chem Soc ; 134(11): 5068-71, 2012 Mar 21.
Article in English | MEDLINE | ID: mdl-22400490

ABSTRACT

Serine hydrolases are one of the largest and most diverse enzyme classes in Nature. Most serine hydrolases lack selective inhibitors, which are valuable probes for assigning functions to these enzymes. We recently discovered a set of aza-ß-lactams (ABLs) that act as potent and selective inhibitors of the mammalian serine hydrolase protein-phosphatase methylesterase-1 (PME-1). The ABLs inactivate PME-1 by covalent acylation of the enzyme's serine nucleophile, suggesting that they could offer a general scaffold for serine hydrolase inhibitor discovery. Here, we have tested this hypothesis by screening ABLs more broadly against cell and tissue proteomes by competitive activity-based protein profiling (ABPP), leading to the discovery of lead inhibitors for several serine hydrolases, including the uncharacterized enzyme α,ß-hydrolase domain-containing 10 (ABHD10). ABPP-guided medicinal chemistry yielded a compound ABL303 that potently (IC(50) ≈ 30 nM) and selectively inactivated ABHD10 in vitro and in living cells. A comparison of optimized inhibitors for PME-1 and ABHD10 indicates that modest structural changes that alter steric bulk can tailor the ABL to selectively react with distinct, distantly related serine hydrolases. Our findings, taken together, designate the ABL as a versatile reactive group for creating first-in-class serine hydrolase inhibitors.


Subject(s)
Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , beta-Lactams/pharmacology , Binding, Competitive/drug effects , Molecular Structure , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry , Stereoisomerism , Structure-Activity Relationship , beta-Lactams/chemistry
9.
J Med Chem ; 54(14): 5229-36, 2011 Jul 28.
Article in English | MEDLINE | ID: mdl-21639134

ABSTRACT

The serine hydrolase protein phosphatase methylesterase-1 (PME-1) regulates the methylesterification state of protein phosphatase 2A (PP2A) and has been implicated in cancer and Alzheimer's disease. We recently reported a fluorescence polarization-activity-based protein profiling (fluopol-ABPP) high-throughput screen for PME-1 that uncovered a remarkably potent and selective class of aza-ß-lactam (ABL) PME-1 inhibitors. Here, we describe a distinct set of sulfonyl acrylonitrile inhibitors that also emerged from this screen. The optimized compound, 28 (AMZ30), selectively inactivates PME-1 and reduces the demethylated form of PP2A in living cells. Considering that 28 is structurally unrelated to ABL inhibitors of PME-1, these agents, together, provide a valuable set of pharmacological probes to study the role of methylation in regulating PP2A function. We furthermore observed that several serine hydrolases were sensitive to analogues of 28, suggesting that more extensive structural exploration of the sulfonyl acrylonitrile chemotype may result in useful inhibitors for other members of this large enzyme class.


Subject(s)
Acrylonitrile/analogs & derivatives , Acrylonitrile/chemical synthesis , Carboxylic Ester Hydrolases/antagonists & inhibitors , Sulfonamides/chemical synthesis , Sulfones/chemical synthesis , Acrylonitrile/chemistry , Acrylonitrile/pharmacology , Carboxylic Ester Hydrolases/metabolism , Cell Line , Humans , Proteome/metabolism , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology , Sulfones/chemistry , Sulfones/pharmacology
10.
J Am Chem Soc ; 132(9): 3009-12, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20148585

ABSTRACT

A total synthesis of the Aspidosperma alkaloids (+)-fendleridine and (+)-1-acetylaspidoalbidine is detailed, providing access to both enantiomers of the natural products and establishing their absolute configuration. Central to the synthetic approach is a powerful intramolecular [4+2]/[3+2] cycloaddition cascade of a 1,3,4-oxadiazole in which the pentacyclic skeleton and all the stereochemistry of the natural products are assembled in a reaction that forms three rings, four C-C bonds, and five stereogenic centers including three contiguous quaternary centers, and introduces the correct oxidation state at C19 in a single synthetic operation. The final tetrahydrofuran bridge is subsequently installed in one step, enlisting an intramolecular alcohol addition to an iminium ion generated by nitrogen-assisted opening of the cycloadduct oxido bridge, with a modification that permits release of useful functionality (a ketone) at the cleavage termini.


Subject(s)
Indole Alkaloids/chemical synthesis , Aspidosperma/chemistry , Cyclization , Indole Alkaloids/chemistry , Molecular Conformation , Stereoisomerism
11.
J Org Chem ; 71(15): 5715-24, 2006 Jul 21.
Article in English | MEDLINE | ID: mdl-16839153

ABSTRACT

A strategy utilizing N-heterocyclic carbenes (NHCs) derived from thiazolium salts has been developed for the generation of carbonyl anions from acylsilanes. Synthetically useful 1,4-diketones and N-phosphinoyl-alpha-aminoketones have been prepared in good to excellent yields via NHC-catalyzed additions of acylsilanes to the corresponding alpha,beta-unsaturated systems and N-phosphinoylimines. These organocatalytic reactions are air- and water-tolerant methods to execute robust carbonyl anion addition reactions. Additionally, polysubstituted aromatic furans and pyrroles have been efficiently synthesized in a one-pot process using this carbonyl anion methodology. The addition of alcohols to the reaction renders the process catalytic in thiazolium salt. In an effort to synthesize a potential intermediate along the proposed reaction pathway, silylated thiazolium carbinols have been identified to provide good yields of carbonyl anion addition products when subjected to the standard reaction conditions in the presence of suitable electrophiles.

12.
J Am Chem Soc ; 128(15): 4932-3, 2006 Apr 19.
Article in English | MEDLINE | ID: mdl-16608309

ABSTRACT

The direct nucleophilic acylation of nitroalkenes is reported utilizing a fluoride-initiated rearrangement of protected thiazolium carbinols. The key combination of fluoride anion and thiourea accesses carbonyl anion reactivity without the use of amine or amide bases typically employed in the generation of these Umpolung nucleophiles. The mild reaction conditions used to generate the reactive carbonyl anion species in situ allow for conjugate additions in good yield to sensitive nitroalkene electrophiles. The process is tolerant of a variety of thiazolium carbinols and nitroalkene substrates and can be rendered enantioselective and diastereoselective by the addition of a chiral thiourea derived from quinine.

SELECTION OF CITATIONS
SEARCH DETAIL