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.
Cell Death Dis ; 14(2): 87, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36750554

ABSTRACT

The metabolic pathways through which p53 functions as a potent tumor suppressor are incompletely understood. Here we report that, by associating with the Vitamin D receptor (VDR), p53 induces numerous genes encoding enzymes for peroxisomal fatty acid ß-oxidation (FAO). This leads to increased cytosolic acetyl-CoA levels and acetylation of the enzyme 5-Aminoimidazole-4-Carboxamide Ribonucleotide Formyltransferase/IMP Cyclohydrolase (ATIC), which catalyzes the last two steps in the purine biosynthetic pathway. This acetylation step, mediated by lysine acetyltransferase 2B (KAT2B), occurs at ATIC Lys 266, dramatically inhibits ATIC activity, and inversely correlates with colorectal cancer (CRC) tumor growth in vitro and in vivo, and acetylation of ATIC is downregulated in human CRC samples. p53-deficient CRCs with high levels of ATIC is more susceptible to ATIC inhibition. Collectively, these findings link p53 to peroxisomal FAO, purine biosynthesis, and CRC pathogenesis in a manner that is regulated by the levels of ATIC acetylation.


Subject(s)
Hydroxymethyl and Formyl Transferases , Tumor Suppressor Protein p53 , Humans , Purines , Fatty Acids
2.
Chem Sci ; 9(27): 5957-5966, 2018 Jul 21.
Article in English | MEDLINE | ID: mdl-30079210

ABSTRACT

Cellular uptake of circulating cholesterol occurs via the low density lipoprotein receptor (LDLR). The E3 ubiquitin ligase IDOL is a mediator of LDLR degradation, with IDOL homodimerization thought to be required for its activity. To probe the possibility of modulating LDLR levels with an inhibitor of IDOL homodimerization, we screened a SICLOPPS library of 3.2 million cyclic peptides for compounds that disrupt this protein-protein interaction. We identified cyclo-CFFLYT as the lead inhibitor, and improved its activity through the incorporation of non-natural amino acids. The activity of the optimized cyclic peptide was assessed in hepatic cells, with a dose-dependent increase in LDLR levels observed in the presence of our IDOL homodimerization inhibitor.

3.
Bioorg Med Chem ; 26(11): 3034-3038, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29555420

ABSTRACT

We report an inhibitor of the homodimeric protein-protein interaction of the BCL6 oncoprotein, identified from a genetically encoded SICLOPPS library of 3.2 million cyclic hexapeptides in combination with a bacterial reverse two-hybrid system. This cyclic peptide is shown to bind the BTB domain of BCL6, disrupts its homodimerization, and subsequent binding of the SMRT2 corepressor peptide.


Subject(s)
Gene Library , Peptides, Cyclic/pharmacology , Proto-Oncogene Proteins c-bcl-6/antagonists & inhibitors , Dimerization , Molecular Structure , Peptides, Cyclic/chemistry , Peptides, Cyclic/genetics , Protein Binding , Proto-Oncogene Proteins c-bcl-6/genetics
4.
Bioorg Med Chem ; 25(14): 3871-3882, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28559059

ABSTRACT

The transcription factor STAT5b is an antitumor target. Recently, we presented the small molecules Stafib-1 and Stafib-2 as potent, selective inhibitors of the STAT5b SH2 domain. Here we report that halogen substitutions on the terminal phenyl ring of Stafib-1 and a close derivative are tolerated and specificity over the STAT5a SH2 domain is maintained, albeit with a slight reduction in activity. Our data demonstrate that the synthetic methodology used for generating Stafib-1 and Stafib-2 can be utilized to synthesize a small library of halogen-substituted derivatives, and extend the panel of catechol bisphosphate-based submicromolar and selective STAT5b inhibitors.


Subject(s)
Catechols/chemistry , Diphosphates/chemistry , Halogens/chemistry , STAT5 Transcription Factor/antagonists & inhibitors , Binding Sites , Catechols/chemical synthesis , Catechols/metabolism , Humans , Inhibitory Concentration 50 , Molecular Docking Simulation , Protein Structure, Tertiary , STAT5 Transcription Factor/metabolism , Structure-Activity Relationship
5.
Sci Rep ; 7(1): 819, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28400581

ABSTRACT

The transcription factor STAT5b is a target for tumour therapy. We recently reported catechol bisphosphate and derivatives such as Stafib-1 as the first selective inhibitors of the STAT5b SH2 domain. Here, we demonstrate STAT5b binding of catechol bisphosphate by solid-state nuclear magnetic resonance, and report on rational optimization of Stafib-1 (Ki = 44 nM) to Stafib-2 (Ki = 9 nM). The binding site of Stafib-2 was validated using combined isothermal titration calorimetry (ITC) and protein point mutant analysis, representing the first time that functional comparison of wild-type versus mutant protein by ITC has been used to characterize the binding site of a small-molecule ligand of a STAT protein with amino acid resolution. The prodrug Pomstafib-2 selectively inhibits tyrosine phosphorylation of STAT5b in human leukaemia cells and induces apoptosis in a STAT5-dependent manner. We propose Pomstafib-2, which currently represents the most active, selective inhibitor of STAT5b activation available, as a chemical tool for addressing the fundamental question of which roles the different STAT5 proteins play in various cell processes.


Subject(s)
Antineoplastic Agents/pharmacology , Catechols/pharmacology , STAT5 Transcription Factor/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Binding Sites , Catechols/chemical synthesis , Catechols/chemistry , Cell Line, Tumor , Humans , Molecular Docking Simulation , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Quantitative Structure-Activity Relationship , STAT5 Transcription Factor/chemistry , STAT5 Transcription Factor/metabolism , src Homology Domains
6.
ACS Chem Biol ; 10(12): 2884-90, 2015 Dec 18.
Article in English | MEDLINE | ID: mdl-26469307

ABSTRACT

Design approaches for inhibitors of protein-protein interactions are rare, but highly sought after. Here, we report that O-phosphorylation of simple derivatives of the natural products dihydrocapsaicin and N-vanillylnonanamide leads to inhibitors of the SH2 domain of the transcription factor STAT5b. The most potent molecule is obtained from dihydrocapsaicin in only three synthetic steps. It has submicromolar affinity for the SH2 domain of STAT5b (Ki = 0.34 µM), while displaying 35-fold selectivity over the highly homologous STAT5a (Ki = 13.0 µM). The corresponding pivaloyloxymethyl ester inhibits STAT5b with selectivity over STAT5a in human tumor cells. Importantly, it inhibits cell viability and induces apoptosis in human tumor cells in a STAT5-dependent manner. Our data validate O-phosphorylation of appropriately preselected natural products or natural product derivatives as a semirational design approach for small molecules that selectively inhibit phosphorylation-dependent protein-protein interaction domains in cultured human tumor cells.


Subject(s)
Capsaicin/chemistry , Capsaicin/pharmacology , Drug Design , STAT5 Transcription Factor/antagonists & inhibitors , Binding Sites , Blotting, Western , Humans , Inhibitory Concentration 50 , K562 Cells , Molecular Structure , Phosphorylation , Protein Binding/drug effects , Proteins/chemistry , STAT5 Transcription Factor/chemistry
7.
Angew Chem Int Ed Engl ; 54(16): 4758-63, 2015 Apr 13.
Article in English | MEDLINE | ID: mdl-25702814

ABSTRACT

Src homology 2 (SH2) domains play a central role in signal transduction. Although many SH2 domains have been validated as drug targets, their structural similarity makes development of specific inhibitors difficult. The cancer-relevant transcription factors STAT5a and STAT5b are particularly challenging small-molecule targets because their SH2 domains are 93% identical on the amino acid level. Here we present the natural product-inspired development of the low-nanomolar inhibitor Stafib-1, as the first small molecule which inhibits the STAT5b SH2 domain (K(i)=44 nM) with more than 50-fold selectivity over STAT5a. The binding site of the core moiety of Stafib-1 was validated by functional analysis of point mutants. A prodrug of Stafib-1 was shown to inhibit STAT5b with high selectivity over STAT5a in tumor cells. Stafib-1 provides the first demonstration that naturally occurring SH2 domains with more than 90% sequence identity can be selectively targeted with small organic molecules.


Subject(s)
STAT5 Transcription Factor/antagonists & inhibitors , Tumor Suppressor Proteins/antagonists & inhibitors , Amino Acid Sequence , Binding Sites , Biological Products/chemistry , Biological Products/metabolism , Catechols/chemistry , Catechols/metabolism , Humans , Molecular Docking Simulation , Peptides/chemistry , Peptides/metabolism , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , STAT5 Transcription Factor/metabolism , Structure-Activity Relationship , Tumor Suppressor Proteins/metabolism , src Homology Domains
8.
ACS Chem Biol ; 6(10): 1008-14, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21797253

ABSTRACT

Phosphorylation-dependent protein binding domains are crucially important for intracellular signaling pathways and thus highly relevant targets in chemical biology. By screening of chemical libraries against 12 structurally diverse phosphorylation-dependent protein binding domains, we have identified fosfosal and dexamethasone-21-phosphate as selective inhibitors of two antitumor targets: the SH2 domain of the transcription factor STAT5b and the substrate-binding domain of the peptidyl-prolyl isomerase Pin1, respectively. Both compounds are phosphate prodrugs with documented clinical use as anti-inflammatory agents in humans and were discovered with a high hit rate from a small subgroup within the screening library. Our study indicates O-phosphorylation of appropriately preselected natural products or natural product derivatives as a generally applicable strategy for the identification of non-reactive and non-peptidic ligands of phosphorylation-dependent protein binding domains. Moreover, our data indicate that it would be advisable to monitor the bioactivities of clinically used prodrugs in their uncleaved state against phosphorylation-dependent protein binding domains.


Subject(s)
Antineoplastic Agents/pharmacology , Dexamethasone/analogs & derivatives , Organophosphates/pharmacology , Peptidylprolyl Isomerase/antagonists & inhibitors , STAT5 Transcription Factor/antagonists & inhibitors , Binding Sites , Dexamethasone/pharmacology , Humans , Models, Molecular , NIMA-Interacting Peptidylprolyl Isomerase , Peptidylprolyl Isomerase/metabolism , Phosphorylation , Protein Binding , STAT5 Transcription Factor/metabolism , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...