Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
1.
Bioorg Med Chem ; 109: 117789, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38870716

ABSTRACT

Targeted protein degradation (TPD), employing proteolysis-targeting chimeras (PROTACs) composed of ligands for both a target protein and ubiquitin ligase (E3) to redirect the ubiquitin-proteasome system (UPS) to the target protein, has emerged as a promising strategy in drug discovery. However, despite the vast number of E3 ligases, the repertoire of E3 ligands utilized in PROTACs remains limited. Here, we report the discovery of a small-molecule degron with a phenylpropionic acid skeleton, derived from a known ligand of S-phase kinase-interacting protein 2 (Skp2), an E3 ligase. We used this degron to design PROTACs inducing proteasomal degradation of HaloTag-fused proteins, and identified key structural relationships. Surprisingly, our mechanistic studies excluded the involvement of Skp2, suggesting that this degron recruits other protein(s) within the UPS.


Subject(s)
S-Phase Kinase-Associated Proteins , Small Molecule Libraries , Humans , S-Phase Kinase-Associated Proteins/metabolism , S-Phase Kinase-Associated Proteins/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemical synthesis , Proteolysis/drug effects , Phenylpropionates/chemistry , Phenylpropionates/pharmacology , Structure-Activity Relationship , Proteasome Endopeptidase Complex/metabolism , Molecular Structure , Ligands , HEK293 Cells , Degrons
2.
Chem Pharm Bull (Tokyo) ; 72(2): 161-165, 2024.
Article in English | MEDLINE | ID: mdl-38296558

ABSTRACT

YM-1, an allosteric modulator of heat-shock 70 kDa protein (Hsp70), inhibits cancer cell growth, but the mechanism is not yet fully understood. Here, we show that YM-1 induces the degradation of bromodomain containing 4 (BRD4), which mediates oncogene expression. Overall, our results indicate that YM-1 promotes the binding of HSP70 to BRD4, and this in turn promotes the ubiquitination of BRD4 by C-terminus of Hsc70-interacting protein (CHIP), an E3 ubiquitin ligase working in concert with Hsp70, leading to proteasomal degradation of BRD4. This YM-1-induced decrease of BRD4 would contribute at least in part to the inhibition of cancer cell growth.


Subject(s)
Doxorubicin/analogs & derivatives , Heat-Shock Proteins , Nuclear Proteins , Heat-Shock Proteins/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Ubiquitination , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Protein Binding
3.
Int J Mol Sci ; 23(19)2022 Oct 02.
Article in English | MEDLINE | ID: mdl-36232972

ABSTRACT

Weak and transient protein interactions are involved in dynamic biological responses and are an important research subject; however, methods to elucidate such interactions are lacking. Proximity labeling is a promising technique for labeling transient ligand-binding proteins and protein-protein interaction partners of analytes via an irreversible covalent bond. Expanding chemical tools for proximity labeling is required to analyze the interactome. We developed several photocatalytic proximity-labeling reactions mediated by two different mechanisms. We found that numerous dye molecules can function as catalysts for protein labeling. We also identified catalysts that selectively modify tyrosine and histidine residues and evaluated their mechanisms. Model experiments using HaloTag were performed to demonstrate photocatalytic proximity labeling. We found that both ATTO465, which catalyzes labeling by a single electron transfer, and BODIPY, which catalyzes labeling by singlet oxygen, catalyze proximity labeling in cells.


Subject(s)
Histidine , Tyrosine , Histidine/chemistry , Ligands , Proteins , Singlet Oxygen/metabolism , Tyrosine/chemistry
4.
J Am Chem Soc ; 143(20): 7726-7731, 2021 05 26.
Article in English | MEDLINE | ID: mdl-33904715

ABSTRACT

While electrophilic reagents for histidine labeling have been developed, we report an umpolung strategy for histidine functionalization. A nucleophilic small molecule, 1-methyl-4-arylurazole, selectively labeled histidine under singlet oxygen (1O2) generation conditions. Rapid histidine labeling can be applied for instant protein labeling. Utilizing the short diffusion distance of 1O2 and a technique to localize the 1O2 generator, a photocatalyst in close proximity to the ligand-binding site, we demonstrated antibody Fc-selective labeling on magnetic beads functionalized with a ruthenium photocatalyst and Fc ligand, ApA. Three histidine residues located around the ApA binding site were identified as labeling sites by liquid chromatography-mass spectrometry analysis. This result suggests that 1O2-mediated histidine labeling can be applied to a proximity labeling reaction on the nanometer scale.

5.
Org Biomol Chem ; 19(2): 446-456, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33331380

ABSTRACT

Aqueous solubility is a key requirement for small-molecule drug candidates. Here, we investigated the regioisomer-physicochemical property relationships of disubstituted benzenes. We found that meta-isomers bearing non-flat substituents tend to possess the lowest melting point and the highest thermodynamic aqueous solubility among the regioisomers. The examination of pharmaceutical compounds containing a disubstituted benzene moiety supported the idea that the introduction of a non-flat substituent at the meta position of a benzene substructure would be a promising approach for medicinal chemists aiming to improve the thermodynamic aqueous solubility of drug candidates, even though it might not be universally effective.


Subject(s)
Drug Design , Small Molecule Libraries/chemistry , Water/chemistry , Isomerism , Solubility , Structure-Activity Relationship , Thermodynamics , Transition Temperature
6.
Bioorg Med Chem ; 41: 116221, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34034148

ABSTRACT

Chemical knockdown of therapeutic targets using proteolysis targeting chimeras (PROTACs) is a rapidly developing field in drug discovery, but PROTACs are bifunctional molecules that generally show poor bioavailability due to their relatively high molecular weight. Recent developments aimed at the development of next-generation PROTACs include the in vivo synthesis of PROTAC molecules, and the exploitation of PROTACs as chemical tools for in vivo synthesis of ubiquitinated proteins. This short review covers recent advances in these areas and discusses the prospects for in vivo synthetic PROTAC technology.


Subject(s)
Drug Discovery , Proteasome Endopeptidase Complex , Ubiquitin-Protein Ligases , Humans , Molecular Targeted Therapy , Proteasome Endopeptidase Complex/physiology , Proteolysis
7.
Int J Mol Sci ; 22(16)2021 Aug 12.
Article in English | MEDLINE | ID: mdl-34445381

ABSTRACT

Human serum albumin (HSA) is a promising drug delivery carrier. Although covalent modification of Cys34 is a well-established method, it is desirable to develop a novel covalent modification method that targets residues other than cysteine to introduce multiple functions into a single HSA molecule. We developed a tyrosine-selective modification of HSA. Three tyrosine selective modification methods, hemin-catalyzed, horseradish peroxidase (HRP)-catalyzed, and laccase-catalyzed reactions were performed, and the modification efficiencies and modification sites of the modified HSAs obtained by these methods were evaluated and compared. We found that the laccase-catalyzed method could efficiently modify the tyrosine residue of HSA under mild reaction conditions without inducing oxidative side reactions. An average of 2.2 molecules of functional groups could be introduced to a single molecule of HSA by the laccase method. Binding site analysis using mass spectrometry suggested Y84, Y138, and Y401 as the main modification sites. Furthermore, we evaluated binding to ibuprofen and found that, unlike the conventional lysine residue modification, the inhibition of drug binding was minimal. These results suggest that tyrosine-residue selective chemical modification is a promising method for covalent drug attachment to HSA.


Subject(s)
Hemin/metabolism , Horseradish Peroxidase/metabolism , Laccase/metabolism , Serum Albumin, Human/chemistry , Tyrosine/chemistry , Binding Sites , Biocatalysis , Click Chemistry , Drug Delivery Systems , Humans , Ibuprofen/chemistry , Mass Spectrometry , Models, Molecular , Molecular Structure , Protein Conformation , Serum Albumin, Human/metabolism
8.
Angew Chem Int Ed Engl ; 60(7): 3346-3354, 2021 02 15.
Article in English | MEDLINE | ID: mdl-32410219

ABSTRACT

Neurodegenerative disorders (NDs) are a group of diseases that cause neural cell damage, leading to motility and/or cognitive dysfunctions. One of the causative agents is misfolded protein aggregates, which are considered as undruggable in terms of conventional tools, such as inhibitors and agonists/antagonists. Indeed, there is currently no FDA-approved drug for the causal treatment of NDs. However, emerging technologies for chemical protein degradation are opening up the possibility of selective elimination of target proteins through physiological protein degradation machineries, which do not depend on the functions of the target proteins. Here, we review recent efforts towards the treatment of NDs using chemical protein degradation technologies, and we briefly discuss the challenges and prospects.


Subject(s)
Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Proteins/agonists , Proteins/antagonists & inhibitors , Proteolysis/drug effects , Humans , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/chemistry , Protein Aggregates/drug effects , Proteins/metabolism
9.
Bioorg Med Chem ; 28(1): 115175, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31767406

ABSTRACT

Polyglutamine diseases are a class of neurodegenerative diseases associated with the accumulation of aggregated mutant proteins. We previously developed a class of degradation-inducing agents targeting the ß-sheet-rich structure typical of such aggregates, and we showed that these agents dose-, time-, and proteasome-dependently decrease the intracellular level of mutant huntingtin with an extended polyglutamine tract, which correlates well with the severity of Huntington's disease. Here, we demonstrate that the same agents also deplete other polyglutamine disease-related proteins: mutant ataxin-3 and ataxin-7 in cells from spino-cerebellar ataxia patients, and mutant atrophin-1 in cells from dentatorubral-pallidoluysian atrophy patients. Targeting cross-ß-sheet structure could be an effective design strategy to develop therapeutic agents for multiple neurodegenerative diseases.


Subject(s)
Ataxin-3/antagonists & inhibitors , Ataxin-7/antagonists & inhibitors , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/pharmacology , Repressor Proteins/antagonists & inhibitors , Ataxin-3/genetics , Ataxin-7/genetics , Cells, Cultured , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Humans , Molecular Structure , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Repressor Proteins/genetics , Structure-Activity Relationship
10.
Bioorg Med Chem Lett ; 28(4): 707-710, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29366651

ABSTRACT

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by aggregation of mutant huntingtin (mHtt), and removal of mHtt is expected as a potential therapeutic option. We previously reported protein knockdown of Htt by using hybrid small molecules (Htt degraders) consisting of BE04, a ligand of ubiquitin ligase (E3), linked to probes for protein aggregates. Here, in order to examine the effect of changing the ligand, we synthesized a similar Htt degrader utilizing MV1, an antagonist of the inhibitor of apoptosis protein (IAP) family (a subgroup of ubiquitin E3 ligases), which is expected to have a higher affinity and specificity for IAP, as compared with BE04. The MV1-based hybrid successfully induced interaction between Htt aggregates and IAP, and reduced mHtt levels in living cells. Its mode of action was confirmed to be the same as that of the BE04-based hybrid. However, although the affinity of MV1 for IAP is greater than that of BE04, the efficacy of Htt degradation by the MV1-based molecule was lower, suggesting that linker length between the ligand and probe might be an important determinant of efficacy.


Subject(s)
Benzothiazoles/pharmacology , Huntingtin Protein/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Oligopeptides/pharmacology , Benzothiazoles/chemical synthesis , Benzothiazoles/chemistry , Fibroblasts/drug effects , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Ligands , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Protein Binding
11.
Bioorg Med Chem Lett ; 28(4): 796-801, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29398545

ABSTRACT

LXRß-selective agonists are promising candidates to improve atherosclerosis without increasing plasma or hepatic TG levels. We have reported a series of tetrachlorophthalimide analogs as an LXRß-selective agonist. However, they exhibited poor aqueous solubility probably due to its high hydrophobicity and highly rigid and plane structure. In this report, we present further structural development of tetrachloro(styrylphenyl)phthalimides as the LXRß-selective agonists with improved aqueous solubility.


Subject(s)
Liver X Receptors/agonists , Phthalimides/pharmacology , Water/chemistry , ATP Binding Cassette Transporter 1/genetics , Animals , Humans , Hydrophobic and Hydrophilic Interactions , Mice , Molecular Docking Simulation , Molecular Structure , Phthalimides/chemical synthesis , Phthalimides/chemistry , RNA, Messenger/genetics , Solubility , Structure-Activity Relationship , THP-1 Cells
12.
Int J Mol Sci ; 19(7)2018 Jul 18.
Article in English | MEDLINE | ID: mdl-30021999

ABSTRACT

BACKGROUND: Nuclear receptors (NRs) are considered as potential drug targets because they control diverse biological functions. However, steroidal ligands for NRs have the potential to cross-react with other nuclear receptors, so development of non-steroidal NR ligands is desirable to obtain safer agents for clinical use. We anticipated that efficient lead finding and enhancement of activity toward nuclear receptors recognizing endogenous steroidal ligands might be achieved by exhaustive evaluation of a steroid surrogate library coupled with examination of structure-activity relationships (SAR). METHOD: We evaluated our library of RORs (retinoic acid receptor-related orphan receptors) inverse agonists and/or PR (progesterone receptor) antagonists based on the phenanthridinone skeleton for antagonistic activities toward liver X receptors (LXRs), androgen receptor (AR) and glucocorticoid receptor (GR) and examined their SAR. RESULTS: Potent LXRß, AR, and GR antagonists were identified. SAR studies led to a potent AR antagonist (IC50: 0.059 µM). CONCLUSIONS: Our approach proved effective for efficient lead finding, activity enhancement and preliminary control of selectivity over other receptors. The phenanthridinone skeleton appears to be a promising steroid surrogate.


Subject(s)
Phenanthridines/chemistry , Phenanthridines/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Androgen Antagonists/chemistry , Androgen Antagonists/pharmacology , Cell Line, Tumor , HEK293 Cells , Humans , Ligands , Structure-Activity Relationship
13.
Mol Pharmacol ; 91(3): 159-166, 2017 03.
Article in English | MEDLINE | ID: mdl-27965304

ABSTRACT

Development of novel small molecules that selectively degrade pathogenic proteins would provide an important advance in targeted therapy. Recently, we have devised a series of hybrid small molecules named SNIPER (specific and nongenetic IAP-dependent protein ERaser) that induces the degradation of target proteins via the ubiquitin-proteasome system. To understand the localization of proteins that can be targeted by this protein knockdown technology, we examined whether SNIPER molecules are able to induce degradation of cellular retinoic acid binding protein II (CRABP-II) proteins localized in subcellular compartments of cells. CRABP-II is genetically fused with subcellular localization signals, and they are expressed in the cells. SNIPER(CRABP) with different IAP-ligands, SNIPER(CRABP)-4 with bestatin and SNIPER(CRABP)-11 with MV1 compound, induce the proteasomal degradation of wild-type (WT), cytosolic, nuclear, and membrane-localized CRABP-II proteins, whereas only SNIPER(CRABP)-11 displayed degradation activity toward the mitochondrial CRABP-II protein. The small interfering RNA-mediated silencing of cIAP1 expression attenuated the knockdown activity of SNIPER(CRABP) against WT and cytosolic CRABP-II proteins, indicating that cIAP1 is the E3 ligase responsible for degradation of these proteins. Against membrane-localized CRABP-II protein, cIAP1 is also a primary E3 ligase in the cells, but another E3 ligase distinct from cIAP2 and X-linked inhibitor of apoptosis protein (XIAP) could also be involved in the SNIPER(CRABP)-11-induced degradation. However, for the degradation of nuclear and mitochondrial CRABP-II proteins, E3 ligases other than cIAP1, cIAP2, and XIAP play a role in the SNIPER-mediated protein knockdown. These results indicate that SNIPER can target cytosolic, nuclear, membrane-localized, and mitochondrial proteins for degradation, but the responsible E3 ligase is different, depending on the localization of the target protein.


Subject(s)
Proteins/metabolism , Proteolysis , Small Molecule Libraries/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Nucleus/metabolism , Cytosol/metabolism , Humans , Mitochondrial Proteins/metabolism , Organelles/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, Retinoic Acid/metabolism , Subcellular Fractions/metabolism , X-Linked Inhibitor of Apoptosis Protein
14.
Bioorg Med Chem Lett ; 27(12): 2776-2780, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28465099

ABSTRACT

Anti-inflammatory effects of peroxisome proliferator-activated receptor gamma (PPRAγ) ligands are thought to be largely due to PPARγ-mediated transrepression. Thus, transrepression-selective PPARγ ligands without agonistic activity or with only partial agonistic activity should exhibit anti-inflammatory properties with reduced side effects. Here, we investigated the structure-activity relationships (SARs) of PPARγ agonist rosiglitazone, focusing on transrepression activity. Alkenic analogs showed slightly more potent transrepression with reduced efficacy of transactivating agonistic activity. Removal of the alkyl group on the nitrogen atom improved selectivity for transrepression over transactivation. Among the synthesized compounds, 3l exhibited stronger transrepressional activity (IC50: 14µM) and weaker agonistic efficacy (11%) than rosiglitazone or pioglitazone.


Subject(s)
PPAR gamma/agonists , Thiazolidinediones/pharmacology , Dose-Response Relationship, Drug , Humans , Models, Molecular , Molecular Structure , PPAR gamma/metabolism , Rosiglitazone , Structure-Activity Relationship , Thiazolidinediones/chemical synthesis , Thiazolidinediones/chemistry
15.
Bioorg Med Chem Lett ; 27(12): 2781-2787, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28465104

ABSTRACT

Niemann-Pick disease type C is a fatal, progressive neurodegenerative disease mostly caused by mutations in Nieamnn-Pick type C1 (NPC1), a late endosomal membrane protein that is essential for intracellular cholesterol transport. The most prevalent mutation, I1061T (Ile to Thr), interferes with the protein folding process. Consequently, mutated but intrinsically functional NPC1 proteins are prematurely degraded via proteasome, leading to loss of NPC1 function. Previously, we reported sterol derivatives as pharmacological chaperones for NPC1, and showed that these derivatives can normalize folding-defective phenotypes of I1061T NPC1 mutant by directly binding to, and stabilizing, the protein. Here, we report a series of compounds containing a phenanthridin-6-one scaffold as the first class of non-steroidal pharmacological chaperones for NPC1. We also examined their structure-activity relationships.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Membrane Glycoproteins/antagonists & inhibitors , Phenanthridines/pharmacology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Dose-Response Relationship, Drug , Humans , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Molecular Structure , Mutation , Niemann-Pick C1 Protein , Phenanthridines/chemical synthesis , Phenanthridines/chemistry , Structure-Activity Relationship
16.
Bioorg Med Chem Lett ; 27(14): 3131-3134, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28539218

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are important drug targets for treatment of dyslipidemia, type 2 diabetes, cardiovascular disease, nonalcoholic fatty liver disease and nonalcoholic steatohepatitis, and great efforts have been made to develop novel PPAR ligands. However, most existing PPAR ligands contain a carboxylic acid (CA) or thiazolidinedione (TZD) structure (acidic head group) that is essential for activity. We recently discovered non-CA/TZD class PPARα/δ partial agonists, which contain an acetamide moiety and adjacent methyl group, linked to a 1,2,4-oxadiazole ring ("fragment a"). We hypothesized that the acetamide structure might interact with the CA/TZD-binding pocket. To test this idea, we firstly replaced fragment a in one of our compounds with the α-alkoxy-CA structure often found in PPAR agonists. Secondly, we replaced the α-alkoxy-CA head group of several reported PPAR agonists with our acetamide-based fragment a. The agonistic activities of the synthesized hybrid compounds toward PPARs (PPARα, PPARγ and PPARδ) were evaluated by means of cell-based reporter gene assays. All the hybrid molecules showed PPAR-agonistic activities, but replacement of the α-alkoxy-CA head group altered the maximum efficacy and the subtype-specificity. The acetamide-based hybrid molecules showed partial agonism toward PPARα and PPARδ, whereas the α-alkoxy-CA-based molecules were generally selective for PPARα and PPARγ, with relatively high activation efficacies. Thus, the fragment replacement strategy appears promising for the development of novel acetamide-based PPARα/δ dual agonists.


Subject(s)
PPAR alpha/agonists , PPAR delta/agonists , Acetamides/chemical synthesis , Acetamides/chemistry , Acetamides/metabolism , Binding Sites , Genes, Reporter , HEK293 Cells , Humans , Ligands , PPAR alpha/metabolism , PPAR delta/metabolism , Protein Binding , Stereoisomerism , Thiazolidinediones/chemistry
17.
Angew Chem Int Ed Engl ; 56(38): 11530-11533, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28703441

ABSTRACT

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by the aggregation of mutant huntingtin (mHtt), and removal of toxic mHtt is expected to be an effective therapeutic approach. We designed two small hybrid molecules (1 and 2) by linking a ligand for ubiquitin ligase (cellular inhibitor of apoptosis protein 1; cIAP1) with probes for mHtt aggregates, anticipating that these compounds would recruit cIAP1 to mHtt and induce selective degradation by the ubiquitin-proteasome system. The synthesized compounds reduced mHtt levels in HD patient fibroblasts and appear to be promising candidates for the development of a treatment for HD.


Subject(s)
Drug Discovery , Huntingtin Protein/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibroblasts/metabolism , HeLa Cells , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/drug therapy , Huntington Disease/metabolism , Ligands , Molecular Structure , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
18.
Bioorg Med Chem ; 24(14): 3144-8, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27236416

ABSTRACT

We previously reported a protein knockdown system for HaloTag-fused proteins using hybrid small molecules consisting of alkyl chloride, which binds covalently to HaloTag, linked to BE04 (2), a bestatin (3) derivative with an affinity for cellular inhibitor of apoptosis protein 1 (cIAP1, a kind of ubiquitin ligase). This system addressed several limitations of prior protein knockdown technology, and was applied to degrade two HaloTag-fused proteins. However, the degradation activity of these hybrid small molecules was not potent. Therefore, we set out to improve this system. We report here the design, synthesis and biological evaluation of novel hybrid compounds 4a and 4b consisting of alkyl chloride linked to IAP antagonist MV1 (5). Compounds 4a and 4b were confirmed to reduce the levels of HaloTag-fused tumor necrosis factor α (HaloTag-TNFα), HaloTag-fused cell division control protein 42 (HaloTag-Cdc42), and unfused HaloTag protein in living cells more potently than did BE04-linked compound 1b. Analysis of the mode of action revealed that the reduction of HaloTag-TNFα is proteasome-dependent, and is also dependent on the linker structure between MV1 (5) and alkyl chloride. These compounds appear to induce ubiquitination at the HaloTag moiety of HaloTag-fused proteins. Our results indicate that these newly synthesized MV1-type hybrid compounds, 4a and 4b, are efficient tools for protein knockdown for HaloTag-fused proteins.


Subject(s)
Recombinant Fusion Proteins/chemistry , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , Carbon-13 Magnetic Resonance Spectroscopy , HEK293 Cells , Humans , Proton Magnetic Resonance Spectroscopy , Recombinant Fusion Proteins/genetics , Spectrometry, Mass, Electrospray Ionization , Tumor Necrosis Factor-alpha/chemistry , cdc42 GTP-Binding Protein/chemistry
19.
Bioorg Med Chem ; 24(21): 5258-5269, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27591006

ABSTRACT

Acetyl-CoA carboxylases (ACCs) catalyze a critical step in de novo lipogenesis, and are considered as promising targets for treatment of obesity, dyslipidemia and type 2 diabetes mellitus. On the other hand, peroxisome proliferator-activated receptors (PPARs) are well-established therapeutic targets for these metabolic syndrome-related diseases. Therefore, we considered that dual modulators of ACC and PPARs would be promising candidates as therapeutic agents. Here, we designed a series of acetamides based on the molecular similarity between ACC inhibitors and PPAR agonists. Screening of the synthesized compounds identified N-(1-(3-(4-phenoxyphenyl)-1,2,4-oxadiazol-5-yl)ethyl)acetamides as novel ACC2 inhibitors with PPARα/PPARδ dual agonistic activity. Structure-activity relationship studies and further structural elaboration afforded compounds with distinct activity profiles. Our findings should be helpful for the discovery of candidate agents with an appropriate balance of ACC-inhibitory and PPAR-activating activities for therapeutic lipid control.


Subject(s)
Acetamides/pharmacology , Acetyl-CoA Carboxylase/antagonists & inhibitors , Drug Discovery , Enzyme Inhibitors/pharmacology , Oxadiazoles/pharmacology , PPAR alpha/agonists , PPAR delta/agonists , Acetamides/chemical synthesis , Acetamides/chemistry , Acetyl-CoA Carboxylase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Oxadiazoles/chemical synthesis , Oxadiazoles/chemistry , Structure-Activity Relationship
20.
Bioorg Med Chem ; 24(21): 5455-5461, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27622746

ABSTRACT

The peroxisome proliferator-activated receptors (PPARs) are ligand-dependent transcription factors that contribute to the regulation of lipid, glucose and cholesterol homeostases. They are considered as therapeutic targets for metabolic diseases such as dyslipidemia and type 2 diabetes mellitus. Various PPAR agonists have been developed, but most of them contain a carboxylic acid (CA) or thiazolidinedione (TZD) moiety, which is essential for the activity. However, we recently discovered non-CA/non-TZD class PPARα/δ dual agonists having an acetamide structure. Here, we describe structure-activity relationship (SAR) studies of these novel acetamide-based PPARα/δ dual agonists. The SAR studies revealed that the acetamide functionality and adjacent methyl group contribute greatly to the agonistic activity. Compound (S)-10 was the most potent PPARα/δ dual agonist among the compounds synthesized (PPARα EC50=17nM, PPARδ EC50=23nM).


Subject(s)
Acetamides/pharmacology , PPAR alpha/agonists , PPAR delta/agonists , Acetamides/chemical synthesis , Acetamides/chemistry , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Molecular Structure , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL