Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 270
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 117(13): 7150-7158, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32170014

ABSTRACT

Cholesterol biosynthesis is a high-cost process and, therefore, tightly regulated by both transcriptional and posttranslational negative feedback mechanisms in response to the level of cellular cholesterol. Squalene monooxygenase (SM, also known as squalene epoxidase or SQLE) is a rate-limiting enzyme in the cholesterol biosynthetic pathway and catalyzes epoxidation of squalene. The stability of SM is negatively regulated by cholesterol via its N-terminal regulatory domain (SM-N100). In this study, using a SM-luciferase fusion reporter cell line, we performed a chemical genetics screen that identified inhibitors of SM itself as up-regulators of SM. This effect was mediated through the SM-N100 region, competed with cholesterol-accelerated degradation, and required the E3 ubiquitin ligase MARCH6. However, up-regulation was not observed with statins, well-established cholesterol biosynthesis inhibitors, and this pointed to the presence of another mechanism other than reduced cholesterol synthesis. Further analyses revealed that squalene accumulation upon treatment with the SM inhibitor was responsible for the up-regulatory effect. Using photoaffinity labeling, we demonstrated that squalene directly bound to the N100 region, thereby reducing interaction with and ubiquitination by MARCH6. Our findings suggest that SM senses squalene via its N100 domain to increase its metabolic capacity, highlighting squalene as a feedforward factor for the cholesterol biosynthetic pathway.


Subject(s)
Squalene Monooxygenase/metabolism , Squalene/metabolism , Allosteric Regulation , Benzylamines , Cholesterol/biosynthesis , Endoplasmic Reticulum/enzymology , HEK293 Cells , Humans , Membrane Proteins/metabolism , Proteostasis , Squalene Monooxygenase/antagonists & inhibitors , Thiophenes , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
2.
Bioorg Med Chem ; 66: 116792, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35576658

ABSTRACT

Liver X receptor (LXR) α and LXRß are nuclear receptors playing key roles in lipid metabolism, and LXR ligands are attractive drug candidates for metabolic disorders. Here we report the structural development of 4-(1,1,1,3,3,3-hexafluoro-2-hydroxyprop-2-yl)phenylsilane derivatives as LXR agonists bearing silyl functionalities as the hydrophobic pharmacophore, based on the structure of the known sulfonamide LXR agonist T0901317. Most of the synthesized compounds exhibit agonistic activity toward LXRs, but the LXR subtype-selectivity differs depending upon the substituents on the silicon atom. Among them, tri(n-propyl) derivative 12 shows potent LXR-agonistic activity with moderate α subtype-selectivity, while dimethylphenylsilyl derivative 19 shows modest ß-selectivity. These results indicate that silanes can serve as an alternative to the sulfonamide moiety of LXR agonists, and are promising structural options for the development of novel subtype-selective LXR agonists.


Subject(s)
Hydrocarbons, Fluorinated , Receptors, Cytoplasmic and Nuclear , Hydrocarbons, Fluorinated/pharmacology , Liver/metabolism , Liver X Receptors/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Structure-Activity Relationship , Sulfonamides/metabolism , Sulfonamides/pharmacology
3.
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
4.
Bioorg Med Chem ; 28(14): 115576, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32616181

ABSTRACT

HMG-CoA reductase (HMGCR) is a rate-limiting enzyme in the cholesterol biosynthetic pathway, and its catalytic domain is the well-known target of cholesterol-lowering drugs, statins. HMGCR is subject to layers of negative feedback loops; excess cholesterol inhibits transcription of the gene, and lanosterols and oxysterols accelerate degradation of HMGCR. A class of synthetic small molecules, bisphosphonate esters exemplified by SR12813, has been known to induce accelerated degradation of HMGCR and reduce the serum cholesterol level. Although genetic and biochemical studies revealed that the accelerated degradation requires the membrane domain of HMGCR and Insig, an oxysterol sensor on the endoplasmic reticulum membrane, the direct target of the bisphosphonate esters remains unclear. In this study, we developed a potent photoaffinity probe of the bisphosphonate esters through preliminary structure-activity relationship study and demonstrated binding of the bisphosphonate esters to the HMGCR membrane domain. These results provide an important clue to understand the elusive mechanism of the SR12813-mediated HMGCR degradation and serve as a basis to develop more potent HMGCR degraders that target the non-catalytic, membrane domain of the enzyme.


Subject(s)
Diphosphonates/pharmacology , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Cells, Cultured , Diphosphonates/chemical synthesis , Diphosphonates/chemistry , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemical synthesis , Hydroxymethylglutaryl-CoA Reductase Inhibitors/chemistry , Molecular Structure , Structure-Activity Relationship
5.
Bioorg Med Chem ; 28(4): 115310, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31980362

ABSTRACT

Increasing structural options in medicinal chemistry is important for the development of novel and distinctive drug candidates. In this study, we focused on phosphorus-containing functionalities. We designed and synthesized a series of phosphinophenol derivatives and determined their physicochemical properties, including hydrophobicity parameter LogP, and their biological activity toward estrogen receptor (ER). Notably, the phosphine borane derivatives (9 and 14) exhibited potent ER-antagonistic activity, exceeding the potency of the corresponding alkane (15) and silane (16) derivatives, despite having a less hydrophobic nature. The determined physicochemical parameters will be helpful for the rational design of phosphorus-containing biologically active compounds. Our results indicate that phosphine boranes are a promising new chemical entry in the range of structural options for drug discovery.


Subject(s)
Boranes/pharmacology , Estrogen Receptor Modulators/pharmacology , Phenols/pharmacology , Phosphines/pharmacology , Receptors, Estrogen/antagonists & inhibitors , Alkanes/chemistry , Boranes/chemistry , Dose-Response Relationship, Drug , Estrogen Receptor Modulators/chemical synthesis , Estrogen Receptor Modulators/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Structure , Phenols/chemical synthesis , Phenols/chemistry , Phosphines/chemistry , Receptors, Estrogen/metabolism , Silanes/chemistry , Structure-Activity Relationship
6.
Bioorg Med Chem ; 28(3): 115298, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31902650

ABSTRACT

HMG-CoA reductase (HMGCR) is the rate-limiting enzyme in the cholesterol biosynthetic pathway, and is the target of cholesterol-lowering drugs, statins. Previous studies have demonstrated that the enzyme activity is regulated by sterol-induced degradation in addition to transcriptional regulation through sterol-regulatory-element-binding proteins (SREBPs). While 25-hydroxycholesterol induces both HMGCR degradation and SREBP inhibition in a nonselective manner, lanosterol selectively induces HMGCR degradation. Here, to clarify the structural determinants of selectivity for the two activities, we established a luciferase-based assay monitoring HMGCR degradation and used it to profile the structure-activity/selectivity relationships of oxysterols and (oxy)lanosterols. We identified several sterols that selectively induce HMGCR degradation and one sterol, 25-hydroxycholest-4-en-3-one, that selectively inhibits the SREBP pathway. These results should be helpful in designing more potent and selective HMGCR degraders.


Subject(s)
Hydroxymethylglutaryl CoA Reductases/metabolism , Lanosterol/metabolism , Oxysterols/metabolism , HEK293 Cells , Humans , Lanosterol/pharmacology , Molecular Structure , Oxysterols/pharmacology , Sterol Regulatory Element Binding Proteins/antagonists & inhibitors , Sterol Regulatory Element Binding Proteins/metabolism , Structure-Activity Relationship
7.
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
8.
Bioorg Med Chem ; 27(10): 1952-1961, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30940565

ABSTRACT

Selective estrogen receptor (ER) down-regulators (SERDs) are pure ER antagonists that also induce ER degradation upon binding to the receptor. Although SERDs have been developed for the treatment of ER-positive breast cancers for nearly a decade, their precise mechanism(s) of action and structure-activity relationship are still unclear. Generally, Western blotting is used to examine the effects of SERDs on ER protein levels, but the methodology is low-throughput and not quantitative. Here, we describe a quantitative, high-throughput, luciferase-based assay for the evaluation of SERDs activity. For this purpose, we established stable recombinant HEK-293 cell lines expressing ERα fused with emerald luciferase. We also designed and synthesized new diphenylmethane derivatives as candidate SERDs, and evaluated their SERDs activity using the developed system in order to examine their structure-activity relationship, taking EC50 as a measure of potency, and Emax as a measure of efficacy.


Subject(s)
Benzhydryl Compounds/chemistry , Down-Regulation/drug effects , Estrogen Receptor alpha/antagonists & inhibitors , Benzhydryl Compounds/pharmacology , Binding Sites , Cyclofenil/chemistry , Cyclofenil/metabolism , Estrogen Antagonists/chemistry , Estrogen Antagonists/metabolism , Estrogen Antagonists/pharmacology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , HEK293 Cells , Humans , Molecular Docking Simulation , Phenols/chemistry , Phenols/pharmacology , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship
9.
Chem Pharm Bull (Tokyo) ; 67(6): 566-575, 2019.
Article in English | MEDLINE | ID: mdl-31155562

ABSTRACT

We report here the development of phenylamino-1,3,5-triazine derivatives as novel nonsteroidal progesterone receptor (PR) antagonists. PR plays key roles in various physiological systems, including the female reproductive system, and PR antagonists are promising candidates for clinical treatment of multiple diseases. By using the phenylamino-1,3,5-triazine scaffold as a template structure, we designed and synthesized a series of 4-cyanophenylamino-1,3,5-triazine derivatives. The synthesized compounds exhibited PR antagonistic activity, and among them, compound 12n was the most potent (IC50 = 0.30 µM); it also showed significant binding affinity to the PR ligand-binding domain. Docking simulation supported the design rationale of the compounds. Our results suggest that the phenylamino-1,3,5-triazine scaffold is a versatile template for development of nonsteroidal PR antagonists and that the developed compounds are promising lead compounds for further structural development of nonsteroidal PR antagonists.


Subject(s)
Antineoplastic Agents/chemical synthesis , Drug Design , Receptors, Progesterone/antagonists & inhibitors , Triazines/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Humans , Inhibitory Concentration 50 , Ligands , Molecular Docking Simulation , Protein Binding , Protein Structure, Tertiary , Receptors, Progesterone/metabolism , Structure-Activity Relationship , Triazines/metabolism , Triazines/pharmacology
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.
Bioorg Med Chem ; 26(15): 4493-4501, 2018 08 15.
Article in English | MEDLINE | ID: mdl-30077610

ABSTRACT

Pregnane X receptor (PXR) is a ligand-dependent transcription factor that is considered to be a potential therapeutic target for multiple diseases. Herein, we report the development and structure-activity relationship studies of a new series of hPXR agonists. Focusing on our recently developed silanol-sulfonamide scaffold, we developed the potent hPXR agonist 28, which shows good selectivity over hLXRα and ß, hFXR, and hRORα and γ. Examination of the structure-activity relationship suggested a possible strategy to manipulate the selectivity. Docking simulation indicated the presence of an additional binding cavity and polar contacts in the ligand-binding pocket of hPXR. This information should be helpful for the future development of more potent and selective hPXR ligands.


Subject(s)
Pregnane X Receptor/agonists , Silanes/chemistry , Binding Sites , Drug Design , Humans , Liver X Receptors/agonists , Liver X Receptors/metabolism , Molecular Docking Simulation , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/metabolism , Pregnane X Receptor/metabolism , Protein Structure, Tertiary , Silanes/chemical synthesis , Silanes/metabolism , Structure-Activity Relationship
13.
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
14.
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
15.
J Biol Chem ; 291(23): 12282-93, 2016 Jun 03.
Article in English | MEDLINE | ID: mdl-27068745

ABSTRACT

A missense mutation (T835M) in the uncoordinated-5C (UNC5C) netrin receptor gene increases the risk of late-onset Alzheimer disease (AD) and also the vulnerability of neurons harboring the mutation to various insults. The molecular mechanisms underlying T835M-UNC5C-induced death remain to be elucidated. In this study, we show that overexpression of wild-type UNC5C causes low-grade death, which is intensified by an AD-linked mutation T835M. An AD-linked survival factor, calmodulin-like skin protein (CLSP), and a natural ligand of UNC5C, netrin1, inhibit this death. T835M-UNC5C-induced neuronal cell death is mediated by an intracellular death-signaling cascade, consisting of death-associated protein kinase 1/protein kinase D/apoptosis signal-regulating kinase 1 (ASK1)/JNK/NADPH oxidase/caspases, which merges at ASK1 with a death-signaling cascade, mediated by amyloid ß precursor protein (APP). Notably, netrin1 also binds to APP and partially inhibits the death-signaling cascade, induced by APP. These results may provide new insight into the amyloid ß-independent pathomechanism of AD.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/genetics , Mutation, Missense , Receptors, Cell Surface/genetics , Signal Transduction/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Apoptosis/genetics , Blotting, Western , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Caspases/metabolism , Cell Line , Cell Line, Tumor , Death-Associated Protein Kinases/metabolism , Humans , MAP Kinase Kinase Kinase 5/genetics , MAP Kinase Kinase Kinase 5/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , NADPH Oxidases/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Netrin Receptors , Netrin-1 , Neurons/cytology , Neurons/metabolism , Protein Binding , Protein Kinase C/metabolism , Receptors, Cell Surface/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
16.
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
17.
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
18.
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
19.
Bioorg Med Chem ; 25(14): 3677-3684, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28549889

ABSTRACT

Epigenetic regulation of gene expression via histone acetylation modulates many cellular processes, including apoptosis, the cell cycle, cell growth and differentiation, and inhibitors are promising drug candidates. We have previously developed inhibitors of BRD4, which recognizes acetylated lysine residue on histones and recruits transcription elongation factor to the transcription start site, while inhibitors of histone deacetylase (HDAC), which catalyzes the removal of acetyl groups on histones, are already in clinical use for cancer treatment. Based on the idea that polypharmacological agents with multiple targets would have a more robust action, we set out to develop dual BRD4/HDAC inhibitors. Here, we describe the design and synthesis of N6-[2-(7-hydroxyamino-7-oxoheptyloxy)benzoyl]adenine (5d) as a BRD4/HDAC dual inhibitor. This compound showed HL-60 cell growth-inhibitory and apoptosis-inducing activity, as well as all-trans retinoic acid (ATRA)-induced HL-60 cell differentiation-enhancing activity, and c-MYC production-inhibitory activity. Interestingly, it also showed growth-inhibitory activity towards BRD4 inhibitor-resistant cells.


Subject(s)
Histone Deacetylase Inhibitors/chemical synthesis , Nuclear Proteins/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Acetylation , Adenine/chemical synthesis , Adenine/chemistry , Adenine/toxicity , Apoptosis/drug effects , Cell Cycle Proteins , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Drug Screening Assays, Antitumor , HL-60 Cells , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylase Inhibitors/toxicity , Histone Deacetylases/chemistry , Histone Deacetylases/metabolism , Humans , Nuclear Proteins/metabolism , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Structure-Activity Relationship , Transcription Factors/metabolism , Tretinoin/pharmacology
20.
Bioorg Med Chem ; 25(13): 3461-3470, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28506584

ABSTRACT

N-Benzyl-N-(4-phenoxyphenyl)benzenesulfonamide derivatives were developed as a novel class of nonsteroidal glucocorticoid receptor (GR) modulators, which are promising drug candidates for treating immune-related disorders. Focusing on the similarity of the GR and progesterone receptor (PR) ligand-binding domain (LBD) structures, we adopted our recently developed PR antagonist 10 as a lead compound and synthesized a series of derivatives. We found that the N-(4-phenoxyphenyl)benzenesulfonamide skeleton serves as a versatile scaffold for GR antagonists. Among them, 4-cyano derivative 14m was the most potent, with an IC50 value of 1.43µM for GR. This compound showed good selectivity for GR; it retained relatively weak antagonistic activity toward PR (IC50 for PR: 8.00µM; 250-fold less potent than 10), but showed no activity toward AR, ERα or ERß. Interestingly, the 4-amino derivative 15a exhibited transrepression activity toward NF-κB in addition to GR-antagonistic activity, whereas 14m did not. The structure-activity relationship for transrepression was different from that for GR-antagonistic activity. Computational docking simulations suggested that 15a might bind to the ligand-binding pocket of GR in a different manner from 14m. These findings open up new possibilities for developing novel nonsteroidal GR modulators with distinctive activity profiles.


Subject(s)
Receptors, Glucocorticoid/antagonists & inhibitors , Sulfonamides/pharmacology , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Molecular Structure , Receptors, Glucocorticoid/metabolism , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL