Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Hepatology ; 76(2): 345-356, 2022 08.
Article in English | MEDLINE | ID: mdl-35108418

ABSTRACT

BACKGROUND AND AIMS: p21-activated kinase 4 (PAK4), an oncogenic protein, has emerged as a promising target for anticancer drug development. Its role in oxidative stress conditions, however, remains elusive. We investigated the effects of PAK4 signaling on hepatic ischemia/reperfusion (I/R) injury. APPROACH AND RESULTS: Hepatocyte- and myeloid-specific Pak4 knockout (KO) mice and their littermate controls were subjected to a partial hepatic I/R (HIR) injury. We manipulated the catalytic activity of PAK4, either through genetic engineering (gene knockout, overexpression of wild-type [WT] or dominant-negative kinase) or pharmacological inhibitor, coupled with a readout of nuclear factor erythroid 2-related factor 2 (Nrf2) activity, to test the potential function of PAK4 on HIR injury. PAK4 expression was markedly up-regulated in liver during HIR injury in mice and humans. Deletion of PAK4 in hepatocytes, but not in myeloid cells, ameliorated liver damages, as demonstrated in the decrease in hepatocellular necrosis and inflammatory responses. Conversely, the forced expression of WT PAK4 aggravated the pathological changes. PAK4 directly phosphorylated Nrf2 at T369, and it led to its nuclear export and proteasomal degradation, all of which impaired antioxidant responses in hepatocytes. Nrf2 silencing in liver abolished the protective effects of PAK4 deficiency. A PAK4 inhibitor protected mice from HIR injury. CONCLUSIONS: PAK4 phosphorylates Nrf2 and suppresses its transcriptional activity. Genetic or pharmacological suppression of PAK4 alleviates HIR injury. Thus, PAK4 inhibition may represent a promising intervention against I/R-induced liver injury.


Subject(s)
Liver Diseases , Reperfusion Injury , p21-Activated Kinases , Animals , Apoptosis , Humans , Ischemia/metabolism , Ischemia/pathology , Liver/pathology , Liver Diseases/etiology , Liver Diseases/metabolism , Liver Diseases/prevention & control , Mice , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Phosphorylation , Reperfusion Injury/metabolism , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism
2.
Nat Chem Biol ; 16(6): 635-643, 2020 06.
Article in English | MEDLINE | ID: mdl-32251410

ABSTRACT

Doublecortin like kinase 1 (DCLK1) is an understudied kinase that is upregulated in a wide range of cancers, including pancreatic ductal adenocarcinoma (PDAC). However, little is known about its potential as a therapeutic target. We used chemoproteomic profiling and structure-based design to develop a selective, in vivo-compatible chemical probe of the DCLK1 kinase domain, DCLK1-IN-1. We demonstrate activity of DCLK1-IN-1 against clinically relevant patient-derived PDAC organoid models and use a combination of RNA-sequencing, proteomics and phosphoproteomics analysis to reveal that DCLK1 inhibition modulates proteins and pathways associated with cell motility in this context. DCLK1-IN-1 will serve as a versatile tool to investigate DCLK1 biology and establish its role in cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Movement , Doublecortin Protein , Doublecortin-Like Kinases , Drug Screening Assays, Antitumor , Gene Expression Regulation , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Molecular Docking Simulation , Molecular Structure , Protein Kinase Inhibitors/pharmacokinetics , Proteomics , Rats , Structure-Activity Relationship , Zebrafish , Pancreatic Neoplasms
3.
Phytother Res ; 36(9): 3601-3618, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35871535

ABSTRACT

Puerarin, the major bioactive ingredient isolated from the root of Pueraria lobata (Willd.), attenuates body weight gain and reduces lipid levels in high-fat diet-induced obese mice; however, the underlying mechanism responsible for regulating lipid metabolism remains unclear. This study investigated the molecular mechanism(s) underlying the role of puerarin in regulating lipogenesis and lipolysis in human HepG2 cells. In this study, puerarin strongly inhibited the expression of fatty acid synthase (FASN) and sterol regulatory element binding protein 1c (SREBP-1c). Moreover, puerarin significantly induced the expression of adipose triglyceride lipase (ATGL), which is responsible for triacylglycerol hydrolase activity in cells. Puerarin enhanced 5' AMP-activated protein kinase (AMPK) activity, which is a central regulator of hepatic lipid metabolism. Furthermore, this AMPK activation could be mediated by sirtuin 1 (SIRT1) and calcium signaling pathways involved in G protein-coupled estrogen receptor (GPER) signaling. GPER blockage significantly reversed the effect of puerarin on lipid accumulation and the related signaling pathways. Docking studies showed that puerarin could bind in the GPER in a similar manner as GPER agonist G1. Our results suggest that puerarin can improve hepatic steatosis by activating GPER; it's signaling cascade sequentially induced calcium and SIRT1 signaling pathways. Thus, puerarin may be a potential therapeutic agent for the treatment of non-alcoholic fatty liver disease.


Subject(s)
Non-alcoholic Fatty Liver Disease , Sirtuin 1 , AMP-Activated Protein Kinases/metabolism , Animals , Calcium/metabolism , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/pharmacology , Hep G2 Cells , Humans , Isoflavones , Lipid Metabolism , Lipids , Liver , Mice , Mice, Obese , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, Estrogen/metabolism , Signal Transduction , Sirtuin 1/metabolism
4.
Int J Mol Sci ; 22(8)2021 Apr 20.
Article in English | MEDLINE | ID: mdl-33923988

ABSTRACT

Quercetin is a well-known plant flavonol and antioxidant; however, there has been some debate regarding the efficacy and safety of native quercetin as a skin-whitening agent via tyrosinase inhibition. Several researchers have synthesized quercetin derivatives as low-toxicity antioxidants and whitening agents. However, no suitable quercetin derivatives have been reported to date. In this study, a novel quercetin derivative was synthesized by the SN2 reaction using quercetin and oleyl bromide. The relationship between the structures and activities of quercetin derivatives as anti-melanogenic agents was assessed using in vitro enzyme kinetics, molecular docking, and quenching studies; cell line experiments; and in vivo zebrafish model studies. Novel 3,7-dioleylquercetin (OQ) exhibited a low cytotoxic concentration level at >100 µg/mL (125 µM), which is five times less toxic than native quercetin. The inhibition mechanism showed that OQ is a competitive inhibitor, similar to native quercetin. Expression of tyrosinase, tyrosinase-related protein 1 (TRP-1) and tyrosinase-related protein 2 (TRP-2), and microphthalmia-associated transcription factor was inhibited in B16F10 melanoma cell lines. mRNA transcription levels of tyrosinase, TRP-1, and TRP-2 decreased in a dose-dependent manner. Melanin formation was confirmed in the zebrafish model using quercetin derivatives. Therefore, OQ might be a valuable asset for the development of novel skin-whitening agents.


Subject(s)
Antineoplastic Agents/pharmacology , Quercetin/chemistry , Animals , Cell Line, Tumor , Humans , Kinetics , Melanins/chemistry , Molecular Docking Simulation , RNA, Messenger/metabolism , Zebrafish
5.
Molecules ; 26(20)2021 Oct 14.
Article in English | MEDLINE | ID: mdl-34684783

ABSTRACT

Signal transducer and activator of transcription 3 (STAT3) plays a critical role in the formation and growth of human cancer. Therefore, STAT3 is a therapeutic target for cancer drug discovery. Acacetin, a flavone present in various plants, inhibits constitutive and inducible STAT3 activation in STAT3-activated DU145 prostate cancer cells. Acacetin inhibits STAT3 activity by directly binding to STAT3, which we confirmed by a pull-down assay with a biotinylated compound and two level-free methods, namely, a drug affinity responsive target stability (DARTS) experiment and a cellular thermal shift assay (CETSA). Acacetin inhibits STAT3 phosphorylation at the tyrosine 705 residue and nuclear translocation in DU145 cells, which leads to the downregulation of STAT3 target genes. Acacetin then induces apoptosis in a time-dependent manner. Interestingly, acacetin induces the production of reactive oxygen species (ROS) that are not involved in the acacetin-induced inhibition of STAT3 activation because the suppressed p-STAT3 level is not rescued by treatment with GSH or NAC, which are general ROS inhibitors. We also found that acacetin inhibits tumor growth in xenografted nude mice. These results suggest that acacetin, as a STAT3 inhibitor, could be a possible drug candidate for targeting STAT3 for the treatment of cancer in humans.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Flavones/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Molecular , Phosphorylation/drug effects , Prostatic Neoplasms/pathology , Protein Binding , Protein-Tyrosine Kinases/metabolism , Reactive Oxygen Species/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Xenograft Model Antitumor Assays
6.
Nature ; 511(7511): 616-20, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25043025

ABSTRACT

Tumour oncogenes include transcription factors that co-opt the general transcriptional machinery to sustain the oncogenic state, but direct pharmacological inhibition of transcription factors has so far proven difficult. However, the transcriptional machinery contains various enzymatic cofactors that can be targeted for the development of new therapeutic candidates, including cyclin-dependent kinases (CDKs). Here we present the discovery and characterization of a covalent CDK7 inhibitor, THZ1, which has the unprecedented ability to target a remote cysteine residue located outside of the canonical kinase domain, providing an unanticipated means of achieving selectivity for CDK7. Cancer cell-line profiling indicates that a subset of cancer cell lines, including human T-cell acute lymphoblastic leukaemia (T-ALL), have exceptional sensitivity to THZ1. Genome-wide analysis in Jurkat T-ALL cells shows that THZ1 disproportionally affects transcription of RUNX1 and suggests that sensitivity to THZ1 may be due to vulnerability conferred by the RUNX1 super-enhancer and the key role of RUNX1 in the core transcriptional regulatory circuitry of these tumour cells. Pharmacological modulation of CDK7 kinase activity may thus provide an approach to identify and treat tumour types that are dependent on transcription for maintenance of the oncogenic state.


Subject(s)
Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Phenylenediamines/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Pyrimidines/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Core Binding Factor Alpha 2 Subunit/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Cysteine/metabolism , Humans , Jurkat Cells , Phosphorylation/drug effects
7.
Bioorg Med Chem Lett ; 29(15): 1985-1993, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31175010

ABSTRACT

The TAIRE family of kinases are an understudied branch of the CDK kinase family, that have been implicated in a number of cancers. This manuscript describes the design, synthesis and SAR of covalent CDK14 inhibitors, culminating in identification of FMF-04-159-2, a potent, covalent CDK14 inhibitor with a TAIRE kinase biased selectivity profile.


Subject(s)
Cyclin-Dependent Kinases/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/chemistry , Cyclin-Dependent Kinases/pharmacology , Humans , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship
8.
Int J Mol Sci ; 19(11)2018 Nov 01.
Article in English | MEDLINE | ID: mdl-30388885

ABSTRACT

Osteoporosis is caused by an imbalance of osteoclast and osteoblast activities and it is characterized by enhanced osteoclast formation and function. Peptidyl-prolyl cis-trans isomerase never in mitosis A (NIMA)-interacting 1 (Pin1) is a key mediator of osteoclast cell-cell fusion via suppression of the dendritic cell-specific transmembrane protein (DC-STAMP). We found that N,N'-1,4-butanediylbis[3-(2-chlorophenyl)acrylamide] (BCPA) inhibited receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclastogenesis in a dose-dependent manner without cytotoxicity. In addition, BCPA attenuated the reduction of Pin1 protein during osteoclast differentiation without changing Pin1 mRNA levels. BCPA repressed the expression of osteoclast-related genes, such as DC-STAMP and osteoclast-associated receptor (OSCAR), without altering the mRNA expression of nuclear factor of activated T cells (NFATc1) and cellular oncogene fos (c-Fos). Furthermore, Tartrate-resistant acid phosphatase (TRAP)-positive mononuclear cells were significantly decreased by BCPA treatment compared to treatment with the Pin1 inhibitor juglone. These data suggest that BCPA can inhibit osteoclastogenesis by regulating the expression of the DC-STAMP osteoclast fusion protein by attenuating Pin1 reduction. Therefore, BCPA may be used to treat osteoporosis.


Subject(s)
Acrylamides/toxicity , Butanes/toxicity , Cell Differentiation , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Osteoclasts/cytology , Osteoclasts/enzymology , Acrylamides/chemistry , Animals , Butanes/chemistry , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Line , Computer Simulation , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred C57BL , NIMA-Interacting Peptidylprolyl Isomerase/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Osteoclasts/drug effects , Osteogenesis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
9.
Int J Mol Sci ; 19(7)2018 Jun 22.
Article in English | MEDLINE | ID: mdl-29932110

ABSTRACT

Interleukin-1β (IL-1β) is a prominent pro-inflammatory cytokine that is implicated in a variety of autoimmune diseases and plays an important role in host defense against infections. IL-1β activity increases with its increasing binding capacity to IL-1 receptors (IL-1Rs). Thus, numerous studies have targeted the discovery of molecules modulating the interactions between IL-1β and IL-1R1. We have conducted an IL-1R1 structure-based virtual screening to identify small molecules that could alter IL-1β activity, using in silico computational analysis. Sixty compounds from commercial libraries were predicted to bind to IL-1R1, and their influence on cytokine production in IL-1β-stimulated gingival fibroblasts (GFs) was determined. Of these, only (2-(1,2-diphenyl-1H-indol-3-yl)ethanamine (DPIE) showed a synergistic increase in inflammatory molecules and cytokine production (IL-6, IL-8, and COX-2) at both mRNA and protein levels in IL-1β-stimulated GFs. The enhancing activity of DPIE in IL-1β-induced cytokine production increased in a dose-dependent manner without cytotoxicity. This pattern was also observed in IL-1β-stimulated primary human periodontal ligament cells (PDLs). Furthermore, we measured the impact of DPIE on the IL-1β⁻IL-1R1 system using surface plasmon resonance and demonstrated that DPIE increased the binding affinity of IL-1β to IL-1R1. These data indicate that DPIE boosts IL-1β signaling by enhancing the binding of IL-1β to IL-1R1 in oral primary cells.


Subject(s)
Amines/pharmacology , Cytokines/metabolism , Fibroblasts/drug effects , Inflammation Mediators/metabolism , Interleukin-1beta/pharmacology , Small Molecule Libraries/pharmacology , Amines/chemistry , Cell Survival/drug effects , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Gingiva/cytology , Humans , Molecular Structure , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Periodontal Ligament/metabolism , Protein Binding/drug effects , Receptors, Interleukin-1 Type I/metabolism , Small Molecule Libraries/chemistry , Surface Plasmon Resonance
10.
Molecules ; 23(12)2018 Nov 29.
Article in English | MEDLINE | ID: mdl-30501117

ABSTRACT

Osteoclasts are large multinucleated cells which are induced by the regulation of the receptor activator of nuclear factor kappa-Β ligand (RANKL), which is important in bone resorption. Excessive osteoclast differentiation can cause pathologic bone loss and destruction. Numerous studies have targeted molecules inhibiting RANKL signaling or bone resorption activity. In this study, 11 compounds from commercial libraries were examined for their effect on RANKL-induced osteoclast differentiation. Of these compounds, only 2-(3-(2-fluoro-4-methoxyphenyl)-6-oxo-1(6H)-pyridazinyl)-N-1H-indol-5-ylacetamide (2N1HIA) caused a significant decrease in multinucleated tartrate-resistant acid phosphatase (TRAP)-positive cell formation in a dose-dependent manner, without inducing cytotoxicity. The 2N1HIA compound neither affected the expression of osteoclast-specific gene markers such as TRAF6, NFATc1, RANK, OC-STAMP, and DC-STAMP, nor the RANKL signaling pathways, including p38, ERK, JNK, and NF-κB. However, 2N1HIA exhibited a significant impact on the expression levels of CD47 and cathepsin K, the early fusion marker and critical protease for bone resorption, respectively. The activity of matrix metalloprotease-9 (MMP-9) decreased due to 2N1HIA treatment. Accordingly, bone resorption activity and actin ring formation decreased in the presence of 2N1HIA. Taken together, 2N1HIA acts as an inhibitor of osteoclast differentiation by attenuating bone resorption activity and may serve as a potential candidate in preventing and/or treating osteoporosis, or other bone diseases associated with excessive bone resorption.


Subject(s)
Acetamides/pharmacology , Cathepsin K/metabolism , Cell Differentiation/drug effects , Osteoclasts/cytology , Osteoclasts/enzymology , Actins/metabolism , Animals , Bone Resorption/genetics , Bone Resorption/pathology , CD47 Antigen/metabolism , Cattle , Gene Expression Regulation/drug effects , Male , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteogenesis/drug effects , RANK Ligand/pharmacology
11.
Protein Expr Purif ; 131: 34-41, 2017 03.
Article in English | MEDLINE | ID: mdl-27838376

ABSTRACT

Many pesticides and chemical warfare nerve agents are highly toxic organophosphorus compounds (OPs), which inhibit acetylcholinesterase activity. Human paraoxonase 1 (PON1) has demonstrated significant potential for use as a catalytic bioscavenger capable of hydrolyzing a broad range of OPs. However, there are several limitations to the use of human PON1 as a catalytic bioscavenger, including the relatively difficult purification of PON1 from human plasma and its dependence on the presence of hydrophobic binding partners to maintain stability. Therefore, research efforts to efficiently produce recombinant human PON1 are necessary. In this study, we developed a Drosophila S2 stable cell line expressing recombinant human PON1. The recombinant human PON1 was fused with the human immunoglobulin Fc domain (PON1-hFc) to improve protein stability and purification efficiency. We purified the recombinant human PON1-hFc from the S2 stable cell line and characterized its enzymatic properties for OP hydrolysis. We purified the recombinant human PON1-hFc from the S2 stable cell line and characterized its enzymatic properties for OP hydrolysis compared with those of the recombinant human PON1 derived from E. coli. We observed that the recombinant human PON1-hFc is functionally more stable for OP hydrolyzing activities compared to the recombinant human PON1. The catalytic efficiency of the recombinant PON1-hFc towards diisopropyl fluorophosphate (DFP, 0.26 × 106 M-1 min-1) and paraoxon hydrolysis (0.015 × 106 M-1 min-1) was 1.63- and 1.24-fold higher, respectively, than the recombinant human PON1. Thus, we report that the recombinant PON1-hFc exerts hydrolytic activity against paraoxon and DFP.


Subject(s)
Aryldialkylphosphatase , Gene Expression , Immunoglobulin Fc Fragments , Recombinant Fusion Proteins , Animals , Aryldialkylphosphatase/biosynthesis , Aryldialkylphosphatase/chemistry , Aryldialkylphosphatase/genetics , Aryldialkylphosphatase/isolation & purification , Cell Line , Drosophila melanogaster , Humans , Immunoglobulin Fc Fragments/biosynthesis , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/genetics , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification
12.
Biochim Biophys Acta Gen Subj ; 1861(4): 947-957, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28161478

ABSTRACT

BACKGROUND: Transient receptor potential melastatin 7 (TRPM7) regulates breast cancer cell proliferation, migration, invasion and metastasis in its ion channel- and kinase domain-dependent manner. The pharmacological effects of TRPM7 ion channel inhibitors on breast cancer cells have been studied, but little is known about the effects of TRPM7 kinase domain inhibitors due to lack of potent TRPM7 kinase inhibitors. METHODS: Screening was performed by using TRPM7 kinase assay. Effects of TG100-115 on breast cancer cell proliferation, migration, invasion, myosin IIA phosphorylation, and TRPM7 ion channel activity were assessed by using MTT, wound healing, transwell assay, Western blotting, and patch clamping, respectively. RESULTS: We found that CREB peptide is a potent substrate for the TR-FRET based TRPM7 kinase assay. Using this method, we discovered a new and potent TRPM7 kinase inhibitor, TG100-115. TG100-115 inhibited TRPM7 kinase activity in an ATP competitive fashion with over 70-fold stronger activity than that of rottlerin, known as a TRPM7 kinase inhibitor. TG100-115 has little effect on proliferation of MDA-MB-231 cells, but significantly decreases cell migration and invasion. Moreover, TG100-115 inhibits TRPM7 kinase regulated phosphorylation of the myosin IIA heavy chain and phosphorylation of focal adhesion kinase. TG100-115 also suppressed TRPM7 ion channel activity. CONCLUSIONS: TG100-115 can be used as a potent TRPM7 kinase inhibitor and a potent inhibitor of breast cancer cell migration. GENERAL SIGNIFICANCE: TG100-115 could be a useful tool for studying the pharmacological effects of TRPM7 kinase activity aimed at providing insight into new therapeutic approaches to the treatment of breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Cell Movement/drug effects , Neoplasm Invasiveness/pathology , Phenols/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pteridines/pharmacology , TRPM Cation Channels/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Nonmuscle Myosin Type IIA/metabolism , Phosphorylation/drug effects
13.
Bioorg Med Chem Lett ; 27(18): 4405-4408, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28818446

ABSTRACT

Aurora kinases play an essential role in mitosis and cell cycle regulation. In recent years Aurora kinases have proved popular cancer targets and many inhibitors have been developed. The majority of these clinical candidates are multi-targeted, rendering them inappropriate as tools for studying Aurora kinase mediated signaling. Here we report discovery of a highly selective inhibitor of Aurora kinases A, B and C, with potent cellular activity and minimal off-target activity (PLK4). The X-ray co-crystal structure of Aurora A in complex with compound 2 is reported, and provides insights into the structural determinants of ligand binding and selectivity.


Subject(s)
Antineoplastic Agents/pharmacology , Aurora Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Aurora Kinases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
14.
Int J Mol Sci ; 18(7)2017 Jul 09.
Article in English | MEDLINE | ID: mdl-28698492

ABSTRACT

The structural information of small therapeutic compounds complexed in biological matrices is important for drug developments. However, structural studies on ligands bound to such a large and dynamic system as microtubules are still challenging. This article reports an application of the solid-state NMR technique to investigating the bioactive conformation of epothilone B, a microtubule stabilizing agent, whose analog ixabepilone was approved by the U.S. Food and Drug Administration (FDA) as an anticancer drug. First, an analog of epothilone B was designed and successfully synthesized with deuterium and fluorine labels while keeping the high potency of the drug; Second, a lyophilization protocol was developed to enhance the low sensitivity of solid-state NMR; Third, molecular dynamics information of microtubule-bound epothilone B was revealed by high-resolution NMR spectra in comparison to the non-bound epothilone B; Last, information for the macrolide conformation of microtubule-bound epothilone B was obtained from rotational-echo double-resonance (REDOR) NMR data, suggesting the X-ray crystal structure of the ligand in the P450epoK complex as a possible candidate for the conformation. Our results are important as the first demonstration of using REDOR for studying epothilones.


Subject(s)
Epothilones/chemistry , Magnetic Resonance Spectroscopy/methods , Crystallography, X-Ray , Epothilones/metabolism , Microtubules/metabolism , Molecular Conformation , United States , United States Food and Drug Administration
15.
Chemotherapy ; 61(3): 159-66, 2016.
Article in English | MEDLINE | ID: mdl-26821172

ABSTRACT

BACKGROUND: The zoonotic transmission of highly pathogenic avian influenza viruses and the global pandemic of H1N1 influenza in 2009 signified the need for a wider coverage of therapeutic options for the control of influenza. METHODS: An in-house compound library was screened using a cytopathic effect inhibition assay. Selected hits were then tested in vivo and used as a core skeleton for derivative synthesis. RESULTS: The hit compound (BMD-2601505) was effective [50% effective concentration (EC50) of 60-70 µM] in reducing the death rate of cells infected with human influenza A and B viruses as well as avian influenza A virus. Furthermore, BMD-2601505 reduced the weight loss and increased the survival after lethal infection. The compound was further modified to enhance its antiviral potency. Results show that one derivative with bromobenzene moiety was most effective (EC50 of 22-37 µM) against the influenza viruses tested. CONCLUSION: We identified a small benzamide compound exhibiting antiviral activity against influenza viruses. The results warrant further evaluation of antiviral activities against drug-resistant influenza isolates.


Subject(s)
Antiviral Agents/pharmacology , Benzamides/pharmacology , Influenza A virus/drug effects , Influenza B virus/drug effects , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Benzamides/administration & dosage , Benzamides/chemistry , Bromobenzenes/administration & dosage , Bromobenzenes/chemistry , Bromobenzenes/pharmacology , Dogs , Female , Humans , Influenza, Human/drug therapy , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacology , Small Molecule Libraries , Zoonoses/drug therapy , Zoonoses/virology
16.
Molecules ; 21(1): 80, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26771593

ABSTRACT

GSK5182 (4) is currently one of the lead compounds for the development of estrogen-related receptor gamma (ERRγ) inverse agonists. Here, we report the design, synthesis, pharmacological and in vitro absorption, distribution, metabolism, excretion, toxicity (ADMET) properties of a series of compounds related to 4. Starting from 4, a series of analogs were structurally modified and their ERRγ inverse agonist activity was measured. A key pharmacophore feature of this novel class of ligands is the introduction of a heterocyclic group for A-ring substitution in the core scaffold. Among the tested compounds, several of them are potent ERRγ inverse agonists as determined by binding and functional assays. The most promising compound, 15g, had excellent binding selectivity over related subtypes (IC50 = 0.44, >10, >10, and 10 µM at the ERRγ, ERRα, ERRß, and ERα subtypes, respectively). Compound 15g also resulted in 95% transcriptional repression at a concentration of 10 µM, while still maintaining an acceptable in vitro ADMET profile. This novel class of ERRγ inverse agonists shows promise in the development of drugs targeting ERRγ-related diseases.


Subject(s)
Estrogens/pharmacology , Receptors, Estrogen/metabolism , Small Molecule Libraries/pharmacology , Tamoxifen/analogs & derivatives , Animals , Binding Sites , Cell Line , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Drug Stability , ERG1 Potassium Channel , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Estrogens/chemical synthesis , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/genetics , Ether-A-Go-Go Potassium Channels/metabolism , Gene Expression , High-Throughput Screening Assays , Humans , Ligands , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Molecular Docking Simulation , Protein Binding , Rats , Receptors, Estrogen/chemistry , Receptors, Estrogen/genetics , Small Molecule Libraries/chemical synthesis , Structure-Activity Relationship , Tamoxifen/chemistry , Tamoxifen/pharmacology , Thermodynamics
17.
J Am Chem Soc ; 137(13): 4358-67, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25785725

ABSTRACT

The mitochondrial pool of Hsp90 and its mitochondrial paralogue, TRAP1, suppresses cell death and reprograms energy metabolism in cancer cells; therefore, Hsp90 and TRAP1 have been suggested as target proteins for anticancer drug development. Here, we report that the actual target protein in cancer cell mitochondria is TRAP1, and current Hsp90 inhibitors cannot effectively inactivate TRAP1 because of their insufficient accumulation in the mitochondria. To develop mitochondrial TRAP1 inhibitors, we determined the crystal structures of human TRAP1 complexed with Hsp90 inhibitors. The isopropyl amine of the Hsp90 inhibitor PU-H71 was replaced with the mitochondria-targeting moiety triphenylphosphonium to produce SMTIN-P01. SMTIN-P01 showed a different mode of action from the nontargeted PU-H71, as well as much improved cytotoxicity to cancer cells. In addition, we determined the structure of a TRAP1-adenylyl-imidodiphosphate (AMP-PNP) complex. On the basis of comparative analysis of TRAP1 structures, we propose a molecular mechanism of ATP hydrolysis that is crucial for chaperone function.


Subject(s)
Benzodioxoles/chemistry , Benzodioxoles/pharmacology , Drug Design , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/chemistry , Mitochondria/drug effects , Purines/chemistry , Purines/pharmacology , Amines/chemistry , Cell Line, Tumor , Crystallography, X-Ray , Humans , Mitochondria/metabolism , Models, Molecular , Organophosphorus Compounds/chemistry , Protein Multimerization , Protein Stability , Protein Structure, Quaternary
18.
Biopharm Drug Dispos ; 36(3): 163-73, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25451157

ABSTRACT

GSK5182 (4-[(Z)-1-[4-(2-dimethylaminoethyloxy)phenyl]-hydroxy-2-phenylpent-1-enyl]phenol) is a specific inverse agonist for estrogen-related receptor γ, a member of the orphan nuclear receptor family that has important functions in development and homeostasis. This study was performed to elucidate the metabolites of GSK5182 and to characterize the enzymes involved in its metabolism. Incubation of human liver microsomes with GSK5182 in the presence of NADPH resulted in the formation of three metabolites, M1, M2 and M3. M1 and M3 were identified as N-desmethyl-GSK5182 and GSK5182 N-oxide, respectively, on the basis of liquid chromatography-tandem mass spectrometric (LC-MS/MS) analysis. M2 was suggested to be hydroxy-GSK5182 through interpretation of its MS/MS fragmentation pattern. In addition, the specific cytochrome P450 (P450) and flavin-containing monooxygenase (FMO) isoforms responsible for GSK5182 oxidation to the three metabolites were identified using a combination of correlation analysis, chemical inhibition in human liver microsomes and metabolism by expressed recombinant P450 and FMO isoforms. GSK5182 N-demethylation and hydroxylation is mainly mediated by CYP3A4, whereas FMO1 and FMO3 contribute to the formation of GSK5182 N-oxide from GSK5182. The present data will be useful for understanding the pharmacokinetics and drug interactions of GSK5182 in vivo.


Subject(s)
Estrogens/pharmacology , Hypoglycemic Agents/pharmacology , Microsomes, Liver/metabolism , Tamoxifen/analogs & derivatives , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Humans , Receptors, Estrogen/metabolism , Recombinant Proteins/metabolism , Tamoxifen/pharmacology
19.
Angew Chem Int Ed Engl ; 53(1): 199-204, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24259466

ABSTRACT

We report the synthesis of a GDP analogue, SML-8-73-1, and a prodrug derivative, SML-10-70-1, which are selective, direct-acting covalent inhibitors of the K-Ras G12C mutant relative to wild-type Ras. Biochemical and biophysical measurements suggest that modification of K-Ras with SML-8-73-1 renders the protein in an inactive state. These first-in-class covalent K-Ras inhibitors demonstrate that irreversible targeting of the K-Ras guanine-nucleotide binding site is potentially a viable therapeutic strategy for inhibition of Ras signaling.


Subject(s)
Catalytic Domain/genetics , ras Proteins/chemistry , ras Proteins/genetics , Drug Design , Signal Transduction , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL