Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Immunol Immunother ; 72(7): 2473-2482, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37017695

RESUMEN

The signal transducer and activator of transcription 3 (STAT3) signaling pathway is a key mediator of cancer cell proliferation, survival, and invasion. We discovered YHO-1701 as a small molecule inhibitor of STAT3 dimerization and demonstrated its potent anti-tumor activity using xenograft mouse models as monotherapy and combination therapy with molecular targeted drugs. STAT3 is also associated with cancer immune tolerance; therefore, we used the female CT26 syngeneic mouse model to examine the effect of combining YHO-1701 administration with PD-1/PD-L1 blockade. Pretreatment of the mice with YHO-1701 before starting anti-PD-1 antibody administration resulted in a significant therapeutic effect. In addition, the effect of monotherapy and combination treatment with YHO-1701 was significantly abolished by depleting natural killer (NK) cell activity. YHO-1701 was also found to restore the activity of mouse NK cells under inhibitory conditions in vitro. Furthermore, this combination therapy significantly inhibited tumor growth in an immunotherapy-resistant model of murine CMS5a fibrosarcoma. These results suggest that the combination of YHO-1701 with PD-1/PD-L1 blockade might be a new candidate for cancer immunotherapy involving the enhancement of NK cell activity in the tumor microenvironment.


Asunto(s)
Anticuerpos , Fibrosarcoma , Células Asesinas Naturales , Receptor de Muerte Celular Programada 1 , Quinolinas , Animales , Ratones , Ratones Endogámicos BALB C , Fibrosarcoma/tratamiento farmacológico , Células Asesinas Naturales/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Quinolinas/administración & dosificación , Anticuerpos/administración & dosificación , Trasplante Isogénico
2.
FEBS Lett ; 597(8): 1138-1148, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36823439

RESUMEN

Centromere-associated protein E (CENP-E) is a kinesin motor protein essential for mitosis and a new target for anticancer agents with less side effects. To rationally design anticancer drug candidates based on structure, it is important to determine the three-dimensional structure of the CENP-E motor domain bound to its inhibitor. Here, we report the first crystal structure of the CENP-E motor domain in complex with a non-hydrolysable ATP analogue, adenylyl-imidodiphosphate (AMPPNP). Furthermore, the structure is compared with the ADP-bound form of the CENP-E motor domain as well as the AMPPNP-bound forms of other kinesins. This study indicates that helix α4 of CENP-E participates in the slow binding of CENP-E to microtubules. These results will contribute to the development of anticancer drugs targeting CENP-E.


Asunto(s)
Antineoplásicos , Microtúbulos , Adenilil Imidodifosfato/análisis , Adenilil Imidodifosfato/metabolismo , Microtúbulos/metabolismo , Mitosis , Antineoplásicos/farmacología , Centrómero/metabolismo
3.
Int J Tryptophan Res ; 15: 11786469221138456, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36467776

RESUMEN

Kynurenine (Kyn), a metabolite of tryptophan (Trp), is a key regulator of mammal immune responses such as cancer immune tolerance. Indoleamine-2,3-dioxygenase (IDO) and tryptophan-2,3-dioxygenase (TDO) are main enzymes regulating the first and rate-limiting step of the Kyn pathway. To identify new small molecule inhibitors of TDO, we selected A172 glioblastoma cell line constitutively expressed TDO. Characterization of this cell line using kinase inhibitor library resulted in identification of MEK/ERK pathway-dependent TDO expression. After knowing the properties for TDO expression, we further proceeded to screen chemical library for TDO inhibitors. We previously determined that S-benzylisothiourea derivatives are enzymatic inhibitors of indoleamine 2,3-dioxygenase 1 (IDO1) and suggested that the isothiourea moiety could be an important pharmacophore for binding to heme. Based on this premise, we screened an in-house library composed of various isothiourea derivatives and identified a bisisothiourea derivative, PVZB3001, as an inhibitor of TDO. Interestingly, PVZB3001 also inhibited the enzymatic activity of IDO1 in both cell-based and cell-free assays but did not inhibit other heme enzymes. Molecular docking studies suggested the importance of isothiourea moieties at the ortho position of the phenyl ring for the inhibition of catalytic activity. PVZB3001 showed competitive inhibition against TDO, and this was supported by the docking simulation. PVZB3001 recovered natural killer (NK) cell viability and functions by inhibiting Kyn accumulation in conditioned medium of both IDO1- and TDO-expressing cells. Furthermore, oral administration of IDO1-overexpressing tumor-bearing mice with PVZB3001 significantly inhibited tumor growth. Thus, we identified a novel selective dual inhibitor of IDO1 and TDO using the Kyn production assay with a glioblastoma cell line. This inhibitor could be a useful pharmacological tool for modulating the Kyn pathway in a variety of experimental systems.

4.
Front Cell Dev Biol ; 10: 884509, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35620056

RESUMEN

Dynamin is an endocytic protein that functions in vesicle formation by scission of invaginated membranes. Dynamin maintains the structure of foot processes in glomerular podocytes by directly and indirectly interacting with actin filaments. However, molecular mechanisms underlying dynamin-mediated actin regulation are largely unknown. Here, biochemical and cell biological experiments were conducted to uncover how dynamin modulates interactions between membranes and actin in human podocytes. Actin-bundling, membrane tubulating, and GTPase activities of dynamin were examined in vitro using recombinant dynamin 2-wild-type (WT) or dynamin 2-K562E, which is a mutant found in Charcot-Marie-Tooth patients. Dynamin 2-WT and dynamin 2-K562E led to the formation of prominent actin bundles with constant diameters. Whereas liposomes incubated with dynamin 2-WT resulted in tubule formation, dynamin 2-K562E reduced tubulation. Actin filaments and liposomes stimulated dynamin 2-WT GTPase activity by 6- and 20-fold, respectively. Actin-filaments, but not liposomes, stimulated dynamin 2-K562E GTPase activity by 4-fold. Self-assembly-dependent GTPase activity of dynamin 2-K562E was reduced to one-third compared to that of dynamin 2-WT. Incubation of liposomes and actin with dynamin 2-WT led to the formation of thick actin bundles, which often bound to liposomes. The interaction between lipid membranes and actin bundles by dynamin 2-K562E was lower than that by dynamin 2-WT. Dynamin 2-WT partially colocalized with stress fibers and actin bundles based on double immunofluorescence of human podocytes. Dynamin 2-K562E expression resulted in decreased stress fiber density and the formation of aberrant actin clusters. Dynamin 2-K562E colocalized with α-actinin-4 in aberrant actin clusters. Reformation of stress fibers after cytochalasin D-induced actin depolymerization and washout was less effective in dynamin 2-K562E-expressing cells than that in dynamin 2-WT. Bis-T-23, a dynamin self-assembly enhancer, was unable to rescue the decreased focal adhesion numbers and reduced stress fiber density induced by dynamin 2-K562E expression. These results suggest that the low affinity of the K562E mutant for lipid membranes, and atypical self-assembling properties, lead to actin disorganization in HPCs. Moreover, lipid-binding and self-assembly of dynamin 2 along actin filaments are required for podocyte morphology and functions. Finally, dynamin 2-mediated interactions between actin and membranes are critical for actin bundle formation in HPCs.

5.
NPJ Precis Oncol ; 6(1): 11, 2022 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-35228642

RESUMEN

Patients with advanced anaplastic lymphoma kinase (ALK)-rearranged non-small cell lung cancer who are prescribed ALK-tyrosine kinase inhibitors (ALK-TKIs) rarely have complete responses, with residual tumors relapsing as heterogeneous resistant phenotypes. Herein, we investigated new therapeutic strategies to reduce and eliminate residual tumors in the early treatment phase. Functional genomic screening using small guide RNA libraries showed that treatment-induced adaptive survival of ALK-rearranged lung cancer cells was predominantly dependent on STAT3 activity upon ALK inhibition. STAT3 inhibition effectively suppressed the adaptive survival of ALK-rearranged lung cancer cells by enhancing ALK inhibition-induced apoptosis. The combined effects were characterized by treatment-induced STAT3 dependence and transcriptional regulation of anti-apoptotic factor BCL-XL. In xenograft study, the combination of YHO-1701 (STAT3 inhibitor) and alectinib significantly suppressed tumor regrowth after treatment cessation with near tumor remission compared with alectinib alone. Hence, this study provides new insights into combined therapeutic strategies for patients with ALK-rearranged lung cancer.

6.
Exp Cell Res ; 413(2): 113079, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35202674

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) plays key roles in cancer cell proliferation, invasion, and immunosuppression. In many human cancer cells, STAT3 is hyperactivated, which leads to tumor progression and drug resistance, and therefore STAT3 and its modulators are considered effective drug targets. However, the complex regulatory mechanisms of STAT3 have made it difficult to develop potent anticancer drugs that suppress its activity. Here, we report serum and glucocorticoid-regulated kinase 1 (SGK1) as a novel regulator of STAT3 signaling and an effective target for combination therapy with Janus kinase (JAK) inhibitors. We screened small molecules using a gain-of-function mutant of STAT3 resistant to JAK inhibition and found that an SGK1 inhibitor suppressed the constitutive activation of STAT3. Importantly, our results revealed that SGK1 also mediated the activation of wild-type STAT3. Further examination suggested that the tuberous sclerosis complex 2 and mammalian target of rapamycin signaling pathway were involved in STAT3 activation by SGK1. Finally, we demonstrated that SGK1 inhibition enhanced the inhibitory effect of a JAK inhibitor on STAT3 phosphorylation and cancer cell proliferation. Our findings provide new insights into the molecular mechanisms of STAT3 activation and suggest SGK1 as a potential target for STAT3-targeted combination cancer therapy.


Asunto(s)
Proteínas Inmediatas-Precoces , Neoplasias , Proteínas Serina-Treonina Quinasas , Factor de Transcripción STAT3 , Línea Celular Tumoral , Humanos , Proteínas Inmediatas-Precoces/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
7.
J Immunother Cancer ; 10(2)2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35115364

RESUMEN

BACKGROUND: Cancer immunotherapy shows insufficient efficacy for low immunogenic tumors. Furthermore, tumors often downregulate antigen and major histocompatibility complex expression to escape recognition by T cells, resulting in insufficient T cell receptor (TCR) stimulation in the tumor microenvironment. Thus, augmenting TCR-mediated recognition of tumor antigens is a useful strategy to improve the efficacy of cancer immunotherapy. METHODS: We screened 310 small molecules from our library and identified PQDN, a small molecule that activates CD8 T cells after TCR engagement, even when antigen stimulation is too weak for their activation. We used inhibitors of mitochondrial functions and Seahorse Flux Analyzer to investigate the mechanism underlying the effect of PQDN on T cells. Effect of PQDN on tumor-infiltrating CD8 T cells was examined using flow cytometry and TCR repertoire analysis. RESULTS: PQDN increased mitochondrial reciprocal capacity through enhancement of electron transport chains (ETCs) and facilitated glycolysis via mTOR/AKT signaling, resulting in augmented CD8 T cell activation, even when antigen stimulation is extremely weak. Intratumoral administration of this compound into tumor-bearing mice tunes inactivated T cell with tumor antigen recognition potent and expanded functional T cell receptor diversity of tumor-infiltrating T cells, augmenting antitumor immune responses and retarding tumor growth. Furthermore, PQDN has a synergistic potent with T cell dependent immunotherapy, such as checkpoint inhibitory therapy or adoptive cell therapy, even in a low immunogenic tumor. We also demonstrated that this compound enhances the activation of human CD8 T cells. CONCLUSIONS: These data suggest that tuning the T cell activation threshold by chemical activation of mitochondrial ETC is a new strategy for improving therapeutic efficacy through the activation of low-avidity tumor-specific T cells.


Asunto(s)
Transporte de Electrón/inmunología , Inmunoterapia/métodos , Redes y Vías Metabólicas/inmunología , Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Microambiente Tumoral
8.
Biochem Pharmacol ; 193: 114789, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34582773

RESUMEN

Kinesin-5 has received considerable attention as a new target for mitosis. Various small-molecule compounds targeting kinesin-5 have been developed in the last few decades. However, the differences in the cellular effects of kinesin-5 inhibitors remain poorly understood. Here, we used two different kinesin-5 inhibitors, biphenyl-type PVZB1194 and S-trityl-L-cysteine-type PVEI0021, to examine their effects on molecular events involving kinesin-5. Our biochemical study of kinesin-5 protein-protein interactions showed that PVZB1194-treated kinesin-5 interacted with TPX2 microtubule nucleation factor, Aurora-A kinase, receptor for hyaluronan-mediated motility, and γ-tubulin, as did untreated mitotic kinesin-5. However, PVEI0021 prevented kinesin-5 from binding to these proteins. In mitotic HeLa cells recovered from nocodazole inhibition, kinesin-5 colocalized with these binding proteins, along with microtubules nucleated near kinetochores. By acting on kinesin-5 interactions with chromatin-associated microtubules, PVZB1194, rather than PVEI0021, not only affected the formation of dispersed microtubule clusters but also enhanced the stability of microtubules. In addition, screening for mitotic inhibitors working synergistically with the kinesin-5 inhibitors revealed that paclitaxel synergistically inhibited HeLa cell proliferation only with PVZB1194. In contrast, the Aurora-A inhibitor MLN8237 exerted a synergistic anti-cell proliferation effect when combined with either inhibitor. Together, these results have provided a better understanding of the molecular action of kinesin-5 inhibitors and indicate their usefulness as molecular tools for the study of mitosis and the development of anticancer agents.


Asunto(s)
Antineoplásicos/farmacología , Compuestos de Bifenilo/farmacología , Cinesinas/antagonistas & inhibidores , Sulfonamidas/farmacología , Azepinas/farmacología , Proliferación Celular/efectos de los fármacos , Células HeLa , Humanos , Estructura Molecular , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología
9.
Acta Crystallogr D Struct Biol ; 77(Pt 3): 280-287, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33645531

RESUMEN

Centromere-associated protein E (CENP-E) plays an essential role in mitosis and is a target candidate for anticancer drugs. However, it is difficult to design small-molecule inhibitors of CENP-E kinesin motor ATPase activity owing to a lack of structural information on the CENP-E motor domain in complex with its inhibitors. Here, the CENP-E motor domain was crystallized in the presence of an ATP-competitive inhibitor and the crystal structure was determined at 1.9 Šresolution. In the determined structure, ADP was observed instead of the inhibitor in the nucleotide-binding site, even though no ADP was added during protein preparation. Structural comparison with the structures of previously reported CENP-E and those of other kinesins indicates that the determined structure is nearly identical except for several loop regions. However, the retention of ADP in the nucleotide-binding site of the structure strengthens the biochemical view that the release of ADP is a rate-limiting step in the ATPase cycle of CENP-E. These results will contribute to the development of anticancer drugs targeting CENP-E and to understanding the function of kinesin motor domains.


Asunto(s)
Proteínas Cromosómicas no Histona/química , Adenosina Difosfato/química , Sitios de Unión , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Dominios Proteicos
10.
Eur J Med Chem ; 215: 113288, 2021 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-33640763

RESUMEN

Kinesin spindle protein (KSP) is expressed only in cells undergoing cell division, and hence represents an attractive target for the treatment of cancer. Several KSP inhibitors have been developed and undergone clinical trial, but their clinical use is limited by their toxicity to rapidly proliferating non-cancerous cells. To create new KSP inhibitors that are highly selective for cancer cells, we optimized the amino acid moiety of S-trityl-l-cysteine (STLC) derivative 1 using in silico modeling. Molecular docking and molecular dynamics simulation were performed to investigate the binding mode of 1 with KSP. Consistent with the structure activity relationship studies, we found that a cysteine amino moiety plays an important role in stabilizing the interaction. Based on these findings and the structure of GSH, a substrate of γ-glutamyltransferase (GGT), we designed and synthesized the prodrug N-γ-glutamylated STLC derivative 9, which could be hydrolyzed by GGT to produce 1. The KSP ATPase inhibitory activity of 9 was lower than that of 1, and LC-MS analysis indicated that 9 was converted to 1 only in the presence of GGT in vitro. In addition, the cytotoxic activity of 9 was significantly attenuated in GGT-knockdown A549 cells. Since GGT is overexpressed on the cell membrane of various cancer cells, these results suggest that compound 9 could be a promising prodrug that selectively inhibits the proliferation of GGT-expressing cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Cisteína/farmacología , Dibenzocicloheptenos/farmacología , Cinesinas/antagonistas & inhibidores , Profármacos/farmacología , Compuestos de Tritilo/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Bovinos , Línea Celular Tumoral , Cisteína/síntesis química , Cisteína/metabolismo , Dibenzocicloheptenos/síntesis química , Dibenzocicloheptenos/metabolismo , Humanos , Cinesinas/metabolismo , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Estructura Molecular , Profármacos/síntesis química , Profármacos/metabolismo , Unión Proteica , Relación Estructura-Actividad , Termodinámica , Compuestos de Tritilo/síntesis química , Compuestos de Tritilo/metabolismo , gamma-Glutamiltransferasa/metabolismo
12.
Physiol Rep ; 8(20): e14627, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33112058

RESUMEN

Kidney fibrosis is a histological hallmark of chronic kidney disease (CKD) and is believed to be involved in the progression of CKD. Therefore, inhibition of kidney fibrosis is a potential strategy for slowing CKD progression. Signal transducer and activator of transcription 3 (STAT3) is a transcription factor that is activated by interleukin-6 and is reported to be involved in fibrosis. Previously, S3I-201, an inhibitor of STAT3 phosphorylation, was shown to inhibit renal fibrosis in a mouse model, but its mechanism was not clarified completely. In this study, we investigated whether STX-0119, a new inhibitor of STAT3 dimerization, suppressed kidney fibrotic gene expression using a mouse model of kidney fibrosis and examined the underlying mechanisms. Kidney fibrosis was induced by unilateral ureteral obstruction (UUO), which was accompanied by upregulation of STAT3 target genes. STX-0119 administration suppressed the expression of fibrotic genes in UUO kidneys without affecting STAT3 phosphorylation. STX-0119 decreased Cxcr4 mRNA in cultured rat kidney fibroblasts and Ccr1 mRNA in blood cells from UUO mice, both of which are reported to be involved in the progression of kidney fibrosis. These results suggest that STX-0119 inhibits fibrotic gene expression in kidney by suppressing Cxcr4 and Ccr1 expression. This is the first report to indicate a part of the mechanism of the antifibrotic effects of a STAT3 inhibitor and suggests that STX-0119 may be a lead compound for the treatment of kidney fibrosis.


Asunto(s)
Riñón/efectos de los fármacos , Oxadiazoles/farmacología , Quinolinas/farmacología , Receptores CCR1/genética , Receptores CXCR4/genética , Insuficiencia Renal Crónica/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores , Animales , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibrosis , Riñón/metabolismo , Riñón/patología , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , Ratas , Receptores CCR1/metabolismo , Receptores CXCR4/metabolismo , Factor de Transcripción STAT3/metabolismo
13.
Cancer Sci ; 111(5): 1774-1784, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32112605

RESUMEN

The signal transducer and activator of transcription 3 (STAT3) signaling pathway is a key mediator of cancer cell proliferation, survival and invasion. Aberrant STAT3 has been demonstrated in various malignant cancers. YHO-1701 is a novel quinolinecarboxamide derivative generated from STX-0119. Here, we examined the effect of YHO-1701 on STAT3 and evaluated antitumor activity of YHO-1701 as a single agent and in combination. YHO-1701 inhibited STAT3-SH2 binding to phospho-Tyr peptide selectively and more potently than STX-0119 in biochemical assays. Molecular docking studies with STAT3 suggested more stable interaction of YHO-1701 with the SH2 domain. YHO-1701 exhibited approximately 10-fold stronger activity than STX-0119 in abrogating the STAT3 signaling pathway of human oral cancer cell line SAS. YHO-1701 also blocked multi-step events by inhibiting STAT3 dimerization and suppressed STAT3 promoter activity. As expected, YHO-1701 exerted strong antiproliferative activity against human cancer cell lines addicted to STAT3 signaling. Orally administered YHO-1701 showed statistically significant antitumor effects with long exposure to high levels of YHO-1701 at tumor sites in SAS xenograft models. Moreover, combination regimen with sorafenib led to significantly stronger antitumor activity. In addition, the suppression level of survivin (a downstream target) was superior for the combination as compared with monotherapy groups within tumor tissues. Thus, YHO-1701 had a favorable specificity for STAT3 and pharmacokinetics after oral treatment; it also contributed to the enhanced antitumor activity of sorafenib. The evidence presented here provides justification using for this approach in future clinical settings.


Asunto(s)
Antineoplásicos/farmacología , Quinolinas/farmacología , Factor de Transcripción STAT3/antagonistas & inhibidores , Administración Oral , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimioterapia Combinada , Humanos , Interleucina-6/sangre , Ratones , Simulación del Acoplamiento Molecular , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Oxadiazoles/química , Oxadiazoles/farmacología , Multimerización de Proteína/efectos de los fármacos , Quinolinas/química , Quinolinas/uso terapéutico , Factor de Transcripción STAT3/química , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Sorafenib/farmacología , Sorafenib/uso terapéutico , Survivin/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Dominios Homologos src
14.
Biochem Biophys Res Commun ; 519(3): 505-511, 2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31530389

RESUMEN

Kinesin centromere-associated protein E (CENP-E) has emerged as a potential target for the development of anticancer drugs due to its involvement in the mitotic progression of the cell cycle. Although several CENP-E inhibitors have been reported, more knowledge of chemical structures and inhibitory mechanisms is necessary for developing CENP-E inhibitors. Here, we describe the identification of new CENP-E inhibitors. Screening of a small-molecule chemical library identified benzo[d]pyrrolo[2,1-b]thiazole derivatives, including 1, as compounds with inhibitory activity against the microtubule-stimulated ATPase of the CENP-E motor domain. Among the mitotic kinesins examined, 1 selectively inhibited the kinesin ATPase activity of CENP-E. In a steady-state ATPase assay, 1 exhibited ATP-competitive behavior, which was different from the CENP-E inhibitor GSK923295. Compound 1 inhibited the proliferation of tumor-derived HeLa and HCT116 cells more efficiently than that of non-cancerous WI-38 cells. The inhibition of cell proliferation was attributed to the ability of 1 to induce apoptotic cell death. The compound showed antimitotic activity, which caused cell cycle arrest at mitosis via interference with proper chromosome alignment. We identified 1 and its derivatives as the lead compounds that target CENP-E, thus providing a new opportunity for the development of anticancer agents targeting kinesins.


Asunto(s)
Antineoplásicos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Sarcosina/análogos & derivados , Antineoplásicos/química , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas Cromosómicas no Histona/metabolismo , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HCT116 , Células HeLa , Humanos , Estructura Molecular , Sarcosina/química , Sarcosina/farmacología , Relación Estructura-Actividad , Células Tumorales Cultivadas
15.
Biol Pharm Bull ; 42(5): 792-800, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31061322

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) is a latent transcription factor that contributes to tumor cell growth and survival and is often constitutively active in several types of cancers, which makes it an attractive target for cancer therapy. We identified 5,5'-(pentane-1,5'-diyl)bis(2-methyl-1,4-benzoquinone) (BPMB) as a new STAT3 inhibitor. BPMB inhibited the transcriptional activities of STAT3, despite its inability to reduce the phosphorylation and nuclear translocation of STAT3. BPMB selectively inhibited the proliferation of human breast cancer cell lines with constitutively activated STAT3. Furthermore, a gel retardation pattern was obtained by immunoblotting only when those STAT3-activated cell lines were treated with BPMB. The shifted bands could be immunoblotted with anti-STAT3 antibody but not with anti-STAT1/STAT5 antibody, and were stable under reducing conditions. The purified recombinant STAT3 protein treated with BPMB afforded a similar band shift pattern. Matrix-assisted laser desorption/ionization-mass spectrometry analysis of the component comprising the main shifted band suggested that the complex is a STAT3 homodimer crosslinked by BPMB through a Michael addition with Cys550 in the linker domain. Alanine replacement at this position resulted in reduction of the STAT3 dimer formation in the gel retardation assay. Thus, our results suggest that BPMB inhibits the proliferation of STAT3-activated cell lines, presumably through acylation of the linker domain and subsequent induction of the inactive STAT3 complexes.


Asunto(s)
Antineoplásicos/farmacología , Factor de Transcripción STAT3/antagonistas & inhibidores , Transporte Biológico , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células HeLa , Humanos , Fosforilación/efectos de los fármacos , Dominios Proteicos , Factor de Transcripción STAT3/química , Factor de Transcripción STAT3/metabolismo
16.
J Exp Bot ; 70(12): 3329-3342, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-30977815

RESUMEN

To identify the upstream signaling of aluminum-induced malate secretion through aluminum-activated malate transporter 1 (AtALMT1), a pharmacological assay using inhibitors of human signal transduction pathways was performed. Early aluminum-induced transcription of AtALMT1 and other aluminum-responsive genes was significantly suppressed by phosphatidylinositol 4-kinase (PI4K) and phospholipase C (PLC) inhibitors, indicating that the PI4K-PLC metabolic pathway activates early aluminum signaling. Inhibitors of phosphatidylinositol 3-kinase (PI3K) and PI4K reduced aluminum-activated malate transport by AtALMT1, suggesting that both the PI3K and PI4K metabolic pathways regulate this process. These results were validated using T-DNA insertion mutants of PI4K and PI3K-RNAi lines. A human protein kinase inhibitor, putatively inhibiting homologous calcineurin B-like protein-interacting protein kinase and/or Ca-dependent protein kinase in Arabidopsis, suppressed late-phase aluminum-induced expression of AtALMT1, which was concomitant with the induction of an AtALMT1 repressor, WRKY46, and suppression of an AtALMT1 activator, Calmodulin-binding transcription activator 2 (CAMTA2). In addition, a human deubiquitinase inhibitor suppressed aluminum-activated malate transport, suggesting that deubiquitinases can regulate this process. We also found a reduction of aluminum-induced citrate secretion in tobacco by applying inhibitors of PI3K and PI4K. Taken together, our results indicated that phosphatidylinositol metabolism regulates organic acid secretion in plants under aluminum stress.


Asunto(s)
Aluminio/metabolismo , Proteínas de Arabidopsis/genética , Arabidopsis/metabolismo , Malatos/metabolismo , Transportadores de Anión Orgánico/genética , Fosfatidilinositoles/metabolismo , Proteínas de Arabidopsis/metabolismo , Transportadores de Anión Orgánico/metabolismo
17.
Sci Rep ; 8(1): 2385, 2018 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-29402895

RESUMEN

Dimerization in signal transduction is a dynamically regulated process and a key regulatory mechanism. Signal transducer and activator of transcription 3 (STAT3) dimerizes after tyrosine phosphorylation upon cytokine stimulation. Because only the STAT3 dimer possesses the trans-activation activity, dimerization is an indispensable process for cytokine signaling. Here we report the detection of dynamic STAT3 dimerization in living cells using the homoFluoppi system. This method allowed us to validate the presence of an intact Src homology 2 domain and STAT3 Tyr705 phosphorylation, which facilitate puncta formation and homodimerization. Puncta formation was reversible, as determined by a decreased punctate signal after washout of oncostatin M. We analyzed STAT3 mutants, which have been reported in patients with hyper IgE syndrome and inflammatory hepatocellular adenoma (IHCA). Analysis of the IHCA mutants using homoFluoppi revealed constitutive activity independent of cytokine stimulation and novel insight into kinetics of dimer dissociation process. Next, we used homoFluoppi to screen for inhibitors of STAT3 dimerization, and identified 3,4-methylenedioxy-ß-nitrostyrene as a novel inhibitor. The results of this study show that homoFluoppi is a useful research tool for the analysis of proteins like STAT3 that dynamically dimerize, and is applicable for the screening of dimerization modulators.


Asunto(s)
Técnicas Citológicas/métodos , Imagen Óptica/métodos , Multimerización de Proteína , Factor de Transcripción STAT3/metabolismo , Coloración y Etiquetado/métodos , Análisis Mutacional de ADN , Células Epiteliales/metabolismo , Células HEK293 , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Factor de Transcripción STAT3/genética
18.
ACS Omega ; 3(9): 12284-12294, 2018 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-31459302

RESUMEN

For a better understanding of protein-inhibitor interactions, we report structural, thermodynamic, and biological analyses of the interactions between S-trityl-l-cysteine (STLC) derivatives and the motor domain of kinesin spindle protein Eg5. Binding of STLC-type inhibitors to Eg5 was enthalpically driven and entropically unfavorable. The introduction of a para-methoxy substituent in one phenyl ring of STLC enhances its inhibitory activity resulting from a larger enthalpy gain possibly due to the increased shape complementarity. The substituent fits to a recess in the binding pocket. To avoid steric hindrance, the substituted STLC is nudged toward the side opposite to the recess, which enhances the interaction of Eg5 with the remaining part of the inhibitor. Further introduction of an ethylene linkage between two phenyl rings enhances Eg5 inhibitory activity by reducing the loss of entropy in forming the complex. This study provides valuable examples of enhancing protein-inhibitor interactions without forming additional hydrogen bonds.

19.
PLoS One ; 12(1): e0170709, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28125678

RESUMEN

Bendamustine (BENDA), which bears the bis(2-chloroethyl)amino moiety, is an alkylating agent that stops the growth of cancer cells by binding to DNA and interfering with its replication. However, the mechanism of action underlying its excellent clinical efficacy remains unclear. In this work, we report that BENDA inhibits signal transducer and activator of transcription 3 (STAT3). In an AlphaScreen-based biochemical assay using recombinant human STAT3, binding of STAT3-Src homology 2 (SH2) to the phosphotyrosine (pTyr, pY) peptide was inhibited by BENDA but not by the inactive metabolite dihydroxy bendamustine (HP2). When a single point mutation of C550A or C712A was introduced into recombinant human STAT3, its sensitivity to BENDA was substantially reduced, suggesting that these cysteine residues are important for BENDA to inhibit STAT3. Furthermore, BENDA suppressed the function of cellular STAT3 as a transcriptional activator in a human breast cancer cell line, MDA-MB-468, with constitutively activated STAT3. A competitive pull-down assay using biotinylated BENDA (Bio-BENDA) revealed that BENDA bound tightly to cellular STAT3, presumably through covalent bonds. Therefore, our results suggest that the anticancer effects of BENDA may be associated, at least in part, with its inhibitory effect on the SH2 domain of STAT3.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Clorhidrato de Bendamustina/farmacología , Cisteína/química , Fosfotirosina/química , Mutación Puntual , Factor de Transcripción STAT3/antagonistas & inhibidores , Alanina/química , Alanina/metabolismo , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/metabolismo , Clorhidrato de Bendamustina/análogos & derivados , Clorhidrato de Bendamustina/metabolismo , Sitios de Unión , Línea Celular Tumoral , Cisteína/metabolismo , Ensayos de Selección de Medicamentos Antitumorales , Expresión Génica , Humanos , Péptidos/antagonistas & inhibidores , Péptidos/síntesis química , Péptidos/metabolismo , Fosfotirosina/metabolismo , Unión Proteica , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Relación Estructura-Actividad , Dominios Homologos src
20.
J Antibiot (Tokyo) ; 70(4): 443-447, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27756911

RESUMEN

Caliciviruses are contagious pathogens of humans and various animals. They are the most common cause of viral gastroenteritis in humans, and can cause lethal diseases in domestic animals such as cats, rabbits and immunocompromised mice. In this study, we conducted cytopathic effect-based screening of 2080 selected compounds from our in-house library to find antiviral compounds against three culturable caliciviruses: feline calicivirus, murine norovirus (MNV) and porcine sapovirus (PoSaV). We identified active six compounds, of which two compounds, both related to theaflavins, showed broad antiviral activities against all three caliciviruses; three compounds (abamectin, a mixture of avermectin B1a and B1b; avermectin B1a; and (-)-epigallocatechin gallate hydrate) were effective against PoSaV only; and a heterocyclic carboxamide derivative (BFTC) specifically inhibited MNV infectivity in cell cultures. Further studies of the antiviral mechanism and structure-activity relationship of theaflavins suggested the following: (1) theaflavins worked before the viral entry step; (2) the effect of theaflavins was time- and concentration-dependent; and (3) the hydroxyl groups of the benzocycloheptenone ring were probably important for the anti-calicivirus activity of theaflavins. Theaflavins could be used for the calicivirus research, and as potential disinfectants and antiviral reagents to prevent and control calicivirus infections in animals and humans.


Asunto(s)
Antivirales/farmacología , Biflavonoides/farmacología , Caliciviridae/efectos de los fármacos , Catequina/farmacología , Flavinas/farmacología , Animales , Infecciones por Caliciviridae , Calicivirus Felino/efectos de los fármacos , Catequina/análogos & derivados , Gatos , Efecto Citopatogénico Viral/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Humanos , Ivermectina/análogos & derivados , Ivermectina/farmacología , Ratones , Norovirus/efectos de los fármacos , Estructura Cuaternaria de Proteína , Sapovirus/efectos de los fármacos , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...