Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Molecules ; 28(21)2023 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-37959760

RESUMEN

This research aimed to determine the target protein and molecular mechanism of trans-(±)-kusunokinin ((±)-KU) derivatives (trans-(±)-ARC and trans-(±)-TTPG-B). Molecular docking was used to predict potential synthesized (±)-KU targets among 22 proteins. The (±)-TTPG-B bound HSP90α better than EC44, native (±)-KU and (-)-KU, and (±)-KU and (-)-ARC. In contrast, (-)-ARC bound PI3K more strongly than any other test compound. CSF1R and AKR1B1 were not supposed to be the target of (±)-TTPG-B and (±)-ARC, unlike native (±)-KU. The (±)-TTPG-B bound Tyr139 and Trp162 of HSP90α. Moreover, (-)-ARC bound PI3K via hydrogen bonds and π-π stacking at distinct amino acids, which was different from the other tested compounds. Using half of the IC50 concentration, (±)-TTPG-B, (±)-KU and (±)-ARC enhanced cell cycle arrest at the G0/G1 phase after 12 h and 24 h on KKU-M213 (CCA) cells. The (±)-TTPG-B showed a stronger inhibitory effect than (±)-ARC and (±)-KU on HSP90α, PI3K, HSP90ß, c-Myc, AKT, MEK1, CyclinB1, CyclinD1, and CDK1 for 24 and 48 h after treatment with the same concentration (0.015 µM). Thus, trans-(±)-TTPG-B, a newly synthesized compound, has pharmacological potential for development as a target therapy for CCA treatment.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Humanos , Simulación del Acoplamiento Molecular , Colangiocarcinoma/patología , Proliferación Celular , División Celular , Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/patología , Neoplasias de los Conductos Biliares/patología , Fosfatidilinositol 3-Quinasas , Línea Celular Tumoral , Apoptosis , Ciclo Celular , Aldehído Reductasa
2.
Molecules ; 27(23)2022 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-36500383

RESUMEN

This study aimed to investigate the cytotoxicity and anticancer activity of (±)-kusunokinin derivatives ((±)-TTPG-A and (±)-TTPG-B). The cytotoxicity effect was performed on human cancer cells, including breast cancer, cholangiocarcinoma, colon and ovarian cancer-cells, compared with normal cells, using the MTT assay. Cell-cycle arrest and apoptosis were detected using flow-cytometry analysis. We found that (±)-TTPG-B exhibited the strongest cytotoxicity on aggressive breast-cancer (MDA-MB-468 and MDA-MB-231) and cholangiocarcinoma (KKU-M213), with an IC50 value of 0.43 ± 0.01, 1.83 ± 0.04 and 0.01 ± 0.001 µM, respectively. Interestingly, (±)-TTPG-A and (±)-TTPG-B exhibited less toxicity than (±)-kusunokinin (9.75 ± 0.39 µM) on L-929 cells (normal fibroblasts). Moreover, (±)-TTPG-A predominated the ell-cycle arrest at the S phase, while (±)-TTPG-B caused cell arrest at the G0/G1 phase, in the same way as (±)-kusunokinin in KKU-M213 cells. Both (±)-TTPG-A and (±)-TTPG-B induced apoptosis and multi-caspase activity more than (±)-kusunokinin. Taken together, we conclude that (±)-TTPG-A and (±)-TTPG-B have a strong anticancer effect on cholangiocarcinoma. Moreover, (±)-TTPG-B could be a potential candidate compound for breast cancer and cholangiocarcinoma in the future.


Asunto(s)
Antineoplásicos , Neoplasias de los Conductos Biliares , Neoplasias de la Mama , Colangiocarcinoma , Humanos , Femenino , Línea Celular Tumoral , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/patología , Apoptosis , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Conductos Biliares Intrahepáticos/patología , Proliferación Celular , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
3.
Antioxidants (Basel) ; 11(12)2022 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-36552555

RESUMEN

Synthetic trans-(±)-kusunokinin ((±)KU), a potential anticancer substance, was revealed to have an inhibitory effect on breast cancer. According to the computational modeling prediction, AKR1B1, an oxidative stress and cancer migration protein, could be a target protein of trans-(-)-kusunokinin. In this study, we determined the binding of (±)KU and AKR1B1 on triple-negative breast and non-serous ovarian cancers. We found that (±)KU exhibited a cytotoxic effect that was significantly stronger than zopolrestat (ZP) and epalrestat (EP) (known AKR1B1 inhibitors) on breast and ovarian cancer cells. (±)KU inhibited aldose reductase activity that was stronger than trans-(-)-arctiin ((-)AR) but weaker than ZP and EP. Interestingly, (±)KU stabilized AKR1B1 on SKOV3 and Hs578T cells after being heated at 60 and 75 °C, respectively. (±)KU decreased malondialdehyde (MDA), an oxidative stress marker, on Hs578T cells in a dose-dependent manner and the suppression was stronger than EP. Furthermore, (±)KU downregulated AKR1B1 and its downstream proteins, including PKC-δ, NF-κB, AKT, Nrf2, COX2, Twist2 and N-cadherin and up-regulated E-cadherin. (±)KU showed an inhibitory effect on AKR1B1 and its downstream proteins, similar to siRNA-AKR1B1. Interestingly, the combination of siRNA-AKR1B1 with EP or (±)KU showed a greater effect on the suppression of AKR1B1, N-cadherin, E-cadherin and NF-κB than single treatments. Taken together, we concluded that (±)KU-bound AKR1B1 leads to the attenuation of cellular oxidative stress, as well as the aggressiveness of breast cancer cell migration.

4.
Molecules ; 26(15)2021 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-34361688

RESUMEN

Trans-(-)-kusunokinin, an anticancer compound, binds CSF1R with low affinity in breast cancer cells. Therefore, finding an additional possible target of trans-(-)-kusunokinin remains of importance for further development. Here, a computational study was completed followed by indirect proof of specific target proteins using small interfering RNA (siRNA). Ten proteins in breast cancer were selected for molecular docking and molecular dynamics simulation. A preferred active form in racemic trans-(±)-kusunokinin was trans-(-)-kusunokinin, which had stronger binding energy on HER2 trans-(+)-kusunokinin; however, it was weaker than the designed HER inhibitors (03Q and neratinib). Predictively, trans-(-)-kusunokinin bound HER2 similarly to a reversible HER2 inhibitor. We then verified the action of (±)-kusunokinin compared with neratinibon breast cancer cells (MCF-7). (±)-Kusunokinin exhibited less cytotoxicity on normal L-929 and MCF-7 than neratinib. (±)-Kusunokinin and neratinib had stronger inhibited cell proliferation than siRNA-HER2. Moreover, (±)-kusunokinin decreased Ras, ERK, CyclinB1, CyclinD and CDK1. Meanwhile, neratinib downregulated HER, MEK1, ERK, c-Myc, CyclinB1, CyclinD and CDK1. Knocking down HER2 downregulated only HER2. siRNA-HER2 combination with (±)-kusunokinin suppressed HER2, c-Myc, CyclinB1, CyclinD and CDK1. On the other hand, siRNA-HER2 combination with neratinib increased HER2, MEK1, ERK, c-Myc, CyclinB1, CyclinD and CDK1 to normal levels. We conclude that trans-(±)-kusunokinin may bind HER2 with low affinity and had a different action from neratinib.


Asunto(s)
Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Lignanos/metabolismo , Lignanos/farmacología , Piper nigrum/química , Extractos Vegetales/metabolismo , Extractos Vegetales/farmacología , Receptor ErbB-2/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Células MCF-7 , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Quinolinas/farmacología , ARN Interferente Pequeño/genética , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transfección
5.
ACS Omega ; 6(1): 606-614, 2021 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-33458512

RESUMEN

(-)-Kusunokinin performed its anticancer potency through CFS1R and AKT pathways. Its ambiguous binding target has, however, hindered the next development phase. Our study thus applied molecular docking and molecular dynamics simulation to predict the protein target from the pathways. Among various candidates, aldo-keto reductase family 1 member B1 (AKR1B1) was finally identified as a (-)-kusunokinin receptor. The predicted binding affinity of (-)-kusunokinin was better than the selected aldose reductase inhibitors (ARIs) and substrates. The compound also had no significant effect on AKR1B1 conformation. An intriguing AKR1B1 efficacy, with respect to the known inhibitors (epalrestat, zenarestat, and minalrestat) and substrates (UVI2008 and prostaglandin H2), as well as a similar interactive insight of the enzyme pocket, pinpointed an ARI equivalence of (-)-kusunokinin. An aromatic ring and a γ-butyrolactone ring shared a role with structural counterparts in known inhibitors. The modeling explained that the aromatic constituent contributed to π-π attraction with Trp111. In addition, the γ-butyrolactone ring bound the catalytic His110 using hydrogen bonds, which could lead to enzymatic inhibition as a consequence of substrate competitiveness. Our computer-based findings suggested that the potential of (-)-kusunokinin could be furthered by in vitro and/or in vivo experiments to consolidate (-)-kusunokinin as a new AKR1B1 antagonist in the future.

6.
Nat Prod Res ; 35(21): 3556-3561, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31933382

RESUMEN

Five new compounds, iranginins A-E (1-5), together with sixteen known compounds were isolated from the insect pathogenic fungus Ophiocordyceps irangiensis BCC 2728. The structures and the absolute configurations of the new compounds were established by spectroscopic analyses, the application of modified Mosher's method (for 2), ECD calculation (for 5), and X-ray crystallographic analysis (for 4). LL-Z1640-5 and mucorisocoumarin C were active against Mycobacterium tuberculosis (MIC 41.7 and 85.0 µM, respectively), while peyroisocoumarin D exhibited cytotoxic activity (IC50 65.6 µM).


Asunto(s)
Antineoplásicos , Hormigas , Hypocreales , Policétidos , Animales , Estructura Molecular
7.
Biomed Pharmacother ; 129: 110361, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32535390

RESUMEN

Kusunokinin, a lignan compound, inhibits cancer cell proliferation and induces apoptosis; however, the role of kusunokinin is not fully understood. Here, we aimed to identify a target protein of (-)-kusunokinin and determine the protein levels of its downstream molecules. We found that (-)-kusunokinin bound 5 possible target proteins, including CSF1R, MMP-12, HSP90-α, CyclinB1 and MEK1 with ΔGbind less than -10.40 kcal/mol. MD simulation indicated (-)-kusunokinin and pexidartinib (P31, a specific CSF1R binding compound) shared some extents of functional similarity in which (-)-kusunokinin bound CSF1R at the juxtamembrane (JM) region with aromatic amino acids similar to pexidartinib using π-π interaction, as well as hydrogen bond. Both P31 and (-)-kusunokinin moved into the same CSF1R region and W7 was a mutual key residue. However, the P31 binding site differed from the (-)-kusunokinin binding site. For in vitro study, the synthetic (±)-kusunokinin exhibited stronger cytotoxicity than picropodophyllotoxin, silibinin and etoposide on MCF-7 cells and represented less toxicity than picropodophyllotoxin and doxorubicin on L-929 and MCF-12A cells. Knocking down CSF1R using a specific siRNA combination with (±)-kusunokinin demonstrated levels of cell proliferation proteins slightly higher than siRNA-CSF1R treatment. However, siRNA-CSF1R combination with P31 represented the number of cell viability and cell proliferation proteins, like in the control groups (Lipofectamine and siRNA-Luciferase). Moreover, (±)-kusunokinin suppressed CSF1R and its downstream proteins, including AKT, CyclinD1 and CDK1. Meanwhile, both P31 and siRNA-CSF1R dramatically suppressed CSF1R, MEK1, AKT, ERK, CyclinB1, CyclinD1 and CDK1. Our overall results indicate that the mechanism of (±)-kusunokinin differed fairly from P31. We have concluded that (±)-kusunokinin inhibited breast cancer cell proliferation partially through the binding and suppression of CSF1R, which consequently affected AKT and its downstream molecules.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Lignanos/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/metabolismo , Sitios de Unión , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteína Quinasa CDC2/genética , Proteína Quinasa CDC2/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Femenino , Humanos , Lignanos/química , Lignanos/metabolismo , Células MCF-7 , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/química , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Transducción de Señal , Relación Estructura-Actividad
8.
J Nat Prod ; 83(4): 905-917, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-32193929

RESUMEN

Fourteen new compounds, oudemansins 1-4, oudemansinols 5-7, favolasins 8-10, favolasinin (12), polyketides 13-15, and (R,E)-2,4-dimethyl-5-phenyl-4-pentene-2,3-diol (16), together with nine known compounds were isolated from the basidiomycete fungus Favolaschia sp. BCC 18686. Two new compounds, favolasin E (11) and 9-oxostrobilurin E (17), were isolated from the closely related organism Favolaschia calocera BCC 36684 along with nine ß-methoxyacrylate-type derivatives. Compounds in the class of oudemansins and strobilurins exhibited moderate to strong antimalarial activity with relatively low cytotoxicity against Vero cells (African green monkey kidney fibroblasts). Potent antimalarial activity was demonstrated for 9-methoxystrobilurins G, K, and E (IC50 values 0.061, 0.089, and 0.14 µM, respectively). The structure-activity relationships (SAR) for antimalarial activity is proposed on the basis of the activity of the new and several known ß-methoxyacrylate derivatives in combination with the data from previously isolated compounds. Furthermore, several compounds showed specific cytotoxicity against NCI-187 cells (human small-cell lung cancer), although the SAR was different from that for antimalarial activity.


Asunto(s)
Agaricales/química , Antimaláricos/química , Antimaláricos/farmacología , Policétidos/química , Policétidos/farmacología , Estrobilurinas/química , Estrobilurinas/farmacología , Acrilatos/química , Acrilatos/farmacología , Animales , Antibióticos Antineoplásicos/farmacología , Línea Celular Tumoral , Chlorocebus aethiops , Ensayos de Selección de Medicamentos Antitumorales , Fermentación , Humanos , Espectroscopía de Resonancia Magnética , Estructura Molecular , Plasmodium falciparum/efectos de los fármacos , Espectrometría de Masa por Ionización de Electrospray , Relación Estructura-Actividad , Células Vero
9.
Biomed Pharmacother ; 117: 109115, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31220743

RESUMEN

Kusunokinin is a potent lignan compound with a several biological properties including antitrypanosomal and anticancer. In this study, (±)-kusunokinin and its derivative, (±)-bursehernin, were synthesized and investigated for their anticancer activities on cell viability, cell cycle arrest and apoptosis in cancer cell lines including breast cancer (MCF-7, MDA-MB-468 and MDA-MB-231), colon cancer (HT-29) and cholangiocarcinoma (KKU-K100, KKU-M213 and KKU-M055) cells. The result showed that (±)-kusunokinin and (±)-bursehernin represented the strongest growth inhibition against breast cancer (MCF-7) and cholangiocarcinoma (KKU-M213) cells with the IC50 values of 4.30 ±â€¯0.65 µM and 3.70 ±â€¯0.79 µM, respectively, both of which were lower than IC50 of normal fibroblast cells (L929). Etoposide was used as a positive control since this chemotherapeutic drug is in the lignan group same as (±)-kusunokinin. Surprisingly, etoposide showed less cytotoxicity than (±)-kusunokinin and its derivative on MCF-7, HT-29, KKU-M213 and KKU-K100. Moreover, (±)-bursehernin induced cell cycle arrest at G2/M phase, meanwhile (±)-kusunokinin tended to increased cell population at G2/M phase but did not show the significant difference compared with non-treated cells. Interestingly, protein levels related to cell proliferation pathway (topoisomerase II, STAT3, cyclin D1, and p21) were significantly decreased at 72 h. Both compounds induced apoptotic cell in time-dependent manner as confirmed by MultiCaspase assay. In conclusion, synthetic compound, (±)-kusunokinin and (±)-bursehernin, showed anticancer effects via the reduction of cell proliferation proteins and induction of apoptosis.


Asunto(s)
Lactonas/farmacología , Lignanos/farmacología , Neoplasias/patología , Antineoplásicos/química , Antineoplásicos/farmacología , Caspasas/metabolismo , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Humanos , Concentración 50 Inhibidora , Lactonas/química , Lignanos/química , Modelos Biológicos , Proteínas de Neoplasias/metabolismo
10.
Org Biomol Chem ; 14(46): 11002-11012, 2016 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-27827507

RESUMEN

F-THENA is designed as an alternative fluorine-containing chiral derivatizing agent (CDA). The fluorine atom functions exclusively as a reporter which can directly sense an anisotropic effect from an aromatic substituent of a chiral alcohol. In combination with chemical shift differences from both 19F NMR and 1H NMR, the F-THENA method can successfully be used for determining the absolute configuration of chiral secondary aromatic alcohols with a self-validating system.

11.
J Nat Prod ; 78(4): 615-22, 2015 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-25734623

RESUMEN

Four new compounds including two eremophilane sesquiterpenes, penicilleremophilanes A (1) and B (2), as well as two sulfur-containing biphenols, penicillithiophenols A (3) and B (4), were isolated from the soil fungus Penicillium copticola PSU-RSPG138 together with 16 known compounds. Their structures were elucidated by spectroscopic methods. Known sporogen AO-1 exhibited significant antimalarial activity against Plasmodium falciparum with an IC50 value of 1.53 µM and cytotoxic activity to noncancerous (Vero) cell lines with an IC50 value of 4.23 µM. Although compound 1 was approximately half as active against P. falciparum with the IC50 value of 3.45 µM, it showed much weaker cytotoxic activity.


Asunto(s)
Compuestos de Bifenilo/aislamiento & purificación , Penicillium/química , Sesquiterpenos/aislamiento & purificación , Sulfuros/aislamiento & purificación , Animales , Antimaláricos/farmacología , Antineoplásicos/farmacología , Compuestos de Bifenilo/química , Chlorocebus aethiops , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Concentración 50 Inhibidora , Estructura Molecular , Plasmodium falciparum/efectos de los fármacos , Sesquiterpenos/química , Microbiología del Suelo , Sulfuros/química , Sulfuros/farmacología , Tailandia , Células Vero
12.
J Nat Prod ; 73(4): 759-62, 2010 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-20329738

RESUMEN

Two strobilurins, 9-methoxystrobilurin B (1) and 9-methoxystrobilurin G (2), two monochlorinated 2,3-dihydro-1-benzoxepin derivatives, 3 and 4a, and butenolide 5, together with four known compounds, strobilurin B, 9-methoxystrobilurin A, and oudemansins A and B, were isolated from culture BCC 18689 of the fungus Favolaschia tonkinensis. 9-Methoxystrobilurins A, B (1), and G (2) and oudemansins A and B exhibited antimalarial, antifungal, and cytotoxic activities, while compounds 3, 4a, and 5 displayed only cytotoxic activity.


Asunto(s)
Basidiomycota/química , Citotoxinas/aislamiento & purificación , Citotoxinas/farmacología , Ácidos Grasos Insaturados/aislamiento & purificación , Ácidos Grasos Insaturados/farmacología , Metacrilatos/aislamiento & purificación , 4-Butirolactona/análogos & derivados , 4-Butirolactona/química , 4-Butirolactona/aislamiento & purificación , Animales , Antifúngicos/aislamiento & purificación , Antifúngicos/farmacología , Antimaláricos/farmacología , Benzoxepinas/química , Benzoxepinas/aislamiento & purificación , Candida albicans/efectos de los fármacos , Chlorocebus aethiops , Citotoxinas/química , Ensayos de Selección de Medicamentos Antitumorales , Ácidos Grasos Insaturados/química , Femenino , Humanos , Células KB , Metacrilatos/química , Metacrilatos/farmacología , Pruebas de Sensibilidad Microbiana , Estructura Molecular , Plasmodium falciparum/efectos de los fármacos , Pirazoles , Pirimidinas , Estrobilurinas , Tailandia , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA