Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 12531, 2024 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822100

RESUMEN

Binding affinity is an important factor in drug design to improve drug-target selectivity and specificity. In this study, in silico techniques based on molecular docking followed by molecular dynamics (MD) simulations were utilized to identify the key residue(s) for CSF1R binding affinity among 14 pan-tyrosine kinase inhibitors and 15 CSF1R-specific inhibitors. We found tryptophan at position 550 (W550) on the CSF1R binding site interacted with the inhibitors' aromatic ring in a π-π way that made the ligands better at binding. Upon W550-Alanine substitution (W550A), the binding affinity of trans-(-)-kusunokinin and imatinib to CSF1R was significantly decreased. However, in terms of structural features, W550 did not significantly affect overall CSF1R structure, but provided destabilizing effect upon mutation. The W550A also did not either cause ligand to change its binding site or conformational changes due to ligand binding. As a result of our findings, the π-π interaction with W550's aromatic ring could be still the choice for increasing binding affinity to CSF1R. Nevertheless, our study showed that the increasing binding to W550 of the design ligand may not ensure CSF1R specificity and inhibition since W550-ligand bound state did not induce significantly conformational change into inactive state.


Asunto(s)
Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Triptófano , Triptófano/química , Triptófano/metabolismo , Ligandos , Sitios de Unión , Humanos , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/química , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química , Mesilato de Imatinib/farmacología , Mesilato de Imatinib/química , Receptor de Factor Estimulante de Colonias de Macrófagos
2.
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
3.
Foods ; 12(10)2023 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-37238871

RESUMEN

A low piperine fractional Piper nigrum extract (PFPE) was prepared by mixing cold-pressed coconut oil and honey in distilled water, namely, PFPE-CH. In this study, PFPE-CH was orally administered as a dietary supplement to decrease the risk of tumor formation and reduce the side effects of chemotherapeutic drugs during breast cancer treatment. The toxicity study demonstrated no mortality or adverse effects after administrating PFPE-CH at 5000 mg/kg during a 14-day observation period. Additionally, PFPE-CH at 86 mg/kg BW/day did not cause any harm to the kidney or liver function of the rats for six months. In a cancer prevention study, treatment with PFPE-CH at 100 mg/kg BW for 101 days induced oxidative stress and increased the immune response by altering the levels of cancer-associated cytokines (IL-4, IL-6, and IFN-g), leading to a reduction in the tumor incidence of up to 71.4% without any adverse effects. In combination with doxorubicin, PFPE-CH did not disrupt the anticancer effects of the drug in rats with mammary tumors. Surprisingly, PFPE-CH reduced chemotherapy-induced toxicity by improving some hematological and biochemical parameters. Therefore, our results suggest that PFPE-CH is safe and effective in reducing breast tumor incidence and toxicity of chemotherapeutic drugs during cancer treatment in mammary tumor rats.

4.
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
5.
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.

6.
Molecules ; 27(13)2022 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-35807438

RESUMEN

Breast cancer cell proliferation and migration are inhibited by naturally extracted trans-(-)-kusunokinin. However, three additional enantiomers of kusunokinin have yet to be investigated: trans-(+)-kusunokinin, cis-(-)-isomer and cis-(+)-isomer. According to the results of molecular docking studies of kusunokinin isomers on 60 breast cancer-related proteins, trans-(-)-kusunokinin was the most preferable and active component of the trans-racemic mixture. Trans-(-)-kusunokinin targeted proteins involved in cell growth and proliferation, whereas the cis-(+)-isomer targeted proteins involved in metastasis. Trans-(-)-kusunokinin targeted CSF1R specifically, whereas trans-(+)-kusunokinin and both cis-isomers may have bound AKR1B1. Interestingly, the compound's stereoisomeric effect may influence protein selectivity. CSF1R preferred trans-(-)-kusunokinin over trans-(+)-kusunokinin because the binding pocket required a ligand planar arrangement to form a π-π interaction with a selective Trp550. Because of its large binding pocket, EGFR exhibited no stereoselectivity. MD simulation revealed that trans-(-)-kusunokinin, trans-(+)-kusunokinin and pexidartinib bound CSF1R differently. Pexidartinib had the highest binding affinity, followed by trans-(-)-kusunokinin and trans-(+)-kusunokinin, respectively. The trans-(-)-kusunokinin-CSF1R complex was found to be stable, whereas trans-(+)-kusunokinin was not. Trans-(±)-kusunokinin, a potential racemic compound, could be developed as a selective CSF1R inhibitor when combined.


Asunto(s)
Neoplasias de la Mama , Proteínas Tirosina Quinasas Receptoras , Aldehído Reductasa , Ciclo Celular , Proliferación Celular , Femenino , Humanos , Isomerismo , Simulación del Acoplamiento Molecular
7.
Molecules ; 27(2)2022 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-35056723

RESUMEN

Triple negative breast cancer (TNBC) is a breast cancer subtype characterized by the absence of estrogen receptor, progesterone receptor and human epidermal growth factor receptor 2 expression. TNBC cells respond poorly to targeted chemotherapies currently in use and the mortality rate of TNBC remains high. Therefore, it is necessary to identify new chemotherapeutic agents for TNBC. In this study, the anti-cancer effects of 7-α-hydroxyfrullanolide (7HF), derived from Grangea maderaspatana, on MCF-7, MDA-MB-231 and MDA-MB-468 breast cancer cells were assessed using MTT assay. The mode of action of 7HF in TNBC cells treated with 6, 12 and 24 µM of 7HF was determined by flow cytometry and propidium iodide (PI) staining for cell cycle analysis and annexin V/fluorescein isothiocyanate + PI staining for detecting apoptosis. The molecular mechanism of action of 7HF in TNBC cells was investigated by evaluating protein expression using proteomic techniques and western blotting. Subsequently, 7HF exhibited the strongest anti-TNBC activity toward MDA-MB-468 cells and a concomitantly weak toxicity toward normal breast cells. The molecular mechanism of action of low-dose 7HF in TNBC cells primarily involved G2/M-phase arrest through upregulation of the expression of Bub3, cyclin B1, phosphorylated Cdk1 (Tyr 15) and p53-independent p21. Contrastingly, the upregulation of PP2A-A subunit expression may have modulated the suppression of various cell survival proteins such as p-Akt (Ser 473), FoxO3a and ß-catenin. The concurrent apoptotic effect of 7HF on the treated cells was mediated via both intrinsic and extrinsic modes through the upregulation of Bax and active cleaved caspase-7-9 expression and downregulation of Bcl-2 and full-length caspase-7-9 expression. Notably, the proteomic approach revealed the upregulation of the expression of pivotal protein clusters associated with G1/S-phase arrest, G2/M-phase transition and apoptosis. Thus, 7HF exhibits promising anti-TNBC activity and at a low dose, it modulates signal transduction associated with G2/M-phase arrest and apoptosis.


Asunto(s)
Neoplasias de la Mama Triple Negativas
8.
Oncol Lett ; 23(2): 59, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34992691

RESUMEN

Ovarian cancer ranks eighth in cancer incidence and mortality among women worldwide. Cisplatin-based chemotherapy is commonly used for patients with ovarian cancer. However, the clinical efficacy of cisplatin is limited due to the occurrence of adverse side effects and development of cancer chemoresistance during treatment. Trans-(±)-kusunokinin has been previously reported to inhibit cell proliferation and induce cell apoptosis in various cancer cell types, including breast, colon and cholangiocarcinoma. However, the potential effects of (±)-kusunokinin on ovarian cancer remains unknown. In the present study, chemosensitive ovarian cancer cell line A2780 and chemoresistant ovarian cancer cell lines A2780cis, SKOV-3 and OVCAR-3 were treated with trans-(±)-kusunokinin to investigate its potential effects. MTT, colony formation, apoptosis and multi-caspase assays were used to determine cytotoxicity, the ability of single cells to form colonies, induction of apoptosis and multi-caspase activity, respectively. Moreover, western blot analysis was performed to determine the proteins level of topoisomerase II, cyclin D1, CDK1, Bax and p53-upregulated modulator of apoptosis (PUMA). The results demonstrated that trans-(±)-kusunokinin exhibited the strongest cytotoxicity against A2780cis cells with an IC50 value of 3.4 µM whilst also reducing the colony formation of A2780 and A2780cis cells. Trans-(±)-kusunokinin also induced the cells to undergo apoptosis and increased multi-caspase activity in A2780 and A2780cis cells. This compound significantly downregulated topoisomerase II, cyclin D1 and CDK1 expression, but upregulated Bax and PUMA expression in both A2780 and A2780cis cells. In conclusion, trans-(±)-kusunokinin suppressed ovarian cancer cells through the inhibition of colony formation, cell proliferation and the induction of apoptosis. This pure compound could be a potential targeted therapy for ovarian cancer treatment in the future. However, studies in an animal model and clinical trial need to be performed to support the efficacy and safety of this new treatment.

9.
Pharm Biol ; 60(1): 96-107, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34962450

RESUMEN

CONTEXT: Many natural extracts have been shown to minimize the toxicity of doxorubicin (Dox). Low piperine Piper nigrum L. (Piperaceae) extract (PFPE) is a natural extract containing many types of antioxidants that may reduce Dox toxicities. OBJECTIVE: To evaluate the effect of PFPE in attenuating the side effects of Dox. MATERIALS AND METHODS: Tumour-bearing Sprague Dawley rats were divided into five groups including normal, vehicle, 100 mg/kg BW of PFPE plus 2 mg/kg BW of Dox (P100 + Dox), 100 mg/kg BW of PFPE plus 2 mg/kg BW of Dox (P200 + Dox) and Dox. Rats were treated with Dox and/or PFPE three times/week for 4 weeks. Tumour burden, blood parameters, weight of internal organs and immunological data were investigated. RESULTS: The addition of 200 mg/kg PFPE significantly restored the levels of AST from 174.60 ± 45.67 U/L in the Dox group near to normal levels at 109.80 ± 4.99 U/L. The combination of PFPE and Dox also decreased the levels of CXCL7, TIMP-1, sICAM-1 and l-selectin about 1.4-1.6-fold compared to Dox group. Feeding rats with 200 mg/kg BW of PFPE combination with Dox slightly increased Th1 from 161.67 ± 14.28 cells in Dox group to 200.75 ± 5.8 cells meanwhile suppressed Treg from 3088 ± 78 cells in Dox to 2561 ± 71 cells. DISCUSSION AND CONCLUSIONS: This study showed that PFPE ameliorated Dox toxicity in many aspects indicating the role of antioxidant and other substances in the extract on toxicity attenuation. This suggested the using of PFPE may be valuable for Dox treated patients.


Asunto(s)
Alcaloides/farmacología , Benzodioxoles/farmacología , Doxorrubicina/toxicidad , Piper nigrum/química , Piperidinas/farmacología , Extractos Vegetales/farmacología , Alcamidas Poliinsaturadas/farmacología , Alcaloides/administración & dosificación , Alcaloides/aislamiento & purificación , Animales , Antibióticos Antineoplásicos/toxicidad , Antioxidantes/administración & dosificación , Antioxidantes/aislamiento & purificación , Antioxidantes/farmacología , Benzodioxoles/administración & dosificación , Benzodioxoles/aislamiento & purificación , Relación Dosis-Respuesta a Droga , Femenino , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Piperidinas/administración & dosificación , Piperidinas/aislamiento & purificación , Extractos Vegetales/administración & dosificación , Extractos Vegetales/química , Alcamidas Poliinsaturadas/administración & dosificación , Alcamidas Poliinsaturadas/aislamiento & purificación , Ratas , Ratas Sprague-Dawley
10.
Planta Med ; 88(7): 527-537, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33902130

RESUMEN

Cancer is one of the major causes of death worldwide. In addition to standard regimens, tumor suppression ability has been demonstrated in many types of natural products, including Piper nigrum, or black pepper. In previous reports, we demonstrated the antitumor effect of low piperine fractional Piper nigrum extract in vitro and in vivo. However, the effects of low piperine fractional P. nigrum extract in the aspect of antitumor immunity has not yet been investigated. In this study, tumor-bearing rats were fed with 100 mg/kg BW or 200 mg/kg BW of low piperine fractional P. nigrum extract 3 times per week for 4 weeks. Tumor burden and hematological data were then evaluated. Immunological data was investigated using a cytokine array and flow cytometry. The results showed that both doses of low piperine fractional P. nigrum extract significantly suppressed tumor progression in N-nitrosomethylurea-induced mammary tumor rats. There were no significant changes observed in the total white blood cells, red blood cells, and hemoglobin. Low piperine fractional P. nigrum extract suppressed some cytokine and chemokine levels including CXCL7, sICAM-1, and L-selectin 0.2- to 0.6-fold. Interestingly, 200 mg/kg BW of low piperine fractional P. nigrum extract significantly promoted type 1 T helper cell, and suppressed neutrophil, basophil, type 2 T helper cell, and regulatory T cell compared to the control group. In summary, these results indicate that low piperine fractional P. nigrum extract had a high efficacy in supporting antitumor activity at immunological levels via regulating Th1/Th2/Treg cells.


Asunto(s)
Piper nigrum , Alcaloides , Animales , Benzodioxoles/farmacología , Carcinogénesis , Citocinas , Piperidinas , Extractos Vegetales/farmacología , Alcamidas Poliinsaturadas/farmacología , Ratas , Linfocitos T Reguladores
11.
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
12.
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.

13.
Eur J Pharmacol ; 882: 173311, 2020 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-32619673

RESUMEN

Natural and synthetic (-)-kusunokinin inhibited breast cancer, colon cancer and cholangiocarcinoma cells at the G2/M phase and induced apoptosis. However, there is no report on the action and adverse effects of (-)-kusunokinin in animal models. In this study, we investigated the cytotoxic effect of (-)-kusunokinin from Piper nigrum on cancer cells. NMU-induced rat mammary tumors, an ER positive breast cancer model, were treated with (-)-kusunokinin. Proteins of interest related to cell cycle, angiogenesis, migration and signaling proteins were detected in tumor tissues. Results showed that (-)-kusunokinin exhibited strong cytotoxicity against breast, colon and lung cancer cells and caused low toxicity against normal fibroblast cells. For in vivo study, 7.0 mg/kg and 14.0 mg/kg of (-)-kusunokinin reduced tumor growth without side effects on body weight, internal organs and bone marrow. Combination of (-)-kusunokinin with a low effective dose of doxorubicin significantly inhibited tumor growth and provoked cell death in cancer tissues. Mechanistically, 14.0 mg/kg of (-)-kusunokinin decreased cell proliferation (c-Src, PI3K, Akt, p-Erk1/2 and c-Myc), cell cycle (E2f-1, cyclin B1 and CDK1), and metastasis (E-cadherin, MMP-2 and MMP-9) proteins in tumor tissues, which supports its anticancer effect. We further confirmed the antimigration effect of (-)-kusunokinin; the results show that this compound inhibited breast cancer cell (MCF-7) migration in a dose-dependent manner. In conclusion, the results suggest that 14 mg/kg of (-)-kusunokinin inhibited tumors through the reduction of signaling proteins and their downstream molecules. Therefore, (-)-kusunokinin becomes an intriguing candidate for cancer treatment as it provides a strong potency in cancer inhibition.


Asunto(s)
Antineoplásicos/uso terapéutico , Lignanos/uso terapéutico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Lignanos/farmacología , Neoplasias Mamarias Experimentales/inducido químicamente , Neoplasias Mamarias Experimentales/patología , Metilnitrosourea , Ratas Sprague-Dawley
14.
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
15.
Oncol Lett ; 17(6): 5283-5291, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31186745

RESUMEN

Breast cancer is the leading cause of female mortality worldwide. Although there are several modern treatments for breast cancer, there is a high rate of recurrence for the majority of treatments; therefore, the search for effective anticancer agents continues. The present study aimed to investigate the anti-breast cancer potential of frullanolide, a compound which is isolated and purified from the Grangea maderaspatana plant, for selected human breast cancer cell lines (MCF-7, MDA-MB-468 and MDA-MB-231). The MTT assay was used to assess cytotoxic activity in breast cancer cell lines of treatment with frullanolide at 1.25, 2.5, 5.0, 10.0 and 20.0 µg/ml. Additionally, the apoptotic induction ability of frullanolide at various concentrations [0.5×, 1× and 2× half maximal inhibitory concentration (IC50)] was investigated by flow cytometry and western blot analysis. Frullanolide exhibited strong anti-breast cancer activity against MDA-MB-468 (IC50, 8.04±2.69 µg/ml) and weak cytotoxicity against the MCF-7 (IC50, 10.74±0.86 µg/ml) and MDA-MB-231 (IC50, 12.36±0.31 µg/ml) cell lines. The IC50 of frullanolide was high in the human normal epithelial breast cell line (MCF-12A) and mouse fibroblast cell line (L-929). Density plot diagrams revealed that frullanolide induced apoptosis in MCF-7, MDA-MB-468 and MDA-MB-231 cells. Notably, a plausible anticancer mechanism was elucidated via cellular apoptosis by p53-independence in the treated MCF-7 cell line and p53-dependence in the treated MDA-MB-468 and MDA-MB-231 cell lines. In conclusion, the present study demonstrated that frullanolide may exert anticancer activity on breast cancer cell lines by inducing apoptosis. Frullanolide offers a possible novel approach to breast cancer therapy.

16.
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
17.
Front Genet ; 10: 61, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30853973

RESUMEN

Autism spectrum disorder (ASD) is a highly heterogeneous neurodevelopmental disorder with many contributing risk genes and loci. To date, several intellectual disability (ID) susceptibility genes have frequently been identified in ASD. Here, whole exome sequencing was carried out on a proband with ASD and identified compound heterozygous mutations of the TRAPPC9, which plays a role in the neuronal NF-κB signaling pathway. These mutations consisted of a novel frameshift mutation (c.2415_2416insC, p.His806Profs∗9) and a rare splice site mutation (c.3349+1G>A) that were segregated from an unaffected father and unaffected mother, respectively. These two heterozygous mutations were also identified in the patient's older brother with ID. Quantitative RT-PCR revealed a significant reduction of TRAPPC9 transcript in two siblings. This study first describes compound heterozygous mutations of the TRAPPC9 gene in two siblings with ASD and ID, which is notable as only homozygous mutations or compound heterozygous for copy number variations and rare variant in this gene have been reported to date and associated with autosomal recessive intellectual disability. The two siblings carrying compound heterozygous TRAPPC9 mutations presented with ID, developmental delay, microcephaly and brain abnormalities similarly to the clinical features found in almost cases with homozygous TRAPPC9 mutation in previous studies. Together this study provides evidence that clinical manifestations of TRAPPC9 mutations as seen in our patients with ID and autism may be broader than previous case reports have indicated.

18.
AAPS PharmSciTech ; 19(2): 730-740, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28975598

RESUMEN

A supersaturatable self-microemulsifying drug delivery system (S-SMEDDS) with a reduced amount of surfactant and incorporation of a polymer precipitation inhibitor, Eudragit® E PO was developed. The optimized S-SMEDDS formulation (SS-15) consisted of 55% surfactants, 40% oils, and 5% Eudragit® E PO (curcumin at 44.4 mg/g of the formulation). The precipitation profiles from the supersaturation assay revealed that the curcumin S-SMEDDS performed as a better inhibitor of curcumin precipitation in simulated gastric fluid over a 240-min study than the normal curcumin SMEDDS and an aqueous curcumin suspension. In addition, the mean droplet size of the curcumin S-SMEDDS (21.6 ± 0.1 nm) was significantly smaller than the SMEDDS (28.1 ± 0.3 nm). The curcumin S-SMEDDS exhibited a threefold reduction of Caco-2 cell toxicity when compared to the curcumin SMEDDS because of the reduced toxic effect of the surfactant present in the SMEDDS formulation. In addition, the absorptive permeability across the Caco-2 monolayer of curcumin in the S-SMEDDS was significantly higher than for the unformulated curcumin (~ 5-folds). The plasma concentration-time profiles from the oral absorption studies in rats dosed with the curcumin S-SMEDDS showed a 1.22- and 53.14-fold increased absorption of curcumin, compared to the SMEDDS and the aqueous suspension, respectively. The curcumin S-SMEDDS was stable under both intermediate and accelerated conditions after 6 months of storage.


Asunto(s)
Curcumina/administración & dosificación , Administración Oral , Animales , Disponibilidad Biológica , Células CACO-2 , Permeabilidad de la Membrana Celular , Curcumina/metabolismo , Curcumina/farmacocinética , Sistemas de Liberación de Medicamentos , Emulsiones , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Masculino , Aceites , Polímeros , Ácidos Polimetacrílicos , Conejos , Ratas , Solubilidad , Tensoactivos
19.
Biomed Pharmacother ; 92: 732-743, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28586745

RESUMEN

Several studies have reported that active compounds isolated from Piper nigrum possess anticancer properties. However, there are no data on anticancer activity of (-)-kusunokinin and piperlonguminine. The purposes of this study were to isolate active compounds from P. nigrum and identify the molecular mechanisms underlying growth and apoptosis pathway in breast cancer cells. Two bioactive compounds, (-)-kusunokinin and piperlonguminine, were isolated from P. nigrum. Cytotoxicity and the molecular mechanism were measured by methyl thiazolyl tetrazolium (MTT) assay, flow cytometry and Western blot analysis. We found that the active compounds, which effect cancer cell lines were (-)-kusunokinin and piperlonguminine. These compounds have potent cytotoxic effects on breast cancer cells (MCF-7 and MDA-MB-468) and colorectal cells (SW-620). (-)-Kusunokinin demonstrated a cytotoxic effect on MCF-7 and MDA-MB-468 with IC50 values of 1.18 and 1.62µg/mL, respectively. Piperlonguminine had a cytotoxic effect on MCF-7 and MDA-MB-468 with IC50 values of 1.63 and 2.19µg/mL, respectively. Both compounds demonstrated lower cytotoxicity against normal breast cell lines with IC50 values higher than 11µg/mL. Cell cycle and apoptotic analysis using flow cytometry, showed that the (-)-kusunokinin and piperlonguminine induced cell undergoing apoptosis and drove cells towards the G2/M phase. Moreover, both compounds decreased topoisomerase II and bcl-2. The increasing of p53 levels further increased p21, bax, cytochrome c, caspase-8, -7 and -3 activities, except caspase-9. These results suggest that the (-)-kusunokinin and piperlonguminine have been shown to have potent anticancer activities through extrinsic pathway and G2/M phase arrest.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Dioxolanos/uso terapéutico , Lignanos/uso terapéutico , Piper nigrum/química , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Densitometría , Dioxolanos/química , Dioxolanos/farmacología , Femenino , Humanos , Lignanos/química , Lignanos/farmacología , Fitoterapia
20.
J Mol Graph Model ; 74: 215-224, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28456097

RESUMEN

Bursehernin (5'-desmethoxyyatein) is a natural lignan, which has anti-tumor activity in vitro. In this study, the binding-inhibitory effects of bursehernin were screening on selected 80 proteins associated with cancer pathway. The computational analysis suggested inhibitory effect due to bursehernin towards proteins related to cancer proliferation, including FMS kinase receptor, heat shock protein 90-α (Hsp90-α), adenylate cyclase 10 (ADCY10), mitogen-activated protein kinase kinase (MEK1), and α-tubulin. Moreover, bursehernin could interfere with cell cycle progression via binding to cyclin B proteins. Among all screened proteins, the compound showed an interesting binding affinity to the FMS kinase receptor. The binding mode studies by molecular dynamic technique showed that aromatic ring of bursehernin compound was responsible for compound-protein interaction through pi-pi stacking with Tyr105 and Phe178 of the FMS kinase receptor. This study suggests that bursehernin has potential for development as an anti-tumor agent with an anti-proliferation, and cell cycle arrest inducing, although further studies are needed.


Asunto(s)
Antineoplásicos/química , Lactonas/química , Lignanos/química , Proteínas de Neoplasias/química , Sitios de Unión , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Neoplasias/tratamiento farmacológico , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios Proteicos , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...