Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Br J Pharmacol ; 181(8): 1221-1237, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37926864

RESUMEN

BACKGROUND AND PURPOSE: The mammalian target of rapamycin (mTOR) pathway plays critical roles in intrinsic chemoresistance by regulating Fanconi anaemia complementation group D2 (FANCD2) expression. However, the mechanisms by which mTOR regulates FANCD2 expression and related inhibitors are not clearly elucidated. Extracts of Centipeda minima (C. minima) showed promising chemosensitizing effects by inhibiting FANCD2 activity. Here, we have aimed to identify the bioactive chemosensitizer in C. minima extracts and elucidate its underlying mechanism. EXPERIMENTAL APPROACH: The chemosensitizing effects of arnicolide C (ArC), a bioactive compound in C. minima, on non-small cell lung cancer (NSCLC) were investigated using immunoblotting, immunofluorescence, flow cytometry, the comet assay, small interfering RNA (siRNA) transfection and animal models. The online SynergyFinder software was used to determine the synergistic effects of ArC and chemotherapeutic drugs on NSCLC cells. KEY RESULTS: ArC had synergistic cytotoxic effects with DNA cross-linking drugs such as cisplatin and mitomycin C in NSCLC cells. ArC treatment markedly decreased FANCD2 expression in NSCLC cells, thus attenuating cisplatin-induced FANCD2 nuclear foci formation, leading to DNA damage and apoptosis. ArC inhibited the mTOR pathway and attenuated mTOR-mediated expression of E2F1, a critical transcription factor of FANCD2. Co-administration of ArC and cisplatin exerted synergistic anticancer effects in the A549 xenograft mouse model by suppressing mTOR/FANCD2 signalling in tumour tissues. CONCLUSION AND IMPLICATIONS: ArC suppressed DNA cross-linking drug-induced DNA damage response by inhibiting the mTOR/E2F1/FANCD2 signalling axis, serving as a chemosensitizing agent. This provides insight into the anticancer mechanisms of ArC and offers a potential combinatorial anticancer therapeutic strategy.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Anemia de Fanconi , Neoplasias Pulmonares , Humanos , Animales , Ratones , Carcinoma de Pulmón de Células no Pequeñas/patología , Cisplatino/farmacología , Neoplasias Pulmonares/patología , Serina-Treonina Quinasas TOR/metabolismo , ADN , Mamíferos/metabolismo , Factor de Transcripción E2F1/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo
2.
Naunyn Schmiedebergs Arch Pharmacol ; 396(4): 749-758, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36472629

RESUMEN

Recently, we have isolated and identified several bioactive flavonoids and stilbenoids with potential anticancer activity from Thai orchids. In this study, we further investigated the cytotoxic and chemosensitizing activities of these phytochemicals (namely, pinocembrin, cardamonin, isalpinin, galangin, pinosylvin monomethyl ether, 2,3'-dihydroxy-5'-methoxystilbene, (E)-2,5'-dihydroxy-2'-(4-hydroxybenzyl)-3'-methoxystilbene, 2,3-dihydroxy-3',5'-dimethoxystilbene, 2,3'-dihydroxy-5,5'-dimethoxystilbene, 3,4'-dihydroxy-5-methoxystilbene and batatasin III) against breast cancer MCF7 cells and its two multidrug resistant (MDR) sublines (MCF7/DOX and MCF7/MX). Cytotoxicity was determined with MTT assay for the estimation of the half maximal cytotoxic concentrations (IC50). Effects of the test compounds on activities of efflux transporters (BCRP, P-gp, MRP1, and MRP2) were evaluated with substrate accumulation assays using fluorometry and flow cytometry analysis. Out of these 11 test compounds, the stilbene pinosylvin monomethyl ether displayed its cytotoxicity specifically toward MCF7 cells (IC50 = 6.2 ± 1.2 µM, 72-h incubation) with 4.96 folds higher than normal fibroblast. Its potency decreased in MCF7/DOX and MCF7/MX cells by 3.94 and 7.38 folds, respectively. Our transporter assay indicated that this stilbene significantly reduced the activities of P-gp, MRP1, and MRP2, but not BCRP. After 48-h co-incubation, this stilbene (at 2 µM) synergistically increased doxorubicin- and mitoxantrone-mediated cytotoxicity in MCF7, MCF7/DOX, and MCF7/MX cells potentially by increasing the intracellular level of cytotoxic drug. Pinosylvin monomethyl ether could sensitize breast cancer cells to chemotherapy and overcome MDR, in part, via the inhibition of drug efflux transporters.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Estilbenos , Humanos , Femenino , Resistencia a Múltiples Medicamentos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/farmacología , Resistencia a Antineoplásicos , Antineoplásicos/farmacología , Doxorrubicina/farmacología , Estilbenos/farmacología , Éteres/farmacología , Neoplasias de la Mama/tratamiento farmacológico
3.
Front Pharmacol ; 11: 518068, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33041789

RESUMEN

Aberrant fibroblast growth factor receptor-1 (FGFR1), a key driver promoting gastric cancer (GC) progression and chemo-resistance, has been increasingly recognized as a potential therapeutic target in GC. Hereon, we designed and synthesized a series of asymmetric analogues using Af23 and NDGA as lead compounds by retaining the basic structural framework (bisaryl-1,4-dien-3-one) and the unilateral active functional groups (3,4-dihydroxyl). Thereinto, Y14 showed considerable inhibitory activity against FGFR1. Next, pharmacological experiments showed that Y14 could significantly inhibit the phosphorylation of FGFR1 and its downstream kinase AKT and ERK, thus inhibiting the growth, survival, and migration of gastric cancer cells. Furthermore, compared with 5-FU treatment alone, the combination of Y14 and 5-FU significantly reduced the phosphorylation level of FGFR1, and enhanced the anti-cancer effect by inhibiting the viability and colony formation in two gastric cancer cell lines. These results confirmed that Y14 exerted anti-gastric activity and chemosensitizing effect by inhibiting FGFR1 phosphorylation and its downstream signaling pathway in vitro. This work also provides evidence that Y14, an effective FGFR1 inhibitor, could be used alone or in combination with chemotherapy to treat gastric cancer in the future.

4.
Cancer Chemother Pharmacol ; 86(2): 257-266, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32696214

RESUMEN

PURPOSE: Inherent and/or acquired multi-drug resistance might be the instigator of treatment failure for acute myeloid leukemia (AML). In the current study, we aimed to explored the chemosensitizing effect of 4-HPR on AML therapy. METHODS: Luciferase reporter assays were used to test the effect of 4-HPR on transcriptional signaling pathways. The quantitative real-time polymerase chain reaction and immunoblots were used to confirm the role of 4-HPR in NF-κB inhibition, apoptosis, and drug resistance. MTT and flow cytometry assays were applied to test the drug response and chemosensitizing effect of 4-HPR with AML cell lines and primary AML samples. RESULTS: 4-HPR suppressed tumor necrosis factor-α- and daunorubin-induced NF-κB activation in AML cell lines. The expression of anti-apoptotic gene, BCL2, was downregulated, while expressions of pro-apoptotic genes, cIAP, XIAP, and BID, were increased after 4-HPR treatment. Immunoblots showed decreased p65-NF-κB, IκBα, and MDR1, but increased cleaved poly (ADP-ribose) polymerase and BIM. A low concentration of 4-HPR chemosensitized AML cells to daunorubin treatment in vitro. CONCLUSION: 4-HPR-induced NF-κB inhibition was the main driver of the chemosensitizing effect observed in AML cell lines and primary AML samples. These results highlight that 4-HPR might be a promising chemosensitizing agent in AML therapy.


Asunto(s)
Daunorrubicina/farmacología , Sinergismo Farmacológico , Fenretinida/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Leucemia Mieloide Aguda/tratamiento farmacológico , FN-kappa B/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/antagonistas & inhibidores , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Antibióticos Antineoplásicos/farmacología , Antineoplásicos/farmacología , Apoptosis , Proliferación Celular , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , FN-kappa B/metabolismo , Células Tumorales Cultivadas
5.
Curr Top Med Chem ; 19(32): 2940-2957, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31738137

RESUMEN

Natural product therapy has been gaining therapeutic importance against various diseases, including cancer. The failure of chemotherapy due to its associated adverse effects promoted adjunct therapy with natural products. Phytochemicals exert anti-carcinogenic activities through the regulation of various cell signaling pathways such as cell survival, inflammation, apoptosis, autophagy and metastasis. The 'small molecule-chemosensitizing agents' from plants induce apoptosis in drug-resistant and host-immune resistant cancer cells in in vitro as well as in vivo models. For example, alkaloids from Nelumbo nucifera, liensinine, isoliensinine and neferine exert the anticancer activity through enhanced ROS generation, activation of MAP kinases, followed by induction of autophagy and apoptotic cell death. Likewise, these alkaloids also exert their cytoprotective action against cerebrovascular stroke/ischemic stroke, diabetes, and chemotherapy-induced cytotoxicity. Therefore, the present review elucidates the pharmacological activities of these bisbenzylisoquinoline alkaloids which include the cytoprotective, anticancer and chemosensitizing abilities against various diseases such as cardiovascular diseases, neurological diseases and cancer.


Asunto(s)
Alcaloides/farmacología , Antineoplásicos/farmacología , Bencilisoquinolinas/farmacología , Citoprotección/efectos de los fármacos , Nelumbo/química , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Humanos , Transducción de Señal/efectos de los fármacos
6.
Life Sci ; 217: 155-163, 2019 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-30528773

RESUMEN

AIMS: Metformin (Met), an essential antidiabetic agent, shows antitumor activity in some cancers. A previous study showed that Met enhanced cytotoxic activity of cisplatin (Cis) in cholangiocarcinoma (CCA) in association with the activation of AMP-activated protein kinase and suppression of Akt-mTOR. However, these effects do not entirely explain the observed chemosensitizing effect. The present study investigated the interaction of Met and Cis over the enhanced antitumor effect. MAIN METHODS: KKU-100 and KKU-M156 cells were used in the study. Cytotoxicity was assessed by acridine orange-ethidium bromide staining. Reactive oxygen species (ROS) and mitochondrial transmembrane potential (Δψm) were measured by dihydroethidium and JC-1 fluorescent methods. Cellular glutathione (GSH) and redox ratio were analyzed by enzymatic coupling assay. Proteins associated with antioxidant system and cell death were evaluated by western immunoblot. KEY FINDINGS: Cytotoxicity of Cis was enhanced by Met in association with ROS formation and GSH redox stress. The antioxidants, N-acetylcysteine and TEMPOL, and MPTP inhibitor, cyclosporine, attenuated cytotoxicity in association with suppression of ROS formation and the losses of Δψm. Met in combination with Cis suppressed expression of Nrf2 and altered the expression of Bcl2 family proteins. SIGNIFICANCE: The chemosensitizing effect of Met in combination with Cis is causally associated with increased oxidative stress-mediated mitochondrial cell death pathway. Met may improve the efficacy of Cis in the treatment of cancer.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Colangiocarcinoma/tratamiento farmacológico , Cisplatino/farmacología , Hipoglucemiantes/farmacología , Metformina/farmacología , Estrés Oxidativo/efectos de los fármacos , Apoptosis/efectos de los fármacos , Neoplasias de los Conductos Biliares/metabolismo , Neoplasias de los Conductos Biliares/patología , Línea Celular Tumoral , Colangiocarcinoma/metabolismo , Colangiocarcinoma/patología , Sinergismo Farmacológico , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Especies Reactivas de Oxígeno/metabolismo
7.
Front Physiol ; 9: 911, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30083105

RESUMEN

Colorectal cancer (CRC) is continuously classified as one of the most incidental and mortal types of cancer worldwide. The positive outcomes of the conventional chemotherapy are frequently associated with high toxicity, which often leads to the suspension of the treatment. Growing evidences consider the use of pharmacological concentrations of ascorbic acid (AA), better known as vitamin C, in the treatment of cancer. The use of AA in a clinical context is essentially related to the adoption of new therapeutic strategies based on combination regimens, where AA plays a chemosensitizing role. The reduced sensitivity of some tumors to chemotherapy and the highly associated adverse effects continue to be some of the major obstacles in the effective treatment of CRC. So, this paper aimed to study the potential of a new therapeutic approach against this neoplasia with diminished side effects for the patient. This approach was based on the study of the combination of high concentrations of AA with reduced concentrations of drugs conventionally used in CRC patients and eligible for first and second line chemotherapeutic regimens, namely 5-fluorouracilo (5-FU), oxaliplatin (Oxa) or irinotecan (Iri). The evaluation of the potential synergy between the compounds was first assessed in vitro in three CRC cell lines with different genetic background and later in vivo using one xenograft animal model of CRC. AA and 5-FU act synergistically in vitro just for longer incubation times, however, in vivo showed no benefit compared to 5-FU alone. In contrast to the lack of synergy seen in in vitro studies with the combination of AA with irinotecan, the animal model revealed the therapeutic potential of this combination. AA also potentiated the effect of Oxa, since a synergistic effect was demonstrated, in almost all conditions and in the three cell lines. Moreover, this combined therapy (CT) caused a stagnation of the tumor growth rate, being the most promising tested combination. Pharmacological concentrations of AA increased the efficacy of Iri and Oxa against CRC, with promising results in cell lines with more aggressive phenotypes, namely, tumors with mutant or null P53 expression and tumors resistant to chemotherapy.

8.
ACS Appl Mater Interfaces ; 10(32): 26870-26881, 2018 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-30039695

RESUMEN

In this work, we investigate whether S-nitrosoglutathione (GSNO)-conjugated hyaluronic acid-based self-assembled nanoparticles (GSNO-HANPs) can be useful as a chemosensitizing agent to improve the anticancer activity of doxorubicin (DOX). The GSNO-HANPs were prepared by aqueous assembly of GSNO-conjugated HA with grafted poly(lactide- co-glycolide). Aqueous GSNO stability shielded within the assembled environments of the GSNO-HANPs was greatly enhanced, compared to that of free GSNO. The NO release from the GSNO-HANPs was facilitated in the presence of hyaluronidase-1 (Hyal-1) and ascorbic acid at intracellular concentrations. Microscopic analysis showed GSNO-HANPs effectively generated NO within the cells. We observed that NO made the human MCF-7 breast cancer cells vulnerable to DOX. This chemosensitizing activity was supported by the observation of an increased level of ONOO- (peroxynitrite), a highly reactive oxygen species, upon co-treatment with the GSNO-HANPs and DOX. Apoptosis assays showed that GSNO-HANP alone exhibited negligible cytotoxic effects and reinforced apoptotic activity of DOX. Animal experiments demonstrated the effective accumulation of GSNO-HANPs in solid MCF-7 tumors and effectively suppressed tumor growth in combination with DOX. This hyaluronic acid-based intracellularly NO-releasing nanoparticles may serve as a significant chemosensitizing agent in treatments of various cancers.


Asunto(s)
Ácido Hialurónico/química , Animales , Citoplasma , Doxorrubicina , Sistemas de Liberación de Medicamentos , Humanos , Células MCF-7 , Nanopartículas , Óxido Nítrico
9.
Life Sci ; 207: 172-183, 2018 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-29847773

RESUMEN

AIMS: AMP-activated protein kinase (AMPK) functions as a cellular energy sensor regulating various aspects of cellular metabolism. Metformin (Met), an activator of AMPK, has been reported to reduce the cancer risk and enhance antitumor effects in certain cancers. Cholangiocarcinoma (CCA) is an aggressive malignancy which rarely responds to chemotherapeutic agents. We investigated the chemosensitizing effects of Met in CCA cells. MATERIALS AND METHODS: KKU-100 and KKU-452 cells were used in the study. Antiproliferation of Met and cisplatin (Cis) was analyzed by sulforhodamine B and colony forming assays. Apoptotic cell death was analyzed by acridine orange and ethidium bromide staining method. Cell cycle analysis was performed by flow cytometric method. Effects on cell migration and invasion were analyzed by wound healing assay and transwell chamber method. Expression of proteins was examined by western blot analysis. KEY FINDINGS: Met enhanced the antiproliferation of Cis, and conferred antimigration and anti-invasion in CCA cells, where Cis alone did not have two latter effects. This chemosensitizing effect is related to the activation of AMPK and suppression of Akt, mTOR and p70S6K. Met and Cis increased expression of p53 and p21 and suppressed expression of cyclin D1. This effect was associated with cell cycle arrest at S phase. The anti-invasion effect was casually associated with the suppression of FAK expression. The cytotoxic effect of the drug combination was mimicked by AICAR, an AMPK agonist. SIGNIFICANCE: Met may be a novel agent to increase the efficacy of Cis to treat CCA.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Neoplasias de los Conductos Biliares/metabolismo , Colangiocarcinoma/metabolismo , Cisplatino/administración & dosificación , Metformina/administración & dosificación , Serina-Treonina Quinasas TOR/metabolismo , Antineoplásicos , Apoptosis , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Ciclo Celular , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Colangiocarcinoma/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Activación Enzimática , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica , Rodaminas/administración & dosificación , Transducción de Señal , Cicatrización de Heridas
10.
J Ethnopharmacol ; 205: 33-40, 2017 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-28456578

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Cryptotanshinone, a natural compound isolated from the roots of Salvia miltiorrhiza Bge. (Danshen), is a commonly used traditional Chinese medicine to treat high blood pressure in some countries. It has been shown that Cryptotanshinone induces cancer cells apoptosis and impairs cell migration and invasion. However, the antiproliferation and chemosensitization effects of Cryptotanshinone on ovarian cancer and the underlying mechanism are not fully elucidated. AIM OF STUDY: In this study, we evaluated the inhibitory effect of Cryptotanshinone on ovarian cancer cells and explored the underlying molecular mechanism. Additionally, the chemosensitization potential of Cryptotanshinone was evaluated in combination with cisplatin. MATERIALS AND METHODS: MTT assay was used for cell viability assessment of ovarian cancer A2780 cells treated with Cryptotanshinone and/ or cisplatin. Flow cytometry was used for apoptosis analysis. Wound healing and transwell assays were used for migratory and invasive potential assessment of Cryptotanshinone-treated ovarian cancer cells. Western blot was used to investigate proteins involved in the mechanisms for metastasis and apoptosis. γH2AX immunocytochemistry was used to detect DNA damage in A2780 cells exposed to Cryptotanshinone and/or cisplatin. RESULTS: Cryptotanshinone significantly induced ovarian cancer A2780 cells apoptosis by activating caspase cascade. Additionally, wound healing and transwell assays revealed that Cryptotanshinone could suppress migration and invasion of ovarian cancer cells and dramatically inhibited MMP-2 and MMP-9 expression. Furthermore, Cryptotanshinone could sensitize A2780 cells to cisplatin treatment in a dose-dependent manner. CONCLUSION: Our data confirmed the anti-tumor effect of Cryptotanshinone on ovarian cancer cells and provided new findings that Cryptotanshinone could sensitize ovarian cancer cells to chemotherapy.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias Ováricas/tratamiento farmacológico , Fenantrenos/uso terapéutico , Salvia miltiorrhiza/química , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/química , Apoptosis , Línea Celular Tumoral , Daño del ADN , Relación Dosis-Respuesta a Droga , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Estructura Molecular , Invasividad Neoplásica , Fenantrenos/administración & dosificación , Fenantrenos/química , Extractos Vegetales/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA