Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
Int J Mol Sci ; 24(4)2023 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-36834960

RESUMEN

Bevacizumab (Bev) a humanized monoclonal antibody that fights vascular endothelial growth factor A (VEGF-A). It was the first specifically considered angiogenesis inhibitor and it has now become the normative first-line therapy for advanced non-small-cell lung cancer (NSCLC). In the current study, polyphenolic compounds were isolated from bee pollen (PCIBP) and encapsulated (EPCIBP) inside moieties of hybrid peptide-protein hydrogel nanoparticles in which bovine serum albumin (BSA) was combined with protamine-free sulfate and targeted with folic acid (FA). The apoptotic effects of PCIBP and its encapsulation (EPCIBP) were further investigated using A549 and MCF-7 cell lines, providing significant upregulation of Bax and caspase 3 genes and downregulation of Bcl2, HRAS, and MAPK as well. This effect was synergistically improved in combination with Bev. Our findings may contribute to the use of EPCIBP simultaneously with chemotherapy to strengthen the effectiveness and minimize the required dose.


Asunto(s)
Antineoplásicos , Bevacizumab , Productos Biológicos , Carcinoma de Pulmón de Células no Pequeñas , Hidrogeles , Animales , Humanos , Células A549/efectos de los fármacos , Células A549/metabolismo , Inhibidores de la Angiogénesis/uso terapéutico , Antineoplásicos/química , Antineoplásicos/farmacología , Abejas/química , Abejas/metabolismo , Bevacizumab/uso terapéutico , Productos Biológicos/química , Productos Biológicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Hidrogeles/química , Hidrogeles/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Células MCF-7/efectos de los fármacos , Células MCF-7/metabolismo , Nanopartículas/química , Nanopartículas/uso terapéutico , Polen/química , Polen/metabolismo , Factor A de Crecimiento Endotelial Vascular/uso terapéutico
2.
Int J Mol Sci ; 23(14)2022 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-35886884

RESUMEN

Claudin-2 (CLDN2), a component of tight junction, is involved in the reduction of anticancer drug-induced toxicity in spheroids of A549 cells derived from human lung adenocarcinoma. Fisetin, a dietary flavonoid, inhibits cancer cell growth, but its effect on chemosensitivity in spheroids is unknown. Here, we found that fisetin (20 µM) decreases the protein level of CLDN2 to 22.3%. Therefore, the expression mechanisms were investigated by real-time polymerase chain reaction and Western blotting. Spheroids were formed in round-bottom plates, and anticancer drug-induced toxicity was measured by ATP content. Fisetin decreased the phosphorylated-Akt level, and CLDN2 expression was decreased by a phosphatidylinositol 3-kinase (PI3K) inhibitor, suggesting the inhibition of PI3K/Akt signal is involved in the reduction of CLDN2 expression. Hypoxia level, one of the hallmarks of tumor microenvironment, was reduced by fisetin. Although fisetin did not change hypoxia inducible factor-1α level, it decreased the protein level of nuclear factor erythroid 2-related factor 2, a stress response factor, by 25.4% in the spheroids. The toxicity of doxorubicin (20 µM) was enhanced by fisetin from 62.8% to 40.9%, which was rescued by CLDN2 overexpression (51.7%). These results suggest that fisetin can enhance anticancer drug toxicity in A549 spheroids mediated by the reduction of CLDN2 expression.


Asunto(s)
Adenocarcinoma del Pulmón , Antineoplásicos , Flavonoles , Neoplasias Pulmonares , Células A549/efectos de los fármacos , Células A549/metabolismo , Adenocarcinoma del Pulmón/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Línea Celular Tumoral , Proliferación Celular , Claudina-2/genética , Claudina-2/metabolismo , Flavonoides/farmacología , Flavonoides/uso terapéutico , Flavonoles/farmacología , Humanos , Hipoxia , Neoplasias Pulmonares/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/metabolismo , Microambiente Tumoral
3.
Int J Exp Pathol ; 102(4-5): 192-199, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34716956

RESUMEN

MicroRNAs (miRNAs) have been demonstrated to play pivotal roles in the pathogenesis of sepsis-induced acute lung injury (ALI). In this work, we aimed to clarify the potential role and the underlying mechanism of miR-942-5p in a lipopolysaccharide (LPS)-induced A549 cell injury model. The cell injury was evaluated by CCK-8 assay, flow cytometry and enzyme-linked immunosorbent assay (ELISA). The expression levels of miR-942-5p and tripartite motif-containing protein 37 (TRIM37) were measured by real-time PCR and Western blot, and their association was then validated by bioinformatics, luciferase reporter assay and RNA pull-down assay. We found that the expression of miR-942-5p was decreased in LPS-treated A549 cells. Furthermore, LPS treatment suppressed A549 cell viability, promoted apoptosis and increased the levels of inflammatory cytokines. Conversely, overexpression of miR-942-5p increased cell viability, reduced apoptosis and alleviated inflammatory cytokine secretion in the presence of LPS. Moreover, miR-942-5p directly targeted TRIM37 by binding to the 3'-UTR of TRIM37 mRNA. Upregulation of TRIM37 effectively reversed the anti-apoptotic and anti-inflammatory effects of miR-942-5p in LPS-induced A549 cells. Our findings suggested that miR-942-5p protected against LPS-induced cell injury through inhibiting apoptosis and inflammation in A549 cells by negatively regulating TRIM37.


Asunto(s)
Lesión Pulmonar Aguda , MicroARNs/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Células A549/efectos de los fármacos , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/metabolismo , Apoptosis , Línea Celular , Citocinas/metabolismo , Humanos , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Sepsis/inducido químicamente , Sepsis/complicaciones
4.
J Appl Toxicol ; 41(3): 470-482, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33022792

RESUMEN

Cetylpyridinium chloride (CPC), a quaternary ammonium compound and cationic surfactant, is used in personal hygiene products such as toothpaste, mouthwash, and nasal spray. Although public exposure to CPC is frequent, its pulmonary toxicity has yet to be fully characterized. Due to high risks of CPC inhalation, we aimed to comprehensively elucidate the in vitro and in vivo toxicity of CPC. The results demonstrated that CPC is highly cytotoxic against the A549 cells with a half-maximal inhibitory concentration (IC50 ) of 5.79 µg/ml. Following CPC exposure, via intratracheal instillation (ITI), leakage of lactate dehydrogenase, a biomarker of cell injury, was significantly increased in all exposure groups. Further, repeated exposure of rats to CPC for 28 days caused a decrease in body weight of the high-exposure group and the relative weights of the lungs and kidneys of the high recovery group, but no changes were evident in the histological and serum chemical analyses. The bronchoalveolar lavage fluid (BALF) analysis showed a significant increase in proinflammatory cytokines interleukin (IL)-6, IL-1ß, and tumor necrosis factor (TNF)-α levels. ITI of CPC induced focal inflammation of the pulmonary parenchyma in rats' lungs. Our study demonstrated that TNF-α was the most commonly secreted proinflammatory cytokine during CPC exposure in both in vitro and in vivo models. Polymorphonuclear leukocytes in the BALF, which are indicators of pulmonary inflammation, significantly increased in a concentration-dependent manner in all in vivo studies including the ITI, acute, and subacute inhalation assays, demonstrating that PMNs are the most sensitive parameters of pulmonary toxicity.


Asunto(s)
Células A549/efectos de los fármacos , Antiinfecciosos Locales/toxicidad , Supervivencia Celular/efectos de los fármacos , Cetilpiridinio/toxicidad , Neumonía/inducido químicamente , Neumonía/fisiopatología , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Ratas , Ratas Sprague-Dawley
5.
Mar Drugs ; 19(8)2021 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-34436252

RESUMEN

Ochraceopetalin (1), a mixed-biogenetic salt compound and its component 2 were isolated from the culture broths of a marine-derived fungus, Aspergillus ochraceopetaliformis. Based on combined spectroscopic and chemical analyses, the structure of 1 was determined to be a sulfonated diphenylether-aminol-amino acid ester guanidinium salt of an unprecedented structural class, while 2 was determined to be the corresponding sulfonated diphenylether. Ochraceopetaguanidine (3), the other guanidine-bearing aminol amino acid ester component, was also prepared and structurally elucidated. Compound 1 exhibited significant cytotoxicity against K562 and A549 cells.


Asunto(s)
Antineoplásicos/farmacología , Aspergillus/química , Células A549/efectos de los fármacos , Organismos Acuáticos , Humanos , Células K562/efectos de los fármacos , Relación Estructura-Actividad
6.
Mar Drugs ; 19(8)2021 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-34436277

RESUMEN

Four new cytochalasans, phychaetoglobins A-D (1-4), together with twelve known cytochalasans (5-16), were isolated from a mangrove-associated fungus Chaetomium globosum kz-19. The new structures were elucidated on the basis of extensive 1D and 2D NMR, HR ESIMS spectroscopic analyses, and electronic circular dichroism (ECD) calculations. The absolute configuration of 2 was established by application of Mosher's method. Compounds 4-8 exhibited moderate cytotoxicities against A549 and HeLa cell lines with the IC50 values less than 20 µM.


Asunto(s)
Antineoplásicos/química , Chaetomium , Citocalasinas/química , Células A549/efectos de los fármacos , Antineoplásicos/farmacología , Organismos Acuáticos , Citocalasinas/farmacología , Células HeLa/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Fitoterapia
7.
Mar Drugs ; 19(11)2021 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-34822512

RESUMEN

Angiogenesis, including the growth of new capillary blood vessels from existing ones and the malignant tumors cells formed vasculogenic mimicry, is quite important for the tumor metastasis. Anti-angiogenesis is one of the significant therapies in tumor treatment, while the clinical angiogenesis inhibitors usually exhibit endothelial cells dysfunction and drug resistance. Bis(2,3,6-tribromo-4,5-dihydroxybenzyl)ether (BTDE), a marine algae-derived bromophenol compound, has shown various biological activities, however, its anti-angiogenesis function remains unknown. The present study illustrated that BTDE had anti-angiogenesis effect in vitro through inhibiting human umbilical vein endothelial cells migration, invasion, tube formation, and the activity of matrix metalloproteinases 9 (MMP9), and in vivo BTDE also blocked intersegmental vessel formation in zebrafish embryos. Moreover, BTDE inhibited the migration, invasion, and vasculogenic mimicry formation of lung cancer cell A549. All these results indicated that BTDE could be used as a potential candidate in anti-angiogenesis for the treatment of cancer.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Microalgas , Fenoles/farmacología , Células A549/efectos de los fármacos , Inhibidores de la Angiogénesis/química , Animales , Organismos Acuáticos , Proliferación Celular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Neovascularización Fisiológica/efectos de los fármacos , Fenoles/química
8.
Pharm Dev Technol ; 26(7): 797-806, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34219578

RESUMEN

The purpose of this study was to investigate the impact of anticancer drug erlotinib-randomly methylated-ß-cyclodextrin complex (ERL-RAMEB CD) on drug solubility and dissolution rate. Phase solubility study showed erlotinib displayed maximum solubility in RAMEB CD solution and the stability constant (Kc) was calculated to be 370 ± 16 M-1. The optimal formulation was obtained with ERL-RAMEB CD in a 1:1 molar ratio using the co-lyophilization technique. Differential scanning calorimetry (DSC) and Scanning electron microscopy (SEM) verified the inclusion of complex formation. In vitro dissolution study confirmed ERL-RAMEB CD as a favorable approach to increase drug dissolution with a 1.5-fold increase than free ERL at 1 h. An improved dissolution with ∼88.4% drug release at 1 h was observed, in comparison with Erlotinib with ∼58.7% release in 45 min. The in vitro cytotoxicity results indicated that the ERL-RAMEB CD inclusion complex reduced cell viability than free erlotinib. Caco-2 cell uptake that is indicative of drug intestinal permeability resulted in a 5-fold higher uptake of ERL-RAMEB CD inclusion complex than the ERL solution. Hence, ERL-RAMEB CD complexation displays a strong potential to increase dissolution and permeability of erlotinib leading eventually to enhanced oral bioavailability.


Asunto(s)
Antineoplásicos/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Clorhidrato de Erlotinib/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Células A549/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Células CACO-2/efectos de los fármacos , Rastreo Diferencial de Calorimetría , Línea Celular Tumoral/efectos de los fármacos , Liberación de Fármacos , Clorhidrato de Erlotinib/uso terapéutico , Humanos , Absorción Intestinal , Metilación , Ratones , Microscopía Electrónica de Rastreo , Solubilidad , Resultado del Tratamiento , beta-Ciclodextrinas/administración & dosificación , beta-Ciclodextrinas/uso terapéutico
9.
Mol Biol Rep ; 47(5): 3765-3778, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32378168

RESUMEN

The importance of microbial natural compounds in drug research is increasing every year and they are used to prevent or treat a variety of diseases. Hypomyces chrysospermus is a cosmopolitan parasite of many Boletaceae members. Since not much work has been conducted to date, this study is undertaken to explore the anticancer effect, including the antiproliferative and antimetastatic activity of Hypomyces chrysospermus. The aim of this study is to determine the antiproliferative and antimetastatic activity of Hypomyces chrysospermus ethyl acetate extract, having antioxidant activity, against A549, Caco2, MCF-7 human cancer and CCD-19 Lu and CCD 841 CoN healthy human cell lines. Firstly, cytotoxic activity was determined by the WST-1 assay. After cell proliferations and anti-metastatic effects were investigated by a real-time cell analysis system (RTCA-DP) and IC50 concentrations were calculated for each cell line. In addition, the expression levels of Apaf-1, TNF and NF-kB mRNA in cancer cells were investigated with RealTime-PCR. The ethyl acetate extract of Hypomyces chrysospermus presented anticancer activities including antiproliferative and antimetastatic effects. Hypomyces chrysospermus as a source of biologically active metabolites can be used as an important resource in the development of new anticancer effective agents.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Hypocreales/metabolismo , Células A549/efectos de los fármacos , Antineoplásicos/farmacología , Antioxidantes/farmacología , Factor Apoptótico 1 Activador de Proteasas/metabolismo , Basidiomycota/patogenicidad , Células CACO-2/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Hypocreales/fisiología , Células MCF-7/efectos de los fármacos , FN-kappa B/metabolismo , Extractos Vegetales/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
10.
Mol Biol Rep ; 47(6): 4155-4168, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32444975

RESUMEN

Plumbagin (PL) is a natural naphthoquinone compound, isolated from Plumbago zeylanica that has cytotoxic and antimigratory potential in many cancer. However, the cytotoxic mechanism of plumbagin in drug resistant lung cancer is poorly understood. To reveal the mechanism, we studied the anticancer effect of plumbagin in both gefitinib-sensitive and resistant A549 lung cancer cells. The anticancer potential of PL was demonstrated by MTT assay and the result suggested that PL showed cytotoxicity in both gefitinib-sensitive (A549) and gefitinib-resistant (A549GR) lung cancer cells. IC50 values of PL in A549 and A549GR were 3.2 µM and 4.5 µM, respectively. Morphological changes were also observed after treatment with PL. Furthermore, PL decreased cell survival by inhibiting colony formation ability, and inhibited cell migration at very low concentrations. From Annexin V-FITC/PI, AO/EtBr, and DAPI staining, we found that increasing concentration of PL leads to increase in apoptosis of lung cancer cells. Furthermore, western blotting results suggested that Bax and Caspase 3 levels were upregulated after PL treatment. In addition, treatment of PL caused DNA damage in a dose-dependent manner. PL arrested the cell cycle at S-G2/M phase, and enhanced reactive oxygen species (ROS) generation. Excess ROS generated by PL disrupted mitochondrial membrane resulted in depletion of mitochondrial membrane potential (MMP). These results conclude that PL decreases lung cancer cell viability by arresting cells at S-G2/M phase, and induces apoptosis by activation of mitochondrial-mediated apoptotic pathway through excess ROS generation. Overall findings suggest that plumbagin shows cytotoxic and therapeutic potential against both A549 and A549GR cell lines.


Asunto(s)
Células A549/efectos de los fármacos , Naftoquinonas/metabolismo , Naftoquinonas/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Gefitinib/farmacología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo
11.
Exp Parasitol ; 218: 107983, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32861680

RESUMEN

Trichinella spiralis (T. spiralis) muscle larvae (ML) excretory/secretory products (ESPs) are antitumor substances extracted from the culture medium of T. spiralis ML. The ESPs inhibit tumor growth and induce tumor cell apoptosis. To explore the effects of these products on the non-small-cell lung cancer (NSCLC) line A549, logarithmically growing A549 cells were co-cultured with different concentrations of T. spiralis ML ESPs for 24, 36 and 48 h. Our results showed that T. spiralis ML ESPs significantly inhibited A549 cells proliferation, which was dose-and time-dependent. To evaluate the inhibition by T. spiralis ML ESPs of the growth of A549 cells, we assayed their apoptosis and cell-cycle distribution by flow cytometry (FCM). To determine whether ESPs induced apoptosis of A549 cells via the mitochondrial pathway, we evaluated the levels of mitochondrion-related factors by Western blotting. The FCM indicated a clear trend toward apoptosis of A549 cells co-cultured with ESPs for 24 h. The cells were blocked in S-phase. Western blotting revealed that the expression levels of the genes encoding Bax, caspase-3, and caspase-9 increased (compared to a control group), and the Bcl-2 gene expression level decreased. Our results suggest that T. spiralis ML ESPs induce apoptosis of the NSCLC line A549 via the mitochondrial pathway; the cells become arrested in S-phase. This may explain the antineoplastic activity of T. spiralis ML ESPs.


Asunto(s)
Células A549/efectos de los fármacos , Antígenos Helmínticos/farmacología , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proteínas del Helminto/farmacología , Fase S/efectos de los fármacos , Trichinella spiralis/química , Células A549/citología , Análisis de Varianza , Animales , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/patología , Proliferación Celular/efectos de los fármacos , Medio de Cultivo Libre de Suero , Femenino , Humanos , Larva/química , Ratones , Trichinella spiralis/inmunología
12.
J Appl Toxicol ; 40(9): 1228-1238, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32220024

RESUMEN

Broad application of reduced graphene oxide (rGO) and ubiquitous lead (Pb) pollution may increase the possibility of combined exposure of humans. Information on the combined effects of rGO and Pb in human cells is scarce. This work was designed to explore the potential effects of rGO on Pb-induced toxicity in human alveolar epithelial (A549) cells. Prepared rGO was polycrystalline in nature. The formation of a few layers of visible creases and silky morphology due to high aspect ratio was confirmed. Low level (25 µg/mL) of rGO was not toxic to A549 cells. However, Pb exposure (25 µg/mL) induced cell viability reduction, lactate dehydrogenase enzyme leakage with rounded morphology in A549 cells. Remarkably, Pb-induced cytotoxicity was significantly mitigated by rGO co-exposure. Pb-induced mitochondrial membrane potential loss, cell cycle arrest and higher activity of caspase-3 and -9 enzymes were also alleviated by rGO co-exposure. Moreover, we observed that Pb exposure causes generation of pro-oxidants (e.g., reactive oxygen species, hydrogen peroxide and lipid peroxidation) and antioxidant depletion (e.g., glutathione and antioxidant enzymes). In addition, the effects of Pb on pro-oxidant and antioxidant markers were significantly reverted by GO co-exposure. Inductively coupled plasma-mass spectrometry suggested that due to the adsorption of Pb on rGO sheets, accessibility of Pb ions for A549 cells was limited. Hence, rGO reduced the toxicity of Pb in A549 cells. This research warrants further study to work on detailed underlying mechanisms of the mitigating effects of rGO against Pb-induced toxicity on a molecular level.


Asunto(s)
Células A549/efectos de los fármacos , Citotoxinas/toxicidad , Grafito/toxicidad , Plomo/toxicidad , Estrés Oxidativo/efectos de los fármacos , Exposición a Riesgos Ambientales , Humanos
13.
Mar Drugs ; 18(3)2020 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-32121035

RESUMEN

Prostaglandin A2-AcMe (1) and Prostaglandin A2 (2) were isolated from the octocoral Plexaura homomalla and three semisynthetic derivatives (3-5) were then obtained using a reduction protocol. All compounds were identified through one- and two-dimensional (1D and 2D) nuclear magnetic resonance (NMR) experiments. Additionally, evaluation of in vitro cytotoxic activity against the breast (MDA-MB-213) and lung (A549) cancer cell lines, in combination with enzymatic activity and molecular docking studies with the enzymes p38α-kinase, Src-kinase, and topoisomerase IIα, were carried out for compounds 1-5 in order to explore their potential as inhibitors of cancer-related molecular targets. Results showed that prostaglandin A2 (2) was the most potent compound with an IC50 of 16.46 and 25.20 µg/mL against MDA-MB-213 and A549 cell lines, respectively. In addition, this compound also inhibited p38α-kinase in 49% and Src-kinase in 59% at 2.5 µM, whereas topoisomerase IIα was inhibited in 64% at 10 µM. Enzymatic activity was found to be consistent with molecular docking simulations, since compound 2 also showed the lowest docking scores against the topoisomerase IIα and Src-kinase (-8.7 and -8.9 kcal/mol, respectively). Thus, molecular docking led to establish some insights into the predicted binding modes. Results suggest that prostaglandin 2 can be considered as a potential lead for development inhibitors against some enzymes present in cancer processes.


Asunto(s)
Antozoos , Antineoplásicos/farmacología , Prostaglandinas/farmacología , Células A549/efectos de los fármacos , Animales , Línea Celular Tumoral/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Océanos y Mares
14.
Mar Drugs ; 18(4)2020 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-32290492

RESUMEN

The anti-inflammatory and anticancer properties of eight meroterpenoids isolated from the brown seaweed Cystoseira usneoides have been evaluated. The algal meroterpenoids (AMTs) 1-8 were tested for their inhibitory effects on the production of the pro-inflammatory cytokines tumor necrosis factor (TNF-α), interleukin-6 (IL-6), and interleukin-1ß (IL-1ß), and the expression of cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS) in LPS-stimulated THP-1 human macrophages. The anticancer effects were assessed by cytotoxicity assays against human lung adenocarcinoma A549 cells and normal lung fibroblastic MRC-5 cells, together with flow cytometry analysis of the effects of these AMTs on different phases of the cell cycle. The AMTs 1-8 significantly reduced the production of TNF-α, IL-6, and IL-1ß, and suppressed the COX-2 and iNOS expression, in LPS-stimulated cells (p < 0.05). The AMTs 1-8 displayed higher cytotoxic activities against A549 cancer cells than against MRC-5 normal lung cells. Cell cycle analyses indicated that most of the AMTs caused the arrest of A549 cells at the G2/M and S phases. The AMTs 2 and 5 stand out by combining significant anti-inflammatory and anticancer activities, while 3 and 4 showed interesting selective anticancer effects. These findings suggest that the AMTs produced by C. usneoides may have therapeutic potential in inflammatory diseases and lung cancer.


Asunto(s)
Antiinflamatorios/química , Antineoplásicos/química , Organismos Acuáticos , Phaeophyceae , Terpenos/química , Células A549/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Antineoplásicos/farmacología , Citocinas/efectos de los fármacos , Humanos , Terpenos/farmacología
15.
Bioorg Med Chem ; 27(13): 2845-2856, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31103402

RESUMEN

Biological activities of a series of fluorescent compounds against human lung cancer cell line A549 were investigated. The results showed that (E)-1,3,3-trimethyl-2-(4-(piperidin-1-yl)styryl)-3H-indol-1-ium iodide (8) and (E)-2-(5,5-dimethyl-3-(4-(piperazin-1-yl)styryl)cyclohex-2-en-1-ylidene) malononitrile (11) could inhibit the growth of A549 cancer cells in a dose and time-dependent manner. Furthermore, compound 8 could trigger autophagy and apoptosis, but not obviously induce necrosis under the stimulatory condition. Therefore, 8 can be used as autophagy activator to investigate the regulatory mechanism of autophagy and may offer a new candidate for the treatment of lung cancer.


Asunto(s)
Células A549/efectos de los fármacos , Autofagia/efectos de los fármacos , Neoplasias Pulmonares/fisiopatología , Humanos , Estructura Molecular
16.
Mol Biol Rep ; 46(1): 133-141, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30374769

RESUMEN

Urease is a potent metalloenzyme with diverse applications. This paper describes the scale up and purification of an extracellular urease from Arthrobacter creatinolyticus MTCC 5604. The urease production was scaled-up in 3.7 L and 20 L fermentor. A maximum activity of 27 and 27.8 U/mL and a productivity of 0.90 and 0.99 U/mL/h were obtained at 30 h and 28 h in 3.7 and 20 L fermentor, respectively. Urease was purified to homogeneity with 49.85-fold purification by gel filtration and anion exchange chromatography with a yield of 36% and a specific activity of 1044.37 U/mg protein. The enzyme showed three protein bands with molecular mass of 72.6, 11.2 and 6.1 kDa on SDS-PAGE and ~ 270 kDa on native PAGE. The cytotoxic effect of urease was assessed in vitro using cancer cell lines (A549 and MG-63) and normal cell line (HEK 293). Urease showed its inhibitory effects on cancer cell lines through the generation of toxic ammonia, which in turn increased the pH of the surrounding medium. This increase in extracellular pH, enhanced the cytotoxic effect of weak base chemotherapeutic drugs, doxorubicin (50 µM) and vinblastine (100 µM) in the presence of urease (5 U/mL) and urea (0-4 mM) significantly.


Asunto(s)
Arthrobacter/enzimología , Ureasa/aislamiento & purificación , Ureasa/farmacología , Células A549/efectos de los fármacos , Amoníaco/metabolismo , Arthrobacter/metabolismo , Arthrobacter/fisiología , Línea Celular , Cromatografía en Gel/métodos , Electroforesis en Gel de Poliacrilamida/métodos , Células HEK293 , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Peso Molecular , Urea/metabolismo , Ureasa/fisiología
17.
Mol Biol Rep ; 46(2): 2059-2066, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30725348

RESUMEN

Derived from rosaceous plant seed, amygdalin belongs to aromatic cyanogenic glycoside group, and its anticancer effects have been supported by mounting evidence. In this study, we objected to investigate amygdalin effect on two antiapoptotic genes (Survivin, XIAP) and two lncRNAs (GAS5, MALAT1) in human cancer cells (A549, MCF7, AGS). Employing RT-qPCR analysis, we compared the mRNA levels of the genes related to apoptosis in A549, MCF7, and AGS cancer cells between amygdalin-treated (24, 48 and 72 h) and un-treated groups. RNA was extracted from both cell groups and then cDNAs were synthesized. The changes in the gene expression levels were specified using ΔΔCt method. RT-qPCR analysis has revealed that the expression of Survivin, XIAP, GAS5 and MALAT1 in amygdala-treated cancer cells were significantly different, compared to the un-treated cells. However, these expressions were different depending on the treatment time. According to the results, amygdalin significantly inhibited the expression level of Survivin, and XIAP genes in treated via untreated group. Our findings suggest that amygdalin might have an anticancer effect due to the various gene expressions in A549, MCF7, and AGS human cancer cells, showing it's potential as a natural therapeutic anticancer drug.


Asunto(s)
Amigdalina/farmacología , Survivin/efectos de los fármacos , Proteína Inhibidora de la Apoptosis Ligada a X/efectos de los fármacos , Células A549/efectos de los fármacos , Amigdalina/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7/efectos de los fármacos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética , ARN Nucleolar Pequeño/genética , ARN Nucleolar Pequeño/metabolismo , Survivin/genética , Proteína Inhibidora de la Apoptosis Ligada a X/genética
18.
J Nat Prod ; 82(6): 1527-1534, 2019 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-31117521

RESUMEN

Six new asperane-type sesterterpenoids, asperunguisins A-F (1-6), were isolated from the endolichenic fungus Aspergillus unguis, together with a known analogue, aspergilloxide (7); these are rare asperane-type sesterterpenoids, characterized by a unique hydroxylated 7/6/6/5 tetracyclic system. The structures of asperunguisins A-F (1-6) were elucidated on the basis of spectroscopic methods (NMR and HRESIMS), X-ray single-crystal diffraction analysis, ECD calculations, and biogenetic considerations. Asperunguisin C (3) showed cytotoxicity against the human cancer cell line A549 with an IC50 value of 6.2 µM. Further investigation revealed that the observed cell death was a result of G0/G1 cell cycle arrest via DNA damage followed by cellular apoptosis.


Asunto(s)
Células A549/efectos de los fármacos , Antineoplásicos/farmacología , Aspergillus/química , Hongos/química , Sesterterpenos/química , Sesterterpenos/farmacología , Células A549/química , Antineoplásicos/química , Cristalografía por Rayos X , Humanos , Espectroscopía de Resonancia Magnética , Estructura Molecular , Sesterterpenos/aislamiento & purificación
19.
Med Sci Monit ; 25: 4110-4121, 2019 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-31154455

RESUMEN

BACKGROUND The tumor microenvironment in lung cancer plays an important role in tumor progression and metastasis. Bone marrow-derived mesenchymal stem cells (MSCs) co-cultured with A549 lung cancer cells show changes in morphology, increase cell proliferation, and cell migration. This study aimed to investigate the effects of Astragalus polysaccharide (APS), a traditional Chinese herbal medicine, on the changes induced in bone marrow-derived MSCs by A549 lung cancer cells in vitro. MATERIAL AND METHODS Bone marrow-derived MSCs were co-cultured with A549 cells (Co-BMSCs). Co-cultured bone marrow-derived MSCs and A549 cells treated with 50 µg/ml of APS (Co-BMSCs + APS) were compared with untreated Co-BMSCs. Cell proliferation was measured using the cell counting kit-8 (CCK-8) assay. Flow cytometry evaluated the cell cycle. Microarray assays for mRNA expression and Western blot for protein expression were used. RESULTS Compared with untreated Co-BMSCs, APS treatment of Co-BMSCs improved cell morphology, reduced cell proliferation, and inhibited cell cycle arrest. The mitogen-activated protein kinase (MAPK)/nuclear factor-kappa B (NF-kappaB) pathway, TP53, caspase-3, acetylated H4K5, acetylated H4K8, and acetylated H3K9 were involved in the regulatory process. CONCLUSIONS APS treatment reduced cell proliferation and morphological changes in bone marrow-derived MSCs that were co-cultured with A549 lung cancer cells in vitro.


Asunto(s)
Células A549/efectos de los fármacos , Planta del Astrágalo/metabolismo , Medicamentos Herbarios Chinos/farmacología , Médula Ósea/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , China , Técnicas de Cocultivo , Humanos , Neoplasias Pulmonares/metabolismo , Medicina Tradicional China/métodos , Células Madre Mesenquimatosas/efectos de los fármacos , Polisacáridos/farmacología , Transducción de Señal/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
20.
Med Sci Monit ; 25: 9509-9516, 2019 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-31833479

RESUMEN

BACKGROUND Cancer stem cells (CSCs) behave as their malignant counterparts, but persist after treatment, and possess properties that allow them to interact with their environment. Itraconazole, an antifungal agent, also has a role in suppressing tumor progression, but its effects in regulating tumor cell stemness remain unclear. This study aimed to evaluate the effects of itraconazole on A549 and NCI-H460 human lung cancer cell stemness in vitro. MATERIAL AND METHODS A549 and NCI-H460 human lung cancer cells and BEAS-2B normal bronchial epithelial cells were cultured with and without itraconazole. Cell viability was evaluated. The expression of stem cell markers, CD133, ATP binding cassette subfamily G member 2 (ABCG2), and aldehyde dehydrogenase 1 (ALDH1), were measured by Western blot and quantitative real-time polymerase chain reaction (qRT-PCR). Sphere-forming cells were evaluated in vitro. RESULTS Itraconazole reduced the expression of stemness molecules CD133, ABCG2, and ALDH1 in A549 and NCI-H460 human lung cancer cells, and the numbers of sphere-forming cells were reduced. However, itraconazole had little effect on cell viability but enhanced the chemosensitivity of A549 and NCI-H460 cells. Itraconazole inhibited Wnt signaling. Re-activation of Wnt signaling restored itraconazole-mediated inhibition on A549 and NCI-H460 cell stemness. CONCLUSIONS Itraconazole altered the stemness characteristics of A549 and NCI-H460 human lung cancer cells by suppressing Wnt signaling but did not affect cell viability.


Asunto(s)
Itraconazol/farmacología , Neoplasias Pulmonares/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células A549/efectos de los fármacos , Antígeno AC133/análisis , Antígeno AC133/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/análisis , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Familia de Aldehído Deshidrogenasa 1/análisis , Familia de Aldehído Deshidrogenasa 1/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , China , Humanos , Células Madre Neoplásicas/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA