Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cytokine ; 158: 155977, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35933851

RESUMEN

Adenosine (ADO) generation in the tumor microenvironment (TME) plays important roles in the promotion of tumor growth, invasion, and metastasis and in suppression of the antitumor immune response. Recently, adenosine deaminase (ADA) activity in the TME has been proposed to be a compensatory mechanism against toxic accumulation of ADO in cancerous tissues. In the present study, the expression and functional activity of ADA in cervical cancer (CeCa) tumor cells were analyzed: C33A (HPV-), CaSki (HPV + ), and HeLa (HPV + ) cells. CeCa tumor cells, as well as activated T lymphocytes (ATLs), which were used as a positive control, showed different ADA contents in the membrane and intracellularly and a strong ability to convert ADO into inosine (INO). Treatment of tumor cells with EHNA, a specific ADA inhibitor, decreased the viability of CeCa tumor cells in a dose-dependent manner. In C33A (EHNA half maximal inhibitory concentration (IC50) = 374 µM), CaSki (EHNA IC50 = 273.6 µM), and HeLa (EHNA IC50 = 252.2 µM) cells, EHNA strongly reversed the resistance of tumor cells to the cytotoxic effect of high concentrations of ADO; 38.82 ± 3.1%, 47.18 ± 4.7%, and 71.63 ± 6.9% of the cells were apoptotic, and 40 ± 4.8%, 52 ± 5.3% and 70 ± 6.8% of the cells had mitochondrial membrane damage, respectively. In ATLs (EHNA IC50 = 391.8 µM) treated with EHNA, 32.4 ± 4.4% were apoptotic, and 32 ± 4.3% had mitochondrial membrane damage. These results suggest that the presence and activity of ADA in CeCa tumor cells can provide protection against the cytotoxic effect of high ADO contents in the TME. Therefore, the inhibition of ADA could be a strategy for the treatment of CeCa.


Asunto(s)
Antineoplásicos , Infecciones por Papillomavirus , Neoplasias del Cuello Uterino , Adenina/farmacología , Adenosina/metabolismo , Adenosina/farmacología , Adenosina Desaminasa/metabolismo , Femenino , Humanos , Microambiente Tumoral , Neoplasias del Cuello Uterino/tratamiento farmacológico
2.
MAGMA ; 33(4): 507-513, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31897902

RESUMEN

OBJECTIVE: A phantom for diffusion-weighted imaging is required to standardize quantitative evaluation. The objectives were to develop a phantom simulating various cell densities and to evaluate repeatability. MATERIALS AND METHODS: The acrylic fine particles with three different diameters were used to simulate human cells. Four-degree cell density components were developed by adjusting the volume of 10-µm particles (5, 20, 35, and 50% volume, respectively). Two-degree components to simulate cell edema were also developed by adjusting the diameter without changing number (17% and 40% volume, respectively). Spearman's rank correlation coefficient was used to find a significant correlation between apparent diffusion coefficient (ADC) and particle density. Coefficient of variation (CV) for ADC was calculated for each component for 6 months. A p value < 0.05 represented a statistically significance. RESULTS: Each component (particle ratio of 5, 17, 20, 35, 40, and 50% volume, respectively) presented ADC values of 1.42, 1.30, 1.30, 1.12, 1.09, and 0.89 (× 10-3 mm2/s), respectively. A negative correlation (r = - 0.986, p < 0.05) was observed between ADC values and particle ratio. CV for ADC was less than 5%. DISCUSSION: A phantom simulating the diffusion restriction correlating with cell density and size could be developed.


Asunto(s)
Imagen de Difusión por Resonancia Magnética/instrumentación , Imagen de Difusión por Resonancia Magnética/métodos , Edema/diagnóstico por imagen , Neoplasias/diagnóstico por imagen , Fantasmas de Imagen , Resinas Sintéticas/química , Detergentes , Difusión , Edema/fisiopatología , Humanos , Modelos Lineales , Ensayo de Materiales , Neoplasias/fisiopatología , Tamaño de la Partícula , Agua/química
3.
Am J Respir Cell Mol Biol ; 58(5): 625-635, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29112457

RESUMEN

The FOSL1/AP-1 transcription factor regulates gene expression, thereby controlling various pathophysiological processes. It is a major effector of RAS-ERK1/2 signaling and is activated in human lung epithelia by tumorigenic stimuli. Recent evidence shows an inverse correlation between FOSL1 expression and the survival of patients with lung cancer and adenocarcinomas; however, its role in lung tumorigenesis remains elusive. In this work, we sought to determine the role of FOSL1 in Kras-induced lung adenocarcinoma in vivo and its downstream effector mechanisms. We used mice expressing the Kras oncogene in the lung with concomitant Fosl1 deletion, Kras-activated murine alveolar epithelial cells (mAECs) with Fosl1 deletion, and KRAS mutant human lung adenocarcinoma (HLAC) cells with FOSL1 deficiency, and performed cell proliferation and gene expression analyses. Mutant Kras induced Fosl1 expression in vitro (mAECs) and in vivo (lung tissue), and mice with Fosl1 deletion showed reduced levels of mutant Kras-induced lung tumorigenesis and survived longer than Fosl1-sufficient mice. Studies with mutant Kras-activated mAECs and KRAS-mutant HLAC cells revealed that FOSL1 regulates mutant KRAS-induced gene expression, thereby controlling cell proliferation and survival. In contrast, FOSL1 depletion in non-KRAS-mutant HLAC cells and nonmalignant human lung epithelia had no effect. Our data support the notion that FOSL1-mediated expression of amphiregulin and apoptotic and antioxidative genes plays a role in regulating HLAC cell proliferation and survival. FOSL1 is a determinant of lung cancer in vivo and regulates HLAC cell proliferation and survival, largely in the context of KRAS mutations. Activation of FOSL1 in adenocarcinomas may be a prognostic marker and potential target for human lung cancer with KRAS mutations.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Anfirregulina/genética , Neoplasias Pulmonares/genética , Mutación , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/patología , Anfirregulina/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal
4.
J Enzyme Inhib Med Chem ; 31(6): 1492-7, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26903444

RESUMEN

OBJECTIVE: The purpose of this study was to investigate the in vitro inhibitory effects of the edible microalga Aphanizomenon flos-aquae (AFA) on human UDP-α-d-glucose 6-dehydrogenase (UGDH) activity, a cytosolic enzyme involved both in tumor progression and in phytochemical bioavailability. METHODS: Both the hydrophilic and ethanolic AFA extracts as well as the constitutive active principles phycocyanin (PC), phycocyanobilin (PCB) and mycosporine-like amino acids (MAAs) were tested. RESULTS: Among AFA components, PCB presented the strongest inhibitory effect on UGDH activity, acting as a competitive inhibitor with respect to UDP-glucose and a non-competitive inhibitor with respect to NAD(+). In preliminary experiments, AFA PCB was also effective in reducing the colony formation capacity of PC-3 prostate cancer cells and FTC-133 thyroid cancer cells. CONCLUSIONS: Overall, these findings confirmed that AFA and its active principles are natural compounds with high biological activity. Further studies evaluating the effects of AFA PCB in reducing tumor cell growth and phytochemical glucuronidation are encouraged.


Asunto(s)
Aphanizomenon/química , Inhibidores Enzimáticos/farmacología , Uridina Difosfato Glucosa Deshidrogenasa/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Masculino
5.
Molecules ; 21(5)2016 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-27164073

RESUMEN

Tuberaria lignosa (Sweet) Samp. is found in European regions, and has antioxidant properties due to its composition in ascorbic acid and phenolic compounds. Given its traditional use and antioxidant properties, the tumor cell growth inhibitory potential of aqueous extracts from T. lignosa (prepared by infusion and decoction) was investigated in three human tumor cell lines: MCF-7 (breast adenocarcinoma), NCI-H460 (non-small cell lung cancer), and HCT-15 (human colorectal adenocarcinoma). Both extracts inhibited the growth of these cell lines; the most potent one being the T. lignosa extract obtained by infusion in the NCI-H460 cells (GI50 of approximately 50 µg/mL). Further assays were carried out with this extract in NCI-H460 cells. At 100 µg/mL or 150 µg/mL it caused an increase in the percentage of cells in the G0/G1 phase and a decrease of cells in S phase of the cell cycle. Additionally, these concentrations caused an increase in the percentage of apoptotic cells. In agreement, a decrease in total poly (ADP-ribose) polymerase (PARP) and pro-caspase 3 levels was found. In conclusion, the T. lignosa extract obtained by infusion was more potent in NCI-H460 cells, altering the cell cycle progression and inducing apoptosis. This work highlights the importance of T. lignosa as a source of bioactive compounds with tumor cell growth inhibitory potential.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Ciclo Celular/efectos de los fármacos , Magnoliaceae/química , Extractos Vegetales/farmacología , Antineoplásicos Fitogénicos/química , Apoptosis , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , Extractos Vegetales/química , Poli(ADP-Ribosa) Polimerasas/metabolismo , Transducción de Señal/efectos de los fármacos
6.
Biochim Biophys Acta ; 1838(8): 2087-98, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24796502

RESUMEN

The peptide KLA (acetyl-(KLAKLAK)2-NH2), which is rather non toxic for eukaryotic cell lines, becomes active when coupled to the cell penetrating peptide, penetratin (Pen), by a disulfide bridge. Remarkably, the conjugate KLA-Pen is cytotoxic, at low micromolar concentrations, against a panel of seven human tumor cell lines of various tissue origins, including cells resistant to conventional chemotherapy agents but not to normal human cell lines. Live microscopy on cells possessing fluorescent labeled mitochondria shows that in tumor cells, KLA-Pen had a strong impact on mitochondria tubular organization instantly resulting in their aggregation, while the unconjugated KLA and pen peptides had no effect. But, mitochondria in various normal cells were not affected by KLA-Pen. The interaction with membrane models of KLA-Pen, KLA and penetratin were studied using dynamic light scattering, calorimetry, plasmon resonance, circular dichroism and ATR-FTIR to unveil the mode of action of the conjugate. To understand the selectivity of the conjugate towards tumor cell lines and its action on mitochondria, lipid model systems composed of zwitterionic lipids were used as mimics of normal cell membranes and anionic lipids as mimics of tumor cell and mitochondria membrane. A very distinct mode of interaction with the two model systems was observed. KLA-Pen may exert its deleterious and selective action on cancer cells by the formation of pores with an oblique membrane orientation and establishment of important hydrophobic interactions. These results suggest that KLA-Pen could be a lead compound for the design of cancer therapeutics.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Portadoras/farmacología , Mitocondrias/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , Neoplasias/patología , Péptidos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica , Rastreo Diferencial de Calorimetría , Péptidos de Penetración Celular , Dicroismo Circular , Humanos , Péptidos y Proteínas de Señalización Intercelular , Liposomas , Lípidos de la Membrana/metabolismo , Neoplasias/tratamiento farmacológico , Péptidos/química , Espectroscopía Infrarroja por Transformada de Fourier , Células Tumorales Cultivadas
7.
Chem Biodivers ; 10(8): 1464-74, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23939794

RESUMEN

The essential oils of Artemisia arborescens growing in Sardinia (Italy), collected during three plant growth stages, i.e., from the vegetative stage to post-blooming time, were characterized. Moreover, the in vitro antiproliferative and antioxidant activities of the oil isolated from aerial parts collected in February were evaluated. The essential oils belonged to the ß-thujone/chamazulene chemotype, notably with the highest amount of chamazulene (ca. 52%) ever detected up to now in the genus Artemisia and, in general, in essential oils. Quantitative variations in the oil composition were observed as the plant passes from the vegetative to the blooming stage. The oil was tested for its potential tumor cell growth-inhibitory effect on T98G, MDA-MB 435S, A375, and HCT116 human cell lines, using the MTT (=3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide) assay. The highest activity was observed on A375 and HCT116 cell lines, with IC50 values of 14 µg/ml. Moreover, the in vitro antioxidant and free radical-scavenging assays revealed the oil to be an effective scavenger of the ABTS radical cation, with an activity comparable to that of Trolox(®) . These results support the use of A. arborescens oil for the treatment of inflamed skin conditions. Finally, the composition of the polar fraction of the A. arborescens aerial parts was also examined, and the main component detected was 5-O-caffeoylquinic acid, which was identified for the first time in this plant.


Asunto(s)
Antineoplásicos/química , Antioxidantes/química , Artemisia/química , Azulenos/química , Depuradores de Radicales Libres/química , Aceites Volátiles/química , Antineoplásicos/farmacología , Antioxidantes/farmacología , Azulenos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ácido Clorogénico/análogos & derivados , Ácido Clorogénico/química , Ácido Clorogénico/farmacología , Depuradores de Radicales Libres/farmacología , Humanos , Concentración 50 Inhibidora , Italia , Espectroscopía de Resonancia Magnética , Estructura Molecular , Componentes Aéreos de las Plantas/química , Ácido Quínico/análogos & derivados , Ácido Quínico/química , Ácido Quínico/farmacología
8.
Cells ; 12(17)2023 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-37681882

RESUMEN

Recently, mesenchymal stem cell (MSC) therapies have been questioned as MSCs are capable of both promoting and inhibiting tumorigenesis. Both MSCs and tumor cells replicate to increase their population size; however, MSCs, but not tumor cells, stop dividing when they reach confluence due to cell-cell contact inhibition and then differentiate. We hypothesized that contact inhibition results in the production of effector molecules by confluent MSCs and these effectors are capable of suppressing tumor cell growth. To test this hypothesis, we co-cultured breast cancer cells (MDA-MB-231) with either confluent or sub-confluent bone-marrow-derived MSCs (BM-MSCs); in addition, we treated various tumor cells with conditioned media (CM) obtained from either confluent or sub-confluent BM-MSCs. The results showed that the growth of tumor cells co-cultured with confluent BM-MSCs or treated with CM obtained from confluent BM-MSCs was inhibited, and this effect was significantly stronger than that seen with tumor cells co-cultured with sub-confluent BM-MSCs or CM obtained from sub-confluent BM-MSCs. Subcutaneous tumor formation was completely prevented by the inoculation of tumor cells mixed with CM. In the future, soluble anti-tumor effectors, produced by confluent MSCs, may be used as cell-free therapeutics; this approach provides a solution to current concerns associated with cell-based therapies.


Asunto(s)
Células Madre Mesenquimatosas , Neoplasias , Humanos , Inhibición de Contacto , Carcinogénesis , Ciclo Celular , Medios de Cultivo Condicionados/farmacología
9.
Foods ; 12(4)2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36832915

RESUMEN

Phaseolus vulgaris L. (common bean) contains high levels of proteins, unsaturated fatty acids, minerals, fibers, and vitamins, and for this reason, it represents an essential component of the diet. More than 40,000 varieties of beans have been recognized and are staple foods in the traditional cuisine of many countries. In addition to its high nutritional value, P. vulgaris is also characterized by its nutraceutical properties and favors environmental sustainability. In this manuscript, we studied two different varieties of P. vulgaris, Cannellino and Piattellino. We investigated the effects of traditional processing (soaking and cooking) and in vitro gastrointestinal digestion of beans on their phytochemical composition and anticancer activity. Using HT29 and HCT116 colon cancer cell lines, we showed that the extract obtained after gastrointestinal digestion of cooked beans (the bioaccessible fraction, BF) induces cell death through the induction of the autophagic process. We demonstrated that the BF of Cannellino and Piattellino beans at the concentration of 100 µg/mL reduces cell vitality, measured by MMT assay, of both HT29 (88.41% ± 5.79 and 94.38% ± 0.47) and HCT116 (86.29% ± 4.3 and 91.23% ± 0.52) cell lines. Consistently, the treatment of HT29 cells with 100 µg/mL of Cannellino and Piattellino BFs reduced clonogenicity by 95% ± 2.14 and 96% ± 0.49, respectively. Moreover, the activity of extracts appeared to be selective for colon cancer cells. The data shown in this work further confirm P. vulgaris to be among foods with beneficial effects for human health.

10.
Biology (Basel) ; 11(5)2022 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-35625427

RESUMEN

Cardoon (Cynara cardunculus var. altilis) blades were collected at sixteen sampling dates (B1-B16) to study the influence of the phenological growth stage on the phenolic composition and biological properties. Twenty phenolic compounds were identified, among which trans 3,4-O-dicaffeoylquinic acid, 5-O-caffeoylquinic acid, and luteolin-O-hexoside (39.6, 42.6, and 101.0 mg/g extract, respectively) were the main compounds. Immature blades (B3) had a higher content of phenolic compounds (178 mg/g extract) and a greater ability to inhibit the formation of thiobarbituric acid reactive substances (IC50 of 1.61 µg/mL). Samples at more advanced growth stages revealed a greater capacity to inhibit oxidative hemolysis (B8, IC50 of 25 and 47.4 µg/mL for Δt of 60 and 120 min, respectively) and higher cytotoxic (B8-B13, GI50 between 7.1 and 17 µg/mL), anti-inflammatory (B13, IC50 of 10 µg/mL), and antibacterial activities. In turn, the antifungal activity varied depending on the tested fungi. All these results suggest that maturity influences the phenolic composition and bioactive properties of cardoon blades, which reveal great potential for the development of bioactive ingredients for food and pharmaceutical applications, among others.

11.
Front Oncol ; 12: 887828, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35686099

RESUMEN

The four and a half LIM domain protein 3, also named the LIM-protein FHL3, belongs to the LIM-only family. Based on the special structure of LIM-only proteins, FHL3 can perform significant functions in muscle proliferation and cardiovascular diseases by regulating cell growth and signal transduction. In recent years, there has been increasing evidence of a relation between FHLs and tumor biology, since FHL3 is often overexpressed or downregulated in different cancers. On the one hand, FHL3 can function as a tumor suppressor and influence the expression of downstream genes. On the other hand, FHL3 can also play a role as an oncoprotein in some cancers to promote tumor progression via phosphorylation. Thus, FHL3 is proposed to have a dual effect on cancer progression, reflecting its complex roles in cancer. This review focuses on the roles of FHL3 in cancer progression and discusses the interaction of FHL3 with other proteins and transcription factors. Finally, the clinical significance of FHL3 for the treatment of cancers is discussed.

12.
Int J Mol Sci ; 12(11): 7581-93, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22174618

RESUMEN

Pu-erh tea is a kind of fermented tea with the incorporation of microorganisms' metabolites. Unlike green tea, the chemical characteristics and bioactivities of Pu-erh tea are still not well understood. Using water extracts of Pu-erh tea, we analyzed the tumor cell growth inhibition activities on several genetically engineered mouse tumor cell lines. We found that at the concentration that did not affect wild type mouse embryo fibroblasts (MEFs) growth, Pu-erh tea extracts could inhibit tumor cell growth by down-regulated S phase and cause G1 or G2 arrest. Further study showed that Pu-erh tea extracts down-regulated the expression of mutant p53 in tumor cells at the protein level as well as mRNA level. The same concentration of Pu-erh tea solution did not cause p53 stabilization or activation of its downstream pathways in wild type cells. We also found that Pu-erh tea treatment could slightly down-regulate both HSP70 and HSP90 protein levels in tumor cells. These data revealed the action of Pu-erh tea on tumor cells and provided the possible mechanism for Pu-erh tea action, which explained its selectivity in inhibiting tumor cells without affecting wild type cells. Our data sheds light on the application of Pu-erh tea as an anti-tumor agent with low side effects.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo , Té/química , Proteína p53 Supresora de Tumor/genética , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Fermentación , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Proteínas del Choque Térmico HSP72/genética , Proteínas del Choque Térmico HSP72/metabolismo , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Extractos Vegetales/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Fase S/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo
13.
Eur J Med Chem ; 210: 112950, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33148494

RESUMEN

Depending on the substitution pattern and stereochemistry, 1,3-dioxanes 1 with an aminoethyl moiety in 4-position represent potent σ1 receptor antagonists. In order to increase the stability, a cyclohexane ring first replaced the acetalic 1, 3-dioxane ring of 1. A large set of aminoethyl substituted cyclohexane derivatives was prepared in a six-step synthesis. All enantiomers and diastereomers were separated by chiral HPLC at the stage of the primary alcohol 7, and their absolute configuration was determined by CD spectroscopy. Neither the relative nor the absolute configuration had a large impact on the σ1 affinity. The highest σ1 affinity was found for cis-configured benzylamines (1R,3S)-11 (Ki = 0.61 nM) and (1S,3R)-11 (Ki = 1.3 nM). Molecular dynamics simulations showed that binding of (1R,3S)-11 at the σ1 receptor is stabilized by the typical polar interaction of the protonated amino moiety with the carboxy group of E172 which is optimally oriented by an H-bond interaction with Y103. The lipophilic interaction of I124 with the N-substituent also contributes to the high σ1 affinity of the benzylamines. The antagonistic activity was determined in a Ca2+ influx assay in retinal ganglion cells. The enantiomeric cis-configured benzylamines (1R,3S)-11 and (1S,3R)-11 were able to inhibit the growth of DU145 cells, a highly aggressive human prostate tumor cell line. Moreover, cis-11 could also inhibit the growth of further human tumor cells expressing σ1 receptors. The experimentally determined logD7.4 value of 3.13 for (1R,3S)-11 is in a promising range regarding membrane penetration. After incubation with mouse liver microsomes and NADPH for 90 min, 43% of the parent (1R,3S)-11 remained unchanged, indicating intermediate metabolic stability. Altogether, nine metabolites including one glutathione adduct were detected by means of LC-MS analysis.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Ciclohexanos/química , Ciclohexanos/farmacología , Receptores sigma/antagonistas & inhibidores , Aminación , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Receptores sigma/metabolismo , Relación Estructura-Actividad , Receptor Sigma-1
14.
Eur J Pharm Sci ; 151: 105302, 2020 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-32407871

RESUMEN

we aimed to explore the role of thalidomide in breast cancer by using a mouse 4T1 breast tumor model. A tumor model was established by injecting logarithmic-phase 4T1 cells into the mammary fat pat.Tumor growth was monitored every day and tumor size was measured periodically. Which were performed to determine the cytokine and related gene mRNA expression, the cytokine production and protein expression, check the vessel formation and necrosis, the CD31 expression,the expression of CD31, F4/80 and CD206,staining the digested cells with F4/80, cd45 and CD11b to determine the M2 type macrophages accumulation by qRT-PCR, Cytokine antibody array, HE staining, Immunohistochemistry, Immunostaining, Flow cytometry, respectively. In the current study, Our results indicated that thalidomide significantly inhibited tumor growth in the mouse model by inhibiting angiogenesis and promoting the necrosis of tumor cells in tumor tissues. Moreover, immunostaining and flow cytometry results demonstrated that M2-type tumor-associated macrophage accumulation and infiltration were profoundly inhibited upon treatment with thalidomide. In addition, thalidomide treatment also regulated cytokine production by inhibiting the production of angiogenesis-related cytokines, such as G-CSF, VEGF-B, VEGFR1, VEGFR3 and IL-10. In conclusion, our studies explored the antitumor effect of thalidomide in a breast tumor model and uncovered that thalidomide inhibited breast tumor growth by inhibiting angiogenesis as a result of reducing TAM accumulation and infiltration and inhibiting angiogenesis-related cytokine production.


Asunto(s)
Neoplasias de la Mama , Talidomida , Animales , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Femenino , Humanos , Macrófagos , Ratones , Ratones Endogámicos BALB C , Neovascularización Patológica/tratamiento farmacológico , Macrófagos Asociados a Tumores
15.
Cancer Biol Med ; 17(2): 270-281, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32587768

RESUMEN

Four and a half LIM domains protein 1 (FHL1), as the name suggests, contains four and a half LIM domains capable of interacting with various molecules, including structural proteins, kinases, and transcriptional machinery. FHL1 contains a zinc-finger domain and performs diverse roles in regulation of gene transcription, cytoarchitecture, cell proliferation, and signal transduction. Several studies have validated the importance of FHL1 in muscle development, myopathy, and cardiovascular diseases. Mutations in the FHL1 gene are associated with various myopathies. Recently, FHL1 was identified as a major host factor for chikungunya virus (CHIKV) infection in both humans and mice. Based on more recent findings over the last decade, FHL1 is proposed to play a dual role in cancer progression. On the one hand, FHL1 expression is suppressed in several cancer types, which correlates with increased metastatic disease and decreased survival. Moreover, FHL1 is reported to inhibit tumor cell growth and migration by associating with diverse signals, such as TGF-ß and ER, and therefore considered a tumor suppressor. On the other hand, FHL1 can function as an oncogenic protein that promotes tumor progression upon phosphorylation, reflecting complex roles in cancer. This review primarily focuses on the dual role and underlying mechanisms of action of FHL1 in human cancer progression and its clinical relevance.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Animales , Proliferación Celular/genética , Genes Supresores de Tumor , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Proteínas con Dominio LIM/química , Proteínas Musculares/química , Enfermedades Musculares/genética , Enfermedades Musculares/metabolismo , Mutación , Procesos Neoplásicos , Fosforilación , Transducción de Señal/genética
16.
Oncol Rep ; 43(4): 1208-1220, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32323835

RESUMEN

Changes in mitochondrial morphology by dysregulated mitochondrial fission­fusion proteins have been increasingly recognized as a hallmark of cancer. MiD49 (mitochondrial dynamics protein of 49 kDa) is a newly identified mitochondrial fission protein involved in the dynamic regulation of mitochondrial morphology. However, the expression pattern and biological functions of MiD49 in human cancers remain largely unexplored, especially in pancreatic cancer (PC). In the present study, the expression and clinical significance of MiD49 was firstly determined by RT­qPCR and western blot analyses in PC cell lines and tumor tissues. In addition, the biologic functions of MiD49 in PC cell growth and metastasis were investigated using gain­ and loss­of­function assays both in vitro and in vivo. Moreover, the underlying mechanisms by which MiD49 regulates PC cell growth and metastasis were further explored. Our results showed that MiD49 was markedly downregulated in both PC cell lines and human PC specimens. Forced expression of MiD49 suppressed PC cell growth and metastasis both in vitro and in vivo, while knockdown of MiD49 exhibited the opposite effect. Mechanistic exploration demonstrated that the tumor­suppressive effect of MiD49 was mediated by decreased mitochondrial fission and subsequent reduced ROS production in PC cells. Our findings suggest a critical tumor­suppressive role played by MiD49 in pancreatic cancer.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/secundario , Dinámicas Mitocondriales , Proteínas Mitocondriales/antagonistas & inhibidores , Neoplasias Pancreáticas/patología , Factores de Elongación de Péptidos/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Biología Computacional/métodos , Bases de Datos Genéticas , Regulación hacia Abajo , Femenino , Genes Supresores de Tumor , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Factores de Elongación de Péptidos/genética , Factores de Elongación de Péptidos/metabolismo , Tasa de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Methods Mol Biol ; 1895: 75-85, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30539530

RESUMEN

Exosomes derived from human mesenchymal stem cells (MSCs) engineered to express the suicide gene yeast cytosine deaminase::uracil phosphoribosyl transferase (yCD::UPRT) represent a new therapeutic approach for tumor-targeted innovative therapy. The yCD::UPRT-MSC-exosomes carry mRNA of the suicide gene in their cargo. Upon internalization by tumor cells, the exosomes inhibit the growth of broad types of cancer cells in vitro, in the presence of a prodrug. Here we describe the method leading to the production and testing of these therapeutic exosomes. The described steps include the preparation of replication-deficient retrovirus possessing the yCD::UPRT suicide gene, and the preparation and selection of MSCs transduced with yCD::UPRT suicide gene. We present procedures to obtain exosomes possessing the ability to induce the death of tumor cells. In addition, we highlight methods for the evaluation of the suicide gene activity of yCD::UPRT-MSC-exosomes.


Asunto(s)
Exosomas , Genes Transgénicos Suicidas , Terapia Genética/métodos , Neoplasias/terapia , Retroviridae/genética , Animales , Línea Celular , Citosina Desaminasa/genética , Portadores de Fármacos , Proteínas Fúngicas/genética , Vectores Genéticos , Humanos , Células Madre Mesenquimatosas , Ratones , Pentosiltransferasa/genética , Levaduras/enzimología
18.
Food Chem Toxicol ; 118: 635-644, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29883784

RESUMEN

The cell growth inhibitory activity of the hydroethanolic extract of Achillea millefolium was studied in human tumor cell lines (NCI-H460 and HCT-15) and its mechanism of action was investigated. The GI50 concentration was determined with the sulforhodamine B assay and cell cycle and apoptosis were analyzed by flow cytometry following incubation with PI or Annexin V FITC/PI, respectively. The expression levels of proteins involved in cell cycle and apoptosis were analyzed by Western blot. The extracts were characterized regarding their phenolic composition by LC-DAD-ESI/MS. 3,5-O-Dicaffeoylquinic acid, followed by 5-O-caffeoylquinic acid, were the main phenolic acids, while, luteolin-O-acetylhexoside and apigenin-O-acetylhexoside were the main flavonoids. This extract decreased the growth of the tested cell lines, being more potent in HCT-15 and then in NCI-H460 cells. Two different concentrations of the extract (75 and 100 µg/mL) caused alterations in cell cycle profile and increased apoptosis levels in HCT-15 and NCI-H460 cells. Moreover, the extract caused an increase in p53 and p21 expression in NCI-H460 cells (which have wt p53), and reduced XIAP levels in HCT-15 cells (with mutant p53). This work enhances the importance of A. millefolium as source of bioactive phenolic compounds, particularly of XIAP inhibitors.


Asunto(s)
Achillea/química , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Etanol/química , Extractos Vegetales/farmacología , Línea Celular Tumoral , Humanos
19.
J Cancer Res Clin Oncol ; 143(8): 1381-1393, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28342001

RESUMEN

PURPOSE: Keratin 17 (KRT17) has been suggested as a potential diagnostic marker of squamous cell carcinoma including oral squamous cell carcinoma (OSCC). The current study was conducted to clarify the function of KRT17 and its expression mechanism in OSCC. METHODS: Immunohistochemical analyses were carried out to examine the expression of KRT17, GLI family zinc finger (GLI)-1, GLI-2, or cleaved caspase-3 in OSCCs. The expression of KRT17, GLI-1, or GLI-2 was investigated among OSCC cell lines, and the effects of loss-of-function of KRT17 or GLI, using siRNA or inhibitor, on the cell growth of the OSCC cell line HSC-2 particularly with respect to apoptosis were examined. RESULTS: Immunohistochemical analyses of tissue specimens obtained from 78 OSCC patients revealed that KRT17 was not observed in non-tumor regions but was strongly expressed at high frequencies in tumor regions. Knockdown of KRT17 increased the number of cleaved caspase-3-positive cells, leading to the reduction of cell number. Loss-of-function of GLI-1 or GLI-2 also increased the cell numbers of apoptotic cells positive for staining of Annexin-V and propidium iodide (PI) and the terminal deoxynucleotidyl transferase dUTP-biotin nick-end labeling (TUNEL) method, and induced DNA fragmentation. This inhibitory effect on cell growth was partially rescued by exogenous KRT17 expression. In the KRT17-positive regions in OSCCs, GLI-1 or GLI-2 was frequently detected, and the number of cells with cleaved caspase-3 positive was decreased. CONCLUSIONS: KRT17 promotes tumor cell growth, at least partially, through its anti-apoptotic effect as a result of the KRT17 overexpression by GLIs in OSCC.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/genética , Queratina-17/genética , Neoplasias de la Boca/genética , Proteína con Dedos de Zinc GLI1/biosíntesis , Apoptosis/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Queratina-17/biosíntesis , Mucosa Bucal/patología , Neoplasias de la Boca/patología , Proteína con Dedos de Zinc GLI1/genética
20.
Eur J Transl Myol ; 26(2): 5958, 2016 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-27478560

RESUMEN

Skeletal muscle tissue is a rare site of tumor metastasis but is the main target of the degenerative processes occurring in cancer-associated cachexia syndrome. Beneficial effects of physical activity in counteracting cancer-related muscle wasting have been described in the last decades. Recently it has been shown that, in tumor xeno-transplanted mouse models, physical activity is able to directly affect tumor growth by modulating inflammatory responses in the tumor mass microenvironment. Here, we investigated the effect of physical activity on tumor cell growth in colon carcinoma C26 cells injected tibialis anterior muscles of BALB/c mice. Histological analyses revealed that 4 days of voluntary wheel running significantly counteracts tumor cell growth in C26-injected muscles compared to the non-injected sedentary controls. Since striated skeletal muscle tissue is the site of voluntary contraction, our results confirm that physical activity can also directly counteract tumor cell growth in a metabolically active tissue that is usually not a target for metastasis.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA