Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 647: 30-36, 2023 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-36709670

RESUMEN

Tumor priming is considered a promising strategy for improving drug distribution in malignant tissues. Multicellular layers (MCLs) of human cancer cells are potentially useful models for evaluating tumor-priming agents. We evaluated the priming effects of paclitaxel (PTX) on doxorubicin (DOX) penetration using MCLs of the human colorectal cancer cell lines including DLD-1, HCT-116, and HT-29. The penetration of DOX treated at 50 µM for 3 h was highly limited in all three MCLs. The penetration of the priming agent PTX into MCLs was determined using rhodamine-labeled PTX and appeared to be cell line-dependent: full penetration was observed in HCT-116 and HT-29 MCLs, whereas only limited penetration occurred in DLD-1 MCLs. PTX pretreatment at 20 µM for 24 or 48 h induced a tumor-priming effect in DOX distribution, with a 3 to 5.6-fold-increase in HCT-116 and HT-29 MCLs but a less than two-fold increase in DLD-1 MCLs. PTX treatment decreased fibronectin expression in HCT-116 and HT-29 MCLs but not in DLD-1, suggesting that the prominent priming effect of PTX in HCT-116 and HT-29 MCLs may be associated with the downregulation of fibronectin expression. Our study demonstrated that MCLs of human cancer cells are a useful model not only for the study of drug penetration into tumor tissues but also for screening and evaluating tumor-priming agents.


Asunto(s)
Neoplasias Colorrectales , Paclitaxel , Humanos , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Fibronectinas , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Células HT29 , Neoplasias Colorrectales/patología , Línea Celular Tumoral
2.
Cancer Cell Int ; 23(1): 219, 2023 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-37759302

RESUMEN

BACKGROUND: Recently, natural killer (NK) cells emerged as a treatment option for various solid tumors. However, the immunosuppressive tumor immune microenvironment (TIME) can reduce the cytotoxic ability of NK cells in pancreatic ductal adenocarcinoma. Cancer-associated fibroblasts within the tumor stroma can suppress immune surveillance by dysregulating factors involved in the cellular activity of NK cells. Herein, the effect of activated pancreatic stellate cells (aPSCs) on NK cell-mediated anticancer efficacy under three-dimensional (3D) coculture conditions was investigated. METHODS: 3D cocultures of PANC-1 tumor spheroids (TSs) with aPSCs and NK-92 cells in a collagen matrix were optimized to identify the occurring cellular interactions and differential cytokine profiles in conditioned media using microchannel chips. PANC-1 TSs and aPSCs were indirectly cocultured, whereas NK-92 cells were allowed to infiltrate the TS channel using convective medium flow. RESULTS: Coculture with aPSCs promoted PANC-1 TSs growth and suppressed the antitumor cytotoxic effects of NK-92 cells. Mutual inhibition of cellular activity without compromising migration ability was observed between aPSCs and NK-92 cells. Moreover, the reduced killing activity of NK-92 cells was found to be related with reduced granzyme B expression in NK cells. CONCLUSIONS: Herein, a novel TIME-on-chip model based on the coculture of PANC-1 TSs, aPSCs, and NK-92 cells was described. This model may be useful for studying the detailed mechanisms underlying NK cells dysregulation and for exploring future therapeutic interventions to restore NK cell activity in the tumor microenvironment.

3.
Int J Mol Sci ; 23(2)2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-35055115

RESUMEN

Circular RNAs (circRNAs) and long noncoding RNAs (lncRNAs) are differentially expressed in gastrointestinal cancers. These noncoding RNAs (ncRNAs) regulate a variety of cellular activities by physically interacting with microRNAs and proteins and altering their activity. It has also been suggested that exosomes encapsulate circRNAs and lncRNAs in cancer cells. Exosomes are then discharged into the extracellular environment, where they are taken up by other cells. As a result, exosomal ncRNA cargo is critical for cell-cell communication within the cancer microenvironment. Exosomal ncRNAs can regulate a range of events, such as angiogenesis, metastasis, immune evasion, drug resistance, and epithelial-to-mesenchymal transition. To set the groundwork for developing novel therapeutic strategies against gastrointestinal malignancies, a thorough understanding of circRNAs and lncRNAs is required. In this review, we discuss the function and intrinsic features of oncogenic circRNAs and lncRNAs that are enriched within exosomes.


Asunto(s)
Exosomas/genética , Neoplasias Gastrointestinales/genética , ARN Circular/genética , ARN Largo no Codificante/genética , Comunicación Celular , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Humanos , Microambiente Tumoral
4.
Biochem Biophys Res Commun ; 575: 78-84, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34461439

RESUMEN

Alterations in sialylation of terminal residues of glycoproteins have been implicated in forming tumor-associated glycans. ST6GALNAC transfers sialyl moiety to N-acetylgalactosamine residue via α2,6 linkage. Although the oncogenic characteristics of ST6GALNACI or II have been demonstrated in various cancer cells, the impact of ST6GALNACIII on tumor progression remains undefined. In this study, we evaluated the effect of ST6GALNACIII knockdown on the growth of A549 non-small cell lung cancer cells. ST6GALNACIII depletion resulted in significant retardation in growth of A549 cells under various culture conditions, including collagen-supported 3D culture and anchorage-independent soft agar culture conditions. Liquid chromatography with tandem mass spectrometry revealed that two glycopeptides of transferrin receptor protein 1 (TFR1) containing N-acetylhexosamine-sialic acid were not detected in ST6GALNACIII-depleted A549 cells compared with control cells. Subsequent lectin binding assay, western blotting, and real-time RT-PCR indicated that TFR1 sialylation was not significantly changed, but TFR1 protein and mRNA expressions were decreased after ST6GALNACIII knockdown. However, cell growth retardation by ST6GALNACIII knockdown was partially rescued by TFR1 overexpression. Additionally, TFR1 mRNA degradation was accelerated following ST6GALNACIII knockdown with concomitant reduction in mRNA levels of iron regulatory protein 1 and 2, the upstream regulators of TFR1 mRNA stability. Therefore, our results indicated an important role of ST6GALNACIII in promoting A549 cell growth through quantitative regulation of TFR1 expression and provided therapeutic implications for ST6GALNACIII targeting in tumor growth suppression in vivo.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/prevención & control , Hierro/metabolismo , Neoplasias Pulmonares/prevención & control , Estabilidad del ARN , Receptores de Transferrina/antagonistas & inhibidores , Sialiltransferasas/deficiencia , Antígenos CD/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Receptores de Transferrina/metabolismo
5.
Int J Mol Sci ; 22(18)2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34575983

RESUMEN

Hypoxia-inducible factor-1 alpha (HIF-1α) is overexpressed in cancer, leading to a poor prognosis in patients. Diverse cellular factors are able to regulate HIF-1α expression in hypoxia and even in non-hypoxic conditions, affecting its progression and malignant characteristics by regulating the expression of the HIF-1α target genes that are involved in cell survival, angiogenesis, metabolism, therapeutic resistance, et cetera. Numerous studies have exhibited the anti-cancer effect of HIF-1α inhibition itself and the augmentation of anti-cancer treatment efficacy by interfering with HIF-1α-mediated signaling. The anti-cancer effect of plant-derived phytochemicals has been evaluated, and they have been found to possess significant therapeutic potentials against numerous cancer types. A better understanding of phytochemicals is indispensable for establishing advanced strategies for cancer therapy. This article reviews the anti-cancer effect of phytochemicals in connection with HIF-1α regulation.


Asunto(s)
Anticarcinógenos/uso terapéutico , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias/tratamiento farmacológico , Fitoquímicos/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias/genética , Hipoxia Tumoral/genética
6.
Int J Mol Sci ; 22(14)2021 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-34298879

RESUMEN

Hypoxia is one of the representative microenvironment features in cancer and is considered to be associated with the dismal prognosis of patients. Hypoxia-driven cellular pathways are largely regulated by hypoxia-inducible factors (HIFs) and notably exert influence on the hallmarks of cancer, such as stemness, angiogenesis, invasion, metastasis, and the resistance towards apoptotic cell death and therapeutic resistance; therefore, hypoxia has been considered as a potential hurdle for cancer therapy. Growing evidence has demonstrated that long noncoding RNAs (lncRNAs) are dysregulated in cancer and take part in gene regulatory networks owing to their various modes of action through interacting with proteins and microRNAs. In this review, we focus attention on the relationship between hypoxia/HIFs and lncRNAs, in company with the possibility of lncRNAs as candidate molecules for controlling cancer.


Asunto(s)
Hipoxia/genética , Neoplasias/genética , ARN Largo no Codificante/genética , Microambiente Tumoral/genética , Animales , Apoptosis/genética , Humanos , MicroARNs/genética
7.
Biochem Biophys Res Commun ; 524(3): 643-648, 2020 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-32029272

RESUMEN

Digoxin, a compound of the cardiac glycoside family, was originally prescribed for heart failure but has recently been rediscovered for its potent antitumor activity. However, it has a narrow therapeutic margin due to its cardiotoxicity, limiting its safe use as an antitumor agent in clinical practice. To widen its therapeutic margin, we investigated whether the antitumor effect of digoxin is potentiated by the depletion of BCL-2-interacting cell death suppressor (BIS) in A549 lung cancer cells. BIS is a multifunctional protein that is frequently overexpressed in most human cancers including lung cancer. Our results demonstrated that the inhibitory potential of digoxin on the migratory behavior of A549 cells is significantly enhanced by BIS depletion as assessed by transwell assay and collagen-incorporated 3D spheroid culture. Western blotting revealed that combination treatment significantly reduces p-STAT3 expression. In addition, a STAT3 inhibitor substantially suppressed the aggressive phenotypes of A549 cells. Thus, our results suggest that loss of STAT3 activity is a possible molecular mechanism for the synergistic effect of digoxin and BIS depletion. Our findings suggest the sensitizing role of BIS silencing to reduce the dose of digoxin for treatment of lung cancer with a high metastatic potential.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Movimiento Celular/efectos de los fármacos , Digoxina/farmacología , Regulación hacia Abajo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Células A549 , Supervivencia Celular/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Invasividad Neoplásica
8.
Molecules ; 25(20)2020 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-33066509

RESUMEN

Cancer is a global health concern and one of the main causes of disease-related death. Even with considerable progress in investigations on cancer therapy, effective anti-cancer agents and regimens have thus far been insufficient. There has been compelling evidence that natural phytochemicals and their derivatives have potent anti-cancer activities. Plant-based anti-cancer agents, such as etoposide, irinotecan, paclitaxel, and vincristine, are currently being applied in medical treatments for patients with cancer. Further, the efficacy of plenty of phytochemicals has been evaluated to discover a promising candidate for cancer therapy. For developing more effective cancer therapy, it is required to apprehend the molecular mechanism deployed by natural compounds. MicroRNAs (miRNAs) have been realized to play a pivotal role in regulating cellular signaling pathways, affecting the efficacy of therapeutic agents in cancer. This review presents a feature of phytochemicals with anti-cancer activity, focusing mainly on the relationship between phytochemicals and miRNAs, with insights into the role of miRNAs as the mediators and the regulators of anti-cancer effects of phytochemicals.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , MicroARNs/genética , Fitoquímicos/farmacología , Animales , Anticarcinógenos/farmacología , Ensayos Clínicos como Asunto , Genes Supresores de Tumor/efectos de los fármacos , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética
9.
Biochem Biophys Res Commun ; 515(1): 183-189, 2019 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-31133378

RESUMEN

Heterotypic cell-cell interaction between cancer cells and pancreatic stellate cells (PSCs) within tumor microenvironment is considered as a key mechanism for epithelial-mesenchymal transition (EMT) that triggers disease progression and chemoresistance in pancreatic ductal adenocarcinoma (PDAC). Hence, PSCs should be incorporated into in vitro co-culture models to maximize clinical relevance of data obtained using these models. In this study, we developed hetero-type spheroids of pancreatic cancer cells (ductal carcinoma cells PANC-1 and primacy sarcomatoid adenocarcinoma 36473 cells) and PSCs. Effect of PSC co-culture on the formation and growth of multicellular spheroids was cell-line dependent in that growth stimulation effect appeared in PANC-1/PSC spheroids, but not in 36473/PSC spheroids. Spatial distribution of PSCs within spheroids was also cell-line dependent. It was either confined to the center region (PANC-1) or evenly distributed (36473). Changes in expression levels of E-cadherin and vimentin revealed EMT induction in PANC-1/PSC hetero-type spheroids, but not in 36473/PSC spheroids. Gemcitabine sensitivity was increased partially by PSC co-culture. However, PSCs showed relative resistance to gemcitabine compared to PANC-1 cells in PANC-1/PSC spheroids. Overall, our hetero-type spheroid model can be used to study cancer-stroma interaction and their mechanism and evaluate anticancer drug activity. We demonstrated that stromal effect by PSC co-culture might be cellular context dependent with regard to growth stimulation and EMT induction. Hence, anti-stromal therapy should take these differences into consideration.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Comunicación Celular , Neoplasias Pancreáticas/metabolismo , Células Estrelladas Pancreáticas/metabolismo , Antimetabolitos Antineoplásicos/farmacología , Cadherinas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Neoplasias Pancreáticas/patología , Células Estrelladas Pancreáticas/citología , Esferoides Celulares/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Vimentina/metabolismo , Gemcitabina
10.
Cancer Sci ; 108(12): 2470-2477, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28940685

RESUMEN

We investigated the combinatorial effects of pentoxifylline (PTX) on the efficacy of gemcitabine (GEM) in a human pancreatic tumor xenograft model. PTX significantly improved the efficacy of GEM, as shown by a 50% reduction in tumor growth rate at 4 weeks of treatment compared with that in animals given GEM alone. The fluorescent drug doxorubicin (DOX) was used to test whether drug delivery was improved by PTX, contributing to the improved efficacy of GEM. PTX given for 2 weeks prior to giving DOX improved drug distribution by 1.8- to 2.2-fold with no changes in vessel density, suggesting that improvement in drug delivery was not related to the vascular mechanism. Instead, collagen I content in tumor stroma was significantly reduced, as was the expression of alpha-smooth muscle actin of cancer-associated fibroblasts and connective tissue growth factor (CTGF) by PTX pretreatment. Overall, our data demonstrated that increased efficacy of GEM by PTX was associated with improved drug delivery to tumor tissue, which may be attributed to decreased expression of CTGF and subsequent reduction in the stromal collagen matrix in the pancreatic ductal adenocarcinoma tumor. These results support the usefulness of PTX in combination with chemotherapy for targeting drug delivery barriers associated with the stromal matrix, which should be further evaluated for clinical development.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/patología , Pentoxifilina/farmacología , Animales , Desoxicitidina/farmacología , Sinergismo Farmacológico , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Inhibidores de Fosfodiesterasa/farmacología , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
11.
Exp Cell Res ; 335(2): 187-96, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26022665

RESUMEN

Epithelial-mesenchymal transition (EMT) acts as a facilitator of metastatic dissemination in the invasive margin of malignant tumors where active tumor-stromal crosstalks take place. Co-cultures of cancer cells with cancer-associated fibroblasts (CAFs) are often used as in vitro models of EMT. We established a tumor-fibroblast proximity co-culture using HT-29 tumor spheroids (TSs) with CCD-18 co fibroblasts. When co-cultured with TSs, CCD-18 co appeared activated, and proliferative activity as well as cell migration increased. Expression of fibronectin increased whereas laminin and type I collagen decreased in TSs co-cultured with fibroblasts compared to TSs alone, closely resembling the margin of in vivo xenograft tissue. Active TGFß1 in culture media significantly increased in TS co-cultures but not in 2D co-cultures of cancer cells-fibroblasts, indicating that 3D context-associated factors from TSs may be crucial to crosstalks between cancer cells and fibroblasts. We also observed in TSs co-cultured with fibroblasts increased expression of α-SMA, EGFR and CTGF; reduced expression of membranous ß-catenin and E-cadherin, together suggesting an EMT-like changes similar to a marginal region of xenograft tissue in vivo. Overall, our in vitro TS-fibroblast proximity co-culture mimics the EMT-state of the invasive margin of in vivo tumors in early metastasis.


Asunto(s)
Transición Epitelial-Mesenquimal , Fibroblastos/fisiología , Esferoides Celulares/fisiología , Animales , Movimiento Celular , Técnicas de Cocultivo , Resistencia a Antineoplásicos , Matriz Extracelular/metabolismo , Femenino , Células HT29 , Humanos , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Factor de Crecimiento Transformador beta1/fisiología
12.
Korean J Physiol Pharmacol ; 17(1): 9-13, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23440944

RESUMEN

The aim of the present study was to examine the effect of micellar systems on the absorption of beta-lapachone (b-lap) through different intestinal segments using a single-pass rat intestinal perfusion technique. B-lap was solubilized in mixed micelles composed of phosphatidylcholine and sodium deoxycholate, and in sodium lauryl sulfate (SLS)-based conventional micelles. Both mixed micelles and SLS micelles improved the in situ permeability of b-lap in all intestinal segments tested although the mixed micellar formulation was more effective in increasing the intestinal absorption of b-lap. The permeability of b-lap was greatest in the large intestinal segments. Compared with SLS micelles, the effective permeability coefficient values measured with mixed micelles were 5- to 23-fold higher depending on the intestinal segment. Our data suggest that b-lap should be delivered to the large intestine using a mixed micellar system for improved absorption.

13.
J Microencapsul ; 29(7): 695-705, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22583128

RESUMEN

The aims of this study were to design and characterise doxorubicin-loaded chitosan microspheres for anti-cancer chemoembolisation. Doxorubicin-loaded chitosan microspheres were prepared by emulsification and cross-linking methods. Doxorubicin-chitosan solution was initially complexed with tripolyphosphate (TPP) to improve drug loading capabilities. Doxorubicin-loaded chitosan microspheres were highly spherical and had approximately diameters of 130-160 µm in size. Drug loading amount and loading efficiency were in the range 3.7-4.0% and 68.5-85.8%, respectively, and affected by TPP concentration, drug levels and cross-linking time. Doxorubicin release was affected by TPP complexation, cross-linking time and release medium. Especially, lysozyme in release media considerably increased drug release. Synergistic anti-cancer activities of doxorubicin-releasing chitosan microspheres were confirmed to VX2 cells in the rabbit auricle model compared with blank microspheres. Doxorubicin-loaded chitosan microspheres can efficiently be prepared by TPP gelation and cross-linking method and developed as multifunctional anti-cancer embolic material.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Quitosano/farmacología , Doxorrubicina/farmacología , Microesferas , Neoplasias Experimentales/tratamiento farmacológico , Animales , Antibióticos Antineoplásicos/química , Antibióticos Antineoplásicos/farmacocinética , Línea Celular Tumoral , Quitosano/química , Quitosano/farmacocinética , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Preparaciones de Acción Retardada/farmacología , Doxorrubicina/química , Doxorrubicina/farmacocinética , Masculino , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Conejos
14.
Pharmazie ; 67(11): 917-24, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23210241

RESUMEN

The aim of this study was to design self-microemulsifying tablets for pH-independent fast release of poorly soluble candesartan cilexetil (CDC). To improve the solubility of CDC, a self-microemulsifying drug delivery system (SMEDDS) was prepared composed of Capryol 90, Tween 80 and tetraglycol at a ratio of 5:35:60. Drug containing SMEDDS was adsorbed onto Fujicalin and Neusilin UFL2, respectively, used as solidification carriers and subsequently compressed into tablets (self-microemulsifying tablet, SMET). SMET using Fujicalin exhibited immediate CDC release in pH 1.2 medium while Neusilin UFL2-based SMET showed fast release, especially at pH 6.5. Thus, optimized SMET could be produced with one layer of Fujicalin and the other layer with Neusilin UFL2, demonstrating CDC release of 75% of the initial dose within 15 min in all pH conditions (1.2, 4.5, and 6.5). The average diameter of emulsion droplets formed from SMET was less than 200 nm. It was thus expected that Fujicalin and Neusilin UFL2-based bi-layer SMET would overcome low oral bioavailability of CDC due to its limited solubility at physiological pH conditions in the gastrointestinal tract.


Asunto(s)
Bloqueadores del Receptor Tipo 1 de Angiotensina II/administración & dosificación , Bencimidazoles/administración & dosificación , Compuestos de Bifenilo/administración & dosificación , Tetrazoles/administración & dosificación , Bloqueadores del Receptor Tipo 1 de Angiotensina II/análisis , Bencimidazoles/análisis , Compuestos de Bifenilo/análisis , Química Farmacéutica , Composición de Medicamentos , Sistemas de Liberación de Medicamentos , Electroquímica , Emulsiones , Concentración de Iones de Hidrógeno , Microscopía Electrónica de Rastreo , Tamaño de la Partícula , Solubilidad , Comprimidos , Tetrazoles/análisis
15.
Cancers (Basel) ; 13(23)2021 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-34885065

RESUMEN

Activated pancreatic stellate cells (aPSCs) and M2 macrophages modulate tumor progression and therapeutic efficacy in pancreatic ductal adenocarcinoma (PDAC) via epithelial-mesenchymal transition (EMT). Here, our aim was to analyze the anti-invasion effects of anti-cancer agents where EMT-inducing cancer-stroma interaction occurs under three-dimensional (3D) culture conditions. We used microfluidic channel chips to co-culture pancreatic tumor spheroids (TSs) with aPSCs and THP-1-derived M2 macrophages (M2 THP-1 cells) embedded in type I collagen. Under stromal cell co-culture conditions, PANC-1 TSs displayed elevated expression of EMT-related proteins and increased invasion and migration. When PANC-1 TSs were exposed to gemcitabine, 5-fluorouracil, oxaliplatin, or paclitaxel, 30-50% cells were found unaffected, with no significant changes in the dose-response profiles under stromal cell co-culture conditions. This indicated intrinsic resistance to these drugs and no further induction of drug resistance by stromal cells. Paclitaxel had a significant anti-invasion effect; in contrast, oxaliplatin did not show such effect despite its specific cytotoxicity in M2 THP-1 cells. Overall, our findings demonstrate that the TS-stroma co-culture model of PDAC is useful for activity profiling of anti-cancer agents against cancer and stromal cells, and analyzing the relationship between anti-stromal activity and anti-invasion effects.

16.
Cancers (Basel) ; 12(12)2020 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-33297288

RESUMEN

Three-dimensional (3D) culture of tumor spheroids (TSs) within the extracellular matrix (ECM) represents a microtumor model that recapitulates human solid tumors in vivo, and is useful for 3D multiplex phenotypic analysis. However, the low efficiency of 3D culture and limited 3D visualization of microtumor specimens impose technical hurdles for the evaluation of TS-based phenotypic analysis. Here, we report a 3D microtumor culture-to-3D visualization system using a minipillar array chip combined with a tissue optical clearing (TOC) method for high-content phenotypic analysis of microtumors. To prove the utility of this method, phenotypic changes in TSs of human pancreatic cancer cells were determined by co-culture with cancer-associated fibroblasts and M2-type tumor-associated macrophages. Significant improvement was achieved in immunostaining and optical transmission in each TS as well as the entire microtumor specimen, enabling optimization in image-based analysis of the morphology, structural organization, and protein expression in cancer cells and the ECM. Changes in the invasive phenotype, including cellular morphology and expression of epithelial-mesenchymal transition-related proteins and drug-induced apoptosis under stromal cell co-culture were also successfully analyzed. Overall, our study demonstrates that a minipillar array chip combined with TOC offers a novel system for 3D culture-to-3D visualization of microtumors to facilitate high-content phenotypic analysis.

17.
Cancers (Basel) ; 12(5)2020 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-32455681

RESUMEN

Invasive cancer cell migration is a key feature of metastatic human pancreatic ductal adenocarcinoma (PDAC), yet the underlying mechanisms remain poorly understood. Here, we investigated modes of cancer cell invasion using two pancreatic cancer cell lines with differential epithelial-mesenchymal status, PANC-1 and BxPC-3, under 3D culture conditions. Multicellular tumor spheroids (TSs) were grown in a collagen matrix co-cultured with pancreatic stellate cells (PSCs) using microchannel chips. PANC-1 cells showed individual migration from TSs via invadopodium formation. BxPC-3 cells showed plasticity between collective and individual migration in either mesenchymal mode, with filopodium-like protrusions, or blebby amoeboid mode. These two cell lines showed significantly different patterns of extracellular matrix (ECM) remodeling, with MMP-dependent degradation in a limited area of ECM around invadopodia for PANC-1 cells, or MMP-independent extensive deformation of ECM for BxPC-3 cells. Cancer cell migration out of the collagen channel significantly increased by PSCs and directional cancer cell migration was mediated by fibronectin deposited by PSCs. Our results highlight the phenotypic heterogeneity and plasticity of PDAC cell migration and ECM remodeling under 3D culture conditions. This 3D co-culture model of pancreatic cancer cells and PSCs offers a useful tool for studying cancer cell migration and ECM remodeling to identify and develop potential molecular targets and anti-cancer agents against human PDAC.

18.
Anticancer Res ; 40(2): 723-731, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32014914

RESUMEN

BACKGROUND/AIM: MicroRNAs (miRNAs) play regulatory roles in pancreatic ductal adenocarcinoma (PDAC). However, it is still required to identify the function of miRNA-301-3p in pancreatic cancer cells. MATERIALS AND METHODS: Effects of luteolin on cell growth, TRAIL cytotoxicity, and miR-301-3p levels were evaluated. The role of miRNA-301-3p in regulating cell proliferation, target gene expression, and TRAIL cytotoxicity were studied. RESULTS: The levels of miR-301-3p were down-regulated in PANC-1 cells exposed to luteolin, which inhibits the growth of PANC-1 cells and sensitizes cells to TRAIL. The knockdown of miR-301-3p attenuates cell proliferation and enhances TRAIL cytotoxicity. In addition, caspase-8 was directly targeted by miR-301-3p. CONCLUSION: Our findings unveil a critical biological function of miR-301-3p in regulating cell proliferation and elevating an antiproliferative effect of TRAIL on cancer cells. Our observation of miR-301-3p/caspase-8 relationship can also serve to clarify the role of miR-301-3p in other cancer types and related diseases.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Caspasa 8/metabolismo , Luteolina/farmacología , MicroARNs/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Carcinoma Ductal Pancreático/genética , Caspasa 8/genética , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Sinergismo Farmacológico , Técnicas de Silenciamiento del Gen , Humanos , Luteolina/administración & dosificación , MicroARNs/genética , Neoplasias Pancreáticas/genética , Proteínas Recombinantes/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Transfección
19.
Biomedicines ; 8(8)2020 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-32756339

RESUMEN

Pancreatic cancer is malignant and the seventh leading cause of cancer-related deaths worldwide. However, chemotherapy and radiotherapy are-at most-moderately effective, indicating the need for new and different kinds of therapies to manage this disease. It has been proposed that the biologic properties of pancreatic cancer cells are finely tuned by the dynamic microenvironment, which includes extracellular matrix, cancer-associated cells, and diverse immune cells. Accumulating evidence has demonstrated that extracellular vesicles (EVs) play an essential role in communication between heterogeneous subpopulations of cells by transmitting multiplex biomolecules. EV-mediated cell-cell communication ultimately contributes to several aspects of pancreatic cancer, such as growth, angiogenesis, metastasis and therapeutic resistance. In this review, we discuss the role of extracellular vesicles and their cargo molecules in pancreatic cancer. We also present the feasibility of the inhibition of extracellular biosynthesis and their itinerary (release and uptake) for a new attractive therapeutic strategy against pancreatic cancer.

20.
Int J Oncol ; 34(6): 1669-79, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19424586

RESUMEN

Similar to arsenic trioxide (As2O3), tetra-arsenic oxide (As4O6, TAO) has shown anti-proliferative and apoptosis-inducing effects against human leukemic and solid tumor cells. In order to assess the increase in efficacy, we evaluated the combinatory interaction of TAO combined with paclitaxel, 5-FU or cisplatin and studied its mechanism of action in the cell lines of human gastric, cervix and head and neck tumors. Two agents were combined at equitoxic ratios based on the IC50 of each drug. Efficacy improvement was evaluated using a combination index and isobologram at 50% inhibition level. Apoptosis induction and expression of apoptosis-related proteins was determined and the effect on microtubule polymerization was monitored. TAO combined with paclitaxel showed synergistic interaction in all three of gastric, cervix and head and neck cancer cell lines. On the other hand, TAO when combined with 5-FU or cisplatin showed an antagonistic interaction in head and neck or cervix cancer cell lines, respectively. Simultaneous treatment with TAO with paclitaxel resulted in an increased percentage of apoptotic cells and a significant increase in PARP cleavage and caspase-3 activation in the gastric and cervix cancer cells compared to TAO alone as well as the antagonistic groups (TAO with 5-FU or cisplatin). TAO suppressed the tubulin polymerization in the presence and absence of paclitaxel in a concentration-dependent manner, suggesting mitotic catastrophe as a potential mechanism of the synergism with paclitaxel. Overall, the present study suggests that TAO may have a greater potential as an anti-cancer agent against human gastric, cervix and head and neck tumors, in combination with paclitaxel. The synergistic interaction with paclitaxel may be associated with increased apoptosis via inhibition of paclitaxel-induced tubulin polymerization. Further detailed studies of combinatory mechanisms and evaluation using in vivo models are warranted.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias del Cuello Uterino/tratamiento farmacológico , Trióxido de Arsénico , Arsenicales/administración & dosificación , Western Blotting , Caspasa 3/metabolismo , Cisplatino/administración & dosificación , Interacciones Farmacológicas , Sinergismo Farmacológico , Femenino , Fluorouracilo/administración & dosificación , Neoplasias de Cabeza y Cuello/patología , Humanos , Técnicas In Vitro , Concentración 50 Inhibidora , Óxidos/administración & dosificación , Paclitaxel/administración & dosificación , Poli(ADP-Ribosa) Polimerasas/metabolismo , Neoplasias Gástricas/patología , Tubulina (Proteína)/metabolismo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias del Cuello Uterino/patología , Proteína X Asociada a bcl-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA