Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros












Intervalo de año de publicación
1.
Int J Mol Sci ; 22(12)2021 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-34205320

RESUMEN

Claudin-2 (CLDN2), a tight junctional protein, is involved in the chemoresistance in a three-dimensional spheroid culture model of human lung adenocarcinoma A549 cells. However, the mechanism has not been fully clarified. We found that the knockdown of CLDN2 expression by siRNA in the spheroid reduces the expression of glucose transporters and metabolic enzymes. In a two-dimensional culture model, the expression of these proteins was increased by glucose deprivation or fasentin, an inhibitor of glucose transporter. In addition, the expression levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and antioxidant enzymes including heme oxygenase-1, NAD(P)H:quinone oxidoreductase-1, and a glutamate-cysteine ligase modifier subunit were increased by fasentin. The fluorescence intensities of JC-1, a probe of mitochondrial membrane potential, and MitoROS 580, a probe of mitochondrial superoxide production, were increased by fasentin. These results suggest that mitochondrial production of reactive oxygen species is increased by glucose deficiency. The knockdown of CLDN2 enhanced the flux of 2-deoxy-2-[(7-nitro-2,1,3-benzoxadiazol-4-yl)amino]-D-glucose (2-NBDG), a fluorescent deoxyglucose derivative, in a transwell assay, and the accumulation of glucose and 2-NBDG in spheroid cells. The expression of Nrf2 was decreased by CLDN2 knockdown, which was inhibited by fasentin and sulforaphane, a typical Nrf2 activator, in spheroid cells. The sensitivity of spheroid cells to doxorubicin, an anthracycline antitumor antibiotic, was enhanced by CLDN2 knockdown, which was inhibited by fasentin and sulforaphane. We suggest that CLDN2 induces chemoresistance in spheroid cells mediated through the inhibition of glucose transport and activation of the Nrf2 signal.


Asunto(s)
Claudinas/fisiología , Resistencia a Antineoplásicos , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Esferoides Celulares/enzimología , Células A549 , Anilidas , Doxorrubicina , Humanos , Isotiocianatos , Especies Reactivas de Oxígeno/metabolismo , Sulfóxidos
2.
Molecules ; 26(2)2021 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-33429981

RESUMEN

Resident cancer cells with stem cell-like features induce drug tolerance, facilitating survival of glioblastoma (GBM). We previously showed that strategies targeting tumor bioenergetics present a novel emerging avenue for treatment of GBM. The objective of this study was to enhance the therapeutic effects of dual inhibition of tumor bioenergetics by combination of gossypol, an aldehyde dehydrogenase inhibitor, and phenformin, a biguanide compound that depletes oxidative phosphorylation, with the chemotherapeutic drug, temozolomide (TMZ), to block proliferation, stemness, and invasiveness of GBM tumorspheres (TSs). Combination therapy with gossypol, phenformin, and TMZ induced a significant reduction in ATP levels, cell viability, stemness, and invasiveness compared to TMZ monotherapy and dual therapy with gossypol and phenformin. Analysis of differentially expressed genes revealed up-regulation of genes involved in programmed cell death, autophagy, and protein metabolism and down-regulation of those associated with cell metabolism, cycle, and adhesion. Combination of TMZ with dual inhibitors of tumor bioenergetics may, therefore, present an effective strategy against GBM by enhancing therapeutic effects through multiple mechanisms of action.


Asunto(s)
Aldehído Deshidrogenasa/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Encefálicas , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Glioblastoma , Proteínas de Neoplasias/antagonistas & inhibidores , Esferoides Celulares/enzimología , Aldehído Deshidrogenasa/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/enzimología , Complejo I de Transporte de Electrón/metabolismo , Inhibidores Enzimáticos/farmacología , Glioblastoma/tratamiento farmacológico , Glioblastoma/enzimología , Humanos , Proteínas de Neoplasias/metabolismo , Temozolomida/farmacología
3.
Sci Rep ; 10(1): 21873, 2020 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-33318517

RESUMEN

Glioblastoma (GBM) is the most common and lethal primary intrinsic tumour of the adult brain and evidence indicates disease progression is driven by glioma stem cells (GSCs). Extensive advances in the molecular characterization of GBM allowed classification into proneural, mesenchymal and classical subtypes, and have raised expectations these insights may predict response to targeted therapies. We utilized GBM neurospheres that display GSC characteristics and found activation of the PI3K/AKT pathway in sphere-forming cells. The PI3Kα selective inhibitor alpelisib blocked PI3K/AKT activation and inhibited spheroid growth, suggesting an essential role for the PI3Kα catalytic isoform. p110α expression was highest in the proneural subtype and this was associated with increased phosphorylation of AKT. Further, employing the GBM BioDP, we found co-expression of PIK3CA with the neuronal stem/progenitor marker NES was associated with poor prognosis in PN GBM patients, indicating a unique role for PI3Kα in PN GSCs. Alpelisib inhibited GSC neurosphere growth and these effects were more pronounced in GSCs of the PN subtype. The antineoplastic effects of alpelisib were substantially enhanced when combined with pharmacologic mTOR inhibition. These findings identify the alpha catalytic PI3K isoform as a unique therapeutic target in proneural GBM and suggest that pharmacological mTOR inhibition may sensitize GSCs to selective PI3Kα inhibition.


Asunto(s)
Neoplasias Encefálicas , Glioma , Células Madre Neoplásicas , Fosfatidilinositol 3-Quinasas/metabolismo , Serina-Treonina Quinasas TOR , Tiazoles/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioma/tratamiento farmacológico , Glioma/enzimología , Glioma/patología , Humanos , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Esferoides Celulares/enzimología , Esferoides Celulares/patología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
4.
Biochem Biophys Res Commun ; 523(2): 398-404, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-31870546

RESUMEN

The role of protein kinase N1 (PKN1) in cell aggregation and spheroid formation was investigated using mouse embryonic fibroblasts (MEFs) deficient in kinase activity caused by a point mutation (T778A) in the activation loop. Wild type (WT) MEFs formed cell aggregates within a few hours in suspension cultures placed in poly-2-hydroxyethylmethacrylate (poly-HEMA) coated flat-bottom dishes. By contrast, PKN1[T778A] (PKN1 T778A/T778A homozygous knock-in) MEFs showed significantly delayed aggregate formation and higher susceptibility to cell death. Video analysis of suspension cultures revealed decreased cell motility and lesser frequency of cell-cell contact in PKN1[T778A] MEFs compared to that in WT MEFs. Aggregate formation of PKN1[T778A] MEFs was compensated by shaking the cell suspension. When cultured in U-shaped ultra-low attachment well plates, initially larger-sized and loosely packed aggregates of WT MEFs underwent compaction resulting in a single round spheroid. On the other hand, image-based quantitative analysis of PKN1[T778A] MEFs revealed irregular compaction with decreased roundness, solidity, and sphericity within 24 h. Flow cytometry of PKN1[T778A] MEFs revealed decreased surface-expression of N-cadherin and integrins α5 and αV. These results suggest that kinase activity of PKN1 controls cell aggregation and spheroid compaction in MEF suspension culture, possibly by regulating the cell migration and cell-surface expression of N-cadherin and integrins.


Asunto(s)
Proteína Quinasa C/metabolismo , Animales , Cadherinas/metabolismo , Agregación Celular/fisiología , Membrana Celular/metabolismo , Supervivencia Celular/fisiología , Células Cultivadas , Fibroblastos/citología , Fibroblastos/enzimología , Técnicas de Sustitución del Gen , Integrina alfa5/metabolismo , Integrina alfaV/metabolismo , Ratones , Ratones Mutantes , Mutación Puntual , Proteína Quinasa C/deficiencia , Proteína Quinasa C/genética , Esferoides Celulares/citología , Esferoides Celulares/enzimología
5.
J Clin Invest ; 128(11): 5034-5055, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30188867

RESUMEN

Lysyl-tRNA synthetase (KRS) functions canonically in cytosolic translational processes. However, KRS is highly expressed in colon cancer, and localizes to distinct cellular compartments upon phosphorylations (i.e., the plasma membranes after T52 phosphorylation and the nucleus after S207 phosphorylation), leading to probably alternative noncanonical functions. It is unknown how other subcellular KRSs crosstalk with environmental cues during cancer progression. Here, we demonstrate that the KRS-dependent metastatic behavior of colon cancer spheroids within 3D gels requires communication between cellular molecules and extracellular soluble factors and neighboring cells. Membranous KRS and nuclear KRS were found to participate in invasive cell dissemination of colon cancer spheroids in 3D gels. Cancer spheroids secreted GAS6 via a KRS-dependent mechanism and caused the M2 polarization of macrophages, which activated the neighboring cells via secretion of FGF2/GROα/M-CSF to promote cancer dissemination under environmental remodeling via fibroblast-mediated laminin production. Analyses of tissues from clinical colon cancer patients and Krs-/+ animal models for cancer metastasis supported the roles of KRS, GAS6, and M2 macrophages in KRS-dependent positive feedback between tumors and environmental factors. Altogether, KRS in colon cancer cells remodels the microenvironment to promote metastasis, which can thus be therapeutically targeted at these bidirectional KRS-dependent communications of cancer spheroids with environmental cues.


Asunto(s)
Neoplasias del Colon/enzimología , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Lisina-ARNt Ligasa/biosíntesis , Macrófagos/enzimología , Proteínas de Neoplasias/biosíntesis , Esferoides Celulares/enzimología , Microambiente Tumoral , Animales , Quimiocina CXCL1/genética , Quimiocina CXCL1/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Fibroblastos/enzimología , Fibroblastos/patología , Células HCT116 , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lisina-ARNt Ligasa/genética , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Esferoides Celulares/patología
6.
Int J Cancer ; 143(12): 3106-3119, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30006927

RESUMEN

Ovarian high-grade serous carcinoma (HGSC) is the most lethal gynecological malignancy. Prevailing evidences suggest that drug resistance and recurrence of ovarian HGSC are caused by the presence of cancer stem cells. Therefore, targeting cancer stems is appealing, however, all attempts to date, have failed. To circumvent this limit, we analyzed differential transcriptomes at early differentiation of ovarian HGSC stem cells and identified the developmental transcription factor GATA3 as highly expressed in stem, compared to progenitor cells. GATA3 expression associates with poor prognosis of ovarian HGSC patients, and was found to recruit the histone H3, lysine 27 (H3K27) demethylase, UTX, activate stemness markers, and promote stem-like phenotypes in ovarian HGSC cell lines. Targeting UTX by its inhibitor, GSKJ4, impeded GATA3-driven stemness phenotypes, and enhanced apoptosis of GATA3-expressing cancer cells. Combinations of gemcitabine or paclitaxel with GSKJ4, resulted in a synergistic cytotoxic effect. Our findings provide evidence for a new role for GATA3 in ovarian HGSC stemness, and demonstrate that GATA3 may serve as a biomarker for precision epigenetic therapy in the future.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Factor de Transcripción GATA3/efectos de los fármacos , Factor de Transcripción GATA3/fisiología , Células Madre Neoplásicas/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Fosfatasa Alcalina/metabolismo , Antígenos CD/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Biomarcadores de Tumor/metabolismo , Cadherinas/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Linaje de la Célula , Desoxicitidina/administración & dosificación , Desoxicitidina/análogos & derivados , Femenino , Factor de Transcripción GATA3/metabolismo , Histona Demetilasas/metabolismo , Humanos , Células Madre Neoplásicas/metabolismo , Proteínas Nucleares/metabolismo , Neoplasias Ováricas/metabolismo , Paclitaxel/administración & dosificación , Pronóstico , Unión Proteica , Esferoides Celulares/enzimología , Esferoides Celulares/metabolismo , Gemcitabina
7.
Biochem Biophys Res Commun ; 502(3): 382-388, 2018 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-29842882

RESUMEN

SETD2 is a histone methyltransferase that catalyzes the trimethylation of lysine 36 on histone 3. SETD2 is frequently found to be mutated or deleted in a variety of human tumors, whereas the role of SETD2 in oncogenesis of osteosarcoma has never been defined. Here in our study, we uncovered that SETD2 regulates tumor growth and chemosensitivity of osteosarcoma. Overexpression of SETD2 significantly inhibited osteosarcoma cell growth in vitro and in vivo. Moreover, SETD2 significantly enhanced cisplatin-induced apoptosis in osteosarcoma cells and inhibited cancer stem cell properties in OS cells. SETD2 regulates Wnt/ß-catenin signaling and its downstream gene c-myc, CD133 and cyclin D1. We further revealed that SETD2 upregulates H3K36me3 modification in GSK3B loci and promotes its transcription, which lead to ß-catenin degradation. Together, our study delineates SETD2 function in osteosarcoma as an important regulator of Wnt/ß-catenin signaling, and suggests SETD2 as a novel target in diagnosis and combined chemotherapy of osteosarcoma.


Asunto(s)
Neoplasias Óseas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Osteosarcoma/metabolismo , Animales , Antineoplásicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Línea Celular Tumoral , Proliferación Celular/fisiología , Cisplatino/farmacología , Regulación hacia Abajo , Resistencia a Antineoplásicos/fisiología , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Xenoinjertos , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Ratones , Ratones Desnudos , Células Madre Neoplásicas/enzimología , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Esferoides Celulares/enzimología , Vía de Señalización Wnt , beta Catenina/metabolismo
8.
BMC Biotechnol ; 17(1): 54, 2017 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-28637431

RESUMEN

BACKGROUND: Three-dimensional (3D) spheroids are frequently used in toxicological study because their morphology and function closely resemble those of tissue. As these properties are maintained over a long term, repeated treatment of the spheroids with a test object is possible. Generally, in the repeated treatment test to assess cytotoxicity in the spheroids, ATP assay, colorimetric measurement using pigments or high-content imaging analysis is performed. However, continuous assessment of cytotoxicity in the same spheroids using the above assays or analysis is impossible because the spheroids must be disrupted or killed. To overcome this technical limitation, we constructed a simple monitoring system in which cytotoxicity in the spheroids can be continuously monitored by nondestructive bioluminescence measurement. RESULTS: Mouse primary hepatocytes were isolated from transchromosomic (Tc) mice harboring a mouse artificial chromosome (MAC) vector expressing beetle luciferase Emerald Luc (ELuc) under the control of cytomegalovirus immediate early enhancer/chicken ß-actin promoter/rabbit ß-globin intron II (CAG) promoter, and used in 3D cultures. We confirmed that both luminescence and albumin secretion from the spheroids seeded in the 96-well format Cell-ableTM were maintained for approximately 1 month. Finally, we repetitively treated the luminescent 3D spheroids with representative hepatotoxicants for approximately 1 month, and continuously and nondestructively measured bioluminescence every day. We successfully obtained daily changes of the dose-response bioluminescence curves for the respective toxicants. CONCLUSIONS: In this study, we constructed a monitoring system in which cytotoxicity in the same 3D spheroids was continuously and sensitively monitored over a long term. Because this system can be easily applied to other cells, such as human primary cells or stem cells, it is expected to serve as the preferred platform for simple and cost-effective long-term monitoring of cellular events, including cytotoxicity.


Asunto(s)
Pruebas de Carcinogenicidad/métodos , Escarabajos/enzimología , Hepatocitos/efectos de los fármacos , Luciferasas/metabolismo , Mediciones Luminiscentes/métodos , Esferoides Celulares/efectos de los fármacos , Toxinas Biológicas/toxicidad , Animales , Bioensayo/métodos , Células Cultivadas , Genes Reporteros/genética , Hepatocitos/enzimología , Hepatocitos/patología , Estudios Longitudinales , Luciferasas/genética , Ratones , Esferoides Celulares/enzimología , Esferoides Celulares/patología
9.
Bull Exp Biol Med ; 161(1): 120-4, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27265124

RESUMEN

We studied the effects of DMSO and fibroblasts during HepaRG cell spheroid formation and conditions of their subsequent culturing on the levels of mRNA of the major cytochromes P450. A protocol of spheroid formation from differentiated HepaRG cells and their culturing in serum- and DMSO-free medium is developed.


Asunto(s)
Sistema Enzimático del Citocromo P-450/genética , Hepatocitos/enzimología , Esferoides Celulares/enzimología , Línea Celular , Medios de Cultivo/química , Sistema Enzimático del Citocromo P-450/metabolismo , Dimetilsulfóxido/química , Expresión Génica , Regulación Enzimológica de la Expresión Génica , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo
10.
Oncol Rep ; 34(5): 2305-10, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26323315

RESUMEN

Malignant mesothelioma is an aggressive tumor arising from mesothelial cells of serous membranes, and forms spheroid-like cell aggregates in pleural and peritoneal effusions. We examined the levels of anoikis, apoptosis induced by the detachment of cells from the extracellur matrix, in suspension culture in the human mesothelioma cell line NCI-H2052. NCI-H2052 cells were adherent in conventional monolayer cultures, but were found to form spheroids in suspension cultures using dishes with ultra-low cell binding capacity. NCI-H2052 cells proliferated in both cultures, but the proliferation rate was markedly lower in suspension cultures than in monolayer cultures. In addition, NCI-H2052 cells in suspension cultures showed little apoptosis, suggesting that the suspension culture induces anoikis resistance. Western blot analysis revealed that suspension cultures induced activation of Src family kinases (SFK) after spheroid formation. Dasatinib, an inhibitor of multi-tyrosine kinases including SFK, abolished anoikis resistance in suspension cultures, indicating that SFK activated by spheroid formation are responsible for anoikis resistance. Cisplatin induced apoptosis in NCI-H2052 cells, but the apoptotic rate was significantly lower in suspension cultures than in monolayer cultures, suggesting that spheroid formation is involved in cisplatin resistance. Furthermore, a combination of dasatinib and cisplatin induced apoptosis more significantly than either alone in suspension cultures. These results suggest that spheroid formation induces resistance to anoikis and to cisplatin through SFK activation and that dasatinib facilitates cisplatin-induced apoptosis in human mesothelioma cells.


Asunto(s)
Anoicis , Antineoplásicos/farmacología , Cisplatino/farmacología , Dasatinib/farmacología , Mesotelioma/enzimología , Familia-src Quinasas/antagonistas & inhibidores , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Mesotelioma/tratamiento farmacológico , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/enzimología , Familia-src Quinasas/metabolismo
11.
Cell Physiol Biochem ; 36(6): 2393-402, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26279442

RESUMEN

BACKGROUND/AIMS: Primary hepatocellular carcinoma (HCC) is highly invasive, and often results in an early distal metastasis resulting in poor prognosis and therapeutic outcome. Cancer cells disseminating from the tumor and entering circulation are termed circulating tumor cells (CTCs). Although substantial progress has been made to identify those CTCs in HCC, no good marker (cocktail) has so far been identified. METHODS: Since only tumorigenic CTCs form metastatic tumor in distal organs, we thus compared the HCC cells that form tumor spheres in culture to those that do not. We transduced HCC cells with a RFP reporter under MMP26 promoter and purified MMP26+CXCR4+ HCC cells. We examined tumor sphere formation in culture, presence of tumor cells in the circulation as well as capability of developing metastatic tumor after transplantation of MMP26+CXCR4+ HCC cells into nude mice, compared to other populations in HCC. RESULTS: Sphere-forming HCC cells expressed high levels of MMP26 and CXCR4. MMP26+CXCR4+ HCC cells formed significantly more tumor spheres in culture, compared to MMP26-CXCR4-, MMP26-CXCR4+ or MMP26+CXCR4- HCC cells. Moreover, tumor cells were more frequently detected in the circulation when MMP26+CXCR4+ HCC cells were subcutaneously transplanted. Further, subcutaneous transplantation of MMP26+CXCR4+ HCC cells, but not transplantation of MMP26-CXCR4-, MMP26-CXCR4+ or MMP26+CXCR4-HCC cells significantly developed distal metastatic tumors. CONCLUSION: MMP26+CXCR4+ cells may be CTCs in HCC. Selective elimination of MMP26+CXCR4+ cells may substantially reduce HCC metastasis after primary tumor resection.


Asunto(s)
Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Metaloproteinasas de la Matriz Secretadas/metabolismo , Células Neoplásicas Circulantes/patología , Receptores CXCR4/metabolismo , Animales , Carcinoma Hepatocelular/enzimología , Proliferación Celular , Células Hep G2 , Humanos , Neoplasias Hepáticas/enzimología , Ratones , Esferoides Celulares/enzimología , Esferoides Celulares/patología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Oncotarget ; 6(28): 24856-70, 2015 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-26259239

RESUMEN

Triple negative, resistant or metastatic disease are major factors in breast cancer mortality, warranting novel approaches. Carbonic anhydrase IX (CAIX) is implicated in survival, migration and invasion of breast cancer cells and inhibition provides an innovative therapeutic strategy. The efficacy of 5 novel ureido-substituted sulfamate CAIX inhibitors were assessed in increasingly complex breast cancer models, including cell lines in normoxia and hypoxia, 3D spheroids and an ex-vivo explant model utilizing fresh biopsy tissue from different breast cancer subtypes. CAIX expression was evaluated in a tissue microarray (TMA) of 92 paired lymph node and primary breast cancers and 2 inhibitors were appraised in vivo using MDA-MB-231 xenografts. FC11409B, FC9398A, FC9403, FC9396A and S4 decreased cell proliferation and migration and inhibited 3D spheroid invasion. S4, FC9398A and FC9403A inhibited or prevented invasion into collagen. FC9403A significantly reversed established invasion whilst FC9398A and DTP348 reduced xenograft growth. TMA analysis showed increased CAIX expression in triple negative cancers. These data establish CAIX inhibition as a relevant therapeutic goal in breast cancer, targeting the migratory, invasive, and metastatic potential of this disease. The use of biopsy tissue suggests efficacy against breast cancer subtypes, and should provide a useful tool in drug testing against invasive cancers.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Inhibidores de Anhidrasa Carbónica/farmacología , Anhidrasas Carbónicas/metabolismo , Terapia Molecular Dirigida/métodos , Animales , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Anhidrasa Carbónica IX , Inhibidores de Anhidrasa Carbónica/química , Hipoxia de la Célula , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales/métodos , Femenino , Humanos , Inmunohistoquímica , Ratones Desnudos , Estructura Molecular , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/enzimología , Análisis de Matrices Tisulares , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Methods Mol Biol ; 1233: 161-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25319898

RESUMEN

While growing cells as a monolayer is the traditional method for cell culture, the incorporation of multicellular spheroids into experimental design is becoming increasingly popular. This is due to the understanding that cells grown as spheroids tend to replicate the in vivo situation more reliably than monolayer cells. Thus, the use of multicellular spheroids may be more clinically relevant than monolayer cell cultures. Here, we describe methods for multicellular 3D spheroid generation that may be used to provide samples for receptor tyrosine kinase (and other protein) detection. Methods described include the forced-floating poly-HEMA method, the hanging-drop method, and the use of ECM to form multicellular 3D spheroids.


Asunto(s)
Técnicas de Cultivo de Célula , Matriz Extracelular/química , Polihidroxietil Metacrilato/química , Receptor ErbB-2/genética , Esferoides Celulares/enzimología , Línea Celular Tumoral , Proliferación Celular , Geles , Expresión Génica , Humanos , Receptor ErbB-2/metabolismo , Esferoides Celulares/patología
14.
PLoS One ; 9(11): e112438, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25375122

RESUMEN

Treatment of advanced ovarian cancer involves platinum-based chemotherapy. However, chemoresistance is a major obstacle. Cancer stem cells (CSCs) are thought to be one of the causes of chemoresistance, but the underlying mechanism remains elusive. Recently, human telomerase reverse transcriptase (hTERT) has been reported to promote CSC-like traits. In this study, we found that a mitotic inhibitor, eribulin mesylate (eribulin), effectively inhibited growth of platinum-resistant ovarian cancer cell lines. Eribulin-sensitive cells showed a higher efficiency for sphere formation, suggesting that these cells possess an enhanced CSC-like phenotype. Moreover, these cells expressed a higher level of hTERT, and suppression of hTERT expression by siRNA resulted in decreased sensitivity to eribulin, suggesting that hTERT may be a target for eribulin. Indeed, we found that eribulin directly inhibited RNA-dependent RNA polymerase (RdRP) activity, but not telomerase activity of hTERT in vitro. We propose that eribulin targets the RdRP activity of hTERT and may be an effective therapeutic option for CSCs. Furthermore, hTERT may be a useful biomarker to predict clinical responses to eribulin.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Furanos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Cetonas/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Transcripción Reversa/efectos de los fármacos , Telomerasa/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Esferoides Celulares/enzimología
15.
Cell Death Dis ; 5: e1502, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25356871

RESUMEN

Nuclear orphan receptor TLX (Drosophila tailless homolog) is essential for the maintenance of neural stem/progenitor cell self-renewal, but its role in neuroblastoma (NB) is not well understood. Here, we show that TLX is essential for the formation of tumor spheres in three different NB cell lines, when grown in neural stem cell media. We demonstrate that the knock down of TLX in IMR-32 cells diminishes its tumor sphere-forming capacity. In tumor spheres, TLX is coexpressed with the neural progenitor markers Nestin, CD133 and Oct-4. In addition, TLX is coexpressed with the migratory neural progenitor markers CD15 and matrix metalloproteinase-2 (MMP-2) in xenografts of primary NB cells from patients. Subsequently, we show the effect of TLX on the proliferative, invasive and migratory properties of IMR-32 cells. We attribute this to the recruitment of TLX to both MMP-2 and Oct-4 gene promoters, which resulted in the respective gene activation. In support of our findings, we found that TLX expression was high in NB patient tissues when compared with normal peripheral nervous system tissues. Further, the Kaplan-Meier estimator indicated a negative correlation between TLX expression and survival in 88 NB patients. Therefore, our results point at TLX being a crucial player in progression of NB, by promoting self-renewal of NB tumor-initiating cells and altering their migratory and invasive properties.


Asunto(s)
Metaloproteinasa 2 de la Matriz/metabolismo , Neuroblastoma/enzimología , Neuroblastoma/patología , Receptores Citoplasmáticos y Nucleares/metabolismo , Esferoides Celulares/patología , Animales , Biomarcadores de Tumor/metabolismo , Hipoxia de la Célula , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Activación Enzimática , Humanos , Ratones SCID , Invasividad Neoplásica , Células Madre Neoplásicas/enzimología , Células Madre Neoplásicas/patología , Factor 3 de Transcripción de Unión a Octámeros/genética , Receptores Nucleares Huérfanos , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Esferoides Celulares/enzimología , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Neurochem Res ; 39(7): 1199-205, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24706070

RESUMEN

Human dental follicle cells (DFCs) are ectomesenchymal multipotent stem cells that form spheroid cell clusters (SCCs) under serum free medium cell culture conditions (SFM). Until today, molecular mechanisms for the formation of SCCs are unknown. In this study a quantitative phosphoproteomics approach revealed regulated phosphorylated proteins in SCCs, which were derived from DFCs after 24 and 48 h in SFM. These regulated proteins were categorized using the Kyoto encyclopedia of genes and genomes program. Here, cellular processes and signaling pathway were identified such as the focal adhesion kinase (FAK) signaling pathway. In addition to the phosphoproteomics approach we showed that a specific phosphorylation of FAK (Y397) was required for the formation of SCCs. In conclusion, this study disclosed the phosphoproteome of SCCs for the first time and showed that the FAK signaling pathway is required for the formation of SCCs.


Asunto(s)
Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Esferoides Celulares/enzimología , Células Cultivadas , Saco Dental/citología , Saco Dental/enzimología , Humanos , Fosforilación/fisiología
17.
Cancer Res ; 74(9): 2487-98, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24626093

RESUMEN

Curcumin is known to induce apoptosis of cancer cells by different mechanisms, but its effects on cancer stem cells (CSC) have been less investigated. Here, we report that curcumin promotes the survival of DCLK1-positive colon CSCs, potentially confounding application of its anticancer properties. At optimal concentrations, curcumin greatly reduced expression levels of stem cell markers (DCLK1/CD44/ALDHA1/Lgr5/Nanog) in three-dimensional spheroid cultures and tumor xenografts derived from colon cancer cells. However, curcumin unexpectedly induced proliferation and autophagic survival of a subset of DCLK1-positive CSCs. Spheroid cultures were disintegrated by curcumin in vitro but regrew within 30 to 40 days of treatment, suggesting a survival benefit from autophagy, permitting long-term persistence of colorectal cancer. Notably, RNA interference-mediated silencing of DCLK1 triggered apoptotic cell death of colon cancer cells in vitro and in vivo, and abolished colorectal cancer survival in response to curcumin; combination of DCLK1-siRNA and curcumin dramatically reversed CSC phenotype, contributing to attenuation of the growth of spheroid cultures and tumor xenografts. Taken together, our findings confirm a role of DCLK1 in colon CSCs and highlight DCLK1 as a target to enhance antitumor properties of curcumin.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias del Colon/enzimología , Curcumina/farmacología , Péptidos y Proteínas de Señalización Intracelular/genética , Células Madre Neoplásicas/fisiología , Proteínas Serina-Treonina Quinasas/genética , Animales , Neoplasias del Colon/patología , Quinasas Similares a Doblecortina , Femenino , Técnicas de Silenciamiento del Gen , Células HCT116 , Humanos , Receptores de Hialuranos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Desnudos , Ratones SCID , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/genética , Esferoides Celulares/enzimología , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Cancer Res ; 74(6): 1857-69, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24448239

RESUMEN

Metastatic colorectal cancer remains largely incurable, although in a subset of patients, survival is prolonged by new targeting agents such as anti-EGF receptor (anti-EGFR) antibodies. This disease is believed to be supported by a subpopulation of stem-like cells termed colon cancer-initiating cell (CCIC), which may also confer therapeutic resistance. However, how CCICs respond to EGFR inhibition has not been fully characterized. To explore this question, we systematically generated CCICs through spheroid cultures of patient-derived xenografts of metastatic colorectal cancer. These cultures, termed "xenospheres," were capable of long-term self-propagation in vitro and phenocopied the original patient tumors in vivo, thus operationally defining CCICs. Xenosphere CCICs retained the genetic determinants for EGFR therapeutic response in vitro and in xenografts; like the original tumors, xenospheres harboring a mutated KRAS gene were resistant to EGFR therapy, whereas those harboring wild-type RAS pathway genes (RAS(wt)) were sensitive. Notably, the effects of EGFR inhibition in sensitive CCICs could be counteracted by cytokines secreted by cancer-associated fibroblasts. In particular, we found that the MET receptor ligand hepatocyte growth factor (HGF) was especially active in supporting in vitro CCIC proliferation and resistance to EGFR inhibition. Ectopic production of human HGF in CCIC xenografts rendered the xenografts susceptible to MET inhibition, which sensitized the response to EGFR therapy. By showing that RAS(wt) CCICs rely on both EGFR and MET signaling, our results offer a strong preclinical proof-of-concept for concurrent targeting of these two pathways in the clinical setting.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Neoplasias del Colon/enzimología , Receptores ErbB/antagonistas & inhibidores , Células Madre Neoplásicas/enzimología , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Proliferación Celular , Supervivencia Celular , Cetuximab , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Receptores ErbB/metabolismo , Femenino , Factor de Crecimiento de Hepatocito/fisiología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Células Madre Neoplásicas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Transducción de Señal , Esferoides Celulares/enzimología , Carga Tumoral/efectos de los fármacos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
PLoS One ; 9(1): e83144, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24416158

RESUMEN

Osteosarcoma is the most common primary bone tumour of both children and dogs. It is an aggressive tumour in both species with a rapid clinical course leading ultimately to metastasis. In dogs and children distant metastasis occurs in >80% of individuals treated by surgery alone. Both canine and human osteosarcoma has been shown to contain a sub-population of cancer stem cells (CSCs), which may drive tumour growth, recurrence and metastasis, suggesting that naturally occurring canine osteosarcoma could act as a preclinical model for the human disease. Here we report the successful isolation of CSCs from primary canine osteosarcoma, as well as established cell lines. We show that these cells can form tumourspheres, and demonstrate relative resistance to chemotherapy. We demonstrate similar results for the human osteosarcma cell lines, U2OS and SAOS2. Utilizing the Affymetrix canine microarray, we are able to definitively show that there are significant differences in global gene expression profiles of isolated osteosarcoma stem cells and the daughter adherent cells. We identified 13,221 significant differences (p = 0.05), and significantly, COX-2 was expressed 141-fold more in CSC spheres than daughter adherent cells. To study the role of COX-2 expression in CSCs we utilized the COX-2 inhibitors meloxicam and mavacoxib. We found that COX-2 inhibition had no effect on CSC growth, or resistance to chemotherapy. However inhibition of COX-2 in daughter cells prevented sphere formation, indicating a potential significant role for COX-2 in tumour initiation.


Asunto(s)
Carcinogénesis/genética , Ciclooxigenasa 2/metabolismo , Enfermedades de los Perros/enzimología , Regulación Neoplásica de la Expresión Génica , Células Madre Neoplásicas/enzimología , Osteosarcoma/genética , Osteosarcoma/veterinaria , Animales , Neoplasias Óseas/enzimología , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Neoplasias Óseas/veterinaria , Carcinogénesis/patología , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Supervivencia Celular/genética , Ciclooxigenasa 2/genética , Inhibidores de la Ciclooxigenasa 2/farmacología , Enfermedades de los Perros/genética , Enfermedades de los Perros/patología , Perros , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Invasividad Neoplásica , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Osteosarcoma/enzimología , Osteosarcoma/patología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/enzimología , Esferoides Celulares/patología , Ensayo de Tumor de Célula Madre
20.
Cell Biochem Biophys ; 68(3): 615-28, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24037715

RESUMEN

Solid tumours undergo considerable alterations in their metabolism of nutrients in order to generate sufficient energy and biomass for sustained growth and proliferation. During growth, the tumour microenvironment exerts a number of influences (e.g. hypoxia and acidity) that affect cellular biology and the flux or utilisation of fuels including glucose. The tumour spheroid model was used to characterise the utilisation of glucose and describe alterations to the activity and expression of key glycolytic enzymes during the tissue growth curve. Glucose was avidly consumed and associated with the production of lactate and an acidified medium, confirming the reliance on glycolytic pathways and a diminution of oxidative phosphorylation. The expression levels and activities of hexokinase, phosphofructokinase-1, pyruvate kinase and lactate dehydrogenase in the glycolytic pathway were measured to assess glycolytic capacity. Similar measurements were made for glucose-6-phosphate dehydrogenase, the entry point and regulatory step of the pentose-phosphate pathway (PPP) and for cytosolic malate dehydrogenase, a key link to TCA cycle intermediates. The parameters for these key enzymes were shown to undergo considerable variation during the growth curve of tumour spheroids. In addition, they revealed that the dynamic alterations were influenced by both transcriptional and posttranslational mechanisms.


Asunto(s)
Neoplasias/patología , Esferoides Celulares/metabolismo , Esferoides Celulares/patología , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular , Metabolismo Energético , Regulación Neoplásica de la Expresión Génica , Glucosa/metabolismo , Glucólisis , Humanos , Concentración de Iones de Hidrógeno , Modelos Biológicos , Neoplasias/enzimología , Neoplasias/metabolismo , Vía de Pentosa Fosfato , Esferoides Celulares/enzimología , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...