Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Int J Mol Sci ; 22(12)2021 Jun 19.
Article in English | MEDLINE | ID: mdl-34205320

ABSTRACT

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.


Subject(s)
Claudins/physiology , Drug Resistance, Neoplasm , Glucose Transport Proteins, Facilitative/metabolism , NF-E2-Related Factor 2/metabolism , Spheroids, Cellular/enzymology , A549 Cells , Anilides , Doxorubicin , Humans , Isothiocyanates , Reactive Oxygen Species/metabolism , Sulfoxides
2.
Molecules ; 26(2)2021 Jan 08.
Article in English | MEDLINE | ID: mdl-33429981

ABSTRACT

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.


Subject(s)
Aldehyde Dehydrogenase/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Brain Neoplasms , Electron Transport Complex I/antagonists & inhibitors , Glioblastoma , Neoplasm Proteins/antagonists & inhibitors , Spheroids, Cellular/enzymology , Aldehyde Dehydrogenase/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/enzymology , Electron Transport Complex I/metabolism , Enzyme Inhibitors/pharmacology , Glioblastoma/drug therapy , Glioblastoma/enzymology , Humans , Neoplasm Proteins/metabolism , Temozolomide/pharmacology
3.
Sci Rep ; 10(1): 21873, 2020 12 14.
Article in English | MEDLINE | ID: mdl-33318517

ABSTRACT

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.


Subject(s)
Brain Neoplasms , Glioma , Neoplastic Stem Cells , Phosphatidylinositol 3-Kinases/metabolism , TOR Serine-Threonine Kinases , Thiazoles/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Cell Line, Tumor , Glioma/drug therapy , Glioma/enzymology , Glioma/pathology , Humans , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Proto-Oncogene Proteins c-akt/metabolism , Spheroids, Cellular/enzymology , Spheroids, Cellular/pathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism
4.
Biochem Biophys Res Commun ; 523(2): 398-404, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31870546

ABSTRACT

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.


Subject(s)
Protein Kinase C/metabolism , Animals , Cadherins/metabolism , Cell Aggregation/physiology , Cell Membrane/metabolism , Cell Survival/physiology , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/enzymology , Gene Knock-In Techniques , Integrin alpha5/metabolism , Integrin alphaV/metabolism , Mice , Mice, Mutant Strains , Point Mutation , Protein Kinase C/deficiency , Protein Kinase C/genetics , Spheroids, Cellular/cytology , Spheroids, Cellular/enzymology
5.
J Clin Invest ; 128(11): 5034-5055, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30188867

ABSTRACT

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.


Subject(s)
Colonic Neoplasms/enzymology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Lysine-tRNA Ligase/biosynthesis , Macrophages/enzymology , Neoplasm Proteins/biosynthesis , Spheroids, Cellular/enzymology , Tumor Microenvironment , Animals , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Fibroblasts/enzymology , Fibroblasts/pathology , HCT116 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Lysine-tRNA Ligase/genetics , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neoplasm Metastasis , Neoplasm Proteins/genetics , Spheroids, Cellular/pathology
6.
Int J Cancer ; 143(12): 3106-3119, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30006927

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , GATA3 Transcription Factor/drug effects , GATA3 Transcription Factor/physiology , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Alkaline Phosphatase/metabolism , Antigens, CD/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Biomarkers, Tumor/metabolism , Cadherins/metabolism , Cell Differentiation , Cell Line, Tumor , Cell Lineage , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Female , GATA3 Transcription Factor/metabolism , Histone Demethylases/metabolism , Humans , Neoplastic Stem Cells/metabolism , Nuclear Proteins/metabolism , Ovarian Neoplasms/metabolism , Paclitaxel/administration & dosage , Prognosis , Protein Binding , Spheroids, Cellular/enzymology , Spheroids, Cellular/metabolism , Gemcitabine
7.
Biochem Biophys Res Commun ; 502(3): 382-388, 2018 07 20.
Article in English | MEDLINE | ID: mdl-29842882

ABSTRACT

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.


Subject(s)
Bone Neoplasms/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Osteosarcoma/metabolism , Animals , Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/physiology , Cisplatin/pharmacology , Down-Regulation , Drug Resistance, Neoplasm/physiology , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Heterografts , Histone-Lysine N-Methyltransferase/genetics , Humans , Mice , Mice, Nude , Neoplastic Stem Cells/enzymology , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Spheroids, Cellular/enzymology , Wnt Signaling Pathway , beta Catenin/metabolism
8.
BMC Biotechnol ; 17(1): 54, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28637431

ABSTRACT

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.


Subject(s)
Carcinogenicity Tests/methods , Coleoptera/enzymology , Hepatocytes/drug effects , Luciferases/metabolism , Luminescent Measurements/methods , Spheroids, Cellular/drug effects , Toxins, Biological/toxicity , Animals , Biological Assay/methods , Cells, Cultured , Genes, Reporter/genetics , Hepatocytes/enzymology , Hepatocytes/pathology , Longitudinal Studies , Luciferases/genetics , Mice , Spheroids, Cellular/enzymology , Spheroids, Cellular/pathology
9.
Bull Exp Biol Med ; 161(1): 120-4, 2016 May.
Article in English | MEDLINE | ID: mdl-27265124

ABSTRACT

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.


Subject(s)
Cytochrome P-450 Enzyme System/genetics , Hepatocytes/enzymology , Spheroids, Cellular/enzymology , Cell Line , Culture Media/chemistry , Cytochrome P-450 Enzyme System/metabolism , Dimethyl Sulfoxide/chemistry , Gene Expression , Gene Expression Regulation, Enzymologic , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism
10.
Oncol Rep ; 34(5): 2305-10, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26323315

ABSTRACT

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.


Subject(s)
Anoikis , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Dasatinib/pharmacology , Mesothelioma/enzymology , src-Family Kinases/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Humans , Mesothelioma/drug therapy , Spheroids, Cellular/drug effects , Spheroids, Cellular/enzymology , src-Family Kinases/metabolism
11.
Cell Physiol Biochem ; 36(6): 2393-402, 2015.
Article in English | MEDLINE | ID: mdl-26279442

ABSTRACT

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.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Matrix Metalloproteinases, Secreted/metabolism , Neoplastic Cells, Circulating/pathology , Receptors, CXCR4/metabolism , Animals , Carcinoma, Hepatocellular/enzymology , Cell Proliferation , Hep G2 Cells , Humans , Liver Neoplasms/enzymology , Mice , Spheroids, Cellular/enzymology , Spheroids, Cellular/pathology , Xenograft Model Antitumor Assays
12.
Oncotarget ; 6(28): 24856-70, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26259239

ABSTRACT

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.


Subject(s)
Antigens, Neoplasm/metabolism , Breast Neoplasms/drug therapy , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Molecular Targeted Therapy/methods , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Carbonic Anhydrase IX , Carbonic Anhydrase Inhibitors/chemistry , Cell Hypoxia , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor/methods , Female , Humans , Immunohistochemistry , Mice, Nude , Molecular Structure , Spheroids, Cellular/drug effects , Spheroids, Cellular/enzymology , Tissue Array Analysis , Xenograft Model Antitumor Assays
13.
Methods Mol Biol ; 1233: 161-8, 2015.
Article in English | MEDLINE | ID: mdl-25319898

ABSTRACT

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.


Subject(s)
Cell Culture Techniques , Extracellular Matrix/chemistry , Polyhydroxyethyl Methacrylate/chemistry , Receptor, ErbB-2/genetics , Spheroids, Cellular/enzymology , Cell Line, Tumor , Cell Proliferation , Gels , Gene Expression , Humans , Receptor, ErbB-2/metabolism , Spheroids, Cellular/pathology
14.
PLoS One ; 9(11): e112438, 2014.
Article in English | MEDLINE | ID: mdl-25375122

ABSTRACT

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.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Furans/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Ketones/pharmacology , Ovarian Neoplasms/drug therapy , Reverse Transcription/drug effects , Telomerase/metabolism , Cell Line, Tumor , Female , Humans , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/pathology , Spheroids, Cellular/enzymology
15.
Cell Death Dis ; 5: e1502, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25356871

ABSTRACT

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.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Neuroblastoma/enzymology , Neuroblastoma/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Spheroids, Cellular/pathology , Animals , Biomarkers, Tumor/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Cell Proliferation , Enzyme Activation , Humans , Mice, SCID , Neoplasm Invasiveness , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Octamer Transcription Factor-3/genetics , Orphan Nuclear Receptors , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Spheroids, Cellular/enzymology , Survival Analysis , Xenograft Model Antitumor Assays
16.
Neurochem Res ; 39(7): 1199-205, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24706070

ABSTRACT

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.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Spheroids, Cellular/enzymology , Cells, Cultured , Dental Sac/cytology , Dental Sac/enzymology , Humans , Phosphorylation/physiology
17.
Cancer Res ; 74(9): 2487-98, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24626093

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cell Survival/drug effects , Colonic Neoplasms/enzymology , Curcumin/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Neoplastic Stem Cells/physiology , Protein Serine-Threonine Kinases/genetics , Animals , Colonic Neoplasms/pathology , Doublecortin-Like Kinases , Female , Gene Knockdown Techniques , HCT116 Cells , Humans , Hyaluronan Receptors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , Spheroids, Cellular/enzymology , Xenograft Model Antitumor Assays
18.
Cancer Res ; 74(6): 1857-69, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24448239

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Colonic Neoplasms/enzymology , ErbB Receptors/antagonists & inhibitors , Neoplastic Stem Cells/enzymology , Proto-Oncogene Proteins c-met/metabolism , Animals , Cell Proliferation , Cell Survival , Cetuximab , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm , Drug Synergism , ErbB Receptors/metabolism , Female , Hepatocyte Growth Factor/physiology , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Neoplastic Stem Cells/drug effects , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Signal Transduction , Spheroids, Cellular/enzymology , Tumor Burden/drug effects , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
PLoS One ; 9(1): e83144, 2014.
Article in English | MEDLINE | ID: mdl-24416158

ABSTRACT

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.


Subject(s)
Carcinogenesis/genetics , Cyclooxygenase 2/metabolism , Dog Diseases/enzymology , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells/enzymology , Osteosarcoma/genetics , Osteosarcoma/veterinary , Animals , Bone Neoplasms/enzymology , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/veterinary , Carcinogenesis/pathology , Cell Death/drug effects , Cell Death/genetics , Cell Survival/genetics , Cyclooxygenase 2/genetics , Cyclooxygenase 2 Inhibitors/pharmacology , Dog Diseases/genetics , Dog Diseases/pathology , Dogs , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasm Invasiveness , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Osteosarcoma/enzymology , Osteosarcoma/pathology , Spheroids, Cellular/drug effects , Spheroids, Cellular/enzymology , Spheroids, Cellular/pathology , Tumor Stem Cell Assay
20.
Cell Biochem Biophys ; 68(3): 615-28, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24037715

ABSTRACT

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.


Subject(s)
Neoplasms/pathology , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Energy Metabolism , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Glycolysis , Humans , Hydrogen-Ion Concentration , Models, Biological , Neoplasms/enzymology , Neoplasms/metabolism , Pentose Phosphate Pathway , Spheroids, Cellular/enzymology , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...