Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 116
Filter
1.
Int J Mol Sci ; 22(13)2021 Jun 22.
Article in English | MEDLINE | ID: mdl-34206240

ABSTRACT

The development of colon cancer, one of the most common malignancies, is accompanied with numerous lipid alterations. However, analyses of whole tumor samples may not always provide an accurate description of specific changes occurring directly in tumor epithelial cells. Here, we analyzed in detail the phospholipid (PL), lysophospholipid (lysoPL), and fatty acid (FA) profiles of purified EpCAM+ cells, isolated from tumor and adjacent non-tumor tissues of colon cancer patients. We found that a number of FAs increased significantly in isolated tumor cells, which also included a number of long polyunsaturated FAs. Higher levels of FAs were associated with increased expression of FA synthesis genes, as well as with altered expression of enzymes involved in FA elongation and desaturation, including particularly fatty acid synthase, stearoyl-CoA desaturase, fatty acid desaturase 2 and ELOVL5 fatty acid elongase 5 We identified significant changes in ratios of specific lysoPLs and corresponding PLs. A number of lysophosphatidylcholine and lysophosphatidylethanolamine species, containing long-chain and very-long chain FAs, often with high numbers of double bonds, were significantly upregulated in tumor cells. Increased de novo synthesis of very long-chain FAs, or, altered uptake or incorporation of these FAs into specific lysoPLs in tumor cells, may thus contribute to reprogramming of cellular phospholipidome and membrane alterations observed in colon cancer.


Subject(s)
Adenocarcinoma/metabolism , Colonic Neoplasms/metabolism , Fatty Acids/metabolism , Gene Expression Regulation, Neoplastic , Lipid Metabolism , Phospholipids/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Aged , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Epithelial Cells/enzymology , Epithelial Cells/metabolism , Fatty Acid Desaturases/genetics , Fatty Acid Desaturases/metabolism , Fatty Acid Elongases/genetics , Fatty Acid Elongases/metabolism , Fatty Acid Synthases/genetics , Fatty Acid Synthases/metabolism , Female , Humans , Lipidomics , Lipogenesis , Male , Stearoyl-CoA Desaturase/genetics , Stearoyl-CoA Desaturase/metabolism
3.
PLoS One ; 15(1): e0228010, 2020.
Article in English | MEDLINE | ID: mdl-31999740

ABSTRACT

Identification of changes of phospholipid (PL) composition occurring during colorectal cancer (CRC) development may help us to better understand their roles in CRC cells. Here, we used LC-MS/MS-based PL profiling of cell lines derived from normal colon mucosa, or isolated at distinct stages of CRC development, in order to study alterations of PL species potentially linked with cell transformation. We found that a detailed evaluation of phosphatidylinositol (PI) and phosphatidylserine (PS) classes allowed us to cluster the studied epithelial cell lines according to their origin: i) cells originally derived from normal colon tissue (NCM460, FHC); ii) cell lines derived from colon adenoma or less advanced differentiating adenocarcinoma cells (AA/C1, HT-29); or, iii) cells obtained by in vitro transformation of adenoma cells and advanced colon adenocarcinoma cells (HCT-116, AA/C1/SB10, SW480, SW620). Although we tentatively identified several PS and PI species contributing to cell line clustering, full PI and PS profiles appeared to be a key to the successful cell line discrimination. In parallel, we compared PL composition of primary epithelial (EpCAM-positive) cells, isolated from tumor and adjacent non-tumor tissues of colon cancer patients, with PL profiles of cell lines derived from normal colon mucosa (NCM460) and from colon adenocarcinoma (HCT-116, SW480) cells, respectively. In general, higher total levels of all PL classes were observed in tumor cells. The overall PL profiles of the cell lines, when compared with the respective patient-derived cells, exhibited similarities. Nevertheless, there were also some notable differences in levels of individual PL species. This indicated that epithelial cell lines, derived either from normal colon tissue or from CRC cells, could be employed as models for functional lipidomic analyses of colon cells, albeit with some caution. The biological significance of the observed PL deregulation, or their potential links with specific CRC stages, deserve further investigation.


Subject(s)
Colon/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Epithelial Cells/metabolism , Lipidomics , Phospholipids/metabolism , Cell Line, Tumor , Epithelial Cells/pathology , Humans , Principal Component Analysis
4.
Int J Mol Sci ; 20(23)2019 Nov 30.
Article in English | MEDLINE | ID: mdl-31801289

ABSTRACT

The development and progression of colorectal cancer (CRC), a major cause of cancer-related death in the western world, is accompanied with alterations of sphingolipid (SL) composition in colon tumors. A number of enzymes involved in the SL metabolism have been found to be deregulated in human colon tumors, in experimental rodent studies, and in human colon cancer cells in vitro. Therefore, the enzymatic pathways that modulate SL levels have received a significant attention, due to their possible contribution to CRC development, or as potential therapeutic targets. Many of these enzymes are associated with an increased sphingosine-1-phosphate/ceramide ratio, which is in turn linked with increased colon cancer cell survival, proliferation and cancer progression. Nevertheless, more attention should also be paid to the more complex SLs, including specific glycosphingolipids, such as lactosylceramides, which can be also deregulated during CRC development. In this review, we focus on the potential roles of individual SLs/SL metabolism enzymes in colon cancer, as well as on the pros and cons of employing the current in vitro models of colon cancer cells for lipidomic studies investigating the SL metabolism in CRC.


Subject(s)
Colonic Neoplasms/enzymology , Gene Expression Regulation, Neoplastic , Lactosylceramides/metabolism , Lipid Metabolism/genetics , Sphingolipids/metabolism , Acid Ceramidase/genetics , Acid Ceramidase/metabolism , Alkaline Ceramidase/genetics , Alkaline Ceramidase/metabolism , Animals , Ceramides/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Disease Models, Animal , Humans , Lysophospholipids/metabolism , Neutral Ceramidase/genetics , Neutral Ceramidase/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine N-Acyltransferase/genetics , Sphingosine N-Acyltransferase/metabolism , Tumor Cells, Cultured
5.
Toxicol Sci ; 172(2): 368-384, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31536130

ABSTRACT

The aryl hydrocarbon receptor (AhR) activation has been shown to alter proliferation, apoptosis, or differentiation of adult rat liver progenitors. Here, we investigated the impact of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-mediated AhR activation on a human model of bipotent liver progenitors, undifferentiated HepaRG cells. We used both intact undifferentiated HepaRG cells, and the cells with silenced Hippo pathway effectors, yes-associated protein 1 (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), which play key role(s) in tissue-specific progenitor cell self-renewal and expansion, such as in liver, cardiac, or respiratory progenitors. TCDD induced cell proliferation in confluent undifferentiated HepaRG cells; however, following YAP, and, in particular, double YAP/TAZ knockdown, TCDD promoted induction of apoptosis. These results suggested that, unlike in mature hepatocytes, or hepatocyte-like cells, activation of the AhR may sensitize undifferentiated HepaRG cells to apoptotic stimuli. Induction of apoptosis in cells with silenced YAP/TAZ was associated with upregulation of death ligand TRAIL, and seemed to involve both extrinsic and mitochondrial apoptosis pathways. Global gene expression analysis further suggested that TCDD significantly altered expression of constituents and/or transcriptional targets of signaling pathways participating in control of expansion or differentiation of liver progenitors, including EGFR, Wnt/ß-catenin, or tumor growth factor-ß signaling pathways. TCDD significantly upregulated cytosolic proapoptotic protein BMF (Bcl-2 modifying factor) in HepaRG cells, which could be linked with an enhanced sensitivity of TCDD-treated cells to apoptosis. Our results suggest that, in addition to promotion of cell proliferation and alteration of signaling pathways controlling expansion of human adult liver progenitors, AhR ligands may also sensitize human liver progenitor cells to apoptosis.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Liver/drug effects , Models, Biological , Polychlorinated Dibenzodioxins/toxicity , Stem Cells/drug effects , Adaptor Proteins, Signal Transducing/genetics , Apoptosis/genetics , Cell Line , Cell Proliferation/genetics , Gene Expression/drug effects , Humans , Liver/pathology , RNA, Small Interfering/genetics , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Stem Cells/pathology , Trans-Activators/genetics , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Transfection , YAP-Signaling Proteins
6.
Toxicology ; 412: 1-11, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30439556

ABSTRACT

Butyrate helps to maintain colon homeostasis and exhibits chemopreventive effects in colon epithelium. We examined the interactive effects of butyrate and benzo[a]pyrene (BaP), dietary carcinogen, in regulation of expression of a panel of phase I and II xenobiotic metabolizing enzymes (XMEs) in human colon cells. In human colon carcinoma HCT-116 and HT-29 cell lines, butyrate alone increased mRNA levels of some enzymes, such as N-acetyltransferases (in particular NAT2). In combination with BaP, butyrate potentiated induction of cytochrome P450 family 1 enzymes (CYP1A1), aldo-keto reductases (AKR1C1) or UDP-glucuronosyltransferases (UGT1A1). There were some notable differences between cell lines, as butyrate potentiated induction of NAD(P)H:quinone oxidoreductase 1 (NQO1) and UGT1A4 only in HCT-116 cells, and it even repressed AKR1C3 induction in HT-29 cells. Butyrate also promoted induction of CYP1, NQO1, NAT2, UGT1A1 or UGT1A4 in human colon Caco-2 cells, in a differentiation-dependent manner. Differentiated Caco-2 cells exhibited a higher inducibility of selected XME genes than undifferentiated cells. Butyrate increased induction of enzymatic activities of NATs, NQO1 and UGTs by BaP in HCT-116 and HT29 cells, whereas in differentiated Caco-2 cells it helped to increase only enzymatic activity of NQO1 and UGTs. Together, the present data suggest that butyrate may modulate expression/activities of several enzymes involved in metabolism of carcinogens in colon. In some cases (NAT2, UGT1 A1), this was linked to inhibition of histone deacetylases (HDAC), as confirmed by using HDAC inhibitor trichostatin A. These results may have implications for our understanding of the role of butyrate in regulation of XMEs and carcinogen metabolism in colon.


Subject(s)
Benzo(a)pyrene/toxicity , Butyrates/pharmacology , Carcinogens/toxicity , Oxidoreductases/metabolism , Transferases/metabolism , Cell Line , Colon/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Oxidoreductases/genetics , Transferases/genetics , Xenobiotics/metabolism
7.
Food Chem Toxicol ; 124: 374-384, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30572064

ABSTRACT

Dietary carcinogens, such as benzo[a]pyrene (BaP), are suspected to contribute to colorectal cancer development. n-3 Polyunsaturated fatty acids (PUFAs) decrease colorectal cancer risk in individuals consuming diets rich in PUFAs. Here, we investigated the impact of eicosapentaenoic (EPA) and docosahexaenoic (DHA) acid on metabolism and genotoxicity of BaP in human cell models derived from the colon: HT-29 and HCT-116 cell lines. Both PUFAs reduced levels of excreted BaP metabolites, in particular BaP-tetrols and hydroxylated BaP metabolites, as well as formation of DNA adducts in HT-29 and HCT-116 cells. However, EPA appeared to be a more potent inhibitor of formation of some intracellular BaP metabolites, including BaP-7,8-dihydrodiol. EPA also reduced phosphorylation of histone H2AX (Ser139) in HT-29 cells, which indicated that it may reduce further forms of DNA damage, including DNA double strand breaks. Both PUFAs inhibited induction of CYP1 activity in colon cells determined as 7-ethoxyresorufin-O-deethylase (EROD); this was at least partly linked with inhibition of induction of CYP1A1, 1A2 and 1B1 mRNAs. The downregulation and/or inhibition of CYP1 enzymes by PUFAs could thus alter metabolism and reduce genotoxicity of BaP in human colon cells, which might contribute to known chemopreventive effects of PUFAs in colon epithelium.


Subject(s)
Anticarcinogenic Agents/pharmacology , Benzo(a)pyrene/metabolism , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Epithelial Cells/drug effects , Mutagens/metabolism , Benzo(a)pyrene/adverse effects , Cell Line, Tumor , Cytochrome P450 Family 1/metabolism , DNA Adducts/metabolism , DNA Damage/drug effects , Histones/metabolism , Humans , Mutagens/adverse effects , S Phase Cell Cycle Checkpoints/drug effects
8.
Biomed Pharmacother ; 99: 511-522, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29665654

ABSTRACT

OBJECTIVE: Cancer stem-like cells (CSLCs) are considered a root of tumorigenicity and resistance. However, their identification remains challenging. The use of the side population (SP) assay as a credible marker of CSLCs remains controversial. The SP assay relies on the elevated activity of ABC transporters that, in turn, can be modulated by hypericin (HYP), a photosensitizer and bioactive compound of St. John's Wort (Hypericum perforatum), a popular over-the-counter antidepressant. Here we aimed to comprehensively characterize the SP phenotype of cancer cells and to determine the impact of HYP on these cells. METHODS: Flow cytometry and sorting-based assays were employed, including CD24-, CD44-, CD133-, and ALDH-positivity, clonogenicity, 3D-forming ability, ABC transporter expression and activity, and intracellular accumulation of HYP/Hoechst 33342. The tumorigenic ability of SP, nonSP, and HYP-treated cells was verified by xenotransplantation into immunodeficient mice. RESULTS: The SP phenotype was associated with elevated expression of several investigated transporters and more intensive growth in non-adherent conditions but not with higher clonogenicity, tumorigenicity or ALDH-positivity. Despite stimulated BCRP level and MRP1 activity, HYP reversibly decreased the SP proportion, presumably via competitive inhibition of BCRP. HYP-selected SP cells acquired additional traits of resistance and extensively eliminated HYP. CONCLUSIONS: Our results suggest that SP is not an unequivocal CSLC-marker. However, SP could play an important role in modulating HYP-treatment and serve as a negative predictive tool for HYP-based therapies. Moreover, the use of supplements containing HYP by cancer patients should be carefully considered, due to its proposed effect on drug efflux and complex impact on tumor cells, which have not yet been sufficiently characterized.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Perylene/analogs & derivatives , Side-Population Cells/pathology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Aldehyde Dehydrogenase/metabolism , Animals , Anthracenes , Biomarkers, Tumor/metabolism , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Clone Cells , Humans , Mice, SCID , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Perylene/pharmacology , Phenotype , Side-Population Cells/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Substrate Specificity/drug effects , Survival Analysis
9.
J Cell Biochem ; 119(6): 4664-4679, 2018 06.
Article in English | MEDLINE | ID: mdl-29274292

ABSTRACT

Docosahexaenoic acid (DHA) and sodium butyrate (NaBt) exhibit a number of interactive effects on colon cancer cell growth, differentiation, or apoptosis; however, the molecular mechanisms responsible for these interactions and their impact on cellular lipidome are still not fully clear. Here, we show that both dietary agents together induce dynamic alterations of lipid metabolism, specific cellular lipid classes, and fatty acid composition. In HT-29 cell line, a model of differentiating colon carcinoma cells, NaBt supported incorporation of free DHA into non-polar lipids and their accumulation in cytoplasmic lipid droplets. DHA itself was not incorporated into sphingolipids; however, it significantly altered representation of individual ceramide (Cer) classes, in particular in combination with NaBt (DHA/NaBt). We observed altered expression of enzymes involved in Cer metabolism in cells treated with NaBt or DHA/NaBt, and exogenous Cer 16:0 was found to promote induction of apoptosis in differentiating HT-29 cells. NaBt, together with DHA, increased n-3 fatty acid synthesis and attenuated metabolism of monounsaturated fatty acids. Finally, DHA and/or NaBt altered expression of proteins involved in synthesis of fatty acids, including elongase 5, stearoyl CoA desaturase 1, or fatty acid synthase, with NaBt increasing expression of caveolin-1 and CD36 transporter, which may further promote DHA incorporation and its impact on cellular lipidome. In conclusion, our results indicate that interactions of DHA and NaBt exert complex changes in cellular lipidome, which may contribute to the alterations of colon cancer cell differentiation/apoptotic responses. The present data extend our knowledge about the nature of interactive effects of dietary fatty acids.


Subject(s)
Apoptosis/drug effects , Butyrates/pharmacology , Cell Differentiation/drug effects , Colonic Neoplasms/metabolism , Docosahexaenoic Acids/pharmacology , Lipid Metabolism/drug effects , Membrane Lipids/metabolism , Colonic Neoplasms/pathology , HCT116 Cells , Humans , Membrane Lipids/classification
10.
Cytometry A ; 93(9): 941-951, 2018 07.
Article in English | MEDLINE | ID: mdl-28383825

ABSTRACT

The identification of fibroblasts and cancer-associated fibroblasts from human cancer tissue using surface markers is difficult, especially because the markers used currently are usually not expressed solely by fibroblasts, and the identification of fibroblast-specific surface molecules is still under investigation. It was aimed to compare three commercially available antibodies in the detection of different surface epitopes of fibroblasts (anti-fibroblast, fibroblast activation protein α, and fibroblast surface protein). The specificity of their expression, employing fibroblast cell lines and tumor-derived fibroblasts from breast and prostate tissues was investigated. Both the established fibroblast cell line HFF-1 and ex vivo primary fibroblasts isolated from breast and prostate cancer tissues expressed the tested surface markers to different degrees. Surprisingly, those markers were expressed also by permanent cell lines of epithelial origin, both benign and cancer-derived (breast-cell lines MCF 10A, HMLE and prostate-cell lines BPH-1, DU 145, and PC-3). The expression of fibroblast activation protein α increased on the surface of previously described models of epithelial cells undergoing epithelial-to-mesenchymal transition in response to treatment with TGF-ß1. To prove the co-expression of the fibroblast markers on cells of epithelial origin, we used freshly dissociated human prostate and breast cancer tissues. The results confirmed the co-expression of anti-fibroblast and fibroblast surface protein on CD31/CD45-negative/EpCAM-positive epithelial cells. In summary, our data support the findings that the tested fibroblast markers are not fibroblast specific and may be expressed also by cells of epithelial origin (e.g., cells undergoing EMT). Therefore, the expression of these markers should be interpreted with caution, and the combination of several epitopes for both positive (anti-fibroblast or fibroblast activation protein α) and negative (EpCAM) identification of fibroblasts from breast and prostate tumor tissues is advised. © 2017 International Society for Advancement of Cytometry.


Subject(s)
Biomarkers/metabolism , Epithelial-Mesenchymal Transition/physiology , Fibroblasts/metabolism , Gelatinases/metabolism , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Endopeptidases , Epithelial Cell Adhesion Molecule/metabolism , Epithelial Cells/metabolism , Female , Humans , Leukocyte Common Antigens/metabolism , Male , PC-3 Cells , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Prostatic Neoplasms/metabolism , Transforming Growth Factor beta1/metabolism
11.
PLoS One ; 12(11): e0188584, 2017.
Article in English | MEDLINE | ID: mdl-29182622

ABSTRACT

Searching for new strategies for effective elimination of human prostate cancer cells, we investigated the cooperative cytotoxic action of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and two platinum-based complexes, cisplatin or LA-12, and related molecular mechanisms. We demonstrated a notable ability of cisplatin or LA-12 to enhance the sensitivity of several human prostate cancer cell lines to TRAIL-induced cell death via an engagement of mitochondrial apoptotic pathway. This was accompanied by augmented Bid cleavage, Bak activation, loss of mitochondrial membrane potential, activation of caspase-8, -10, -9, and -3, and XIAP cleavage. RNAi-mediated silencing of Bid or Bak in Bax-deficient DU 145 cells suppressed the drug combination-induced cytotoxicity, further underscoring the involvement of mitochondrial signaling. The caspase-10 was dispensable for enhancement of cisplatin/LA-12 and TRAIL combination-induced cell death and stimulation of Bid cleavage. Importantly, we newly demonstrated LA-12-mediated enhancement of TRAIL-induced cell death in cancer cells derived from human patient prostate tumor specimens. Our results provide convincing evidence that employing TRAIL combined with cisplatin/LA-12 could contribute to more effective killing of prostate cancer cells compared to the individual action of the drugs, and offer new mechanistic insights into their cooperative anticancer action.


Subject(s)
Amantadine/analogs & derivatives , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspase 10/metabolism , Cisplatin/pharmacology , Mitochondria/drug effects , Organoplatinum Compounds/pharmacology , Prostatic Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Amantadine/pharmacology , Humans , Male , Mitochondria/metabolism , Prostatic Neoplasms/metabolism
12.
Neoplasia ; 19(10): 830-841, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28888100

ABSTRACT

Although Chk1 kinase inhibitors are currently under clinical investigation as effective cancer cell sensitizers to the cytotoxic effects of numerous chemotherapeutics, there is still a considerable uncertainty regarding their role in modulation of anticancer potential of platinum-based drugs. Here we newly demonstrate the ability of one of the most specific Chk1 inhibitors, SCH900776 (MK-8776), to enhance human colon cancer cell sensitivity to the cytotoxic effects of platinum(II) cisplatin and platinum(IV)- LA-12 complexes. The combined treatment with SCH900776 and cisplatin or LA-12 results in apparent increase in G1/S phase-related apoptosis, stimulation of mitotic slippage, and senescence of HCT116 cells. We further show that the cancer cell response to the drug combinations is significantly affected by the p21, p53, and PTEN status. In contrast to their wt counterparts, the p53- or p21-deficient cells treated with SCH900776 and cisplatin or LA-12 enter mitosis and become polyploid, and the senescence phenotype is strongly suppressed. While the cell death induced by SCH900776 and cisplatin or LA-12 is significantly delayed in the absence of p53, the anticancer action of the drug combinations is significantly accelerated in p21-deficient cells, which is associated with stimulation of apoptosis beyond G2/M cell cycle phase. We also show that cooperative killing action of the drug combinations in HCT116 cells is facilitated in the absence of PTEN. Our results indicate that SCH900776 may act as an important modulator of cytotoxic response triggered by platinum-based drugs in colon cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Checkpoint Kinase 1/antagonists & inhibitors , Checkpoint Kinase 1/metabolism , Colonic Neoplasms/metabolism , Platinum Compounds/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cell Survival/drug effects , Cellular Senescence/drug effects , Checkpoint Kinase 1/genetics , Cisplatin/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/drug effects , Gene Knockout Techniques , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Eur J Nutr ; 56(4): 1493-1508, 2017 Jun.
Article in English | MEDLINE | ID: mdl-26983609

ABSTRACT

PURPOSE: Although beneficial effects of the dietary n-3 docosahexaenoic acid (DHA) or butyrate in colon carcinogenesis have been implicated, the mechanisms of their action are not fully clear. Here, we investigated modulations of composition of individual phospholipid (PL) classes, with a particular emphasis on cardiolipins (CLs), in colon cells treated with DHA, sodium butyrate (NaBt), or their combination (DHA/NaBt), and we evaluated possible associations between lipid changes and cell fate after fatty acid treatment. METHODS: In two distinct human colon cell models, foetal colon (FHC) and adenocarcinoma (HCT-116) cells, we compared patterns and composition of individual PL classes following the fatty acid treatment by HPLC-MS/MS. In parallel, we measured the parameters reflecting cell proliferation, differentiation and death. RESULTS: In FHC cells, NaBt induced primarily differentiation, while co-treatment with DHA shifted their response towards cell death. In contrast, NaBt induced apoptosis in HCT-116 cells, which was not further affected by DHA. DHA was incorporated in all main PL types, increasing their unsaturation, while NaBt did not additionally modulate these effects in either cell model. Nevertheless, we identified an unusually wide range of CL species to be highly increased by NaBt and particularly by DHA/NaBt, and these effects were more pronounced in HCT-116 cells. DHA and DHA/NaBt enhanced levels of high molecular weight and more unsaturated CL species, containing DHA, which was specific for either differentiation or apoptotic responses. CONCLUSIONS: We identified a wide range of CL species in the colon cells which composition was significantly modified after DHA and NaBt treatment. These specific CL modulations might contribute to distinct cellular differentiation or apoptotic responses.


Subject(s)
Cell Differentiation/drug effects , Colon/drug effects , Docosahexaenoic Acids/pharmacology , Phospholipids/chemistry , Apoptosis/drug effects , Butyric Acid/pharmacology , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colon/cytology , HCT116 Cells , Humans , Tandem Mass Spectrometry
14.
Arch Toxicol ; 91(5): 2135-2150, 2017 May.
Article in English | MEDLINE | ID: mdl-27830268

ABSTRACT

Butyrate, a short-chain fatty acid produced by fermentation of dietary fiber, is an important regulator of colonic epithelium homeostasis. In this study, we investigated the impact of this histone deacetylase (HDAC) inhibitor on expression/activity of cytochrome P450 family 1 (CYP1) and on metabolism of carcinogenic polycyclic aromatic hydrocarbon, benzo[a]pyrene (BaP), in colon epithelial cells. Sodium butyrate (NaBt) strongly potentiated the BaP-induced expression of CYP1A1 in human colon carcinoma HCT116 cells. It also co-stimulated the 7-ethoxyresorufin-O-deethylase (EROD) activity induced by the 2,3,7,8-tetrachlorodibenzo-p-dioxin, a prototypical ligand of the aryl hydrocarbon receptor. Up-regulation of CYP1A1 expression/activity corresponded with an enhanced metabolism of BaP and formation of covalent DNA adducts. NaBt significantly potentiated CYP1A1 induction and/or metabolic activation of BaP also in other human colon cell models, colon adenoma AA/C1 cells, colon carcinoma HT-29 cells, or in NCM460D cell line derived from normal colon mucosa. Our results suggest that the effects of NaBt were due to its impact on histone acetylation, because additional HDAC inhibitors (trichostatin A and suberanilohydroxamic acid) likewise increased both the induction of EROD activity and formation of covalent DNA adducts. NaBt-induced acetylation of histone H3 (at Lys14) and histone H4 (at Lys16), two histone modifications modulated during activation of CYP1A1 transcription, and it reduced binding of HDAC1 to the enhancer region of CYP1A1 gene. This in vitro study suggests that butyrate, through modulation of histone acetylation, may potentiate induction of CYP1A1 expression, which might in turn alter the metabolism of BaP within colon epithelial cells.


Subject(s)
Benzo(a)pyrene/pharmacokinetics , Butyric Acid/pharmacology , Colon/drug effects , Cytochrome P-450 CYP1A1/metabolism , Benzo(a)pyrene/metabolism , Colon/metabolism , Cytochrome P-450 CYP1A1/genetics , DNA Adducts/drug effects , DNA Adducts/metabolism , Enhancer Elements, Genetic/drug effects , HCT116 Cells , HT29 Cells , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Inactivation, Metabolic , beta Catenin/metabolism
15.
J Nutr Biochem ; 39: 145-155, 2017 01.
Article in English | MEDLINE | ID: mdl-27840291

ABSTRACT

The short-chain and n-3 polyunsaturated fatty acids exhibit anticancer properties, and they may mutually interact within the colon. However, the molecular mechanisms of their action in colon cancer cells are still not fully understood. Our study focused on the mechanisms responsible for the diverse effects of sodium butyrate (NaBt), in particular when interacting with docosahexaenoic acid (DHA), in distinct colon cancer cell types, in which NaBt either induces cell differentiation or activates programmed cell death involving mitochondrial pathway. NaBt activated autophagy both in HT-29 cells, which are sensitive to induction of differentiation, and in nondifferentiating HCT-116 cells. However, autophagy supported cell survival only in HT-29 cells. Combination of NaBt with DHA-promoted cell death, especially in HCT-116 cells and after longer time intervals. The inhibition of autophagy both attenuated differentiation and enhanced apoptosis in HT-29 cells treated with NaBt and DHA, but it had no effect in HCT-116 cells. NaBt, especially in combination with DHA, activated PPARγ in both cell types. PPARγ silencing decreased differentiation and increased apoptosis only in HT-29 cells, therefore we verified the role of caspases in apoptosis, differentiation and also PPARγ activity using a pan-caspase inhibitor. In summary, our data suggest that diverse responses of colon cancer cells to fatty acids may rely on their sensitivity to differentiation, which may in turn depend on distinct engagement of autophagy, caspases and PPARγ. These results contribute to understanding of mechanisms underlying differential effects of NaBt, when interacting with other dietary fatty acids, in colon cancer cells.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Butyrates/pharmacology , Colonic Neoplasms/pathology , Docosahexaenoic Acids/pharmacology , PPAR gamma/metabolism , Antineoplastic Agents/pharmacology , Butyric Acid/pharmacology , Caspase 3/genetics , Caspase 3/metabolism , Cell Differentiation/drug effects , HCT116 Cells , HT29 Cells , Humans , Mitochondria/drug effects , Mitochondria/metabolism , PPAR gamma/genetics
16.
Br J Haematol ; 175(5): 851-859, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27651098

ABSTRACT

The canonical Wnt pathway, dependent on ß-catenin-controlled transcription, is the most explored Wnt pathway, known to drive the malignant transformation of multiple cell types. Several reports have suggested that this pathway also participates in chronic lymphocytic leukaemia (CLL) pathogenesis. To get a better insight into the role of the Wnt/ß-catenin pathway in CLL we analysed in detail the expression of the most overexpressed Wnt ligand, encoded by the WNT3 gene, in a well-defined cohort of 137 CLL patients. Our analysis demonstrated that (i) untreated patients with more aggressive disease (with a notable exception of patients with 11q deletion) express less WNT3, (ii) WNT3 declines with disease progression in a significant proportion of patients and (iii) low WNT3 was identified as a strong independent marker indicating shorter treatment-free survival in CLL patients with IGHV mutation. Interestingly, CLL-related lymphoid cell lines, but not stromal cells, failed to respond to the ligand-induced activation of the Wnt/ß-catenin pathway. This opens the possibility that CLL cells use Wnt-3 to communicate with the cells in the microenvironment. We thus propose that the Wnt/ß-catenin pathway plays a more complex role in CLL pathogenesis than previously anticipated.


Subject(s)
Gene Expression Regulation, Neoplastic , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Wnt3 Protein/genetics , Cell Communication , Cell Line , Cohort Studies , Disease Progression , Female , Genes, Immunoglobulin Heavy Chain/genetics , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Male , Mutation , Prognosis , Wnt Signaling Pathway
17.
Hum Mol Genet ; 25(1): 9-23, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26494904

ABSTRACT

Activating mutations in the fibroblast growth factor receptor 3 (FGFR3) cause the most common genetic form of human dwarfism, achondroplasia (ACH). Small chemical inhibitors of FGFR tyrosine kinase activity are considered to be viable option for treating ACH, but little experimental evidence supports this claim. We evaluated five FGFR tyrosine kinase inhibitors (TKIs) (SU5402, PD173074, AZD1480, AZD4547 and BGJ398) for their activity against FGFR signaling in chondrocytes. All five TKIs strongly inhibited FGFR activation in cultured chondrocytes and limb rudiment cultures, completely relieving FGFR-mediated inhibition of chondrocyte proliferation and maturation. In contrast, TKI treatment of newborn mice did not improve skeletal growth and had lethal toxic effects on the liver, lungs and kidneys. In cell-free kinase assays as well as in vitro and in vivo cell assays, none of the tested TKIs demonstrated selectivity for FGFR3 over three other FGFR tyrosine kinases. In addition, the TKIs exhibited significant off-target activity when screened against a panel of 14 unrelated tyrosine kinases. This was most extensive in SU5402 and AZD1480, which inhibited DDR2, IGF1R, FLT3, TRKA, FLT4, ABL and JAK3 with efficiencies similar to or greater than those for FGFR. Low target specificity and toxicity of FGFR TKIs thus compromise their use for treatment of ACH. Conceptually, different avenues of therapeutic FGFR3 targeting should be investigated.


Subject(s)
Achondroplasia/drug therapy , Pyrroles/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Animals , Benzamides/pharmacology , Cartilage/drug effects , Cartilage/metabolism , Catalysis/drug effects , Cells, Cultured , Chick Embryo , Chondrocytes/metabolism , Humans , Mice , Phenylurea Compounds/pharmacology , Piperazines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Syndrome
18.
Clin Cancer Res ; 22(2): 459-69, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26240275

ABSTRACT

PURPOSE: ROR1, a receptor in the noncanonical Wnt/planar cell polarity (PCP) pathway, is upregulated in malignant B cells of chronic lymphocytic leukemia (CLL) patients. It has been shown that the Wnt/PCP pathway drives pathogenesis of CLL, but which factors activate the ROR1 and PCP pathway in CLL cells remains unclear. EXPERIMENTAL DESIGN: B lymphocytes from the peripheral blood of CLL patients were negatively separated using RosetteSep (StemCell) and gradient density centrifugation. Relative expression of WNT5A, WNT5B, and ROR1 was assessed by quantitative real-time PCR. Protein levels, protein interaction, and downstream signaling were analyzed by immunoprecipitation and Western blotting. Migration capacity of primary CLL cells was analyzed by the Transwell migration assay. RESULTS: By analyzing the expression in 137 previously untreated CLL patients, we demonstrate that WNT5A and WNT5B genes show dramatically (five orders of magnitude) varying expression in CLL cells. High WNT5A and WNT5B expression strongly associates with unmutated IGHV and shortened time to first treatment. In addition, WNT5A levels associate, independent of IGHV status, with the clinically worst CLL subgroups characterized by dysfunctional p53 and mutated SF3B1. We provide functional evidence that WNT5A-positive primary CLL cells have increased motility and attenuated chemotaxis toward CXCL12 and CCL19 that can be overcome by inhibitors of Wnt/PCP signaling. CONCLUSIONS: These observations identify Wnt-5a as the crucial regulator of ROR1 activity in CLL and suggest that the autocrine Wnt-5a signaling pathway allows CLL cells to overcome natural microenvironmental regulation.


Subject(s)
Autocrine Communication/physiology , Chemotaxis/physiology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Proto-Oncogene Proteins/metabolism , Wnt Proteins/metabolism , Wnt Signaling Pathway/physiology , B-Lymphocytes/metabolism , Cell Movement/physiology , Gene Expression Regulation, Neoplastic/physiology , HEK293 Cells , Humans , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Up-Regulation/physiology , Wnt-5a Protein
19.
PLoS One ; 10(10): e0141020, 2015.
Article in English | MEDLINE | ID: mdl-26492315

ABSTRACT

We demonstrated for the first time an outstanding ability of rosiglitazone to mediate a profound enhancement of LA-12-induced apoptosis associated with activation of mitochondrial pathway in human colon cancer cells. This effect was preferentially observed in the G1 cell cycle phase, independent on p53 and PPARγ proteins, and accompanied with significant changes of selected Bcl-2 family protein levels. Further stimulation of cooperative synergic cytotoxic action of rosiglitazone and LA-12 was demonstrated in the cells deficient for PTEN, where mitochondrial apoptotic pathway was more stimulated and G1-phase-associated dying was reinforced. Our results suggest that combined treatment with rosiglitazone and LA-12 might be promising anticancer strategy in colon-derived tumours regardless of their p53 status, and also favourable in those defective in PTEN function.


Subject(s)
Amantadine/analogs & derivatives , Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Organoplatinum Compounds/pharmacology , PTEN Phosphohydrolase/genetics , Thiazolidinediones/pharmacology , Amantadine/pharmacology , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Energy Metabolism/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , HCT116 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , PPAR gamma/genetics , RNA Interference , RNA, Small Interfering , Rosiglitazone , Tumor Suppressor Protein p53/genetics
20.
Oncotarget ; 6(34): 36156-71, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26416355

ABSTRACT

Plasticity of cancer cells, manifested by transitions between epithelial and mesenchymal phenotypes, represents a challenging issue in the treatment of neoplasias. Both epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) are implicated in the processes of metastasis formation and acquisition of stem cell-like properties. Mouse double minute (MDM) 2 and MDMX are important players in cancer progression, as they act as regulators of p53, but their function in EMT and metastasis may be contradictory. Here, we show that the EMT phenotype in multiple cellular models and in clinical prostate and breast cancer samples is associated with a decrease in MDM2 and increase in MDMX expression. Modulation of EMT-accompanying changes in MDM2 expression in benign and transformed prostate epithelial cells influences their migration capacity and sensitivity to docetaxel. Analysis of putative mechanisms of MDM2 expression control demonstrates that in the context of defective p53 function, MDM2 expression is regulated by EMT-inducing transcription factors Slug and Twist. These results provide an alternative context-specific role of MDM2 in EMT, cell migration, metastasis, and therapy resistance.


Subject(s)
Breast Neoplasms/metabolism , Epithelial-Mesenchymal Transition/physiology , Nuclear Proteins/biosynthesis , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-mdm2/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Proteins , Cell Line, Tumor , Female , Heterografts , Humans , Male , Mice , Mice, Nude , Phenotype , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...