Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
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.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Biochem Pharmacol ; 92(3): 415-24, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25285768

ABSTRACT

In search for novel strategies in colon cancer treatment, we investigated the unique ability of platinum(IV) complex LA-12 to efficiently enhance the killing effects of tumor necrosis factor-related apoptosis inducing ligand (TRAIL), and compared it with the sensitizing action of cisplatin. We provide the first evidence that LA-12 primes human colon cancer cells for TRAIL-induced cytotoxicity by p53-independent activation of the mitochondrial apoptotic pathway. The cooperative action of LA-12 and TRAIL was associated with stimulation of Bax/Bak activation, drop of mitochondrial membrane potential, caspase-9 activation, and a shift of the balance among Bcl-2 family proteins in favor of the pro-apoptotic members. In contrast to cisplatin, LA-12 was a potent inducer of ERK-mediated Noxa and BimL protein upregulation, and more effectively enhanced TRAIL-induced apoptosis in the absence of Bax. The cooperative action of LA-12 and TRAIL was augmented following the siRNA-mediated silencing of Mcl-1 in both Bax proficient/deficient cells. We newly demonstrated that LA-12 induced ERK-mediated c-Myc upregulation, and proved that c-Myc silencing inhibited the mitochondrial activation and apoptosis in colon cancer cells treated with LA-12 and TRAIL. The LA-12-mediated sensitization to TRAIL-induced apoptosis was demonstrated in several colon cancer cell lines, further underscoring the general relevance of our findings. The selective action of LA-12 was documented by preferential priming of cancer but not normal colon cancer cells to TRAIL killing effects. Our work highlights the promising potential of LA-12 over cisplatin to enhance the colon cancer cell sensitivity to TRAIL-induced apoptosis, and provides new mechanistic insights into their cooperative action.


Subject(s)
Amantadine/analogs & derivatives , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Mitochondria/drug effects , Organoplatinum Compounds/pharmacology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Amantadine/pharmacology , Apoptosis/genetics , Colonic Neoplasms/pathology , Genes, p53 , HCT116 Cells/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Cells, Cultured , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
13.
Biochim Biophys Acta ; 1841(9): 1308-17, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24953781

ABSTRACT

Docosahexaenoic acid (DHA), an n-3 polyunsaturated fatty acid present in fish oil, may exert cytotoxic and/or cytostatic effects on colon cancer cells when applied individually or in combination with some anticancer drugs. Here we demonstrate a selective ability of subtoxic doses of DHA to enhance antiproliferative and apoptotic effects of clinically useful cytokine TRAIL (tumor necrosis factor-related apoptosis inducing ligand) in cancer but not normal human colon cells. DHA-mediated stimulation of TRAIL-induced apoptosis was associated with extensive engagement of mitochondrial pathway (Bax/Bak activation, drop of mitochondrial membrane potential, cytochrome c release), activation of endoplasmic reticulum stress response (CHOP upregulation, changes in PERK level), decrease of cellular inhibitor of apoptosis protein (XIAP, cIAP1) levels and significant changes in sphingolipid metabolism (intracellular levels of ceramides, hexosyl ceramides, sphingomyelines, sphingosines; HPLC/MS/MS). Interestingly, we found significant differences in representation of various classes of ceramides (especially C16:0, C24:1) between the cancer and normal colon cells treated with DHA and TRAIL, and suggested their potential role in the regulation of the cell response to the drug combination. These study outcomes highlight the potential of DHA for a new combination therapy with TRAIL for selective elimination of colon cancer cells via simultaneous targeting of multiple steps in apoptotic pathways.


Subject(s)
Docosahexaenoic Acids/pharmacology , Gene Expression Regulation, Neoplastic , Mitochondria/drug effects , Sphingolipids/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cytochromes c/metabolism , Drug Synergism , Endoplasmic Reticulum Stress/drug effects , Humans , Inhibitor of Apoptosis Proteins , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Signal Transduction , Sphingolipids/chemistry , Sphingolipids/classification , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
14.
Mediators Inflamm ; 2014: 848632, 2014.
Article in English | MEDLINE | ID: mdl-24876678

ABSTRACT

Intestinal homeostasis is precisely regulated by a number of endogenous regulatory molecules but significantly influenced by dietary compounds. Malfunction of this system may result in chronic inflammation and cancer. Dietary essential n-3 polyunsaturated fatty acids (PUFAs) and short-chain fatty acid butyrate produced from fibre display anti-inflammatory and anticancer activities. Both compounds were shown to modulate the production and activities of TNF family cytokines. Cytokines from the TNF family (TNF- α, TRAIL, and FasL) have potent inflammatory activities and can also regulate apoptosis, which plays an important role in cancer development. The results of our own research showed enhancement of apoptosis in colon cancer cells by a combination of either docosahexaenoic acid (DHA) or butyrate with TNF family cytokines, especially by promotion of the mitochondrial apoptotic pathway and modulation of NF κ B activity. This review is focused mainly on the interaction of dietary PUFAs and butyrate with these cytokines during colon inflammation and cancer development. We summarised recent knowledge about the cellular and molecular mechanisms involved in such effects and outcomes for intestinal cell behaviour and pathologies. Finally, the possible application for the prevention and therapy of colon inflammation and cancer is also outlined.


Subject(s)
Colon/pathology , Fatty Acids, Unsaturated/metabolism , Inflammation/metabolism , Neoplasms/metabolism , Tumor Necrosis Factors/metabolism , Animals , Apoptosis , Butyrates/metabolism , Cytokines/metabolism , Diet , Docosahexaenoic Acids/metabolism , Humans , Intestinal Mucosa/metabolism , Mice , Mitochondria/pathology , NF-kappa B/metabolism
15.
Curr Pharm Biotechnol ; 14(3): 274-88, 2013.
Article in English | MEDLINE | ID: mdl-22201596

ABSTRACT

Experimental and epidemiological evidence supports the idea that dietary fat and fiber influence colon carcinogenesis. Particularly, their components, n-3 polyunsaturated fatty acids (PUFAs) and butyrate, have been proven to exhibit beneficial effects on colon epithelial cell metabolism, signaling, and kinetics, thus preventing colon inflammation and cancer. Moreover, these effects may be strengthened by PUFA and butyrate combination. It appears that administration of these compounds might be a relatively nontoxic form of supportive therapy improving cancer treatment outcomes and slowing down or preventing recurrence of certain types of cancer. However, their efficient application has to be based on solid scientific evidence of their mechanisms of action from the molecular and cellular to the organismal level. In this review, we emphasize the role of lipids and their metabolism during tumor development, describe some important mechanisms considering cellular and molecular levels of PUFA and butyrate action in colon epithelial cells, and particularly focus on the interaction of their metabolism and the signaling pathways with respect to the differences in response of normal and cancer colon cells.


Subject(s)
Butyrates/metabolism , Colonic Neoplasms/metabolism , Fatty Acids, Unsaturated/metabolism , Animals , Humans , Lipid Metabolism
16.
Article in English | MEDLINE | ID: mdl-23069883

ABSTRACT

BACKGROUND: Experimental, epidemiological and clinical data substantiate the beneficial role of n-3 polyunsaturated fatty acids (PUFAs) in preventing inflammation and cancer of the colon. This review covers the unsaturated docosahexaenoic fatty acid (DHA), describes some of its important cellular and molecular mechanisms, its interaction with another dietary lipid, butyrate and with endogenous apoptotic regulators of the tumour necrosis factor (TNF) family. We also discuss the clinical impact of this knowledge and the use of these lipids in colon cancer prevention and treatment. RESULTS: From the literature, DHA has been shown to suppress the growth, induce apoptosis in colon cancer cells in vitro and decrease the incidence and growth of experimental tumours in vivo. Based on these data and our own experimental results, we describe and discuss the possible mechanisms of DHA anticancer effects at various levels of cell organization. We show that DHA can sensitize colon cancer cells to other chemotherapeutic/chemopreventive agents and affect the action of physiological apoptotic regulators of the TNF family. CONCLUSION: Use of n-3 PUFAs could be a relatively non-toxic form of supportive therapy for improving colon cancer treatment and slowing down or preventing its recurrence. However, it is necessary to use them with caution, based on solid scientific evidence of their mechanisms of action from the molecular to the cellular and organism levels.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Colon/cytology , Docosahexaenoic Acids/pharmacology , Colon/drug effects , Colonic Neoplasms/pathology , Docosahexaenoic Acids/physiology , Humans
17.
Oncol Lett ; 3(4): 913-916, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22741017

ABSTRACT

Apoptosis inhibitor 5 (API-5) is a 55 kDa nuclear protein with potent anti-apoptotic signaling in tumor cells in vitro. In this study, we analyzed the expression of the API-5 protein in vivo in a broad spectrum of human carcinomas, including those of the colon, lung, liver, kidney, pancreas, stomach and esophagus using tumor tissues obtained during tumor resection. The results showed significant upregulation of API-5 expression in biopsies of lung (23%, n=13) and colorectal tumors (33%, n=27) in comparison with biopsies from the adjacent normal tissue. Colon cancer biopsies were used to study the cell populations with an upregulated level of expression of API-5 more closely. Using a magnetic bead-based selection for the epithelial cell marker EpCAM, we purified epithelial cells from the tumor and control tissues and analyzed these cells for API-5 expression by western immunoblotting. We observed that EpCAM-positive tumor cells expressed API-5 in all three colorectal cancer cases tested, in contrast to the control EpCAM-positive and EpCAM-negative cells isolated from the control or tumor tissues. These data suggest that the expression of the API-5 protein is upregulated in tumor epithelial cells and may serve as a prognostic marker in colorectal cancer.

18.
J Nutr Biochem ; 23(6): 539-48, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21775115

ABSTRACT

The present study highlights the important association between lipid alterations and differentiation/apoptotic responses in human colon differentiating (FHC) and nondifferentiating (HCT-116) cell lines after their treatment with short-chain fatty acid sodium butyrate (NaBt), polyunsaturated fatty acids (PUFAs), and/or their combination. Our data from GC/MS and LC/MS/MS showed an effective incorporation and metabolization of the supplemented arachidonic acid (AA) or docosahexaenoic acid (DHA), resulting in an enhanced content of the respective PUFA in individual phospholipid (PL) classes and an altered composition of the whole cellular fatty acid spectrum in both FHC and HCT-116 cells. We provide novel evidence that NaBt combined with PUFAs additionally modulated AA and DHA cellular levels and caused their shift from triacylglycerol to PL fractions. NaBt increased, while AA, DHA and their combination with NaBt decreased endogenous fatty acid synthesis in FHC but not in HCT-116 cells. Fatty acid treatment also altered membrane lipid structure, augmented cytoplasmic lipid droplet accumulation, reactive oxygen species (ROS) production and dissipation of the mitochondrial membrane potential. All these parameters were significantly enhanced by combined NaBt/PUFA treatment, but only in FHC cells was this accompanied by highly increased apoptosis and suppressed differentiation. Moreover, the most significant changes of ROS production, differentiation and apoptosis among the parameters studied, the highest effects of combined NaBt/PUFA treatment and a lower sensitivity of HCT-116 cells were confirmed using two-way ANOVA. Our results demonstrate an important role of fatty acid-induced lipid alterations in the different apoptotic/differentiation response of colon cells with various carcinogenic potential.


Subject(s)
Apoptosis/drug effects , Arachidonic Acid/pharmacology , Butyrates/pharmacology , Cell Differentiation/drug effects , Docosahexaenoic Acids/pharmacology , Epithelial Cells/drug effects , Chromatography, Liquid , Colon/cytology , Epithelial Cells/metabolism , HCT116 Cells , Humans , Phospholipids/metabolism , Reactive Oxygen Species/metabolism , Tandem Mass Spectrometry , Triglycerides/metabolism
19.
Cytokine ; 55(1): 34-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21482133

ABSTRACT

The resistance of transformed epithelial cells to a detachment-induced apoptosis (anoikis) can significantly affect their susceptibility to anticancer therapy. We showed that detachment of both fetal (FHC) and adenocarcinoma (HT-29) human colon epithelial cells resulted in the activation of the pro-survival Akt pathway, and significant changes in integrin-linked kinase (ILK) and focal adhesive kinase (FAK) phosphorylation. We demonstrated a detachment-induced and PI3K/Akt-mediated resistance to apoptotic effects of TRAIL, which was not associated with any changes in the cell surface TRAIL death receptor levels. Instead, a modulation of downstream intracellular signaling events was suggested to be involved. Our results may have important implications for optimization of new strategies in treatment of cancers at different stages of development.


Subject(s)
Anoikis/drug effects , Drug Resistance, Neoplasm/drug effects , Epithelial Cells/pathology , Fetus/cytology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Cell Adhesion/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , HT29 Cells , Humans , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Receptors, Death Domain/metabolism , Signal Transduction/drug effects
20.
Photochem Photobiol Sci ; 10(4): 626-32, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21298151

ABSTRACT

Preferential uptake of photosensitizer by tumour tissue is an elementary prerequisite of effective and successful photodynamic therapy (PDT). Therefore intracellular concentration of photosensitizer is one of the limiting factors affecting PDT efficiency. Hypericin (HY) has found applications in photodynamic diagnostics solely due to its high specificity for tumour cells and tissues. However, here we suggest that not only HY uptake, but importantly also the cell ability to manage oxidative stress induced by HY-PDT can be important decisive factors finally affecting the cell death response. We showed that despite the higher accumulation of HY in FHC human fetal colon epithelial cells compared to HT-29 colon adenocarcinoma cells, the cytotoxic effects of this photosensitizer were more pronounced in the latter cell line, and this was associated with enhanced accumulation of HY-PDT-induced reactive oxygen species (ROS).


Subject(s)
Adenocarcinoma/drug therapy , Colonic Neoplasms/drug therapy , Epithelial Cells/drug effects , Perylene/analogs & derivatives , Photochemotherapy , Photosensitizing Agents/therapeutic use , Anthracenes , Cell Line , Colon/cytology , Epithelial Cells/radiation effects , Fetus , Humans , Microscopy, Fluorescence , Perylene/therapeutic use , Perylene/toxicity , Photosensitizing Agents/toxicity , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...