1
|
Castangia I, Fulgheri F, Leyva-Jimenez FJ, Alañón ME, Cádiz-Gurrea MDLL, Marongiu F, Meloni MC, Aroffu M, Perra M, Allaw M, Abi Rached R, Oliver-Simancas R, Escribano Ferrer E, Asunis F, Manca ML, Manconi M. From Grape By-Products to Enriched Yogurt Containing Pomace Extract Loaded in Nanotechnological Nutriosomes Tailored for Promoting Gastro-Intestinal Wellness. Antioxidants (Basel) 2023; 12:1285. [PMID: 37372015 DOI: 10.3390/antiox12061285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Grape pomace is the main by-product generated during the winemaking process; since it is still rich in bioactive molecules, especially phenolic compounds with high antioxidant power, its transformation in beneficial and health-promoting foods is an innovative challenge to extend the grape life cycle. Hence, in this work, the phytochemicals still contained in the grape pomace were recovered by an enhanced ultrasound assisted extraction. The extract was incorporated in liposomes prepared with soy lecithin and in nutriosomes obtained combining soy lecithin and Nutriose FM06®, which were further enriched with gelatin (gelatin-liposomes and gelatin-nutriosomes) to increase the samples' stability in modulated pH values, as they were designed for yogurt fortification. The vesicles were sized ~100 nm, homogeneously dispersed (polydispersity index < 0.2) and maintained their characteristics when dispersed in fluids at different pH values (6.75, 1.20 and 7.00), simulating salivary, gastric and intestinal environments. The extract loaded vesicles were biocompatible and effectively protected Caco-2 cells against oxidative stress caused by hydrogen peroxide, to a better extent than the free extract in dispersion. The structural integrity of gelatin-nutriosomes, after dilution with milk whey was confirmed, and the addition of vesicles to the yogurt did not modify its appearance. The results pointed out the promising suitability of vesicles loading the phytocomplex obtained from the grape by-product to enrich the yogurt, offering a new and easy strategy for healthy and nutritional food development.
Collapse
Affiliation(s)
- Ines Castangia
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, Pad. A, S.P. Monserrato-Sestu Km 0.700, 09042 Monserrato, CA, Italy
| | - Federica Fulgheri
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, Pad. A, S.P. Monserrato-Sestu Km 0.700, 09042 Monserrato, CA, Italy
| | - Francisco Javier Leyva-Jimenez
- Regional Institute for Applied Scientific Research (IRICA), University of Castilla-La Mancha, Avda. Camilo José Cela 10, 13071 Ciudad Real, Spain
- Department of Analytical Chemistry and Food Science and Technology, University of Castilla-La Mancha, Ronda de Calatrava 7, 13071 Ciudad Real, Spain
| | - Maria Elena Alañón
- Regional Institute for Applied Scientific Research (IRICA), University of Castilla-La Mancha, Avda. Camilo José Cela 10, 13071 Ciudad Real, Spain
- Department of Analytical Chemistry and Food Science and Technology, University of Castilla-La Mancha, Ronda de Calatrava 7, 13071 Ciudad Real, Spain
| | | | - Francesca Marongiu
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, Pad. A, S.P. Monserrato-Sestu Km 0.700, 09042 Monserrato, CA, Italy
| | - Maria Cristina Meloni
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, Pad. A, S.P. Monserrato-Sestu Km 0.700, 09042 Monserrato, CA, Italy
| | - Matteo Aroffu
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, Pad. A, S.P. Monserrato-Sestu Km 0.700, 09042 Monserrato, CA, Italy
| | - Matteo Perra
- Biomedical and Tissue Engineering Laboratory, Fundación de Investigación Hospital General Universitario, 46022 Valencia, Spain
| | - Mohamad Allaw
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, Pad. A, S.P. Monserrato-Sestu Km 0.700, 09042 Monserrato, CA, Italy
| | - Rita Abi Rached
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, Pad. A, S.P. Monserrato-Sestu Km 0.700, 09042 Monserrato, CA, Italy
| | - Rodrigo Oliver-Simancas
- Regional Institute for Applied Scientific Research (IRICA), University of Castilla-La Mancha, Avda. Camilo José Cela 10, 13071 Ciudad Real, Spain
- Department of Analytical Chemistry and Food Science and Technology, University of Castilla-La Mancha, Ronda de Calatrava 7, 13071 Ciudad Real, Spain
| | - Elvira Escribano Ferrer
- Biopharmaceutics and Pharmacokinetics Unit, Institute for Nanoscience and Nanotechnology, University of Barcelona, 08028 Barcelona, Spain
| | - Fabiano Asunis
- Department of Civil, Environmental Engineering and Architecture (DICAAR), University of Cagliari, Piazza D'Armi 1, 09123 Cagliari, Italy
| | - Maria Letizia Manca
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, Pad. A, S.P. Monserrato-Sestu Km 0.700, 09042 Monserrato, CA, Italy
| | - Maria Manconi
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, Pad. A, S.P. Monserrato-Sestu Km 0.700, 09042 Monserrato, CA, Italy
| |
Collapse
|
2
|
A Planar Culture Model of Human Absorptive Enterocytes Reveals Metformin Increases Fatty Acid Oxidation and Export. Cell Mol Gastroenterol Hepatol 2022; 14:409-434. [PMID: 35489715 PMCID: PMC9305019 DOI: 10.1016/j.jcmgh.2022.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/06/2022] [Accepted: 04/18/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Fatty acid oxidation by absorptive enterocytes has been linked to the pathophysiology of type 2 diabetes, obesity, and dyslipidemia. Caco-2 and organoids have been used to study dietary lipid-handling processes including fatty acid oxidation, but are limited in physiological relevance or preclude simultaneous apical and basal access. Here, we developed a high-throughput planar human absorptive enterocyte monolayer system for investigating lipid handling, and then evaluated the role of fatty acid oxidation in fatty acid export, using etomoxir, C75, and the antidiabetic drug metformin. METHODS Single-cell RNA-sequencing, transcriptomics, and lineage trajectory was performed on primary human jejunum. In vivo absorptive enterocyte maturational states informed conditions used to differentiate human intestinal stem cells (ISCs) that mimic in vivo absorptive enterocyte maturation. The system was scaled for high-throughput drug screening. Fatty acid oxidation was modulated pharmacologically and BODIPY (Thermo Fisher Scientific, Waltham, MA) (B)-labeled fatty acids were used to evaluate fatty acid handling via fluorescence and thin-layer chromatography. RESULTS Single-cell RNA-sequencing shows increasing expression of lipid-handling genes as absorptive enterocytes mature. Culture conditions promote ISC differentiation into confluent absorptive enterocyte monolayers. Fatty acid-handling gene expression mimics in vivo maturational states. The fatty acid oxidation inhibitor etomoxir decreased apical-to-basolateral export of medium-chain B-C12 and long-chain B-C16 fatty acids, whereas the CPT1 agonist C75 and the antidiabetic drug metformin increased apical-to-basolateral export. Short-chain B-C5 was unaffected by fatty acid oxidation inhibition and diffused through absorptive enterocytes. CONCLUSIONS Primary human ISCs in culture undergo programmed maturation. Absorptive enterocyte monolayers show in vivo maturational states and lipid-handling gene expression profiles. Absorptive enterocytes create strong epithelial barriers in 96-Transwell format. Fatty acid export is proportional to fatty acid oxidation. Metformin enhances fatty acid oxidation and increases basolateral fatty acid export, supporting an intestine-specific role.
Collapse
|
3
|
Enz N, Vliegen G, De Meester I, Jungraithmayr W. CD26/DPP4 - a potential biomarker and target for cancer therapy. Pharmacol Ther 2019; 198:135-159. [PMID: 30822465 DOI: 10.1016/j.pharmthera.2019.02.015] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CD26/dipeptidyl peptidase (DPP)4 is a membrane-bound protein found in many cell types of the body, and a soluble form is present in body fluids. There is longstanding evidence that various primary tumors and also metastases express CD26/DPP4 to a variable extent. By cleaving dipeptides from peptides with a proline or alanine in the penultimate position at the N-terminus, it regulates the activity of incretin hormones, chemokines and many other peptides. Due to these effects and interactions with other molecules, a tumor promoting or suppressing role can be attributed to CD26/DPP4. In this review, we discuss the existing evidence on the expression of soluble or membrane-bound CD26/DPP4 in malignant diseases, along with the most recent findings on CD26/DPP4 as a therapeutic target in specific malignancies. The expression and possible involvement of the related DPP8 and DPP9 in cancer are also reviewed. A higher expression of CD26/DPP4 is found in a wide variety of tumor entities, however more research on CD26/DPP4 in the tumor microenvironment is needed to fully explore its use as a tumor biomarker. Circulating soluble CD26/DPP4 has also been studied as a cancer biomarker, however, the observed decrease in most cancer patients does not seem to be cancer specific. Encouraging results from experimental work and a recently reported first phase clinical trial targeting CD26/DPP4 in mesothelioma, renal and urological tumors pave the way for follow-up clinical studies, also in other tumor entities, possibly leading to the development of more effective complementary therapies against cancer.
Collapse
Affiliation(s)
- Njanja Enz
- Department of Thoracic Surgery, University Hospital Rostock, Schillingallee 35, 18057 Rostock, Germany
| | - Gwendolyn Vliegen
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Ingrid De Meester
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium.
| | - Wolfgang Jungraithmayr
- Department of Thoracic Surgery, University Hospital Rostock, Schillingallee 35, 18057 Rostock, Germany.
| |
Collapse
|
4
|
Zapletal O, Procházková J, Dubec V, Hofmanová J, Kozubík A, Vondráček J. Butyrate interacts with benzo[a]pyrene to alter expression and activities of xenobiotic metabolizing enzymes involved in metabolism of carcinogens within colon epithelial cell models. Toxicology 2018; 412:1-11. [PMID: 30439556 DOI: 10.1016/j.tox.2018.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 10/30/2018] [Accepted: 11/08/2018] [Indexed: 01/24/2023]
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.
Collapse
Affiliation(s)
- Ondřej Zapletal
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 61265 Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, 61137 Brno, Czech Republic
| | - Jiřina Procházková
- Department of Chemistry and Toxicology, Veterinary Research Institute, 62100 Brno, Czech Republic
| | - Vít Dubec
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 61265 Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, 61137 Brno, Czech Republic
| | - Jiřina Hofmanová
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 61265 Brno, Czech Republic
| | - Alois Kozubík
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 61265 Brno, Czech Republic
| | - Jan Vondráček
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, 61265 Brno, Czech Republic.
| |
Collapse
|
5
|
Butyrate alters expression of cytochrome P450 1A1 and metabolism of benzo[a]pyrene via its histone deacetylase activity in colon epithelial cell models. Arch Toxicol 2016; 91:2135-2150. [PMID: 27830268 DOI: 10.1007/s00204-016-1887-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 11/02/2016] [Indexed: 01/24/2023]
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.
Collapse
|
6
|
Souazé F, Bou-Hanna C, Kandel C, Leclair F, Devallière J, Charreau B, Bézieau S, Mosnier JF, Laboisse CL. Differential roles of Hath1, MUC2 and P27Kip1 in relation with gamma-secretase inhibition in human colonic carcinomas: a translational study. PLoS One 2013; 8:e55904. [PMID: 23409082 PMCID: PMC3569436 DOI: 10.1371/journal.pone.0055904] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 01/04/2013] [Indexed: 11/18/2022] Open
Abstract
Hath1, a bHLH transcription factor negatively regulated by the γ-secretase-dependent Notch pathway, is required for intestinal secretory cell differentiation. Our aim was fourfold: 1) determine whether Hath1 is able to alter the phenotype of colon cancer cells that are committed to a differentiated phenotype, 2) determine whether the Hath1-dependent alteration of differentiation is coupled to a restriction of anchorage-dependent growth, 3) decipher the respective roles of three putative tumor suppressor genes Hath1, MUC2 and P27kip1 in this coupling and, 4) examine how our findings translate to primary tumors. Human colon carcinoma cell lines that differentiate along a mucin secreting (MUC2/MUC5AC) and/or enterocytic (DPPIV) lineages were maintained on inserts with or without a γ-secretase inhibitor (DBZ). Then the cells were detached and their ability to survive/proliferate in the absence of substratum was assessed. γ-secretase inhibition led to a Hath1-mediated preferential induction of MUC2 over MUC5AC, without DPPIV modification, in association with a decrease in anchorage-independent growth. While P27kip1 silencing relieved the cells from the Hath1-induced decrease of anchorage-independent growth, MUC2 silencing did not modify this parameter. Hath1 ectopic expression in the Hath1 negative enterocytic Caco2 cells led to a decreased anchorage-independent growth in a P27kip1-independent manner. In cultured primary human colon carcinomas, Hath1 was up-regulated in 7 out of 10 tumors upon DBZ treatment. Parallel MUC2 up-regulation occurred in 4 (4/7) and P27kip1 in only 2 (2/7) tumors. Interestingly, the response patterns of primary tumors to DBZ fitted with the hierarchical model of divergent signalling derived from our findings on cell lines.
Collapse
|
7
|
Leoni BD, Natoli M, Nardella M, Bucci B, Zucco F, D'Agnano I, Felsani A. Differentiation of Caco-2 cells requires both transcriptional and post-translational down-regulation of Myc. Differentiation 2012; 83:116-27. [DOI: 10.1016/j.diff.2011.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2011] [Revised: 10/18/2011] [Accepted: 10/20/2011] [Indexed: 01/25/2023]
|
8
|
Abe M, Havre PA, Urasaki Y, Ohnuma K, Morimoto C, Dang LH, Dang NH. Mechanisms of confluence-dependent expression of CD26 in colon cancer cell lines. BMC Cancer 2011; 11:51. [PMID: 21284881 PMCID: PMC3038146 DOI: 10.1186/1471-2407-11-51] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 02/01/2011] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND CD26 (dipeptidyl peptidase IV, DPPIV) is a 110 kDa surface glycoprotein expressed in most normal tissues, and is a potential novel therapeutic target for selected cancers. Our work evaluates the mechanism involved in confluence-dependent CD26 expression in colon cancer. METHODS Colon adenocarcinoma cells were grown to confluence, and expression of CD26 and transcription factors implicated in its regulation was confirmed by immunofluorescence and Western blotting. Real-time PCR was also performed to evaluate CD26 upregulation at the transcriptional level. The influence of c-Myc on CD26 expression during different growth conditions was further evaluated following transient transfection of a c-Myc-expressing plasmid and a c-Myc specific siRNA. RESULTS We found that the colon cancer cell lines HCT-116 and HCT-15 exhibited a confluence-dependent increase in CD26 mRNA and protein, associated with decreased expression of c-Myc, increased USF-1 and Cdx 2 levels, and unchanged HNF-1α expression. Meanwhile, ectopic expression of c-Myc in both cell lines led to decreased CD26 expression. In contrast, transfection of a siRNA targeted to Cdx2 resulted in decreased CD26 level. Importantly, culturing of cells in serum-depleted media, but not acidic conditions, upregulated CD26. While HIF-1α level also increased when cells were cultured in serum-depleted media, its expression was required but not sufficient for CD26 upregulation. CONCLUSIONS CD26 mRNA and protein levels increase in a confluence-dependent manner in colon carcinoma cell lines, with c-Myc acting as a repressor and Cdx2 acting as an enhancer of CD26 expression. The enhanced expression of CD26 in serum-depleted media and a requirement for HIF-1α suggest a role for nutrients or growth factors in the regulation of CD26 protein expression.
Collapse
Affiliation(s)
- Masako Abe
- Department of Hematologic Malignancies, Nevada Cancer Institute, Las Vegas, Nevada, 89135, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Badouel C, Chartrain I, Blot J, Tassan JP. Maternal embryonic leucine zipper kinase is stabilized in mitosis by phosphorylation and is partially degraded upon mitotic exit. Exp Cell Res 2010; 316:2166-73. [PMID: 20420823 DOI: 10.1016/j.yexcr.2010.04.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2010] [Revised: 03/22/2010] [Accepted: 04/19/2010] [Indexed: 12/18/2022]
Abstract
MELK (maternal embryonic leucine zipper kinase) is a cell cycle dependent protein kinase involved in diverse cell processes including cell proliferation, apoptosis, cell cycle and mRNA processing. Noticeably, MELK expression is increased in cancerous tissues, upon cell transformation and in mitotically-blocked cells. The question of how MELK protein level is controlled is therefore important. Here, we show that MELK protein is restricted to proliferating cells derived from either cancer or normal tissues and that MELK protein level is severely decreased concomitantly with other cell cycle proteins in cells which exit the cell cycle. Moreover, we demonstrate in human HeLa cells and Xenopus embryos that approximately half of MELK protein is degraded upon mitotic exit whereas another half remains stable during interphase. We show that the stability of MELK protein in M-phase is dependent on its phosphorylation state.
Collapse
Affiliation(s)
- Caroline Badouel
- CNRS UMR 6061 Génétique et Développement, Université de Rennes 1, IFR140 GFAS, Faculté de médecine, 2 avenue du Professeur Léon Bernard, CS 34317, 35043 Rennes Cedex, France
| | | | | | | |
Collapse
|
10
|
Mayo C, Mayol X. Cycling D1 negatively regulates the expression of differentiation genes in HT-29 M6 mucus-secreting colon cancer cells. Cancer Lett 2009; 281:183-7. [DOI: 10.1016/j.canlet.2009.02.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2009] [Revised: 02/16/2009] [Accepted: 02/17/2009] [Indexed: 02/02/2023]
|
11
|
Borlak J, Zwadlo C. Expression of drug-metabolizing enzymes, nuclear transcription factors and ABC transporters in Caco-2 cells. Xenobiotica 2008; 33:927-43. [PMID: 14514442 DOI: 10.1080/00498250310001614286] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
1. Caco-2 cells are frequently used in intestinal drug absorption and metabolism studies, but little is known about the effects of drugs on the simultaneous expression of genes coding for drug-metabolizing enzymes (DMEs), nuclear transcription factors and ABC transporters. 2. The gene expression and enzyme activities of control and Aroclor 1254-treated cultures were therefore explored, the latter being a powerful inducer of DMEs. Fourteen- and 80-fold induction of CYP1A1 and CYP1A2 mRNA were shown, whereas expression of other DMEs was either increased (CYP2C8-2C19, 10-fold; CYP3A5, twofold; FMO1, 2 and 5, twofold; epoxide hydrolase, threefold) or repressed (CYP2D6 and CYP2E1 to 75% of control values). 3. Notably, gene copies of CYP3A4 and CYP2B6/7 were below the limit of detection, but a three- and 10-fold induction of HNF 1alpha + beta, HNF-4alpha4 and a similar 10-fold increase in STAT 3 and 4 was observed. 4. Similarly, c/EBP transcripts were only detected in treated cell cultures, but MRP1, its isoforms 3-5 as well as MDR-1 were increased threefold after dosing with Aroclor 1254. 5. Overall, CYP gene expression correlated well with the cognate enzyme activity using testosterone as a marker substrate.
Collapse
Affiliation(s)
- J Borlak
- Fraunhofer Institute of Toxicology and Experimental Medicine, Center for Drug Research and Medical Biotechnology, Nicolai-Fuchs-Str. 1 D-30659, Hannover, Germany.
| | | |
Collapse
|
12
|
Gross I, Duluc I, Benameur T, Calon A, Martin E, Brabletz T, Kedinger M, Domon-Dell C, Freund JN. The intestine-specific homeobox gene Cdx2 decreases mobility and antagonizes dissemination of colon cancer cells. Oncogene 2007; 27:107-15. [PMID: 17599044 DOI: 10.1038/sj.onc.1210601] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The gravity of colorectal cancer is mainly due to the capacity of tumor cells to migrate out of the tumor mass to invade the stroma and disseminate as metastases. The acquisition of a migratory phenotype also occurs during wound healing. Here, we show that several features characterizing invasive colon tumor cells are shared by migrating cells during wound repair in vitro. In particular, the expression of the intestine-specific transcription factor Cdx2, a key gene for intestinal identity downregulated in invasive cancer cells, is reduced during wound healing in vitro. Transcription factors involved in epithelial-mesenchymal transition such as Snail and Slug are upregulated during wound healing and are able to repress Cdx2 transcription. In vitro, forced expression of Cdx2 in human colon cancer cell lines retarded wound repair and reduced migration, whereas inhibition of Cdx2 expression by RNA interference enhanced migration. In vivo, forced expression of Cdx2 opposed tumor cells spreading in nude mice xenografted at three different sites. These data provide evidence that Cdx2 antagonizes the process of tumor cell dissemination, and they suggest that this homeobox gene might represent a new therapeutic target against metastatic spreading of colon cancer.
Collapse
Affiliation(s)
- I Gross
- INSERM, U682, Strasbourg, France
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Le Hégarat L, Jacquin AG, Bazin E, Fessard V. Genotoxicity of the marine toxin okadaic acid, in human Caco-2 cells and in mice gut cells. ENVIRONMENTAL TOXICOLOGY 2006; 21:55-64. [PMID: 16463260 DOI: 10.1002/tox.20154] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Micronucleus induction by the diarrhetic shellfish toxin okadaic acid (OA) was investigated in two intestinal models, cultured human Caco-2 cells and colon epithelial cells of mice treated in vivo. Exposure to OA for 4 and 24 h induced dose-responsive increases in the frequency of micronucleated Caco-2 cells; the minimum OA doses increasing micronucleus frequency were 20 nM for the 4 h treatment and 5 nM for the 24 h treatment. OA treatment of Caco-2 cells also resulted in dose- and time-dependent increases in mitotic arrest and multinucleated cells. Two experiments were conducted in which mice were treated with single oral gavages of 435-610 and 115-1341 microg/kg OA. In the first experiment, samples were taken 24 h after the treatment, and the frequencies of both micronucleated and mitotic gut cells were increased after treatment with 525 microg/kg OA. In the second experiment, no increases in micronucleus frequency were detected at 24, 36, or 48 h following OA doses of 230 and 115 microg/kg; however, an increase in the mitotic index was observed 36 h after a gavage with 115 microg/kg OA. In this experiment, doses higher than 230 microg/kg were rapidly lethal to the mice. Immunohistology with monoclonal OA antibodies showed that OA was distributed into the liver at all the sampling times and in the small intestine at 24 and 36 h; OA was not detected in the colon. In addition, the TUNEL assay indicated that OA induced apoptosis in mouse ileum, liver, and kidney. The results of our investigations suggest that OA is aneugenic in Caco-2 cells, whereas the in vivo data were inconclusive. Further studies should be performed in mice using intragastric doses of 230-525 microg/kg OA. Moreover, the apoptosis and cell proliferation results indicate that OA can reach organs other than colon, indicating further evaluation of the genotoxic potential of OA in these organs is warranted.
Collapse
Affiliation(s)
- Ludovic Le Hégarat
- AFSSA, Unité de Toxicologie Génétique des Contaminants Alimentaires, La Haute Marche, BP 90203, 35133 Fougères, France
| | | | | | | |
Collapse
|
14
|
Papp B, Brouland JP, Gélébart P, Kovàcs T, Chomienne C. Endoplasmic reticulum calcium transport ATPase expression during differentiation of colon cancer and leukaemia cells. Biochem Biophys Res Commun 2004; 322:1223-36. [PMID: 15336970 DOI: 10.1016/j.bbrc.2004.08.030] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2004] [Indexed: 11/19/2022]
Abstract
The calcium homeostasis of the endoplasmic reticulum (ER) is connected to a multitude of cell functions involved in intracellular signal transduction, control of proliferation, programmed cell death, or the synthesis of mature proteins. Calcium is accumulated in the ER by various biochemically distinct sarco/endoplasmic reticulum calcium transport ATPase isoenzymes (SERCA isoforms). Experimental data indicate that the SERCA composition of some carcinoma and leukaemia cell types undergoes significant changes during differentiation, and that this is accompanied by modifications of SERCA-dependent calcium accumulation in the ER. Because ER calcium homeostasis can also influence cell differentiation, we propose that the modulation of the expression of various SERCA isoforms, and in particular, the induction of the expression of SERCA3-type proteins, is an integral part of the differentiation program of some cancer and leukaemia cell types. The SERCA content of the ER may constitute a new parameter by which the calcium homeostatic characteristics of the organelle are adjusted. The cross-talk between ER calcium homeostasis and cell differentiation may have some implications for the better understanding of the signalling defects involved in the acquisition and maintenance of the malignant phenotype.
Collapse
Affiliation(s)
- Béla Papp
- INSERM EMI-00-03 Laboratoire de Biologie Cellulaire Hématopoïétique, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, 1, Avenue Claude Vellefaux, 75010 Paris, France.
| | | | | | | | | |
Collapse
|
15
|
Cai J, Chen Y, Murphy TJ, Jones DP, Sartorelli AC. Role of caspase activation in butyrate-induced terminal differentiation of HT29 colon carcinoma cells. Arch Biochem Biophys 2004; 424:119-27. [PMID: 15047183 DOI: 10.1016/j.abb.2004.02.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2003] [Revised: 02/03/2004] [Indexed: 10/26/2022]
Abstract
Colon epithelial cells have a defined life span and undergo terminal differentiation as they mature and migrate to the luminal surface. The differentiation process can be induced in cultured colon cancer cells by sodium butyrate, which induces expression of various differentiation markers followed subsequently by cell death. In the present study, HT29 colorectal carcinoma cells were shown to undergo butyrate-induced caspase activation that was mainly produced through a mitochondrial pathway. Inhibition of caspase activation, either by peptide pan caspase inhibitor Z-VAD-FMK, by caspase 9 inhibitor Z-LEHD-FMK, or by overexpression of Bcl-XL, also inhibited the expression of differentiation markers. These findings suggest (a) that terminal differentiation of HT29 colon carcinoma cells is tightly linked to caspase activation and (b) that increased expression of anti-apoptotic members of the Bcl-2 family of proteins, as well as other inhibitors of caspase activation, has the potential to inhibit terminal differentiation and thereby may contribute to the progression of colon cancer.
Collapse
Affiliation(s)
- Jiyang Cai
- The Department of Biochemistry, Emory University School of Medicine, 4157 Rollins Research Center, 1510 Clifton Road, Atlanta, GA 30322, USA.
| | | | | | | | | |
Collapse
|
16
|
Arnold KA, Eichelbaum M, Burk O. Alternative splicing affects the function and tissue-specific expression of the human constitutive androstane receptor. NUCLEAR RECEPTOR 2004; 2:1. [PMID: 15043764 PMCID: PMC406421 DOI: 10.1186/1478-1336-2-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2004] [Accepted: 03/25/2004] [Indexed: 12/26/2022]
Abstract
Background The constitutive androstane receptor (CAR) plays a key role in the control of drug metabolism and transport by mediating the phenobarbital-type induction of many phase I and II drug metabolizing enzymes and drug transporters. Results We identified transcripts generated by four different alternative splicing events in the human CAR gene. Two of the corresponding ligand binding domain isoforms demonstrated novel functional properties: First, CAR(SV3), which is encoded by a transcript containing an lengthened exon 7, differentially transactivated target gene promoters. Second, CAR(SV2), which results from the use of an alternative 3' splice site lengthening exon 8, showed ligand-dependent instead of constitutive interaction with coactivators. Furthermore, alternatively spliced transcripts demonstrated a tissue-specific expression pattern. In most tissues, only transcripts generated by alternative splicing within exon 9 were expressed. The encoded variant demonstrated a loss-of-function phenotype. Correct splicing of exon 8 to exon 9 is restricted to only a few tissues, among them liver and small intestine for which CAR function has been demonstrated, and is associated with the induction of CAR expression during differentiation of intestinal cells. Conclusion Due to their specific activities, CAR variant proteins SV2 and SV3 may modulate the activity of reference CAR(SV1). Furthermore, we propose that transcriptional activation and regulation of splicing of exon 9 may be coupled to ensure appropriate tissue- and differentiation state-specific expression of transcripts encoding functional CAR protein. Altogether, alternative splicing seems to be of utmost importance for the regulation of CAR expression and function.
Collapse
Affiliation(s)
- Katja A Arnold
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstrasse 112, D-70376 Stuttgart, Germany
| | - Michel Eichelbaum
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstrasse 112, D-70376 Stuttgart, Germany
| | - Oliver Burk
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Auerbachstrasse 112, D-70376 Stuttgart, Germany
| |
Collapse
|
17
|
Rösmann S, Hahn D, Lottaz D, Kruse MN, Stöcker W, Sterchi EE. Activation of human meprin-alpha in a cell culture model of colorectal cancer is triggered by the plasminogen-activating system. J Biol Chem 2002; 277:40650-8. [PMID: 12189145 DOI: 10.1074/jbc.m206203200] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The activation of latent proenzymes is an important mechanism for the regulation of localized proteolytic activity. Human meprin-alpha, an astacin-like zinc metalloprotease expressed in normal colon epithelial cells, is secreted as a zymogen into the intestinal lumen. Here, meprin is activated after propeptide cleavage by trypsin. In contrast, colorectal cancer cells secrete meprin-alpha in a non-polarized way, leading to accumulation and increased activity of meprin-alpha in the tumor stroma. We have analyzed the activation mechanism of promeprin-alpha in colorectal cancer using a co-culture model of the intestinal mucosa composed of colorectal adenocarcinoma cells (Caco-2) cultivated on filter supports and intestinal fibroblasts grown in the companion dish. We provide evidence that meprin-alpha is activated by plasmin and show that the presence of plasminogen in the basolateral compartment of the co-cultures is sufficient for promeprin-alpha activation. Analysis of the plasminogen-activating system in the co-cultures revealed that plasminogen activators produced and secreted by fibroblasts converted plasminogen to active plasmin, which in turn generated active meprin-alpha. This activation mechanism offers an explanation for the observed meprin-alpha activity in the tumor stroma, a prerequisite for a potential role of this protease in colorectal cancer.
Collapse
Affiliation(s)
- Sandra Rösmann
- Institute of Biochemistry and Molecular Biology, Department of Pediatrics, University of Berne, Bühlstrasse 28, CH-3012 Berne, Switzerland
| | | | | | | | | | | |
Collapse
|
18
|
Carnesecchi S, Langley K, Exinger F, Gosse F, Raul F. Geraniol, a component of plant essential oils, sensitizes human colonic cancer cells to 5-Fluorouracil treatment. J Pharmacol Exp Ther 2002; 301:625-30. [PMID: 11961066 DOI: 10.1124/jpet.301.2.625] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Differentiation of human colonic cancer cells at confluency has been correlated to their increased resistance to chemotherapeutic agents. The aim of this study was to determine whether blocking Caco-2 cell differentiation could sensitize the cells to 5-fluorouracil (5-FU) treatment. We show that in cells at confluency, geraniol (400 microM) prevented the formation of brush-border membranes and inhibited the expression of intestinal hydrolases (sucrase, lactase, alkaline phosphatase). The antiproliferative effect of geraniol (400 microM) together with 5-FU (5 microM) was twice that of 5-FU alone. The cytotoxicity induced by 5-FU was enhanced in the presence of geraniol, as shown by a 50% increase of lactate dehydrogenase release in the culture medium. These effects are related to enhanced intracellular accumulation of 5-FU in the presence of geraniol as shown by a 2-fold increase in intracellular 5-[6-(3)H]FU (1.5 microCi/ml). It is concluded that geraniol sensitizes colonic cancer cells to 5-FU treatment, by increasing the cytotoxicity of the drug, and that this results from the facilitated transport of 5-FU and the blockade of the morphological and functional differentiation of the cancer cells.
Collapse
Affiliation(s)
- S Carnesecchi
- Laboratory of Cancer Nutritional Prevention, Institut de Recherche contre les Cancers de l'Appareil Digestif, Strasbourg, France
| | | | | | | | | |
Collapse
|