1
|
Evaluation of the Anticancer and Probiotic Potential of Autochthonous (Wild) Lacticaseibacillus paracasei Strains from New Ecological Niches as a Possible Additive for Functional Dairy Foods. Foods 2023; 12:foods12010185. [PMID: 36613399 PMCID: PMC9818674 DOI: 10.3390/foods12010185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Probiotics such as Lactobacillus spp. could modulate the intestinal microbiota composition, supporting gastrointestinal tract barrier function and benefiting human health. To evaluate the anticancer and probiotic properties of potentially active autochthonous Lacticaseibacillus paracasei strains on proliferating and differentiated enterocytes, human colon adenocarcinoma cell line HT29 was used as a model. The lactic acid bacteria (LAB) were isolated from new ecological niches—mountain anthills populated by redwood ants (Formica rufa L.). Human colorectal adenocarcinoma cells (HT29, ATCC, HTB-38™) were treated for twenty-four hours with supernatants (SNs) derived from four strains of Lacticaseibacillus paracasei: P4, C8, C15 and M2.1. An MTT assay, alkaline phosphatase activity, IAP, Bax and Bcl-2 gene expression analysis (RT-qPCR) and the Bax/Bcl-2 ratio were evaluated. The MTT assay revealed that the observed effects varied among groups. However, 10% neutralized supernatants from P4, C8, C15 and M2.1 strains did not show cytotoxic effects. In contrast to non-differentiated cells, a significant (p < 0.001) rise in ALP activity in all treatments, with an average of 18%, was established in differentiated cells. The IAP expression was remarkably downregulated in the differentiated M2.1 group (p < 0.05) and upregulated in the non-differentiated P4 (p < 0.05) and M2.1 (p < 0.05) groups. The Bax/Bcl-2 quantity expression ratio in P4 was significantly (p < 0.05) upregulated in proliferating cancer cells, but in P4- and M2.1-differentiated cells these values were downregulated (p < 0.05). The obtained results indicate that the isolated L. paracasei strains possess anticancer and probiotic properties and could be used as additives for functional dairy foods and thus benefit human health.
Collapse
|
2
|
Mattinzoli D, Cacioppo M, Ikehata M, Armelloni S, Alfieri CM, Castellano G, Barilani M, Arcudi F, Messa P, Prato M. Carbon dots conjugated to SN38 for improved colorectal anticancer therapy. Mater Today Bio 2022; 16:100286. [PMID: 36186846 PMCID: PMC9523396 DOI: 10.1016/j.mtbio.2022.100286] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/30/2022] [Accepted: 05/09/2022] [Indexed: 11/18/2022] Open
Abstract
Irinotecan (CTP-11) is one of the standard therapies for colorectal cancer (CRC). CTP-11 is enzymatically converted to the hydrophobic 7-ethyl-10-hydroxycamptothecin (SN38), a one hundred-fold more active metabolite. Conjugation of hydrophobic anticancer drugs to nanomaterials is a strategy to improve their solubility, efficacy, and selectivity. Carbon dots (CDs) have garnered interest for their small sizes (<10 nm), low toxicity, high water solubility, and bright fluorescence. This paper describes the use of CDs to improve drug vehiculation, stability, and chemotherapeutic efficiency of SN38 through a direct intracellular uptake in CRC. The covalent conjugation of SN38 to CDs via a carbamate bond provides a CD-SN38 hybrid material for slow, sustained, and pH-responsive drug release. CD-SN38 successfully penetrates the CRC cells with a release in the nucleus affecting first the cell cycle and then the cytoskeleton. Moreover, CD-SN38 leads to a deregulation of the extracellular matrix (ECM), one of the major components of the cancer niche considered a possible target therapy for reducing the cancer progression. This work shows the combined therapeutic and imaging potential of CD-based hybrid materials for the treatment of CRC. Future efforts for targeted therapy of chronic diseases characterized by altered ECM deposition, such as chronic kidney disease and chronic allograft nephropathy in kidney transplant patients are envisaged.
Collapse
Affiliation(s)
- Deborah Mattinzoli
- Renal Research Laboratory, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Via Pace 9, Milan, 20122, Italy
- Corresponding author.
| | - Michele Cacioppo
- Department of Chemical and Pharmaceutical Sciences, INSTM UdR Trieste, University of Trieste, Via Licio Giorgieri 1, Trieste, 34127, Italy
- Center for Cooperative Research in Biomaterials (CIC BiomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 182, Donostia-San Sebastián, 20014, Spain
| | - Masami Ikehata
- Renal Research Laboratory, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Via Pace 9, Milan, 20122, Italy
| | - Silvia Armelloni
- Renal Research Laboratory, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Via Pace 9, Milan, 20122, Italy
| | - Carlo Maria Alfieri
- Unit of Nephrology, Dialysis and Renal Transplant Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Via Della Commenda 15, Milan, 20122, Italy
- University of Study of Milan, Via Festa Del Perdono 7, 20122, Milan, Italy
- Corresponding author. University of Study of Milan, via Festa Del Perdono 7, 20122, Milan, Italy.
| | - Giuseppe Castellano
- Unit of Nephrology, Dialysis and Renal Transplant Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Via Della Commenda 15, Milan, 20122, Italy
- University of Study of Milan, Via Festa Del Perdono 7, 20122, Milan, Italy
| | - Mario Barilani
- EPIGET LAB, Department of Clinical Sciences and Community Health, University of Milan, Milan, 20122, Italy
- Department of Transfusion Medicine and Hematology, Cell Factory, Regenerative Medicine Laboratory, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Francesca Arcudi
- Department of Chemical and Pharmaceutical Sciences, INSTM UdR Trieste, University of Trieste, Via Licio Giorgieri 1, Trieste, 34127, Italy
- Corresponding author.
| | - Piergiorgio Messa
- Unit of Nephrology, Dialysis and Renal Transplant Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Via Della Commenda 15, Milan, 20122, Italy
- University of Study of Milan, Via Festa Del Perdono 7, 20122, Milan, Italy
| | - Maurizio Prato
- Department of Chemical and Pharmaceutical Sciences, INSTM UdR Trieste, University of Trieste, Via Licio Giorgieri 1, Trieste, 34127, Italy
- Center for Cooperative Research in Biomaterials (CIC BiomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 182, Donostia-San Sebastián, 20014, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, 48013, Spain
- Corresponding author. Department of Chemical and Pharmaceutical Sciences, INSTM UdR Trieste, University of Trieste, via Licio Giorgieri 1, Trieste, 34127, Italy.
| |
Collapse
|
3
|
Veenstra JP, Vemu B, Tocmo R, Nauman MC, Johnson JJ. Pharmacokinetic Analysis of Carnosic Acid and Carnosol in Standardized Rosemary Extract and the Effect on the Disease Activity Index of DSS-Induced Colitis. Nutrients 2021; 13:nu13030773. [PMID: 33673488 PMCID: PMC7997407 DOI: 10.3390/nu13030773] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/17/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Rosemary extract (RE) is an approved food preservative in the European Union and contains dietary phytochemicals that are beneficial for gastrointestinal health. This study investigated the effects of RE on dextran sodium sulfate (DSS)-induced colitis and also determined the pharmacokinetics of dietary phytochemicals administered to mice via oral gavage. Individual components of rosemary extract were separated and identified by LC–MS/MS. The pharmacokinetics of two major diterpenes from RE, carnosic acid (CA) and carnosol (CL), administered to mice via oral gavage were determined. Then, the effect of RE pre-treatment on the disease activity index (DAI) of DSS-induced colitis in mice was investigated. The study determined that 100 mg/kg RE significantly improved DAI in DSS-induced colitis compared to negative control. Sestrin 2 protein expression, which increased with DSS exposure, was reduced with RE treatment. Intestinal barrier integrity was also shown to improve via fluorescein isothiocyanate (FITC)–dextran administration and Western blot of zonula occludens-1 (ZO-1), a tight junction protein. Rosemary extract was able to improve the DAI of DSS-induced colitis in mice at a daily dose of 100 mg/kg and showed improvement in the intestinal barrier integrity. This study suggests that RE can be an effective preventative agent against IBD.
Collapse
|
4
|
Tocmo R, Le B, Heun A, van Pijkeren JP, Parkin K, Johnson JJ. Prenylated xanthones from mangosteen (Garcinia mangostana) activate the AhR and Nrf2 pathways and protect intestinal barrier integrity in HT-29 cells. Free Radic Biol Med 2021; 163:102-115. [PMID: 33310139 PMCID: PMC8647718 DOI: 10.1016/j.freeradbiomed.2020.11.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 11/04/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022]
Abstract
Xanthones from the tropical fruit mangosteen (Garcinia mangostana) display anti-inflammatory and anti-oxidative activities. Here, we isolate and identify potential inducers of the aryl hydrocarbon receptor (AhR) and nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathways from mangosteen using a bioassay-guided strategy. Mangosteen fruit pericarp extracts were subjected to sequential solvent extractions, followed by chromatography coupled with NMR spectroscopy and mass spectrometric analyses for identification and isolation of pure compounds. Isolation of active fractions led to seven prenylated xanthones that were identified and subsequently evaluated for bioactivity. In vitro luciferase reporter cellular assays using H1L6.1c3 (AhR induction) and HepG2-ARE (Nrf2 induction) were used to identify AhR and Nrf2 activators. All seven prenylated xanthones displayed AhR inducing activity, whereas only five xanthones activated Nrf2. Garcinone D (GarD) significantly upregulated AhR/Cyp1a1 and Nrf2/HO-1 protein expression and enhanced zonula occludens-1 and occludin protein levels in HT-29 cells. In addition, GarD inhibited oxidative stress-induced intestinal epithelial barrier dysfunction by enhancing tight junction (TJ) proteins and inhibition of reactive oxygen species production. Inhibition of AhR by pretreating cells with an AhR antagonist revealed that the AhR pathway is required for the improved epithelial barrier functions of GarD. These results highlight a dual mechanism by GarD that confers protection against intestinal epithelial barrier dysfunction.
Collapse
Affiliation(s)
- Restituto Tocmo
- Department of Pharmacy Practice, University of Illinois-Chicago, 833 South Wood Street, Chicago, IL, 60612, USA
| | - Bryan Le
- Department of Food Science, University of Wisconsin-Madison, Babcock Hall, 1605 Linden Drive, Madison, WI, 53706, USA
| | - Amber Heun
- Department of Food Science, University of Wisconsin-Madison, Babcock Hall, 1605 Linden Drive, Madison, WI, 53706, USA
| | - Jan Peter van Pijkeren
- Department of Food Science, University of Wisconsin-Madison, Babcock Hall, 1605 Linden Drive, Madison, WI, 53706, USA
| | - Kirk Parkin
- Department of Food Science, University of Wisconsin-Madison, Babcock Hall, 1605 Linden Drive, Madison, WI, 53706, USA
| | - Jeremy James Johnson
- Department of Pharmacy Practice, University of Illinois-Chicago, 833 South Wood Street, Chicago, IL, 60612, USA.
| |
Collapse
|
5
|
Maltseva D, Raygorodskaya M, Knyazev E, Zgoda V, Tikhonova O, Zaidi S, Nikulin S, Baranova A, Turchinovich A, Rodin S, Tonevitsky A. Knockdown of the α5 laminin chain affects differentiation of colorectal cancer cells and their sensitivity to chemotherapy. Biochimie 2020; 174:107-116. [PMID: 32334043 DOI: 10.1016/j.biochi.2020.04.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023]
Abstract
The interaction of tumor cells with the extracellular matrix (ECM) may affect the rate of cancer progression and metastasis. One of the major components of ECM are laminins, the heterotrimeric glycoproteins consisting of α-, β-, and γ-chains (αβγ). Laminins interact with their cell surface receptors and, thus, regulate multiple cellular processes. In this work, we demonstrate that shRNA-mediated knockdown of the α5 laminin chain results in Wnt- and mTORC1-dependent partial dedifferentiation of colorectal cancer cells. Furthermore, we showed that this dedifferentiation involved activation of ER-stress signaling, pathway promoting the sensitivity of cells to 5-fluorouracil.
Collapse
Affiliation(s)
- Diana Maltseva
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Myasnitskaya str. 13/4, 117997, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya str. 16/10, 117997, Moscow, Russia; Scientific Research Center Bioclinicum, Ugreshskaya str. 2/85, 115088, Moscow, Russia.
| | - Maria Raygorodskaya
- Scientific Research Center Bioclinicum, Ugreshskaya str. 2/85, 115088, Moscow, Russia
| | - Evgeny Knyazev
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Myasnitskaya str. 13/4, 117997, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya str. 16/10, 117997, Moscow, Russia
| | - Victor Zgoda
- Institute of Biomedical Chemistry, Pogodinskaya str. 10, 119121, Moscow, Russia
| | - Olga Tikhonova
- Institute of Biomedical Chemistry, Pogodinskaya str. 10, 119121, Moscow, Russia
| | - Shan Zaidi
- School of Systems Biology, George Mason University, Fairfax, VA, 22030, USA
| | - Sergey Nikulin
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Myasnitskaya str. 13/4, 117997, Moscow, Russia; Moscow Institute of Physics and Technology, Institutskiy per. 9, 141700, Dolgoprudny, Russia
| | - Ancha Baranova
- School of Systems Biology, George Mason University, Fairfax, VA, 22030, USA; Moscow Institute of Physics and Technology, Institutskiy per. 9, 141700, Dolgoprudny, Russia; Research Center of Medical Genetics, Moskvorechye str. 1, 115522, Moscow, Russia
| | | | - Sergey Rodin
- Department of Surgical Sciences, Ångström Laboratory, Uppsala University, 752 37, Uppsala, Sweden
| | - Alexander Tonevitsky
- Faculty of Biology and Biotechnology, National Research University Higher School of Economics, Myasnitskaya str. 13/4, 117997, Moscow, Russia; Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Miklukho-Maklaya str. 16/10, 117997, Moscow, Russia; Scientific Research Center Bioclinicum, Ugreshskaya str. 2/85, 115088, Moscow, Russia.
| |
Collapse
|
6
|
Cizkova K, Birke P, Malohlava J, Tauber Z, Huskova Z, Ehrmann J. HT-29 and Caco2 Cell Lines Are Suitable Models for Studying the Role of Arachidonic Acid-Metabolizing Enzymes in Intestinal Cell Differentiation. Cells Tissues Organs 2020; 208:37-47. [PMID: 32248197 DOI: 10.1159/000506735] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 02/23/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Cytochrome (CYP) epoxygenases (CYP2C and CYP2J) and soluble epoxide hydrolase (sEH) participate in the metabolism of arachidonic acid and may also have a potential role in enterocyte differentiation. The first critical step in the study of intestinal cell differentiation is the determination of a suitable in vitro model, which must be as similar as possible to the conditions of a living organism. It is known that HT-29 and Caco2 cell lines derived from human colorectal carcinomas can differentiate into enterocyte-like cells in appropriate culture conditions. MATERIAL AND METHODS We tested 4 different approaches of enterocyte-like differentiation and determined the most appropriate culture conditions for each model. Subsequently, the changes in the expression of CYP epoxygenases and sEH in undifferentiated and differentiated cells were measured by In-Cell ELISA. These results were compared with immunohistochemical profiles of expression of CYP epoxygenases and sEH in samples of human embryonic and fetal intestines as well as adult duodenum and colon. RESULTS Our results show that sodium butyrate (NaBt)-differentiated HT-29 cells and spontaneously differentiated Caco2 cells resemble CYP epoxygenases and sEH profiles, corresponding with different types of intestines. CONCLUSION Our study revealed that the most suitable models for the study of the role of CYP epoxygenases and sEH expression in differentiation of intestinal epithelium are NaBt-differentiated HT-29 cells and spontaneously differentiated Caco2 cells.
Collapse
Affiliation(s)
- Katerina Cizkova
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia,
| | - Petr Birke
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
| | - Jakub Malohlava
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia.,Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
| | - Zdenek Tauber
- Department of Histology and Embryology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
| | - Zlata Huskova
- Department of Clinical and Molecular Pathology and Laboratory of Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
| | - Jiri Ehrmann
- Department of Clinical and Molecular Pathology and Laboratory of Molecular Pathology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czechia
| |
Collapse
|
7
|
Alabi BR, LaRanger R, Shay JW. Decellularized mice colons as models to study the contribution of the extracellular matrix to cell behavior and colon cancer progression. Acta Biomater 2019; 100:213-222. [PMID: 31562987 DOI: 10.1016/j.actbio.2019.09.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 09/11/2019] [Accepted: 09/21/2019] [Indexed: 12/13/2022]
Abstract
Current 3D culture models to study colorectal cancer lack architectural support and signaling proteins provided by the tissue extracellular matrix (ECM) which may influence cell behavior and cancer progression. Therefore, the ability to study cancer cells in the context of a matrix that is physiologically more relevant and to understand how the ECM affects cancer progression has been understudied. To address this, we developed an ex-vivo 3D system, provided by intact wild type (WT) and colon cancer susceptible decellularized mouse colons (DMC), to support the growth of human cancer cells. DMC are free of viable cells but still contain extracellular matrix proteins including subsets of collagens. Stiffness, an important mechanical property, is also maintained in DMCs. Importantly, we observed that the DMC is permissive for cell proliferation and differentiation of a human colon cancer cell line (HT-29). Notably, the ability of cells in the WT DMC to differentiate was also greater when compared to Matrigel™, an extracellular matrix extract from a mouse tumor cell line. Additionally, we observed in invasion assays that DMC obtained from polyps from a colon cancer susceptible mouse model facilitated increased cell migration/invasion of colorectal cancer cells and immortalized non-tumor colonic epithelial cells compared to DMC from WT mice. Finally, using mass spectrometry, we identified extracellular matrix proteins that are more abundant in DMC from a colorectal cancer mouse model compared to age and sex-matched WT mice. We propose that these abundantly expressed proteins in the tumor microenvironment are potentially involved in colorectal cancer progression. STATEMENT OF SIGNIFICANCE: Decellularized matrices, when properly produced, are attractive biomaterials for tissue regeneration and replacement. We show here that the mouse decellularized matrices can also be repurposed to elucidate how the extracellular matrix influences human cell behavior and cancer progression. To do this we produce decellularized matrices, from mice colonic tissue, that have preserved tissue mechanical and structural properties. We demonstrate that the matrix better supports the differentiation of HT-29 cells, a colonic cancer cell line, compared to Matrigel™. Additionally, we show that the extracellular matrix contributes to colon cancer progression via invasion assays using extracellular matrix extracts. Finally, we use mass spectrometry to identify ECM proteins that are more abundant in colonic polyps compared to adjacent tissue regions. This model system may have therapeutic implications for colorectal cancer patients.
Collapse
Affiliation(s)
- Busola R Alabi
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9039, United States
| | - Ryan LaRanger
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9039, United States
| | - Jerry W Shay
- Department of Cell Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9039, United States.
| |
Collapse
|
8
|
Swain S, Roe MM, Sebrell TA, Sidar B, Dankoff J, VanAusdol R, Smythies LE, Smith PD, Bimczok D. CD103 (αE Integrin) Undergoes Endosomal Trafficking in Human Dendritic Cells, but Does Not Mediate Epithelial Adhesion. Front Immunol 2018; 9:2989. [PMID: 30622531 PMCID: PMC6308147 DOI: 10.3389/fimmu.2018.02989] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/04/2018] [Indexed: 12/20/2022] Open
Abstract
Dendritic cell (DC) expression of CD103, the α subunit of αEβ7 integrin, is thought to enable DC interactions with E-cadherin-expressing gastrointestinal epithelia for improved mucosal immunosurveillance. In the stomach, efficient DC surveillance of the epithelial barrier is crucial for the induction of immune responses to H. pylori, the causative agent of peptic ulcers and gastric cancer. However, gastric DCs express only low levels of surface CD103, as we previously showed. We here tested the hypothesis that intracellular pools of CD103 in human gastric DCs can be redistributed to the cell surface for engagement of epithelial cell-expressed E-cadherin to promote DC-epithelial cell adhesion. In support of our hypothesis, immunofluorescence analysis of tissue sections showed that CD103+ gastric DCs were preferentially localized within the gastric epithelial layer. Flow cytometry and imaging cytometry revealed that human gastric DCs expressed intracellular CD103, corroborating our previous findings in monocyte-derived DCs (MoDCs). Using confocal microscopy, we show that CD103 was present in endosomal compartments, where CD103 partially co-localized with clathrin, early endosome antigen-1 and Rab11, suggesting that CD103 undergoes endosomal trafficking similar to β1 integrins. Dynamic expression of CD103 on human MoDCs was confirmed by internalization assay. To analyze whether DC-expressed CD103 promotes adhesion to E-cadherin, we performed adhesion and spreading assays on E-cadherin-coated glass slides. In MoDCs generated in the presence of retinoic acid, which express increased CD103, intracellular CD103 significantly redistributed toward the E-cadherin-coated glass surface. However, DCs spreading and adhesion did not differ between E-cadherin-coated slides and slides coated with serum alone. In adhesion assays using E-cadherin-positive HT-29 cells, DC binding was significantly improved by addition of Mn2+ and decreased in the presence of EGTA, consistent with the dependence of integrin-based interactions on divalent cations. However, retinoic acid failed to increase DC adhesion, and a CD103 neutralizing antibody was unable to inhibit DC binding to the E-cadherin positive cells. In contrast, a blocking antibody to DC-expressed E-cadherin significantly reduced DC binding to the epithelium. Overall, these data indicate that CD103 engages in DC-epithelial cell interactions upon contact with epithelial E-cadherin, but is not a major driver of DC adhesion to gastrointestinal epithelia.
Collapse
Affiliation(s)
- Steve Swain
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Mandi M. Roe
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Thomas A. Sebrell
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Barkan Sidar
- Chemical and Biological Engineering Department, Montana State University, Bozeman, MT, United States
| | - Jennifer Dankoff
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Rachel VanAusdol
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| | - Lesley E. Smythies
- Division of Gastroenterology and Hepatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Phillip D. Smith
- Division of Gastroenterology and Hepatology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Diane Bimczok
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, United States
| |
Collapse
|
9
|
Ducarouge B, Pelissier-Rota M, Powell R, Buisson A, Bonaz B, Jacquier-Sarlin M. Involvement of CRF2 signaling in enterocyte differentiation. World J Gastroenterol 2017; 23:5127-5145. [PMID: 28811708 PMCID: PMC5537180 DOI: 10.3748/wjg.v23.i28.5127] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/06/2017] [Accepted: 07/12/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To determine the role of corticotropin releasing factor receptor (CRF2) in epithelial permeability and enterocyte cell differentiation.
METHODS For this purpose, we used rat Sprague Dawley and various colon carcinoma cell lines (SW620, HCT8R, HT-29 and Caco-2 cell lines). Expression of CRF2 protein was analyzed by fluorescent immunolabeling in normal rat colon and then by western blot in dissociated colonic epithelial cells and in the lysates of colon carcinoma cell lines or during the early differentiation of HT-29 cells (ten first days). To assess the impact of CRF2 signaling on colonic cell differentiation, HT-29 and Caco-2 cells were exposed to Urocortin 3 recombinant proteins (Ucn3, 100 nmol/L). In some experiments, cells were pre-exposed to the astressin 2b (A2b) a CRF2 antagonist in order to inhibit the action of Ucn3. Intestinal cell differentiation was first analyzed by functional assays: the trans-cellular permeability and the para-cellular permeability were determined by Dextran-FITC intake and measure of the transepithelial electrical resistance respectively. Morphological modifications associated to epithelial dysfunction were analyzed by confocal microscopy after fluorescent labeling of actin (phaloidin-TRITC) and intercellular adhesion proteins such as E-cadherin, p120ctn, occludin and ZO-1. The establishment of mature adherens junctions (AJ) was monitored by following the distribution of AJ proteins in lipid raft fractions, after separation of cell lysates on sucrose gradients. Finally, the mRNA and the protein expression levels of characteristic markers of intestinal epithelial cell (IEC) differentiation such as the transcriptional factor krüppel-like factor 4 (KLF4) or the dipeptidyl peptidase IV (DPPIV) were performed by RT-PCR and western blot respectively. The specific activities of DPPIV and alkaline phosphatase (AP) enzymes were determined by a colorimetric method.
RESULTS CRF2 protein is preferentially expressed in undifferentiated epithelial cells from the crypts of colon and in human colon carcinoma cell lines. Furthermore, CRF2 expression is down regulated according to the kinetic of HT-29 cell differentiation. By performing functional assays, we found that Ucn3-induced CRF2 signaling alters both para- and trans-cellular permeability of differentiated HT-29 and Caco-2 cells. These effects are partly mediated by Ucn3-induced morphological changes associated with the disruption of mature AJ in HT-29 cells and tight junctions (TJ) in Caco-2 cells. Ucn3-mediated activation of CRF2 decreases mRNA and protein expression levels of KLF4 a transcription factor involved in IEC differentiation. This signaling is correlated to a down-regulation of key IEC markers such as DPPIV and AP, at both transcriptional and post-transcriptional levels.
CONCLUSION Our findings suggest that CRF2 signaling could modulate IEC differentiation. These mechanisms could be relevant to the stress induced epithelial alterations found in inflammatory bowel diseases.
Collapse
|
10
|
Comparison of in vitro methods for carboxylesterase activity determination in immortalized cells representative of the intestine, liver and kidney. Mol Cell Probes 2015; 29:215-22. [DOI: 10.1016/j.mcp.2015.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 04/17/2015] [Accepted: 05/04/2015] [Indexed: 12/24/2022]
|
11
|
Ye J, Song L, Liu Y, Pan Q, Zhong X, Li S, Shang Y, Tian Y, He Y, Chen L, Chen W, Peng Z, Wang R. Core 2 Mucin-Type O-Glycan Is Related to EPEC and EHEC O157:H7 Adherence to Human Colon Carcinoma HT-29 Epithelial Cells. Dig Dis Sci 2015; 60:1977-90. [PMID: 25701318 DOI: 10.1007/s10620-015-3548-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/19/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM The roles of host glycosylation in interactions with EPEC and EHEC O157:H7 are largely unclear; this study examined whether O-glycans are involved in EPEC and EHEC O157:H7 adherence to HT-29 cells. METHODS Bacterial adherence to the cultured cells was determined using the direct co-staining of adherent bacteria and host cells, the adherent bacteria plating, and/or the direct fluorescent observation of the adherent GFP-labeled bacteria. RESULTS A comparison of the adherence of EPEC and EHEC O157:H7 to HT-29-Gal and HT-29 cells indicated that the differentiation of HT-29 cells led to a reduction in the adherence of EPEC and EHEC O157:H7. EPEC and EHEC O157:H7 adhesion decreased after the abrogation of O-glycan biosynthesis mediated by benzyl-α-GalNAc treatment. Core 2 O-glycan-deficient HT-29 cells induced by C2GnT2 knockdown had a significant reduction in EPEC and EHEC O157:H7 adhesion in C2GnT2-sh2/HT-29 cells compared with HT-29 and shRNA-Ctr/HT-29 cells. MUC2 expression in benzyl-α-GalNAc-treated HT-29 cells was significantly reduced but unchanged in C2GnT2-deficient HT-29 cells. EPEC or EHEC O157:H7 infection in C2GnT2-deficient HT-29 cells deteriorated the epithelial barrier function. The occludin expression in the shRNA-Ctr/HT-29 and C2GnT2-sh2/HT-29 cells after infection with EPEC or EHEC O157:H7 was pyknic and discontinuous at the cell surface compared with its continuous distribution of control cells. These data indicate that EPEC and EHEC O157:H7 adherence to HT-29 cells is related to mucin-type core 2 O-glycan. CONCLUSIONS This study provides the concepts toward the design of carbohydrate-dependent inhibition of EPEC and EHEC O157:H7 adhesion to human intestinal epithelial cells.
Collapse
Affiliation(s)
- Jun Ye
- Department of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Pelissier-Rota MA, Chartier NT, Jacquier-Sarlin MR. Dynamic Regulation of Adherens Junctions: Implication in Cell Differentiation and Tumor Development. INTERCELLULAR COMMUNICATION IN CANCER 2015:53-149. [DOI: 10.1007/978-94-017-7380-5_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
13
|
Herr R, Köhler M, Andrlová H, Weinberg F, Möller Y, Halbach S, Lutz L, Mastroianni J, Klose M, Bittermann N, Kowar S, Zeiser R, Olayioye MA, Lassmann S, Busch H, Boerries M, Brummer T. B-Raf inhibitors induce epithelial differentiation in BRAF-mutant colorectal cancer cells. Cancer Res 2014; 75:216-29. [PMID: 25381152 DOI: 10.1158/0008-5472.can-13-3686] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
BRAF mutations are associated with aggressive, less-differentiated and therapy-resistant colorectal carcinoma. However, the underlying mechanisms for these correlations remain unknown. To understand how oncogenic B-Raf contributes to carcinogenesis, in particular to aspects other than cellular proliferation and survival, we generated three isogenic human colorectal carcinoma cell line models in which we can dynamically modulate the expression of the B-Raf(V600E) oncoprotein. Doxycyclin-inducible knockdown of endogenous B-Raf(V600E) decreases cellular motility and invasion in conventional and three-dimensional (3D) culture, whereas it promotes cell-cell contacts and induces various hallmarks of differentiated epithelia. Importantly, all these effects are recapitulated by B-Raf (PLX4720, vemurafenib, and dabrafenib) or MEK inhibitors (trametinib). Surprisingly, loss of B-Raf(V600E) in HT29 xenografts does not only stall tumor growth, but also induces glandular structures with marked expression of CDX2, a tumor-suppressor and master transcription factor of intestinal differentiation. By performing the first transcriptome profiles of PLX4720-treated 3D cultures of HT29 and Colo-205 cells, we identify several upregulated genes linked to epithelial differentiation and effector functions, such as claudin-1, a Cdx-2 target gene encoding a critical tight junction component. Thereby, we provide a mechanism for the clinically observed correlation between mutant BRAF and the loss of Cdx-2 and claudin-1. PLX4720 also suppressed several metastasis-associated transcripts that have not been implicated as targets, effectors or potential biomarkers of oncogenic B-Raf signaling so far. Together, we identify a novel facet of clinically applied B-Raf or MEK inhibitors by showing that they promote cellular adhesion and differentiation of colorectal carcinoma cells.
Collapse
Affiliation(s)
- Ricarda Herr
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University (ALU), Freiburg, Germany. Faculty of Biology, ALU, Freiburg, Germany
| | - Martin Köhler
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University (ALU), Freiburg, Germany. Faculty of Biology, ALU, Freiburg, Germany. Spemann Graduate School of Biology and Medicine (SGBM), ALU, Freiburg, Germany
| | - Hana Andrlová
- Department of Hematology and Oncology, University Medical Center, ALU, Freiburg, Germany
| | - Florian Weinberg
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University (ALU), Freiburg, Germany. Faculty of Biology, ALU, Freiburg, Germany
| | - Yvonne Möller
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Sebastian Halbach
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University (ALU), Freiburg, Germany. Faculty of Biology, ALU, Freiburg, Germany. Spemann Graduate School of Biology and Medicine (SGBM), ALU, Freiburg, Germany
| | - Lisa Lutz
- Department of Pathology, University Medical Center, ALU, Freiburg, Germany
| | - Justin Mastroianni
- Faculty of Biology, ALU, Freiburg, Germany. Department of Hematology and Oncology, University Medical Center, ALU, Freiburg, Germany
| | - Martin Klose
- Systems Biology of the Cellular Microenvironment Group, IMMZ, ALU, Freiburg, Germany
| | - Nicola Bittermann
- Department of Pathology, University Medical Center, ALU, Freiburg, Germany
| | - Silke Kowar
- Systems Biology of the Cellular Microenvironment Group, IMMZ, ALU, Freiburg, Germany
| | - Robert Zeiser
- Department of Hematology and Oncology, University Medical Center, ALU, Freiburg, Germany. Centre for Biological Signalling Studies BIOSS, ALU Freiburg
| | - Monilola A Olayioye
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Silke Lassmann
- Department of Pathology, University Medical Center, ALU, Freiburg, Germany. Centre for Biological Signalling Studies BIOSS, ALU Freiburg. German Cancer Consortium (DKTK), Freiburg, Germany. German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hauke Busch
- Systems Biology of the Cellular Microenvironment Group, IMMZ, ALU, Freiburg, Germany. German Cancer Consortium (DKTK), Freiburg, Germany. German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Melanie Boerries
- Systems Biology of the Cellular Microenvironment Group, IMMZ, ALU, Freiburg, Germany. German Cancer Consortium (DKTK), Freiburg, Germany. German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Tilman Brummer
- Signal Transduction in Tumour Development and Drug Resistance Group, Institute of Molecular Medicine and Cell Research (IMMZ), Albert-Ludwigs-University (ALU), Freiburg, Germany. Centre for Biological Signalling Studies BIOSS, ALU Freiburg.
| |
Collapse
|
14
|
Fan L, Li A, Li W, Cai P, Yang B, Zhang M, Gu Y, Shu Y, Sun Y, Shen Y, Wu X, Hu G, Wu X, Xu Q. Novel role of Sarco/endoplasmic reticulum calcium ATPase 2 in development of colorectal cancer and its regulation by F36, a curcumin analog. Biomed Pharmacother 2014; 68:1141-8. [DOI: 10.1016/j.biopha.2014.10.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 10/16/2014] [Indexed: 11/29/2022] Open
|
15
|
Cohen L, Sekler I, Hershfinkel M. The zinc sensing receptor, ZnR/GPR39, controls proliferation and differentiation of colonocytes and thereby tight junction formation in the colon. Cell Death Dis 2014; 5:e1307. [PMID: 24967969 PMCID: PMC4611734 DOI: 10.1038/cddis.2014.262] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/11/2014] [Accepted: 05/05/2014] [Indexed: 12/21/2022]
Abstract
The intestinal epithelium is a renewable tissue that requires precise balance between proliferation and differentiation, an essential process for the formation of a tightly sealed barrier. Zinc deficiency impairs the integrity of the intestinal epithelial barrier and is associated with ulcerative and diarrheal pathologies, but the mechanisms underlying the role of Zn2+ are not well understood. Here, we determined a role of the colonocytic Zn2+ sensing receptor, ZnR/GPR39, in mediating Zn2+-dependent signaling and regulating the proliferation and differentiation of colonocytes. Silencing of ZnR/GPR39 expression attenuated Zn2+-dependent activation of ERK1/2 and AKT as well as downstream activation of mTOR/p70S6K, pathways that are linked with proliferation. Consistently, ZnR/GPR39 silencing inhibited HT29 and Caco-2 colonocyte proliferation, while not inducing caspase-3 cleavage. Remarkably, in differentiating HT29 colonocytes, silencing of ZnR/GPR39 expression inhibited alkaline phosphatase activity, a marker of differentiation. Furthermore, Caco-2 colonocytes showed elevated expression of ZnR/GPR39 during differentiation, whereas silencing of ZnR/GPR39 decreased monolayer transepithelial electrical resistance, suggesting compromised barrier formation. Indeed, silencing of ZnR/GPR39 or chelation of Zn2+ by the cell impermeable chelator CaEDTA was followed by impaired expression of the junctional proteins, that is, occludin, zonula-1 (ZO-1) and E-cadherin. Importantly, colon tissues of GPR39 knockout mice also showed a decrease in expression levels of ZO-1 and occludin compared with wildtype mice. Altogether, our results indicate that ZnR/GPR39 has a dual role in promoting proliferation of colonocytes and in controlling their differentiation. The latter is followed by ZnR/GPR39-dependent expression of tight junctional proteins, thereby leading to formation of a sealed intestinal epithelial barrier. Thus, ZnR/GPR39 may be a therapeutic target for promoting epithelial function and tight junction barrier integrity during ulcerative colon diseases.
Collapse
Affiliation(s)
- L Cohen
- Department of Physiology and Cell Biology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - I Sekler
- Department of Physiology and Cell Biology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - M Hershfinkel
- Department of Physiology and Cell Biology, Faculty of Health Science, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
16
|
Pan Q, Tian Y, Li X, Ye J, Liu Y, Song L, Yang Y, Zhu R, He Y, Chen L, Chen W, Mao X, Peng Z, Wang R. Enhanced membrane-tethered mucin 3 (MUC3) expression by a tetrameric branched peptide with a conserved TFLK motif inhibits bacteria adherence. J Biol Chem 2013; 288:5407-16. [PMID: 23316049 DOI: 10.1074/jbc.m112.408245] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
We investigated whether a synthetic tetrameric branched peptide based on the conserved TFLK motif from mammary-associated serum amyloid A3 (M-SAA3) is more efficient than the monomeric peptide at up-regulating MUC3 expression and examined the possible mechanism(s) and biological significance of this process. We used standard solid-phase methods to synthesize a tetrameric branched peptide (sequence GWLTFLKAAG) containing a trilysine core, termed the TFLK-containing 10-mer BP. The aberrant expression of transcription factors was analyzed using a transcription factor protein/DNA array. MUC3 and relevant transcription factors were detected using real-time PCR and/or Western blots. The luciferase assay, EMSA, and ChIP assays were used to analyze the activity of the human MUC3 promoter. The bacterial adherence assay was used to evaluate the in vitro inhibition of enteropathogenic Escherichia coli or enterohemorrhage E. coli serotype O157:H7 (EHEC O157:H7) adherence to HT-29-Gal cells after treatment with the TFLK-containing 10-mer BP. In HT-29-Gal cells, the TFLK-containing 10-mer BP induced higher levels of MUC3 expression than the M-SAA3-derived N-terminal 10-mer monomeric peptide, and MUC3 expression was activated through transcriptional mechanisms, including the induction of multiple transcription factors and further binding with their cis-elements between nucleotides -242 and -62 within MUC3 promoter. Interestingly, the TFLK-containing 10-mer BP dramatically inhibited enteropathogenic E. coli and EHEC O157:H7 adherence to the HT-29-Gal cells compared with the controls. This finding suggests a potential therapeutic use for this peptide to prevent gastrointestinal infection.
Collapse
Affiliation(s)
- Qiong Pan
- Department of Gastroenterology, Southwest Hospital, Chongqing, 400038 China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Pelissier-Rota M, Lainé M, Ducarouge B, Bonaz B, Jacquier-Sarlin M. Role of Cholinergic Receptors in Colorectal Cancer: Potential Therapeutic Implications of Vagus Nerve Stimulation? ACTA ACUST UNITED AC 2013. [DOI: 10.4236/jct.2013.46128] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
18
|
Scherbakov AM, Andreeva OE, Shatskaya VA, Krasil'nikov MA. The relationships between snail1 and estrogen receptor signaling in breast cancer cells. J Cell Biochem 2012; 113:2147-55. [PMID: 22307688 DOI: 10.1002/jcb.24087] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The loss of hormonal dependency of breast tumor cells is often accompanied with the appearance of epithelial-mesenchymal transition (EMT) features and increase in cell metastasis and invasiveness. The central role in the EMT belongs to transcription factors Snail responded for the decrease in E-cadherin expression and cell contacts, stimulation of cell mobility and invasiveness. Aim was to study the relationships between estrogen receptor machinery and Snail1 signaling, and mechanism of Snail1 regulation in hormone-resistant breast cancer cells. The experiments were performed on the estrogen-dependent MCF-7 breast cancer cells, estrogen-hyposensitive MCF-7/LS subline generated through long-term cultivation of the parental cells in steroid-free medium, and ER-negative estrogen-resistant HBL-100 cells. Snail1, estrogen receptor, p65 NF-κB, E-cadherin levels were analyzed by Western blot. We found that decrease in the estrogen dependency is correlated with increase in Snail1 expression and activity, we demonstrated the Snail1 involvement in the negative regulation of ER, and showed that Snail1 inhibition partially restores the sensitivity of the estrogen-hyposensitive cells to antiestrogen tamoxifen. Furthermore, NF-κB was found to serve as a positive regulator of Snail1 in breast cancer cells, and simultaneous inhibition of NF-κB and Snail1 resulted in additional increase in cell response to tamoxifen. In general, the results obtained demonstrate the phenomenon of Snail1 activation in the hormone-resistant breast cancer cells, and show that Snail1 and NF-κB may serve as an important targets in the treatment of breast cancer, both estrogen-dependent and estrogen-independent tumors.
Collapse
Affiliation(s)
- Alexander M Scherbakov
- Laboratory of Clinical Biochemistry, Institute of Clinical Oncology, N.N. Blokhin Cancer Research Centre, Kashirskoye sh. 24, Moscow 115478, Russia.
| | | | | | | |
Collapse
|
19
|
Chartier NT, Lainé MG, Ducarouge B, Oddou C, Bonaz B, Albiges-Rizo C, Jacquier-Sarlin MR. Enterocytic differentiation is modulated by lipid rafts-dependent assembly of adherens junctions. Exp Cell Res 2011; 317:1422-36. [PMID: 21419117 DOI: 10.1016/j.yexcr.2011.03.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 02/18/2011] [Accepted: 03/09/2011] [Indexed: 01/29/2023]
Abstract
Integrity of the epithelial barrier is determined by apical junctional complexes which also participate in the signalling pathways inducing intestinal cell differentiation. Lipid rafts (LR) have been proposed to play a role in the organization and the function of these intercellular complexes. This study investigated potential mechanisms by which LR could participate in the establishment of adherens junctions (AJ) and the initiation of enterocytic differentiation. We showed that the differentiation of epithelial cells in rat colons correlates with the emergence of LR. Using HT-29 cells we demonstrated that during the differentiation process, LR are required for the recruitment and the association of p120ctn to E-cadherin. Silencing of flotillin-1, a LR component, alters the recruitment of AJ proteins in LR and delays the expression of differentiation markers. Furthermore, the ability of p120ctn/E-cadherin complexes to support cell differentiation is altered in HT-29 Rac1N17 cells. These results show a contributory role of LR in the enterocytic differentiation process, which serve as signalling platforms for Rac1-mediated organization of AJ. A better understanding of the mechanism involved in the establishment of junctional complex and their role in enterocytic differentiation provides new insights into the regulation of intestinal homeostasis.
Collapse
Affiliation(s)
- Nicolas T Chartier
- Centre de Recherche Inserm U823, Institut Albert Bonniot, Equipe de Dynamique des Systèmes d'Adhérence et de Différenciation, Site Santé BP 170 La Tronche F-38042 Grenoble Cedex 9, France
| | | | | | | | | | | | | |
Collapse
|
20
|
Tiang JM, Butcher NJ, Cullinane C, Humbert PO, Minchin RF. RNAi-mediated knock-down of arylamine N-acetyltransferase-1 expression induces E-cadherin up-regulation and cell-cell contact growth inhibition. PLoS One 2011; 6:e17031. [PMID: 21347396 PMCID: PMC3036737 DOI: 10.1371/journal.pone.0017031] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Accepted: 01/09/2011] [Indexed: 01/06/2023] Open
Abstract
Arylamine N-acetyltransferase-1 (NAT1) is an enzyme that catalyzes the biotransformation of arylamine and hydrazine substrates. It also has a role in the catabolism of the folate metabolite p-aminobenzoyl glutamate. Recent bioinformatics studies have correlated NAT1 expression with various cancer subtypes. However, a direct role for NAT1 in cell biology has not been established. In this study, we have knocked down NAT1 in the colon adenocarcinoma cell-line HT-29 and found a marked change in cell morphology that was accompanied by an increase in cell-cell contact growth inhibition and a loss of cell viability at confluence. NAT1 knock-down also led to attenuation in anchorage independent growth in soft agar. Loss of NAT1 led to the up-regulation of E-cadherin mRNA and protein levels. This change in E-cadherin was not attributed to RNAi off-target effects and was also observed in the prostate cancer cell-line 22Rv1. In vivo, NAT1 knock-down cells grew with a longer doubling time compared to cells stably transfected with a scrambled RNAi or to parental HT-29 cells. This study has shown that NAT1 affects cell growth and morphology. In addition, it suggests that NAT1 may be a novel drug target for cancer therapeutics.
Collapse
Affiliation(s)
- Jacky M Tiang
- School of Biomedical Sciences, University of Queensland, St. Lucia, Queensland, Australia
| | | | | | | | | |
Collapse
|
21
|
Biazik JM, Jahn KA, Su Y, Wu YN, Braet F. Unlocking the ultrastructure of colorectal cancer cells in vitro using selective staining. World J Gastroenterol 2010; 16:2743-53. [PMID: 20533594 PMCID: PMC2883130 DOI: 10.3748/wjg.v16.i22.2743] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To characterise differences between three widely used colorectal cancer cell lines using ultrastructural selective staining for glycogen to determine variation in metastatic properties.
METHODS: Transmission electron microscopy was used in this investigation to help identify intracellular structures and morphological features which are precursors of tumor invasion. In addition to morphological markers, we used selective staining of glycogen as a marker for neoplastic cellular proliferation and determined whether levels of glycogen change between the three different cell lines.
RESULTS: Ultrastructural analysis revealed morphological differences between the cell lines, as well as differentiation into two sub-populations within each cell line. Caco-2 cells contained large glycogen deposits as well as showing the most obvious morphological changes between the two sub-populations. SW480 cells also contained large glycogen stores as well as deep cellular protrusions when grown on porous filter membranes. HT-29 cells had trace amounts of glycogen stores with few cellular projections into the filter pores and no tight junction formation.
CONCLUSION: Morphology indicative of metastatic properties coincided with larger glycogen deposits, providing strong evidence for the use of selective staining to determine the neoplastic properties of cells.
Collapse
|
22
|
Nin V, Hernández JA, Chifflet S. Hyperpolarization of the plasma membrane potential provokes reorganization of the actin cytoskeleton and increases the stability of adherens junctions in bovine corneal endothelial cells in culture. ACTA ACUST UNITED AC 2009; 66:1087-99. [DOI: 10.1002/cm.20416] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
23
|
Galván EM, Roth GA, Monferran CG. Functional interaction of Escherichia coli heat-labile enterotoxin with blood group A-active glycoconjugates from differentiated HT29 cells. FEBS J 2006; 273:3444-53. [PMID: 16884490 DOI: 10.1111/j.1742-4658.2006.05368.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Human colon adenocarcinoma cells (HT29-ATCC) and the clone HT29-5F7 were cultured under conditions that differentiate cells to a polarized intestinal phenotype. Differentiated cells showed the presence of junctional complexes and intercellular lumina bordered by microvilli. Intestinal brush border hydrolase activities (sucrase, aminopeptidase N, lactase and maltase) were detected mainly in differentiated HT29-ATCC cells compared with the differentiated clone, HT29-5F7. The presence of non-GM1 receptors of Escherichia coli heat-labile enterotoxin (LT-I) on both types of differentiated HT29 cells was indicated by the inability of cholera toxin B subunit to block LT-I binding to the cells. Binding of LT-I to cells, when GM1 was blocked by the cholera toxin B subunit, was characterized by an increased number of LT-I receptors with respect to undifferentiated control cells. Moreover, both types of differentiated cells accumulated higher amounts of cyclic AMP in response to LT-I than undifferentiated cells. Helix pomatia lectin inhibited the binding of LT-I to cells and the subsequent production of cyclic AMP. LT-I recognized blood group A-active glycosphingolipids as functional receptors in both HT29 cell lines and the active pro-sucrase form of the glycoprotein carrying A-blood group activity present in HT29-ATCC cells. These results strongly suggest that LT-I can elicit an enhanced functional response using blood group A-active glycoconjugates as additional receptors on polarized intestinal epithelial cells.
Collapse
Affiliation(s)
- Estela M Galván
- Departamento de Química Biológica-CIQUIBIC (CONICET), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Argentina
| | | | | |
Collapse
|
24
|
Chartier NT, Lainé M, Gout S, Pawlak G, Marie CA, Matos P, Block MR, Jacquier-Sarlin MR. Laminin-5-integrin interaction signals through PI 3-kinase and Rac1b to promote assembly of adherens junctions in HT-29 cells. J Cell Sci 2006; 119:31-46. [PMID: 16339173 DOI: 10.1242/jcs.02698] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Human intestinal cell differentiation is mediated by signaling pathways that remain largely undefined. We and others have shown that cell migration and differentiation along the crypt-villus axis is associated with temporal and spatial modulations of the repertoire, as well as with the function of integrins and E-cadherins and their substrates. Cross-talk between integrin and cadherin signaling was previously described and seems to coordinate this differentiation process. Here, we report that engagement of alpha6 and, to a lesser extent, alpha3 integrin subunits after HT-29 cell adhesion on laminin 5 increases the expression of E-cadherin, which then organizes into nascent adherens junctions. We further identify that phosphoinositide 3-kinase (PI 3-kinase) activation plays a key role in this cross-talk. Indeed, integrin-dependent adhesion on laminin 5 stimulates PI 3-kinase activity. Immunofluorescence and immunoprecipitation experiments revealed that activated PI 3-kinase is recruited at cell-cell contacts. Using LY294002, an inhibitor of PI 3-kinase activity, we found that this activation is essential for E-cadherin connection with the cytoskeleton and for biogenesis of adherens junctions. Finally, we demonstrated that PI 3-kinase could signal through Rac1b activation to control adherens junction assembly. Our results provide a mechanistic insight into integrin-cadherin cross-talk and identify a novel role for PI 3-kinase in the establishment of adherens junctions.
Collapse
Affiliation(s)
- Nicolas T Chartier
- Laboratoire d'Etude de la Différenciation et de l'Adhérence Cellulaires, UMR UJF/CNRS 5538, Institut Albert Bonniot, Faculté de Médecine de Grenoble, Domaine de la Merci, 38706 La Tronche Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Taniuchi K, Nakagawa H, Hosokawa M, Nakamura T, Eguchi H, Ohigashi H, Ishikawa O, Katagiri T, Nakamura Y. Overexpressed P-cadherin/CDH3 promotes motility of pancreatic cancer cells by interacting with p120ctn and activating rho-family GTPases. Cancer Res 2005; 65:3092-9. [PMID: 15833838 DOI: 10.1158/0008.5472.can-04-3646] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
P-Cadherin/CDH3 belongs to the family of classic cadherins that are engaged in various cellular activities including motility, invasion, and signaling of tumor cells, in addition to cell adhesion. However, the biological roles of P-cadherin itself are not fully characterized. Based on information derived from a previous genome-wide cDNA microarray analysis of microdissected pancreatic ductal adenocarcinoma (PDAC), we focused on P-cadherin as one of the genes most strongly overexpressed in the great majority of PDACs. To investigate the consequences of overexpression of P-cadherin in terms of pancreatic carcinogenesis and tumor progression, we used a P-cadherin-deficient PDAC cell line, Panc-1, to construct a cell line (Panc1-CDH3) that stably overexpressed P-cadherin. Induction of P-cadherin in Panc1-CDH3 increased the motility of the cancer cells, but a blocking antibody against P-cadherin suppressed the motility in vitro. Overexpression of P-cadherin was strongly associated with cytoplasmic accumulation of one of the catenins, p120ctn, and cadherin switching in PDAC cells. Moreover, P-cadherin-dependent activation of cell motility was associated with activation of Rho GTPases, Rac1 and Cdc42, through accumulation of p120ctn in cytoplasm and cadherin switching. These findings suggest that overexpression of P-cadherin is likely to be related to the biological aggressiveness of PDACs; blocking of P-cadherin activity or its associated signaling could be a novel therapeutic approach for treatment of aggressive pancreatic cancers.
Collapse
Affiliation(s)
- Keisuke Taniuchi
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Inhibition of defined molecular steps of tumorigenesis by natural nontoxic compounds may be an efficient means to tackle the population cancer burden. Extensive research has addressed the chemotherapeutic potential of curcumin (diferuloylmethane), a relatively nontoxic plant-derived polyphenol. This review considers the following properties of curcumin: anticancer effects in animal model systems; metabolism; biological structure and pharmacokinetics; biological properties implicated in chemoprevention; antioxidant properties; influences upon Phase I and II carcinogen-metabolizing enzymes; signal transduction properties and the neoplastic phenotype; apoptosis evasion, cell proliferation, de-differentiation, migration and invasion and clinical studies. This review will summarize the unique properties of curcumin that may be exploited for successful clinical cancer prevention.
Collapse
Affiliation(s)
- Frederick C Campbell
- Queen's University of Belfast, Department of Surgery, Centre for Cancer and Cell Biology, Belfast, BT12 6BJ, Ireland.
| | | |
Collapse
|