1
|
Ferreira RM, Figueiredo J, Pinto-Ribeiro I, Gullo I, Sgouras DN, Carreto L, Castro P, Santos MA, Carneiro F, Seruca R, Figueiredo C. Activation of Laminin γ2 by Helicobacter pylori Promotes Invasion and Survival of Gastric Cancer Cells With E-Cadherin Defects. J Infect Dis 2022; 226:2226-2237. [PMID: 36173814 DOI: 10.1093/infdis/jiac397] [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: 08/01/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Helicobacter pylori infection induces cellular phenotypes relevant for cancer progression, namely cell motility and invasion. We hypothesized that the extracellular matrix (ECM) could be involved in these deleterious effects. METHODS Microarrays were used to uncover ECM interactors in cells infected with H. pylori. LAMC2, encoding laminin γ2, was selected as a candidate gene and its expression was assessed in vitro and in vivo. The role of LAMC2 was investigated by small interference RNA (siRNA) combined with a set of functional assays. Laminin γ2 and E-cadherin expression patterns were evaluated in gastric cancer cases. RESULTS Laminin γ2 was found significantly overexpressed in gastric cancer cells infected with H. pylori. This finding was validated in vitro by infection with clinical isolates and in vivo by using gastric biopsies of infected and noninfected individuals. We showed that laminin γ2 overexpression is dependent on the bacterial type IV secretion system and on the CagA. Functionally, laminin γ2 promotes cell invasion and resistance to apoptosis, through modulation of Src, JNK, and AKT activity. These effects were abrogated in cells with functional E-cadherin. CONCLUSIONS These data highlight laminin γ2 and its downstream effectors as potential therapeutic targets, and the value of H. pylori eradication to delay gastric cancer onset and progression.
Collapse
Affiliation(s)
- Rui M Ferreira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Joana Figueiredo
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Ines Pinto-Ribeiro
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Irene Gullo
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Pathology, Centro Hospitalar Universitário São João, Porto, Portugal
| | | | - Laura Carreto
- Department of Biology, University of Aveiro, Aveiro, Portugal.,Centre of Environmental and Marine Studies, University of Aveiro, Aveiro, Portugal
| | - Patricia Castro
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | - Manuel A Santos
- Institute of Biomedicine, University of Aveiro, Aveiro, Portugal.,Multidisciplinary Institute of Ageing, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Fatima Carneiro
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Pathology, Centro Hospitalar Universitário São João, Porto, Portugal
| | - Raquel Seruca
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Ceu Figueiredo
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Department of Pathology, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
2
|
Skipping Exon-v6 from CD44v6-Containing Isoforms Influences Chemotherapy Response and Self-Renewal Capacity of Gastric Cancer Cells. Cancers (Basel) 2020; 12:cancers12092378. [PMID: 32842638 PMCID: PMC7564355 DOI: 10.3390/cancers12092378] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/13/2020] [Accepted: 08/18/2020] [Indexed: 12/12/2022] Open
Abstract
De novo expressed CD44 isoforms containing exon-v6 are frequently associated with gastric cancer (GC) aggressiveness, and may predict chemotherapy response in vitro. Whether exon-v6 itself is responsible for conferring these properties to CD44v6-containing isoforms remains to be elucidated. CRISPR/Cas9 and Phosphorodiamidate Morpholino oligomers (PMOs) were used to induce specific exon-v6 skipping, maintaining the CD44 reading frame, in two GC cell lines endogenously expressing CD44v6. Cisplatin and 5-fluorouracil treatment response, and self-renewal ability was compared between CRISPR/Cas9-edited, CD44v6 knockdown and mock cells. We obtained homozygous genome-edited cell lines with exon-v6 deletion. Edited cells transcribed CD44v isoforms presenting in frame v5–v7 splicing, mimicking exon-v6 skipping. Results showed that removing specifically exon-v6 sensitizes cells to cisplatin and impairs cells’ self-renewal ability, similarly to CD44v6 knockdown. In parallel, we also tested a clinically feasible approach for transient exon-v6 skipping with a PMO-based strategy. We demonstrate that exon-v6 specific removal from CD44v isoforms increases cell sensitivity to cisplatin and impairs GC cells self-renewal. We trust that a PMO approach designed towards CD44v6 overexpressing GC cells may be a suitable approach to sensitize tumor cells for conventional therapy.
Collapse
|
3
|
Pereira C, Ferreira D, Mendes N, Granja PL, Almeida GM, Oliveira C. Expression of CD44v6-Containing Isoforms Influences Cisplatin Response in Gastric Cancer Cells. Cancers (Basel) 2020; 12:cancers12040858. [PMID: 32252293 PMCID: PMC7226224 DOI: 10.3390/cancers12040858] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/22/2020] [Accepted: 03/31/2020] [Indexed: 12/17/2022] Open
Abstract
CD44v6-containing isoforms are frequently de novo expressed in gastric cancer (GC). Whether CD44v6 has a central role in GC transformation and/or progression, whether it conditions response to therapy or whether it is only a bystander marker is still not known. Therefore, we aimed to clarify the role of CD44v6 in GC. We generated GC isogenic cell lines stably expressing CD44s or CD44v6 and tested them for different cancer hallmarks and response to cisplatin, and we further confirmed our findings in cells that endogenously express CD44v6. No correlation between overexpression of CD44v6 and the tested cancer hallmarks was observed, suggesting CD44v6 is not a driver of GC progression. Upon cisplatin treatment, CD44v6+ cells survive better and have lower apoptosis levels than CD44v6− cells, possibly due to concomitant activation of STAT3 and P38. In co-culture experiments, we discovered that CD44v6+ cells are involved in GC cell overgrowth after cisplatin treatment. In conclusion, we show that CD44v6 expression increases cell survival in response to cisplatin treatment in GC cells and that these cells override CD44v6-negative cells after cisplatin-treatment. This suggests that tumor expression of CD44v6-containing variants may condition the outcome of GC patients treated with chemotherapy.
Collapse
Affiliation(s)
- Carla Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (D.F.); (N.M.); (P.L.G.)
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Daniel Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (D.F.); (N.M.); (P.L.G.)
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-180 Porto, Portugal
| | - Nuno Mendes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (D.F.); (N.M.); (P.L.G.)
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
| | - Pedro L. Granja
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (D.F.); (N.M.); (P.L.G.)
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-180 Porto, Portugal
- ICBAS - Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313 Porto, Portugal
| | - Gabriela M. Almeida
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (D.F.); (N.M.); (P.L.G.)
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- FMUP - Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
- Correspondence: (G.M.A.); (C.O.); Tel.: +351-225-570-785 (C.O.)
| | - Carla Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (C.P.); (D.F.); (N.M.); (P.L.G.)
- IPATIMUP - Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal
- FMUP - Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
- Correspondence: (G.M.A.); (C.O.); Tel.: +351-225-570-785 (C.O.)
| |
Collapse
|
4
|
Rocha S, Teles SP, Azevedo M, Oliveira P, Carvalho J, Oliveira C. Gastric Cancer Extracellular Vesicles Tune the Migration and Invasion of Epithelial and Mesenchymal Cells in a Histotype-Dependent Manner. Int J Mol Sci 2019; 20:ijms20112608. [PMID: 31141946 PMCID: PMC6600627 DOI: 10.3390/ijms20112608] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 12/26/2022] Open
Abstract
Extracellular vesicles (EVs) secreted by tumor cells modulate recipient cells' behavior, but their effects in normal cells from the tumor microenvironment remain poorly known. In this study, we dissected the functional impact of gastric cancer cell-derived EVs (GC-EVs), representative of distinct GC histotypes, on the behavior of normal isogenic epithelial and mesenchymal cells. GC-EVs were isolated by differential centrifugation and characterized by transmission electron microscopy, nanoparticle tracking analysis, and imaging flow-cytometry. Epithelial and mesenchymal cells were challenged with GC-EVs and submitted to proliferation, migration, and invasion assays. Expression of epithelial and mesenchymal markers was followed by immunofluorescence and flow-cytometry. Our results indicated that GC-EVs secreted by diffuse-type cancer cells decrease the migration of recipient cells. This effect was more prominent and persistent for mesenchymal recipient cells, which also increased Fibronectin expression in response to EVs. GC-EVs secreted by cancer cells derived from tumors with an intestinal component increased invasion of recipient epithelial cells, without changes in EMT markers. In summary, this study demonstrated that GC-EVs modulate the migration and invasion of epithelial and mesenchymal cells from the tumor microenvironment, in a histotype-dependent manner, highlighting new features of intestinal and diffuse-type GC cells, which may help explaining differential metastasis patterns and aggressiveness of GC histotypes.
Collapse
Affiliation(s)
- Sara Rocha
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Ipatimup-Institute of Molecular Pathology and Immunology of University of Porto, 4200-135 Porto, Portugal.
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal.
| | - Sara Pinto Teles
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Ipatimup-Institute of Molecular Pathology and Immunology of University of Porto, 4200-135 Porto, Portugal.
| | - Mafalda Azevedo
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal.
- Program in Developmental Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Patrícia Oliveira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Ipatimup-Institute of Molecular Pathology and Immunology of University of Porto, 4200-135 Porto, Portugal.
| | - Joana Carvalho
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Ipatimup-Institute of Molecular Pathology and Immunology of University of Porto, 4200-135 Porto, Portugal.
| | - Carla Oliveira
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal.
- Ipatimup-Institute of Molecular Pathology and Immunology of University of Porto, 4200-135 Porto, Portugal.
- Department Pathology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal.
| |
Collapse
|
5
|
Tsai MH, Lin X, Shumilov A, Bernhardt K, Feederle R, Poirey R, Kopp-Schneider A, Pereira B, Almeida R, Delecluse HJ. The biological properties of different Epstein-Barr virus strains explain their association with various types of cancers. Oncotarget 2018; 8:10238-10254. [PMID: 28052012 PMCID: PMC5354655 DOI: 10.18632/oncotarget.14380] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/15/2016] [Indexed: 12/27/2022] Open
Abstract
The Epstein-Barr virus (EBV) is etiologically associated with the development of multiple types of tumors, but it is unclear whether this diversity is due to infection with different EBV strains. We report a comparative characterization of SNU719, GP202, and YCCEL1, three EBV strains that were isolated from gastric carcinomas, M81, a virus isolated in a nasopharyngeal carcinoma and several well-characterized laboratory type A strains. We found that B95-8, Akata and GP202 induced cell growth more efficiently than YCCEL1, SNU719 and M81 and this correlated positively with the expression levels of the viral BHRF1 miRNAs. In infected B cells, all strains except Akata and B95-8 induced lytic replication, a risk factor for carcinoma development, although less efficiently than M81. The panel of viruses induced tumors in immunocompromised mice with variable speed and efficacy that did not strictly mirror their in vitro characteristics, suggesting that additional parameters play an important role. We found that YCCEL1 and M81 infected primary epithelial cells, gastric carcinoma cells and gastric spheroids more efficiently than Akata or B95-8. Reciprocally, Akata and B95-8 had a stronger tropism for B cells than YCCEL1 or M81. These data suggest that different EBV strains will induce the development of lymphoid tumors with variable efficacy in immunocompromised patients and that there is a parallel between the cell tropism of the viral strains and the lineage of the tumors they induce. Thus, EBV strains can be endowed with properties that will influence their transforming abilities and the type of tumor they induce.
Collapse
Affiliation(s)
- Ming-Han Tsai
- German Cancer Research Centre (DKFZ), Unit F100, 69120 Heidelberg, Germany.,Inserm unit U1074, DKFZ, 69120 Heidelberg, Germany
| | - Xiaochen Lin
- German Cancer Research Centre (DKFZ), Unit F100, 69120 Heidelberg, Germany.,Inserm unit U1074, DKFZ, 69120 Heidelberg, Germany
| | - Anatoliy Shumilov
- German Cancer Research Centre (DKFZ), Unit F100, 69120 Heidelberg, Germany.,Inserm unit U1074, DKFZ, 69120 Heidelberg, Germany
| | - Katharina Bernhardt
- German Cancer Research Centre (DKFZ), Unit F100, 69120 Heidelberg, Germany.,Inserm unit U1074, DKFZ, 69120 Heidelberg, Germany
| | - Regina Feederle
- German Cancer Research Centre (DKFZ), Unit F100, 69120 Heidelberg, Germany.,Inserm unit U1074, DKFZ, 69120 Heidelberg, Germany.,Institute for Diabetes and Obesitas, Monoclonal Antibody Core Facility, German Research Center for Environmental Health, Helmholtz Zentrum München, 81377 Munich, Germany
| | - Remy Poirey
- German Cancer Research Centre (DKFZ), Unit F100, 69120 Heidelberg, Germany.,Inserm unit U1074, DKFZ, 69120 Heidelberg, Germany
| | | | - Bruno Pereira
- Differentiation and Cancer Group, IPATIMUP, Rua Dr Roberto Frias s/n, 4200 - 465 Porto, Portugal
| | - Raquel Almeida
- Differentiation and Cancer Group, IPATIMUP, Rua Dr Roberto Frias s/n, 4200 - 465 Porto, Portugal
| | - Henri-Jacques Delecluse
- German Cancer Research Centre (DKFZ), Unit F100, 69120 Heidelberg, Germany.,Inserm unit U1074, DKFZ, 69120 Heidelberg, Germany
| |
Collapse
|
6
|
Korsak B, Almeida GM, Rocha S, Pereira C, Mendes N, Osório H, Pereira PMR, Rodrigues JMM, Schneider RJ, Sarmento B, Tomé JPC, Oliveira C. Porphyrin modified trastuzumab improves efficacy of HER2 targeted photodynamic therapy of gastric cancer. Int J Cancer 2017; 141:1478-1489. [PMID: 28639285 DOI: 10.1002/ijc.30844] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 05/04/2017] [Accepted: 06/12/2017] [Indexed: 01/03/2023]
Abstract
Gastric cancer (GC) is the 3rd deadliest cancer worldwide, due to limited treatment options and late diagnosis. Human epidermal growth factor receptor-2 (HER2) is overexpressed in ∼20% of GC cases and anti-HER2 antibody trastuzumab in combination with conventional chemotherapy, is recognized as standard therapy for HER2-positive metastatic GC. This strategy improves GC patients' survival by 2-3 months, however its optimal results in breast cancer indicate that GC survival may be improved. A new photoimmunoconjugate was developed by conjugating a porphyrin with trastuzumab (Trast:Porph) for targeted photodynamic therapy in HER2-positive GC. Using mass spectrometry analysis, the lysine residues in the trastuzumab structure most prone for porphyrin conjugation were mapped. The in vitro data demonstrates that Trast:Porph specifically binds to HER2-positive cells, accumulates intracellularly, co-localizes with lysosomal marker LAMP1, and induces massive HER2-positive cell death upon cellular irradiation. The high selectivity and cytotoxicity of Trast:Porph based photoimmunotherapy is confirmed in vivo in comparison with trastuzumab alone, using nude mice xenografted with a HER2-positive GC cell line. In the setting of human disease, these data suggest that repetitive cycles of Trast:Porph photoimmunotherapy may be used as an improved treatment strategy in HER2-positive GC patients.
Collapse
Affiliation(s)
- Barbara Korsak
- Instituto de Investigação e Inovação em Saúde- i3S, Universidade do Porto, Portugal
- Ipatimup, Institute of Molecular Pathology and Immunology at the University of Porto, Porto, Portugal
- QOPNA and Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Gabriela M Almeida
- Instituto de Investigação e Inovação em Saúde- i3S, Universidade do Porto, Portugal
- Ipatimup, Institute of Molecular Pathology and Immunology at the University of Porto, Porto, Portugal
| | - Sara Rocha
- Instituto de Investigação e Inovação em Saúde- i3S, Universidade do Porto, Portugal
- Ipatimup, Institute of Molecular Pathology and Immunology at the University of Porto, Porto, Portugal
| | - Carla Pereira
- Instituto de Investigação e Inovação em Saúde- i3S, Universidade do Porto, Portugal
- Ipatimup, Institute of Molecular Pathology and Immunology at the University of Porto, Porto, Portugal
| | - Nuno Mendes
- Instituto de Investigação e Inovação em Saúde- i3S, Universidade do Porto, Portugal
- Ipatimup, Institute of Molecular Pathology and Immunology at the University of Porto, Porto, Portugal
| | - Hugo Osório
- Instituto de Investigação e Inovação em Saúde- i3S, Universidade do Porto, Portugal
- Ipatimup, Institute of Molecular Pathology and Immunology at the University of Porto, Porto, Portugal
- Department of Pathology and Oncology, Faculty of Medicine, University of Porto, Porto, Portugal
| | | | - João M M Rodrigues
- QOPNA and Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Rudolf J Schneider
- Department of Analytical Chemistry, Reference Materials, BAM Federal Institute for Materials Research and Testing, Berlin, Germany
| | - Bruno Sarmento
- Instituto de Investigação e Inovação em Saúde- i3S, Universidade do Porto, Portugal
- INEB, National Institute of Biomedical Engineering-University of Porto, Porto, Portugal
- Inovapotek Pharmaceutical Research and Development, Porto, Portugal
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde and Instituto Universitário de Ciências da Saúde, Gandra, Portugal
| | - João P C Tomé
- QOPNA and Department of Chemistry, University of Aveiro, Aveiro, Portugal
- Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
- Departamento de Engenharia Química, Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Carla Oliveira
- Instituto de Investigação e Inovação em Saúde- i3S, Universidade do Porto, Portugal
- Ipatimup, Institute of Molecular Pathology and Immunology at the University of Porto, Porto, Portugal
- Department of Pathology and Oncology, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
7
|
Dies1/VISTA expression loss is a recurrent event in gastric cancer due to epigenetic regulation. Sci Rep 2016; 6:34860. [PMID: 27721458 PMCID: PMC5056517 DOI: 10.1038/srep34860] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 09/16/2016] [Indexed: 12/18/2022] Open
Abstract
Dies1/VISTA induces embryonic stem-cell differentiation, via BMP-pathway, but also acts as inflammation regulator and immune-response modulator. Dies1 inhibition in a melanoma-mouse model led to increased tumour-infiltrating T-cells and decreased tumour growth, emphasizing Dies1 relevance in tumour-microenvironment. Dies1 is involved in cell de/differentiation, inflammation and cancer processes, which mimic those associated with Epithelial-to-Mesenchymal-Transition (EMT). Despite this axis linking Dies1 with EMT and cancer, its expression, modulation and relevance in these contexts is unknown. To address this, we analysed Dies1 expression, its regulation by promoter-methylation and miR-125a-5p overexpression, and its association with BMP-pathway downstream-effectors, in a TGFβ1-induced EMT-model, cancer cell-lines and primary samples. We detected promoter-methylation as a mechanism controlling Dies1 expression in our EMT-model and in several cancer cell-lines. We showed that the relationship between Dies1 expression and BMP-pathway effectors observed in the EMT-model, was not present in all cell-lines, suggesting that Dies1 has other cell-specific effectors, beyond the BMP-pathway. We further demonstrated that: Dies1 expression loss is a recurrent event in GC, caused by promoter methylation and/or miR-125a-5p overexpression and; GC-microenvironment myofibroblasts overexpress Dies1. Our findings highlight Dies1 as a novel player in GC, with distinct roles within tumour cells and in the tumour-microenvironment.
Collapse
|
8
|
Costa AM, Ferreira RM, Pinto-Ribeiro I, Sougleri IS, Oliveira MJ, Carreto L, Santos MA, Sgouras DN, Carneiro F, Leite M, Figueiredo C. HelicobacterpyloriActivates Matrix Metalloproteinase 10 in Gastric Epithelial Cells via EGFR and ERK-mediated Pathways. J Infect Dis 2016; 213:1767-1776. [DOI: 10.1093/infdis/jiw031] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
9
|
Sadio A, Gustafsson JK, Pereira B, Gomes CP, Hansson GC, David L, Pêgo AP, Almeida R. Modified-chitosan/siRNA nanoparticles downregulate cellular CDX2 expression and cross the gastric mucus barrier. PLoS One 2014; 9:e99449. [PMID: 24925340 PMCID: PMC4055692 DOI: 10.1371/journal.pone.0099449] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 05/15/2014] [Indexed: 11/18/2022] Open
Abstract
Development of effective non-viral vectors is of crucial importance in the implementation of RNA interference in clinical routine. The localized delivery of siRNAs to the gastrointestinal mucosa is highly desired but faces specific problems such as the stability in gastric acidity conditions and the presence of the mucus barrier. CDX2 is a transcription factor critical for intestinal differentiation being involved in the initiation and maintenance of gastrointestinal diseases. Specifically, it is the trigger of gastric intestinal metaplasia which is a precursor lesion of gastric cancer. Its expression is also altered in colorectal cancer, where it may constitute a lineage-survival oncogene. Our main objective was to develop a nanoparticle-delivery system of siRNA targeting CDX2 using modified chitosan as a vector. CDX2 expression was assessed in gastric carcinoma cell lines and nanoparticles behaviour in gastrointestinal mucus was tested in mouse explants. We show that imidazole-modified chitosan and trimethylchitosan/siRNA nanoparticles are able to downregulate CDX2 expression and overpass the gastric mucus layer but not colonic mucus. This system might constitute a potential therapeutic approach to treat CDX2-dependent gastric lesions.
Collapse
Affiliation(s)
- Ana Sadio
- IPATIMUP- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Gastroenterology Department, Unidade Local Saúde da Guarda, Guarda, Portugal
- Gulbenkian Programme for Advanced Medical Education, Lisboa, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Jenny K. Gustafsson
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Bruno Pereira
- IPATIMUP- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Carla Pereira Gomes
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
| | - Gunnar C. Hansson
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Leonor David
- IPATIMUP- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - Ana Paula Pêgo
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Raquel Almeida
- IPATIMUP- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- * E-mail:
| |
Collapse
|
10
|
Carvalho J, van Grieken NC, Pereira PM, Sousa S, Tijssen M, Buffart TE, Diosdado B, Grabsch H, Santos MAS, Meijer G, Seruca R, Carvalho B, Oliveira C. Lack of microRNA-101 causes E-cadherin functional deregulation through EZH2 up-regulation in intestinal gastric cancer. J Pathol 2012; 228:31-44. [PMID: 22450781 DOI: 10.1002/path.4032] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 03/10/2012] [Accepted: 03/20/2012] [Indexed: 12/21/2022]
Abstract
E-cadherin expression disruption is commonly observed in metastatic epithelial cancers and is a crucial step in gastric cancer (GC) initiation and progression. As aberrant expression of microRNAs often perturb the normal expression/function of pivotal cancer-related genes, we characterized and dissected a pathway that causes E-cadherin dysfunction via loss of microRNA-101 and up-regulation of EZH2 expression in GC. MicroRNA microarray expression profiling and array-CGH were used to reinforce miR-101 involvement in GC. By using quantitative real-time PCR and quantitative SNaPshot genomic PCR, we confirmed that miR-101 was significantly down-regulated in GC (p < 0.0089) in comparison with normal gastric mucosas and, at least in 65% of the GC cases analysed, this down-regulation was caused by deletions and/or microdeletions at miR-101 genomic loci. Moreover, around 40% of cases showing miR-101 down-regulation displayed concomitant EZH2 over-expression (at the RNA and protein levels), which, in turn, was associated with loss/aberrant expression of E-cadherin. Interestingly, this occurred preferentially in intestinal-type GCs, retaining allele(s) untargeted by classical CDH1-inactivating mechanisms. We also demonstrated that miR-101 gain of function or direct inhibition of EZH2 in Kato III GC cells led to a strong depletion of endogenous EZH2 and consequent rescue of E-cadherin membranous localization, mimicking results obtained in clinical GC samples. In conclusion, we show that deletions and/or microdeletions at both miR-101 genomic loci cause mature miR-101 down-regulation, subsequent EZH2 over-expression and E-cadherin dysfunction, specifically in intestinal-type GC.
Collapse
Affiliation(s)
- Joana Carvalho
- Institute of Molecular Pathology and Immunology, University of Porto (IPATIMUP), Portugal; Faculty of Medicine, University of Porto, Portugal
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Padró M, Cobler L, Garrido M, de Bolós C. Down-regulation of FUT3 and FUT5 by shRNA alters Lewis antigens expression and reduces the adhesion capacities of gastric cancer cells. Biochim Biophys Acta Gen Subj 2011; 1810:1141-9. [PMID: 21978830 DOI: 10.1016/j.bbagen.2011.09.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 09/16/2011] [Accepted: 09/19/2011] [Indexed: 01/02/2023]
Abstract
BACKGROUND Lewis antigens are fucosylated glycoconjugates involved in the development of several pathologies. The adhesion of sialyl-Lewis antigens to E-selectin is a key step in the development of metastasis and the glycosidic component of CD44 plays a key role in the binding to hyaluronic acid, a component of the extracellular matrix associated to tumor development and invasion. Fucosyltransferases are enzymes that add fucose to precursor glycan structures: FUT3 and FUT5 catalyze the addition of fucose to the α1-3,4 position and are detected in epithelial cells. In this study, we have analyzed the effects of silencing FUT3, FUT5 or FUT3/FUT5, in two gastric cancer cell lines, in the expression of Lewis antigens and in the adhesive and migratory capacities of the cells. METHODS FUT3, FUT5 and FUT3/FUT5 were down-regulated using lentiviral delivery of shRNAs in MKN45 and GP220 gastric cancer cells. RESULTS In the infected cells, decreased levels of FUT3 and FUT5 mRNA detected by quantitative RT-PCR; and lower levels of sialyl-Lewis antigens, evaluated by flow cytometry, were observed. The adhesion to endothelial cells trough the binding to E-selectin, and the binding to hyaluronic acid were reduced in the shFUT3, shFUT5 and shFUT3/FUT5, whereas the levels of CD44, analyzed by western blot, did not change. GENERAL SIGNIFICANCE The down-regulation of FUT3, FUT5 and FUT3/FUT5 reduces the expression of sialyl-Lewis antigens and the adhesion and binding capacities of gastric cancer cells; and allows to identify the specific α1-3,4 fucosyltransferases implicated in the Lewis antigens synthesis in this cellular model.
Collapse
Affiliation(s)
- Mercè Padró
- Programa de Recerca en Càncer, IMIM-Hospital del Mar, Parc de Recerca Biomèdica de Barcelona, Spain
| | | | | | | |
Collapse
|
12
|
Mejías-Luque R, Lindén SK, Garrido M, Tye H, Najdovska M, Jenkins BJ, Iglesias M, Ernst M, de Bolós C. Inflammation modulates the expression of the intestinal mucins MUC2 and MUC4 in gastric tumors. Oncogene 2010; 29:1753-62. [PMID: 20062084 DOI: 10.1038/onc.2009.467] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Infection of gastric mucosa by Helicobacter pylori induces an inflammatory response with increased levels of proinflammatory cytokines. Among them, tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta and IL-6 induce the activation of signaling pathways that regulate genes expression, such as MUC2 and MUC4 intestinal mucins ectopically detected in gastric tumors. This study evaluated if the predominant inflammatory cell type correlates with MUC2 and MUC4 expression in human intestinal gastric tumors (n=78). In addition, we analyzed the regulatory effects of the associated inflammatory signaling pathways on their expression in gastric cancer cell lines, and in a mouse model with hyperactivated STAT3 signaling pathway. Tumors with predominant lymphoplasmocytic infiltrate (chronic inflammation), presented higher levels of MUC2 and were more differentiated than tumors with predominant polymorphonuclear infiltrate (acute inflammation). These differences can be attributed to specific cytokines, because TNF-alpha and IL-1beta induced MUC2 but no MUC4 expression in gastric cancer cell lines. The two groups of tumors expressed similar levels of MUC4 that correlated with the expression of STAT3 transcription factor, implicated in the activation of genes through the IL-6 pathway. In gastric tissues from gp130(+/+), gp130(Y757F/Y757F) and gp130(Y757F/Y757F) Stat3(-/+) mice, Muc2 was not detected, whereas Muc4 was found in the gastric tumors developed in the gp130(Y757F/Y757F) mice, with hyperactivated STAT3. These data indicate that the signaling pathways associated with the inflammatory response can modulate the expression of MUC2 and MUC4 intestinal mucin genes, in human and mouse gastric tumors.
Collapse
Affiliation(s)
- R Mejías-Luque
- IMIM-Hospital del Mar, Programa de Recerca en Càncer, Parc de Recerca Biomèdica de Barcelona, Barcelona 08003, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Carvalho AS, Harduin-Lepers A, Magalhães A, Machado E, Mendes N, Costa LT, Matthiesen R, Almeida R, Costa J, Reis CA. Differential expression of alpha-2,3-sialyltransferases and alpha-1,3/4-fucosyltransferases regulates the levels of sialyl Lewis a and sialyl Lewis x in gastrointestinal carcinoma cells. Int J Biochem Cell Biol 2009; 42:80-9. [PMID: 19781661 DOI: 10.1016/j.biocel.2009.09.010] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 09/16/2009] [Accepted: 09/17/2009] [Indexed: 11/26/2022]
Abstract
Sialyl Lewis x and sialyl Lewis a expression depends on sialyltransferases and fucosyltransferases. In this study, we screened for major variations of sialyltransferases and fucosyltransferases involved in the synthesis and regulation of sialyl Lewis x and sialyl Lewis a epitopes in gastrointestinal carcinoma cells. Our results show that expression of ST3Gal IV in several gastrointestinal cell lines is correlated with the expression of sialyl Lewis x at the cell surface. ST3Gal IV overexpressed in the gastric MKN45 cell line, showed exclusive enzymatic activity towards glycoproteins containing terminal Galbeta1-4GlcNAc structure. On the other hand, when ST3Gal III was overexpressed in MKN45, an increase in the expression levels of both sialyl Lewis epitopes was observed. ST3Gal III and ST3Gal IV lead to de novo synthesis of sialyl Lewis x determinant on different molecular weight glycoproteins of MKN45 cells suggesting that each enzyme used different substrates within the available glycoproteome. The final glycosylation step in sialyl Lewis x and sialyl Lewis a biosynthesis in MKN45 cell line was shown to be associated to FUT5, which efficiently fucosylated sialyl Lewis precursors on glycoproteins. Moreover we demonstrate that the expression of sialyl Lewis epitopes in the MKN45 was induced by cell confluence, which can be regarded as a model to study altered glycosylation during tumour progression. This increase was observed together with an increase in mRNA levels of ST3GAL3, FUT5 and FUT6, and a decrease in FUT4 transcript levels in MKN45 confluent cells, suggesting a possible control at the transcriptional level.
Collapse
Affiliation(s)
- A S Carvalho
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Buffart TE, Tijssen M, El-Bchiri J, Duval A, van de Wiel MA, Ylstra B, Meijer GA, Carvalho B. NMD inhibition fails to identify tumour suppressor genes in microsatellite stable gastric cancer cell lines. BMC Med Genomics 2009; 2:39. [PMID: 19563644 PMCID: PMC2709900 DOI: 10.1186/1755-8794-2-39] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Accepted: 06/29/2009] [Indexed: 12/05/2022] Open
Abstract
Background Gastric cancers frequently show chromosomal alterations which can cause activation of oncogenes, and/or inactivation of tumour suppressor genes. In gastric cancer several chromosomal regions are described to be frequently lost, but for most of the regions, no tumour suppressor genes have been identified yet. The present study aimed to identify tumour suppressor genes inactivated by nonsense mutation and deletion in gastric cancer by means of GINI (gene identification by nonsense mediated decay inhibition) and whole genome copy number analysis. Methods Two non-commercial gastric cancer cell lines, GP202 and IPA220, were transfected with siRNA directed against UPF1, to specifically inhibit the nonsense mediated decay (NMD) pathway, and with siRNA directed against non-specific siRNA duplexes (CVII) as a control. Microarray expression experiments were performed in triplicate on 4 × 44 K Agilent arrays by hybridizing RNA from UPF1-transfected cells against non-specific CVII-transfected cells. In addition, array CGH of the two cell lines was performed on 4 × 44K agilent arrays to obtain the DNA copy number profiles. Mutation analysis of GINI candidates was performed by sequencing. Results UPF1 expression was reduced for >70% and >80% in the GP202 and IPA220 gastric cancer cell lines, respectively. Integration of array CGH and microarray expression data provided a list of 134 and 50 candidate genes inactivated by nonsense mutation and deletion for GP202 and IPA220, respectively. We selected 12 candidate genes for mutation analysis. Of these, sequence analysis was performed on 11 genes. One gene, PLA2G4A, showed a silent mutation, and in two genes, CTSA and PTPRJ, missense mutations were detected. No nonsense mutations were detected in any of the 11 genes tested. Conclusion Although UPF1 was substantially repressed, thus resulting in the inhibition of the NMD system, we did not find genes inactivated by nonsense mutations. Our results show that the GINI strategy leads to a high number of false positives.
Collapse
Affiliation(s)
- Tineke E Buffart
- Dept Pathology, VU University Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Abstract
T-lymphocyte maturation associated protein, MAL, has been described as a tumour-suppressor gene with diagnostic value in colorectal and oesophageal cancers, and can be inactivated by promoter hypermethylation. The aim of this study was to analyse the prevalence of MAL promoter hypermethylation and the association with mRNA expression in gastric cancers and to correlate methylation status to clinicopathological data. Bisulphite-treated DNA isolated from formalin-fixed and paraffin-embedded samples of 202 gastric adenocarcinomas and 22 normal gastric mucosae was subjected to real-time methylation-specific PCR (Q-MSP). Two regions within the MAL promoter (M1 and M2) were analysed. In addition, 17 frozen gastric carcinomas and two gastric cancer cell lines were analysed both by Q-MSP and real-time RT–PCR. Methylation of M1 and M2 occurred in 71 and 80% of the gastric cancers, respectively, but not in normal gastric mucosa tissue. Hypermethylation of M2, but not M1, correlated with significantly better disease-free survival in a univariate (P=0.03) and multivariate analysis (P=0.03) and with downregulation of expression (P=0.01). These results indicate that MAL has a putative tumour-suppressor gene function in gastric cancer, and detection of promoter hypermethylation may be useful as a prognostic marker.
Collapse
|
16
|
IL-6 induces MUC4 expression through gp130/STAT3 pathway in gastric cancer cell lines. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:1728-36. [DOI: 10.1016/j.bbamcr.2008.05.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2007] [Revised: 04/25/2008] [Accepted: 05/16/2008] [Indexed: 02/04/2023]
|
17
|
Barros R, Pereira B, Duluc I, Azevedo M, Mendes N, Camilo V, Jacobs RJ, Paulo P, Santos-Silva F, van Seuningen I, van den Brink GR, David L, Freund JN, Almeida R. Key elements of the BMP/SMAD pathway co-localize with CDX2 in intestinal metaplasia and regulate CDX2 expression in human gastric cell lines. J Pathol 2008; 215:411-20. [PMID: 18498120 DOI: 10.1002/path.2369] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Helicobacter pylori infection induces intestinal metaplasia of the stomach, a preneoplastic lesion associated with an increased risk for gastric cancer development. Intestinal metaplasia is induced by the intestine-specific transcription factor CDX2 but the mechanisms responsible for this ectopic expression have never been described. We hypothesized that the BMP/SMAD pathway has a role in CDX2 regulation, in this context, for the following reasons: (1) the BMP pathway is crucial for normal intestinal differentiation and (2) there is an influx of BMP2 and BMP4-producing cells to the stomach upon Helicobacter pylori infection. We evaluated the expression of key elements of the BMP pathway in human stomach specimens with IM. Growth factor treatments, with BMP2 and BMP4, were performed in cultured cells and a knock-down experiment of SMAD4 was done using RNAi. We showed overexpression in IM of BMP2/4, BMPR1A, and SMAD4 in 56% of IM foci, and pSMAD1/5/8 in 100% of IM foci as compared to adjacent mucosa. In vitro, treatment of AGS cells with BMP2 and BMP4 increased endogenous CDX2 expression as well as the intestinal differentiation markers MUC2 and LI-cadherin. On the other hand, SMAD4 knock-down led to decreased endogenous CDX2, MUC2, and LI-cadherin in AGS. Treatment of the SMAD4 knock-down cells had no influence on CDX2 expression as opposed to wild-type cells. A 9.3 kb CDX2 promoter could be transactivated by SMAD4 and SMAD1 in a cell-dependent manner. In conclusion, we identified for the first time that the BMP pathway is active in intestinal metaplasia and that BMP2 and BMP4 regulate CDX2 expression and promote intestinal differentiation through the canonical signal transducers.
Collapse
Affiliation(s)
- R Barros
- Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Costa NR, Mendes N, Marcos NT, Reis CA, Caffrey T, Hollingsworth MA, Santos-Silva F. Relevance of MUC1 mucin variable number of tandem repeats polymorphism in H pylori adhesion to gastric epithelial cells. World J Gastroenterol 2008; 14:1411-4. [PMID: 18322957 PMCID: PMC2693691 DOI: 10.3748/wjg.14.1411] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [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 evaluate the influence of MUC1 mucin variable number of tandem repeats (VNTR) variability on H pylori adhesion to gastric cells.
METHODS: Enzyme linked immunosorbent assay (ELISA)-based adhesion assays were performed to measure the adhesion of different H pylori strains (HP26695 and HPTx30a) to gastric carcinoma cell lines (GP202 and MKN45) and GP202 clones expressing recombinant MUC1 with different VNTR lengths.
RESULTS: Evaluation of adhesion results shows that H pylori pathogenic strain HP26695 has a significantly higher (P < 0.05) adhesion to all the cell lines and clones tested, when compared to the non-pathogenic strain HPTx30a. Bacteria showed a significantly higher (P < 0.05) adhesion to the GP202 cell line, when compared to the MKN45 cell line. Furthermore, both strains showed a significantly higher (P < 0.05) adhesion to GP202 clones with larger MUC1 VNTR domains.
CONCLUSION: This work shows that MUC1 mucin variability conditions H pylori binding to gastric cells. The extent of bacterial adhesion depends on the size of the MUC1 VNTR domain. The adhesion is further dependent on bacterial pathogenicity and the gastric cell line. MUC1 mucin variability may contribute to determine H pylori colonization of the gastric mucosa.
Collapse
|
19
|
Serpa J, Mesquita P, Mendes N, Oliveira C, Almeida R, Santos-Silva F, Reis CA, LePendu J, David L. Expression of Lea in gastric cancer cell lines depends on FUT3 expression regulated by promoter methylation. Cancer Lett 2006; 242:191-7. [PMID: 16427187 DOI: 10.1016/j.canlet.2005.11.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2005] [Revised: 10/28/2005] [Accepted: 11/04/2005] [Indexed: 02/05/2023]
Abstract
Aberrant expression of Lewis antigens has been demonstrated in gastric lesions, namely gastritis, intestinal metaplasia (IM) and gastric carcinoma (GC), and can be partly due to overexpression of the Lewis (FUT3) enzyme. Our aim was to evaluate the role of promoter methylation in FUT3 and Le(a) expression in gastric carcinoma cell lines. MKN45 cell line showed low amounts of Le(a), in the absence of FUT3; GP220 expressed high levels of Le(a) and FUT3. After 5aza-2'deoxycytidine MKN45 showed increased levels of FUT3 and Le(a), by immunohistochemistry and Real-Time PCR, whereas GP220 showed an increase in FUT3 without increase of Le(a). Enzyme activity assays confirmed an increase in alpha-1,4 fucosyltransferase activity in both cell lines by 5aza-2'deoxycytidine. Luciferase reporter gene assays, using methylated and unmethylated deletion constructs of FUT3 promoter, showed that FUT3 expression is regulated by methylation. Summing up, we showed that FUT3 overexpression in gastric cells depends upon promoter hypomethylation and that FUT3 is responsible for overexpression of Le(a) in gastric cells, in vitro. FUT3, Lea, Methylation.
Collapse
Affiliation(s)
- Jacinta Serpa
- Institute of Molecular Pathology and Immunology of the University of Porto-IPATIMUP, Porto, Portugal
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Ferreira B, Marcos NT, David L, Nakayama J, Reis CA. Terminal α1,4-linked N-acetylglucosamine in Helicobacter pylori-associated Intestinal Metaplasia of the Human Stomach and Gastric Carcinoma Cell Lines. J Histochem Cytochem 2006; 54:585-91. [PMID: 16618943 DOI: 10.1369/jhc.5a6836.2006] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori (Hp) infection is associated with the development of gastric lesions including gastritis, intestinal metaplasia (IM), and gastric carcinoma. In humans, Hp is found almost exclusively in the foveolar epithelium of the gastric mucosa and rarely colonizes the deeper portions where mucous cells of the glands produce mucins with terminal α1, 4-GlcNAc O-glycans. This structure exerts antimicrobial activity against Hp. The development of IM in the stomach is characterized by Hp clearance from the metaplastic glands and by major alterations in the expression of mucins and mucin-carbohydrates. The present work evaluated whether terminal α1,4-GlcNAc and sialyl-Tn antigen are implicated in the process of Hp clearance from metaplastic glands by analyzing the expression of these antigens in different types of IM—complete ( n=12) and incomplete ( n=8)—and in gastric cell lines. Terminal α1,4-GlcNAc was not detected in IM except in a single foci of one case, indicating that this structure is not implicated in the clearance of Hp from IM, in contrast to what is observed in normal gastric mucosa. None of the gastric carcinoma cell lines studied showed terminal α1,4-GlcNAc, suggesting that they do not display a gastric gland mucous cell phenotype and therefore are useful models for in vitro Hp studies. Finally, sialyl-Tn antigen colocalizes with MUC2 mucin and is present in all cases of complete and incomplete IM, suggesting that either or both can be implicated in Hp clearance from IM. (J Histochem Cytochem 54:585-591, 2006)
Collapse
Affiliation(s)
- Bibiana Ferreira
- Institute of Molecular Pathology and Immunology, University of Porto, Portugal
| | | | | | | | | |
Collapse
|
21
|
Milne ANA, Sitarz R, Carvalho R, Polak MM, Ligtenberg M, Pauwels P, Offerhaus GJA, Weterman MAJ. Promoter hypermethylation and silencing of CHFR mitotic stress checkpoint gene in human gastric cancers. Oncol Rep 2005; 38:903-13. [PMID: 17376510 DOI: 10.1016/j.humpath.2006.12.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Revised: 12/06/2006] [Accepted: 12/08/2006] [Indexed: 02/05/2023] Open
Abstract
CHFR is a recently identified mitotic stress check-point gene. CHFR is ubiquitously expressed in normal human tissues, whereas loss of CHFR expression has been observed in human tumors. Silencing of CHFR has been associated with aberrant promoter methylation and histone deacetylation in several cancer types. In this study, we investigated epigenetic CHFR inactivation in human gastric cancers by examining CHFR expression and methylation status in gastric cancer cell lines with RT-PCR analysis, bisulfite PCR and sequencing. A series of primary gastric tumors were also analyzed for CHFR methylation. Eight of 12 (66.7%) gastric cancer cell lines and 19/43 (44.2%) primary gastric tumors showed CHFR methylation. In addition, CpG methylation status correlated well with CHFR expression in the human gastric cancer cell lines, in which treatment with 5-aza-dC resulted in de novo or enhanced expression of CHFR. Combination treatment of 5-aza-dC with trichostatin A showed a synergistic effect on CHFR expression in some cases. Our results indicate that aberrant promoter methylation of the CHFR gene was observed in a significant proportion of human gastric cancers and was responsible for the inactivation of the CHFR gene in gastric cancers.
Collapse
Affiliation(s)
- Anya N A Milne
- Department of Pathology, University Medical Centre, 3508 GA Utrecht, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Santos-Silva F, Fonseca A, Caffrey T, Carvalho F, Mesquita P, Reis C, Almeida R, David L, Hollingsworth MA. Thomsen-Friedenreich antigen expression in gastric carcinomas is associated with MUC1 mucin VNTR polymorphism. Glycobiology 2004; 15:511-7. [PMID: 15604091 DOI: 10.1093/glycob/cwi027] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Aberrant glycosylation of mucins is a common phenomenon associated with oncogenic transformation. We investigated the association between expression of the tumor-associated antigens T, Tn, and sialyl-Tn and polymorphism in the length of the MUC1 mucin tandem repeat in a series of gastric carcinomas. We further evaluated the relevance of MUC1 tandem repeat length on the expression of these tumor-associated carbohydrate antigens (TACAs) using a gastric carcinoma cell line model expressing recombinant MUC1 constructs carrying 0, 3, 9, and 42 repeats. Gastric carcinomas showed a high prevalence of Tn and sialyl-Tn antigens, whereas T antigen was less frequently expressed. The expression of T antigen was significantly higher in gastric carcinomas from patients homozygous for MUC1 large tandem repeat alleles. No significant associations were found for Tn and sialyl-Tn antigens. This novel association was reinforced by the gastric carcinoma cell line model experiments, where de novo expression of T antigen was detected in clones transfected with larger VNTR regions. Our results indicate that polymorphism in the MUC1 tandem repeat influences the expression of TACAs in gastric cancer cells and may therefore allow the identification of subgroups of patients that develop more aggressive tumors expressing T antigen.
Collapse
Affiliation(s)
- F Santos-Silva
- Institute of Molecular Pathology and Immunology of the University of Porto, IPATIMUP, 4200-465 Porto, Portugal.
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Takemura S, Yashiro M, Sunami T, Tendo M, Hirakawa K. Novel models for human scirrhous gastric carcinoma in vivo. Cancer Sci 2004; 95:893-900. [PMID: 15546507 PMCID: PMC11159367 DOI: 10.1111/j.1349-7006.2004.tb02199.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2004] [Revised: 09/24/2004] [Accepted: 09/27/2004] [Indexed: 11/27/2022] Open
Abstract
Human scirrhous gastric carcinoma, a diffusely infiltrating type of poorly differentiated gastric carcinoma also known as linitis plastica type carcinoma, is characterized by cancer cell infiltration and proliferation accompanied with extensive stromal fibrosis. We established two new gastric cancer cell lines, designated OUCM-8 and OCUM-11, which developed the characteristic biology of scirrhous gastric carcinoma upon orthotopic implantation in mice. Involvement of lymph nodes and liver metastasis was also found in both orthotopic models. Histologically, these orthotopic models showed proliferation with extensive fibrosis, resembling human scirrhous gastric cancer. Both cell lines were derived from ascites of patients with scirrhous gastric cancer. The growth of OCUM-8 and OCUM-11 cells following the addition of KGF, FGF, and EGF was increased significantly relative to untreated cells. An increase in the number of attached and spreading cells occurred following the addition of TGF-beta 1 in both cell lines. OCUM-11 cells showed microsatellite instability. Although subcutaneous scirrhous gastric cancer cells show medullary growth, most in vivo studies of scirrhous gastric cancer have used xenografted tumors implanted subcutaneously. Only in a few cases was it confirmed that these scirrhous gastric cancer cell lines retained the original histologic characteristics. Our orthotopic models should contribute to the elucidation of disease progression in situ and to the development of therapy for scirrhous gastric cancer.
Collapse
Affiliation(s)
- Satoru Takemura
- The Department of Surgical Oncology, Osaka City University Graduate School of Medicine, Osaka 545-8585
| | | | | | | | | |
Collapse
|
24
|
Shen L, Li HX, Luo HS, Shen ZX, Tan SY, Guo J, Sun J. CDw75 is a significant histopathological marker for gastric carcinoma. World J Gastroenterol 2004; 10:1682-5. [PMID: 15162552 PMCID: PMC4572781 DOI: 10.3748/wjg.v10.i11.1682] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM: To study the expression of CDw75 in patients with gastric carcinoma and to correlate CDw75 expression with progression of the tumor.
METHODS: Immunohistochemical method was used to examine the expression of CDw75 in 72 cases of the gastric carcinoma and adjacent normal gastric mucosa, and the percentage of the cells positively stained with CDw75 was calculated using a computer-aided microscopic image analysis system.
RESULTS: CDw75 was not expressed in normal gastric mucosa but detected in 37 of the 72 neoplastic gastric lesions. The expression of CDw75 was associated with the tumor progression as indicated by its close correlation with the depth of the tumor infiltration (χ2 = 18.415, P < 0.01), TNM stage (χ2 = 10.419, P < 0.05) and lymph node metastasis (χ2 = 6.675, P < 0.01). The overall survival rate of the patients with positive CDw75 expression (32.4%) was significantly lower than that of the patients without CDw75 expression (71.4%) (P < 0.01). There was no significant correlation between the expression of CDw75 and the sex and age and histological type of patients (P > 0.05).
CONCLUSION: These findings suggest that the expression of CDw75 is a significant histopathological marker for more advanced stage of gastric carcinoma and indicates a poor prognosis for the patients.
Collapse
Affiliation(s)
- Lei Shen
- Department of Gastroenterology, Renmin Hospital, Wuhan University, 238 Jie-fang Road, Wuhan 430060, Hubei Province, China.
| | | | | | | | | | | | | |
Collapse
|
25
|
Mesquita P, Jonckheere N, Almeida R, Ducourouble MP, Serpa J, Silva E, Pigny P, Silva FS, Reis C, Silberg D, Van Seuningen I, David L. Human MUC2 mucin gene is transcriptionally regulated by Cdx homeodomain proteins in gastrointestinal carcinoma cell lines. J Biol Chem 2003; 278:51549-56. [PMID: 14525978 DOI: 10.1074/jbc.m309019200] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In intestinal metaplasia and 30% of gastric carcinomas, MUC2 intestinal mucin and the intestine-specific transcription factors Cdx-1 and Cdx-2 are aberrantly expressed. The involvement of Cdx-1 and Cdx-2 in the intestinal development and their role in transcription of several intestinal genes support the hypothesis that Cdx-1 and/or Cdx-2 play important roles in the aberrant intestinal differentiation program of intestinal metaplasia and gastric carcinoma. To clarify the mechanisms of transcriptional regulation of the MUC2 mucin gene in gastric cells, pGL3 deletion constructs covering 2.6 kb of the human MUC2 promoter were used in transient transfection assays, enabling us to identify a relevant region for MUC2 transcription in all gastric cell lines. To evaluate the role of Cdx-1 and Cdx-2 in MUC2 transcription we performed co-transfection experiments with expression vectors encoding Cdx-1 and Cdx-2. In two of the four gastric carcinoma cell lines and in all colon carcinoma cell lines we observed transactivation of the MUC2 promoter by Cdx-2. Using gel shift assays we identified two Cdx-2 binding sites at -177/-171 and -191/-187. Only simultaneous mutation of the two sites resulted in inhibition of Cdx-2-mediated transactivation of MUC2 promoter, implying that both Cdx-2 sites are active. Finally, stable expression of Cdx-2 in a gastric cell line initially not expressing Cdx-2, led to induction of MUC2 expression. In conclusion, this work demonstrates that Cdx-2 activates the expression of MUC2 mucin gene in gastric cells, inducing an intestinal transdifferentiation phenotype that parallels what is observed both in intestinal metaplasia and some gastric carcinomas.
Collapse
Affiliation(s)
- Patrícia Mesquita
- Institute of Molecular Pathology and Immunology (IPATIMUP), University of Porto, Rua Dr. Roberto Frias s/n, 4200 Porto, Portugal.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Marcos NT, Cruz A, Silva F, Almeida R, David L, Mandel U, Clausen H, Von Mensdorff-Pouilly S, Reis CA. Polypeptide GalNAc-transferases, ST6GalNAc-transferase I, and ST3Gal-transferase I expression in gastric carcinoma cell lines. J Histochem Cytochem 2003; 51:761-71. [PMID: 12754287 DOI: 10.1177/002215540305100607] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Mucin O-glycosylation in cancer is characterized by aberrant expression of immature carbohydrate structures leading to exposure of simple mucin-type carbohydrate antigens and peptide epitopes. Glycosyltransferases controlling the initial steps of mucin O-glycosylation are responsible for the altered glycosylation observed in cancer. We studied the expression in gastric cell lines of six UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferases (GalNAc-T1, T2, T3, T4, T6, T11) that catalyze the initial key step in the regulation of mucin O-glycosylation, the transfer of GalNAc from UDP-GalNAc to serine and threonine residues. We also studied the expression of ST6GalNAc-I, the enzyme responsible for the synthesis of Sialyl-Tn antigen (NeuAcalpha2,6GalNAc) and the ST3Gal-I, the enzyme responsible for the synthesis of Sialyl-T antigen (NeuAcalpha2,3Galbeta1,3GalNAc). This study was done using specific monoclonal antibodies, enzymatic assays, and RT-PCR. Our results showed that GalNAc-T1, -T2, and -T3 have an ubiquitous expression in all gastric cell lines, whereas GalNAc-T4, -T6, and -T11 show a restricted expression pattern. The immunoreactivity with MAb VU-2-G7 suggests that, apart from GalNAc-T4, another GalNAc transferase is involved in the glycosylation of the Thr in the PDTR region of the MUC1 tandem repeat. The expression of ST3Gal-I correlates with the expression of the Sialyl-T antigen in gastric cell lines and in the control cell lines studied. The expression of ST6GalNAc-I is low in gastric cell lines, in accordance with the low/absent expression of the Sialyl-Tn antigen.
Collapse
Affiliation(s)
- Nuno T Marcos
- Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Carvalho B, van der Veen A, Gärtner F, Carneiro F, Seruca R, Buys CH, Kok K. Allelic gains and losses in distinct regions of chromosome 6 in gastric carcinoma. CANCER GENETICS AND CYTOGENETICS 2001; 131:54-9. [PMID: 11734319 DOI: 10.1016/s0165-4608(01)00514-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In gastric cancer, alterations in the long arm of chromosome 6 are a frequent event. Two regions of heterozygous loss have been described: 6q16.3-6q23 and 6q26-6q27. We have evaluated by microsatellite and FISH analyses the 6q status of three cell lines that we established from primary gastric carcinomas xenografted in nude mice, in order to elucidate the underlying mechanisms of 6q alterations. Alterations of the long arm of chromosome 6 were found in all three xenografts and corresponding cell lines. Allelic imbalance (AI) was found in the three cases, by microsatellite analysis. Fluorescence in situ hybridization analyses clearly demonstrated a duplication of the larger part of the 6q arm in all three cell lines. Two of the cell lines and the corresponding xenografts showed, in addition, complete loss of one of the parental alleles at the terminal part of 6q. Thus, the AI observed along the long arm of chromosome 6 is represented by gain of alleles in one distinct chromosomal segment and loss of alleles in another distinct segment.
Collapse
Affiliation(s)
- B Carvalho
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto, IPATIMUP, Porto, Portugal
| | | | | | | | | | | | | |
Collapse
|
28
|
Beck S, Sommer P, dos Santos Silva E, Blin N, Gött P. Hepatocyte nuclear factor 3 (winged helix domain) activates trefoil factor gene TFF1 through a binding motif adjacent to the TATAA box. DNA Cell Biol 1999; 18:157-64. [PMID: 10073575 DOI: 10.1089/104454999315547] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The winged helix transcription factors HNF-3/FKH (forkhead homologs) activate endodermal-derived and acute-phase gene expression and control gut development in Drosophila. Trefoil factor family (TFFs) peptides are vertebrate products secreted by mucin-producing epithelial cells of the gastrointestinal tract involved in restitution and repair of the mucosa. They are positively regulated in ulcerative and neoplastic conditions. We describe a consensus sequence in human and rodent TFF promoters close to the TATAA box showing striking similarity to the binding site of the HNF-3/FKH family. In gel retardation assays, HNF-3 alpha and beta bound predominantly to the site in TFF1 (formerly pS2) and, to a lesser extent, to the sites in TFF2 or TFF3. Mutations generated in this motif severely impaired transcription of TFF1 reporter genes. Cotransfection with expression vectors of HNF-3alpha and beta, but not the related HFH 11A and B, specifically activated the wild-type TFF1 reporter genes. Activation of endogenous expression of TFF1 by HNF-3 alpha and beta gene products was more than 1000 fold in the pancreatic cell line Capan-2 and fivefold in the gastric cell line MKN-45, whereas the intestinal cell lines HUTU 80 and HT-29 displayed no effect. Thus, HNF-3/FKH factors contribute causally to cell-specific regulation of TFF genes and may explain the acute-phase response of TFF peptides.
Collapse
Affiliation(s)
- S Beck
- Division of Molecular Genetics, Institute of Anthropology and Human Genetics, University of Tübingen, Germany
| | | | | | | | | |
Collapse
|