51
|
Gadea G, Arsic N, Fernandes K, Diot A, Joruiz SM, Abdallah S, Meuray V, Vinot S, Anguille C, Remenyi J, Khoury MP, Quinlan PR, Purdie CA, Jordan LB, Fuller-Pace FV, de Toledo M, Cren M, Thompson AM, Bourdon JC, Roux P. TP53 drives invasion through expression of its Δ133p53β variant. eLife 2016; 5:e14734. [PMID: 27630122 PMCID: PMC5067115 DOI: 10.7554/elife.14734] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 09/13/2016] [Indexed: 12/28/2022] Open
Abstract
TP53 is conventionally thought to prevent cancer formation and progression to metastasis, while mutant TP53 has transforming activities. However, in the clinic, TP53 mutation status does not accurately predict cancer progression. Here we report, based on clinical analysis corroborated with experimental data, that the p53 isoform Δ133p53β promotes cancer cell invasion, regardless of TP53 mutation status. Δ133p53β increases risk of cancer recurrence and death in breast cancer patients. Furthermore Δ133p53β is critical to define invasiveness in a panel of breast and colon cell lines, expressing WT or mutant TP53. Endogenous mutant Δ133p53β depletion prevents invasiveness without affecting mutant full-length p53 protein expression. Mechanistically WT and mutant Δ133p53β induces EMT. Our findings provide explanations to 2 long-lasting and important clinical conundrums: how WT TP53 can promote cancer cell invasion and reciprocally why mutant TP53 gene does not systematically induce cancer progression.
Collapse
Affiliation(s)
- Gilles Gadea
- UM 134 Processus Infectieux en Milieu Insulaire Tropical (PIMIT), INSERM U1187, CNRS UMR9192, IRD UMR249, Université de la Réunion, Sainte Clotilde, France
| | - Nikola Arsic
- Université Montpellier, Montpellier, France
- CRBM, CNRS, Centre de Recherche de Biologie cellulaire de Montpellier, Montpellier, France
| | - Kenneth Fernandes
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Alexandra Diot
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Sébastien M Joruiz
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Samer Abdallah
- Université Montpellier, Montpellier, France
- CRBM, CNRS, Centre de Recherche de Biologie cellulaire de Montpellier, Montpellier, France
| | - Valerie Meuray
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Stéphanie Vinot
- Université Montpellier, Montpellier, France
- CRBM, CNRS, Centre de Recherche de Biologie cellulaire de Montpellier, Montpellier, France
| | - Christelle Anguille
- Université Montpellier, Montpellier, France
- CRBM, CNRS, Centre de Recherche de Biologie cellulaire de Montpellier, Montpellier, France
| | - Judit Remenyi
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Marie P Khoury
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Philip R Quinlan
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Colin A Purdie
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Lee B Jordan
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Frances V Fuller-Pace
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Marion de Toledo
- Université Montpellier, Montpellier, France
- CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | - Maïlys Cren
- Université Montpellier, Montpellier, France
- IRB, Institut de Recherche en Biothérapie, Montpellier, France
| | - Alastair M Thompson
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
- Department of Surgical Oncology, MD Anderson Cancer Centre, Houston, United States
| | - Jean-Christophe Bourdon
- Division of Cancer Research, University of Dundee, Ninewells Hospital and Medical School, Dundee, United Kingdom
| | - Pierre Roux
- Université Montpellier, Montpellier, France
- CRBM, CNRS, Centre de Recherche de Biologie cellulaire de Montpellier, Montpellier, France
| |
Collapse
|
52
|
Chowdhury AR, Long A, Fuchs SY, Rustgi A, Avadhani NG. Mitochondrial stress-induced p53 attenuates HIF-1α activity by physical association and enhanced ubiquitination. Oncogene 2016; 36:397-409. [PMID: 27345397 PMCID: PMC5192009 DOI: 10.1038/onc.2016.211] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 03/22/2016] [Accepted: 04/26/2016] [Indexed: 12/17/2022]
Abstract
Retrograde signaling is a mechanism by which mitochondrial dysfunction is communicated to the nucleus for inducing a metabolic shift essential for cell survival. Previously we showed that partial mtDNA depletion in different cell types induced mitochondrial retrograde signaling pathway (MtRS) involving Ca+2 sensitive Calcineurin (Cn) activation as an immediate upstream event of stress response. In multiple cell types, this stress signaling was shown to induce tumorigenic phenotypes in immortalized cells. In this study we show that MtRS also induces p53 expression which was abrogated by Ca2+ chelators and shRNA mediated knock down of CnAβ mRNA. Mitochondrial dysfunction induced by mitochondrial ionophore, carbonyl cyanide m-chlorophenyl hydrazone (CCCP) and other respiratory inhibitors, which perturb the transmembrane potential, were equally efficient in inducing the expression of p53 and downregulation of MDM2. Stress-induced p53 physically interacted with HIF-1α and attenuated the latter’s binding to promoter DNA motifs. Additionally, p53 promoted ubiquitination and degradation of HIF-1α in partial mtDNA depleted cells. The mtDNA depleted cells, with inhibited HIF-1α, showed upregulation of glycolytic pathway genes, glucose transporter 1–4 (Glut1–4), phosphoglycerate kinase 1 (PGK1) and Glucokinase (GSK) but not of prolyl hydroxylase (PHD) isoforms. For the first time we show that p53 is induced as part of MtRS and it renders HIF-1α inactive by physical interaction. In this respect our results show that MtRS induces tumor growth independent of HIF-1α pathway.
Collapse
Affiliation(s)
- A Roy Chowdhury
- Department of Biomedical Sciences and Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - A Long
- Division of Gastroenterology, Department of Medicine and Genetics, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - S Y Fuchs
- Department of Biomedical Sciences and Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - A Rustgi
- Division of Gastroenterology, Department of Medicine and Genetics, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - N G Avadhani
- Department of Biomedical Sciences and Mari Lowe Center for Comparative Oncology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
53
|
Jin MS, Park IA, Kim JY, Chung YR, Im SA, Lee KH, Moon HG, Han W, Noh DY, Ryu HS. New insight on the biological role of p53 protein as a tumor suppressor: re-evaluation of its clinical significance in triple-negative breast cancer. Tumour Biol 2016; 37:11017-24. [DOI: 10.1007/s13277-016-4990-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 02/10/2016] [Indexed: 01/04/2023] Open
|
54
|
Králová V, Hanušová V, Rudolf E, Čáňová K, Skálová L. Flubendazole induces mitotic catastrophe and senescence in colon cancer cells in vitro. J Pharm Pharmacol 2016; 68:208-18. [DOI: 10.1111/jphp.12503] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 10/28/2015] [Indexed: 12/31/2022]
Abstract
Abstract
Objectives
Flubendazole (FLU), a member of benzimidazole family of anthelmintic drugs, is able to inhibit proliferation of various cancer cells. The aim of present study was to elucidate the mechanisms of antiproliferative effect of FLU on colorectal cancer cells in vitro.
Methods
The effect of FLU on proliferation, microtubular network, DNA content, caspase activation and senescence induction was studied in SW480 and SW620 cell lines.
Key findings
Flubendazole significantly affected cell proliferation in a pattern typical for mitotic inhibitor. This was accompanied by decrease in cyclin D1 levels, increase in cyclin B1 levels, activation of caspase 2 and caspase 3/7 and PARP cleavage. Morphological observations revealed disruption of microtubular network, irregular mitotic spindles, formation of giant multinucleated cells and increase in nuclear area and DNA content. In SW620 cell line, 37.5% giant multinucleated cells induced by FLU treatment showed positivity for SA-β-galactosidase staining. Cell lines were able to recover from the treatment and this process was faster in SW480 cells.
Conclusion
Flubendazole in low concentration temporarily inhibits cell proliferation and induces mitotic catastrophe and premature senescence in human colon cancer cells in vitro.
Collapse
Affiliation(s)
- Věra Králová
- Department of Medical Biology and Genetics, Charles University in Prague, Hradec Králové, Czech Republic
| | - Veronika Hanušová
- Department of Medical Biology and Genetics, Charles University in Prague, Hradec Králové, Czech Republic
| | - Emil Rudolf
- Department of Medical Biology and Genetics, Charles University in Prague, Hradec Králové, Czech Republic
| | - Kristýna Čáňová
- Department of Medical Biology and Genetics, Charles University in Prague, Hradec Králové, Czech Republic
| | - Lenka Skálová
- Department of Biochemical Sciences, Charles University in Prague, Hradec Králové, Czech Republic
| |
Collapse
|
55
|
Tsou SH, Hou MH, Hsu LC, Chen TM, Chen YH. Gain-of-function p53 mutant with 21-bp deletion confers susceptibility to multidrug resistance in MCF-7 cells. Int J Mol Med 2015; 37:233-42. [PMID: 26572087 DOI: 10.3892/ijmm.2015.2406] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 10/30/2015] [Indexed: 11/06/2022] Open
Abstract
The majority of p53 mutations, which are responsible for gain of oncogenic function, are missense mutations in hotspot codons. However, in our previous study, we demonstrated that a deletion spanning codons 127-133 in the p53 gene (designated as del p53) was detected in doxorubicin-resistant MCF-7 cell lines following various induction processes. In the present study, we aimed to investigate the role of del p53 and its association with the proliferation, metastasis and drug resistance of MCF-7 cells. The MCF-7/del p53 cell line is a representative of the del p53 stably expressed clones which were constructed by transfection of the del p53-containing construct into MCF-7/wt cells. Markers of multidrug resistance (MDR), epithelial-mesenchymal transition (EMT) and stem cell-like properties were examined in the MCF-7/del p53 cells. The results revealed that the MCF-7/del p53 cells expressed full-length p53 and del p53 mRNA and protein, as well as P-glycoprotein (P-gp). The MCF-7/del p53 cells acquired resistance to doxorubicin with increased P-gp efflux function. Using a transient expression assay, the mdr1 promoter was found to be significantly activated by external or integrated del p53 (P<0.001). The inhibition of nuclear factor (NF)-κB by cyclosporine sensitized the MCF-7/del p53 cells to doxorubicin toxicity. In addition, the morphological characteristics of the MCF-7/del p53 and MCF-7/adr were similar. EMT was observed in the MCF-7/del p53 cells as demonstrated by the presence of the mesenchymal markers, Slug and vimentin, and the decrease in the epithelial marker, cadherin 1, type 1, E-cadherin (CDH1), as well as an enhanced migration ability (P<0.001). Furthermore, the number of cells expressing the cancer stem cell-like marker, CD44, increased, accompanied by mammosphere formation. Taken together, these findings indicate that the expression of del p53 in MCF-7/del p53 cells enables the cells to partially acquire doxorubicin resistance characteristics of the MCF-7/adr cells. Thus, del p53 may be an important factor in non-invasive MCF-7 cells, activating NF-κB signaling and the mdr1 promoter and partially attributing to EMT; the cells thus acquire stem cell‑like properties, which facilitates drug resistance. Therefore, the 21-bp deletion of p53 may prove to be a therapeutic strategy with which to prevent cancer cells from acquiring resistance to drugs.
Collapse
Affiliation(s)
- Shang-Hsun Tsou
- Graduate Institute of Pharmaceutical Sciences, School of Pharmacy, College of Medicine, National Taiwan University, Taipei 10050, Taiwan, R.O.C
| | - Ming-Hung Hou
- Graduate Institute of Pharmaceutical Sciences, School of Pharmacy, College of Medicine, National Taiwan University, Taipei 10050, Taiwan, R.O.C
| | - Lih-Ching Hsu
- Graduate Institute of Pharmaceutical Sciences, School of Pharmacy, College of Medicine, National Taiwan University, Taipei 10050, Taiwan, R.O.C
| | - Tzer-Ming Chen
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei 10050, Taiwan, R.O.C
| | - Yen-Hui Chen
- Graduate Institute of Pharmaceutical Sciences, School of Pharmacy, College of Medicine, National Taiwan University, Taipei 10050, Taiwan, R.O.C
| |
Collapse
|
56
|
GARUFI ALESSIA, D'ORAZI VALERIO, CRISPINI ALESSANDRA, D'ORAZI GABRIELLA. Zn(II)-curc targets p53 in thyroid cancer cells. Int J Oncol 2015; 47:1241-1248. [PMID: 26314369 PMCID: PMC4583539 DOI: 10.3892/ijo.2015.3125] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 07/13/2015] [Indexed: 02/06/2023] Open
Abstract
TP53 mutation is a common event in many cancers, including thyroid carcinoma. Defective p53 activity promotes cancer resistance to therapies and a more malignant phenotype, acquiring oncogenic functions. Rescuing the function of mutant p53 (mutp53) protein is an attractive anticancer therapeutic strategy. Zn(II)-curc is a novel small molecule that has been shown to target mutp53 protein in several cancer cells, but its effect in thyroid cancer cells remains unclear. Here, we investigated whether Zn(II)-curc could affect p53 in thyroid cancer cells with both p53 mutation (R273H) and wild-type p53. Zn(II)-curc induced mutp53H273 downregulation and reactivation of wild-type functions, such as binding to canonical target promoters and target gene transactivation. This latter effect was similar to that induced by PRIMA-1. In addition, Zn(II)-curc triggered p53 target gene expression in wild-type p53-carrying cells. In combination treatments, Zn(II)-curc enhanced the antitumor activity of chemotherapeutic drugs, in both mutant and wild-type-carrying cancer cells. Taken together, our data indicate that Zn(II)-curc promotes the reactivation of p53 in thyroid cancer cells, providing in vitro evidence for a potential therapeutic approach in thyroid cancers.
Collapse
Affiliation(s)
- ALESSIA GARUFI
- Department of Experimental Oncology, Regina Elena National Cancer Institute, Rome, Italy
- Department of Medical, Oral and Biotechnological Sciences, University ‘G. d’Annunzio’, Chieti, Italy
| | - VALERIO D'ORAZI
- Department of Surgical Sciences, Sapienza University, Rome, Italy
| | - ALESSANDRA CRISPINI
- Department of Chemistry and Technologic Chemistry, University of Calabria, Cosenza, Italy
| | - GABRIELLA D'ORAZI
- Department of Experimental Oncology, Regina Elena National Cancer Institute, Rome, Italy
- Department of Medical, Oral and Biotechnological Sciences, University ‘G. d’Annunzio’, Chieti, Italy
| |
Collapse
|
57
|
Liu X, Ji Q, Fan Z, Li Q. Cellular signaling pathways implicated in metastasis of colorectal cancer and the associated targeted agents. Future Oncol 2015; 11:2911-22. [PMID: 26414153 DOI: 10.2217/fon.15.235] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is the third leading cancer worldwide and CRC-related death is mainly attributed to metastasis. Many cellular signaling pathways have been demonstrated to be aberrant in colorectal tumors, and some of them lead to the acquisition of malignant phenotypes. Therefore, the evaluation of signaling pathways implicated in CRC metastasis is urgent for further understanding of CRC progression and pharmacotherapy. This review focuses on several novel cellular signaling pathways associated with CRC metastasis, including Wnt/β-catenin, p53, COX, TGF-β/Smad, NF-κB, Notch, VEGF and JAKs/STAT3 signaling pathways. Targeted agents developed based on these pathways are also briefly discussed.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Qing Ji
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Zhongze Fan
- Interventional Cancer Institute, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Qi Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| |
Collapse
|
58
|
Huang J, Li H, Ren G. Epithelial-mesenchymal transition and drug resistance in breast cancer (Review). Int J Oncol 2015. [PMID: 26202679 DOI: 10.3892/ijo.2015.3084] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is the leading cause of cancer death in women worldwide. Insensitivity of tumor cells to drug therapies is an essential reason arousing such high mortality. Epithelial-mesenchymal transition (EMT) is defined by the loss of epithelial characteristics and the acquisition of a mesenchymal phenotype. It is well known that EMT plays an important role in breast cancer progression. Recently, mounting evidence has demonstrated involvement of EMT in antagonizing chemotherapy in breast cancer. Here, we discuss the biological significance and clinical implications of these findings, with an emphasis on novel approaches that effectively target EMT to increase the efficacy of anticancer therapies.
Collapse
Affiliation(s)
- Jing Huang
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Hongzhong Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Guosheng Ren
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
59
|
Arsic N, Gadea G, Lagerqvist EL, Busson M, Cahuzac N, Brock C, Hollande F, Gire V, Pannequin J, Roux P. The p53 isoform Δ133p53β promotes cancer stem cell potential. Stem Cell Reports 2015; 4:531-40. [PMID: 25754205 PMCID: PMC4400643 DOI: 10.1016/j.stemcr.2015.02.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Revised: 02/03/2015] [Accepted: 02/03/2015] [Indexed: 11/18/2022] Open
Abstract
Cancer stem cells (CSC) are responsible for cancer chemoresistance and metastasis formation. Here we report that Δ133p53β, a TP53 splice variant, enhanced cancer cell stemness in MCF-7 breast cancer cells, while its depletion reduced it. Δ133p53β stimulated the expression of the key pluripotency factors SOX2, OCT3/4, and NANOG. Similarly, in highly metastatic breast cancer cells, aggressiveness was coupled with enhanced CSC potential and Δ133p53β expression. Like in MCF-7 cells, SOX2, OCT3/4, and NANOG expression were positively regulated by Δ133p53β in these cells. Finally, treatment of MCF-7 cells with etoposide, a cytotoxic anti-cancer drug, increased CSC formation and SOX2, OCT3/4, and NANOG expression via Δ133p53, thus potentially increasing the risk of cancer recurrence. Our findings show that Δ133p53β supports CSC potential. Moreover, they indicate that the TP53 gene, which is considered a major tumor suppressor gene, also acts as an oncogene via the Δ133p53β isoform. The Δ133p53β isoform promotes stemness of breast cancer cells The Δ133p53β isoform regulates SOX2, OCT3/4, and NANOG expression, but not C-MYC Etoposide promotes cancer cell stemness through Δ133p53β induction Δ133p53β expression, like p53 mutations, promotes cancer cell stemness
Collapse
Affiliation(s)
- Nikola Arsic
- Centre National de la Recherche Scientifique, UMR 5237, Centre de Recherche en Biochimie Macromoléculaire, Université Montpellier, 1919 route de Mende, 34293 Montpellier Cedex 5, France
| | - Gilles Gadea
- Centre National de la Recherche Scientifique, UMR 5237, Centre de Recherche en Biochimie Macromoléculaire, Université Montpellier, 1919 route de Mende, 34293 Montpellier Cedex 5, France
| | - E Louise Lagerqvist
- Centre National de la Recherche Scientifique, UMR5203, Institut de Génomique Fonctionnelle, Institut National de la Santé et de la Recherche Médicale, U661, Université Montpellier, route de Cardonille, 34094 Montpellier, France
| | - Muriel Busson
- Plateforme Imagerie du Petit Animal de Montpellier (IPAM), Institut de Recherche en Cancérologie de Montpellier Inserm U896, Université Montpellier, ICM Val d'Aurelle Campus Val d'Aurelle, 208 Rue des Apothicaires, 34298 Montpellier Cedex 5, France
| | - Nathalie Cahuzac
- Eurobiodev, 2040 avenue du Père Soulas, 34090 Montpellier, France
| | - Carsten Brock
- Eurofins Cerep, Le bois L'Evèque, 86600 Celle L'Evescault, France
| | - Frederic Hollande
- Department of Pathology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Veronique Gire
- Centre National de la Recherche Scientifique, UMR 5237, Centre de Recherche en Biochimie Macromoléculaire, Université Montpellier, 1919 route de Mende, 34293 Montpellier Cedex 5, France
| | - Julie Pannequin
- Centre National de la Recherche Scientifique, UMR5203, Institut de Génomique Fonctionnelle, Institut National de la Santé et de la Recherche Médicale, U661, Université Montpellier, route de Cardonille, 34094 Montpellier, France
| | - Pierre Roux
- Centre National de la Recherche Scientifique, UMR 5237, Centre de Recherche en Biochimie Macromoléculaire, Université Montpellier, 1919 route de Mende, 34293 Montpellier Cedex 5, France.
| |
Collapse
|
60
|
Yan ZH, Cui LH, Wang XH, Li C, He X. Comparative study of mutations in SNP loci of K-RAS, hMLH1 and hMSH2 genes in neoplastic intestinal polyps and colorectal cancer. World J Gastroenterol 2014; 20:18338-18345. [PMID: 25561800 PMCID: PMC4277970 DOI: 10.3748/wjg.v20.i48.18338] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 07/12/2014] [Accepted: 08/28/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To clarify the molecular mechanism involved in pathogenesis of colorectal cancer as well as clinical significance of genetic analysis of histological samples.
METHODS: A total of 480 blood and tissue specimens were collected in our hospital from January 2011 to October 2012. In the observation group, there were 120 blood specimens and 120 intestinal tract tissue specimens collected from patients with neoplastic intestinal polyps. In the control group I there were 80 blood specimens and 80 intestinal tract tissue specimens collected from patients with colorectal cancer. In the control group II there were 40 blood specimens and 40 intestinal tract tissue specimens collected from healthy individuals. The gene segments were amplified using PCR and DNA gel electrophoresis along with DNA sequence analysis were employed for the detection of the following single nucleotide polymorphisms (SNPs): K-RAS codons 12 and 13; hMLH1 (human mutS homolog 1) gene missense mutation at Va1384Asp; hMSH2 (human mutS homolog 2) gene missense mutation at 2783C/A.
RESULTS: The mutation rate of the SNP at Va1384Asp locus of the hMLH1 gene from blood and tissue specimens in the observation group showed no statistical difference from those in the control group I. The mutation rates of SNPs in codons 12 and 13 of K-RAS and at 2783C/A locus of the hMSH2 gene were significantly lower in the observation group than in the control group I (χ2 = 15.476, 29.670, 10.811, 16.618, 33.538, 7.898, P < 0.05). The mutation rate of SNP at Va1384Asp locus of the hMLH1 gene was significantly higher in the observation group when compared to the control group II (χ2 = 10.486, 4.876, P < 0.05). The mutation rates of SNPs in codons 12 and 13 of K-RAS and at 2783C/A locus of the hMSH2 gene did not show any statistical difference from those in the control group II.
CONCLUSION: There may be important clinical significance and relevance between neoplastic intestinal polyps and colorectal cancer in terms of the mechanisms involved in the pathogenesis.
Collapse
|
61
|
Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, Lee SJ. Herbimycin A inhibits cell growth with reversal of epithelial–mesenchymal transition in anaplastic thyroid carcinoma cells. Biochem Biophys Res Commun 2014; 455:363-70. [DOI: 10.1016/j.bbrc.2014.11.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 11/09/2014] [Indexed: 11/28/2022]
|
62
|
Fernandez SV, Bingham C, Fittipaldi P, Austin L, Palazzo J, Palmer G, Alpaugh K, Cristofanilli M. TP53 mutations detected in circulating tumor cells present in the blood of metastatic triple negative breast cancer patients. Breast Cancer Res 2014; 16:445. [PMID: 25307991 PMCID: PMC4303125 DOI: 10.1186/s13058-014-0445-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 09/11/2014] [Indexed: 01/24/2023] Open
Abstract
Introduction Circulating tumor cells (CTCs) are tumor cells shed from either primary tumors or its metastases that circulate in the peripheral blood of patients with metastatic cancers. The molecular characterization of the CTCs is critical to identifying the key drivers of cancer metastasis and devising therapeutic approaches. However, the molecular characterization of CTCs is difficult to achieve because their isolation is a major technological challenge. Methods CTCs from two triple negative breast cancer patients were enriched using CellSearch and single cells selected by DEPArray™. A TP53 R110 fs*13 mutation identified by next generation sequencing in the breast and chest skin biopsies of both patients was studied in single CTCs. Results From 6 single CTC isolated from one patient, 1 CTC had TP53 R110 delC, 1 CTC showed the TP53 R110 delG mutation, and the remaining 4 single CTCs showed the wild type p53 sequence; a pool of 14 CTCs isolated from the same patient also showed TP53 R110 delC mutation. In the tumor breast tissue of this patient, only the TP53 R110 delG mutation was detected. In the second patient a TP53 R110 delC mutation was detected in the chest wall skin biopsy; from the peripheral blood of this patient, 5 single CTC and 6 clusters of 2 to 6 CTCs were isolated; 3 of the 5 single CTCs showed the TP53 R110 delC mutation and 2 CTCs showed the wild type TP53 allele; from the clusters, 5 showed the TP53 R110 delC mutation, and 1 cluster the wild type TP53 allele. Single white blood cells isolated as controls from both patients only showed the wild type TP53 allele. Conclusions We are able to isolate uncontaminated CTCs and achieve single cell molecular analysis. Our studies showed the presence of different CTC sub-clones in patients with metastatic breast cancer. Some CTCs had the same TP53 mutation as their matching tumor samples although others showed either a different TP53 mutation or the wild type allele. Our results indicate that CTCs could represent a non-invasive source of cancer cells from which to determine genetic markers of the disease progression and potential therapeutic targets. Electronic supplementary material The online version of this article (doi:10.1186/s13058-014-0445-3) contains supplementary material, which is available to authorized users.
Collapse
|
63
|
Lenfert E, Maenz C, Heinlein C, Jannasch K, Schumacher U, Pantel K, Tolstonog GV, Deppert W, Wegwitz F. Mutant p53 promotes epithelial-mesenchymal plasticity and enhances metastasis in mammary carcinomas of WAP-T mice. Int J Cancer 2014; 136:E521-33. [PMID: 25195563 DOI: 10.1002/ijc.29186] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 07/31/2014] [Accepted: 08/21/2014] [Indexed: 12/20/2022]
Abstract
To study the postulated mutant p53 (mutp53) "gain of function" effects in mammary tumor development, progression and metastasis, we crossed SV40 transgenic WAP-T mice with mutant p53 transgenic WAP-mutp53 mice. Compared to tumors in monotransgenic WAP-T mice, tumors in bitransgenic WAP-T x WAP-mutp53 mice showed higher tumor grading, enhanced vascularization, and significantly increased metastasis. Bitransgenic tumors revealed a gene signature associated with the oncogenic epithelial-mesenchymal transition pathway (EMT gene signature). In cultures of WAP-T tumor-derived G-2 cancer cells, which are comprised of subpopulations displaying "mesenchymal" and "epithelial" phenotypes, this EMT gene signature was associated with the "mesenchymal" compartment. Furthermore, ectopic expression of mutp53 in G-2 cells sufficed to induce a strong EMT phenotype. In contrast to these in vitro effects, monotransgenic and bitransgenic tumors were phenotypically similar suggesting that in vivo the tumor cell phenotype might be under control of the tumor microenvironment. In support, orthotopic transplantation of G-2 cells as well as of G-2 cells expressing ectopic mutp53 into syngeneic mice resulted in tumors with a predominantly epithelial phenotype, closely similar to that of endogenous primary tumors. We conclude that induction of an EMT gene signature by mutp53 in bitransgenic tumors primarily promotes tumor cell plasticity, that is, the probability of tumor cells to undergo EMT processes under appropriate stimuli, thereby possibly increasing their potential to disseminate and metastasize.
Collapse
Affiliation(s)
- Eva Lenfert
- Department for Tumor Biology, University Medical Center Hamburg-Eppendorf (UKE), D-20246, Hamburg, Germany; Department of Tumor Virology, Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, D-20251, Hamburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Phospholipase Cδ1 induces E-cadherin expression and suppresses malignancy in colorectal cancer cells. Proc Natl Acad Sci U S A 2014; 111:13505-10. [PMID: 25197077 DOI: 10.1073/pnas.1405374111] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common causes of cancer-related deaths worldwide, and Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations in CRC predict the ineffectiveness of EGF receptor-targeted therapy. Previous transcriptional microarray analysis suggests the association between phospholipase Cδ1 (PLCδ1) expression and KRAS mutation status in CRC. However, both the roles and the regulatory mechanisms of PLCδ1 in CRC are not known. Here, we found that the expression of PLCδ1, one of the most basal PLCs, is down-regulated in CRC specimens compared with normal colon epithelium by immunohistochemistry. Furthermore, we examined the roles of PLCδ1 in CRC cell lines that harbor an activating KRAS mutation. Ectopic expression of PLCδ1 in CRC cells induced the expression of E-cadherin, whereas knockdown of PLCδ1 repressed the expression of E-cadherin. Moreover, the overexpression of PLCδ1 suppressed the expression of several mesenchymal genes and reduced cell motility, invasiveness, and in vivo tumorigenicity of SW620 CRC cells. We also showed that PLCδ1 expression is repressed by the KRAS/mitogen-activated protein kinase kinase (MEK) pathway. Furthermore, PLCδ1 suppressed the phosphorylation of extracellular signal-regulated kinase (ERK)1/2 through E-cadherin induction in CRC cells, suggesting the presence of a negative regulatory loop between KRAS/MEK/ERK signaling and PLCδ1. These data indicate that PLCδ1 has tumor-suppressive functions in CRC through E-cadherin induction and KRAS/MEK/ERK signal attenuation.
Collapse
|
65
|
|
66
|
Song Y, Zhang C, Cao Z, Xu J, Wang L, Lin X. [Significance of epithelial-mesenchaymal transition phenotype in invasive tumor front cells of lung squamous cell carcinoma]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2014; 17:315-20. [PMID: 24758906 PMCID: PMC6000016 DOI: 10.3779/j.issn.1009-3419.2014.04.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
背景与目的 肿瘤浸润前沿(invasive tumor front, ITF)细胞是指肿瘤与宿主组织交界处的细胞或浸润的细胞团,对判断患者预后具有较高的价值。本研究旨在探讨肺鳞状细胞癌(squamous cell carcinoma, SCC)ITF细胞的上皮-间叶转化(epithelial-mesenchaymal transition, EMT)表型特点,并分析与临床病理特征和预后的关系。 方法 采用免疫组织化学SP法检测104例肺SCC ITF细胞中上皮性标志物E-cadherin/β-catenin和间叶性标志物vimentin的表达。 结果 E-cadherin在53.8%(56/104)的肺SCC ITF细胞中表达下调,较非ITF细胞表达降低(P=0.04),而vimentin在42.3%(44 /104)ITF细胞中表达,较非ITF肿瘤细胞表达升高;两者均与肿瘤浸润方式、肺门淋巴结转移和患者预后有相关性(P < 0.01)。β-catenin在肺SCC ITF细胞的表达阳性率为67.3%(70/104),低于非ITF细胞(P < 0.01),在ITF细胞呈胞质和胞核阳性表达,并与肺门淋巴结转移密切相关。 结论 肺SCC ITF细胞中E-cadherin/β-catenin表达缺失和vimentin高表达可能与患者的不良预后有关。
Collapse
Affiliation(s)
- Yinghua Song
- Department of Respiratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Ji'nan 250014, China
| | - Caiqing Zhang
- Department of Respiratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Ji'nan 250014, China
| | - Zhixin Cao
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Ji'nan 250021, China
| | - Jiawen Xu
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Ji'nan 250021, China
| | - Lingcheng Wang
- Department of Respiratory Medicine, the Forth People's Hospital of Jinan, Ji'nan 250031, China
| | - Xiaoyan Lin
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Ji'nan 250021, China
| |
Collapse
|
67
|
Muller PAJ, Vousden KH. Mutant p53 in cancer: new functions and therapeutic opportunities. Cancer Cell 2014; 25:304-17. [PMID: 24651012 PMCID: PMC3970583 DOI: 10.1016/j.ccr.2014.01.021] [Citation(s) in RCA: 1149] [Impact Index Per Article: 104.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/13/2013] [Accepted: 01/13/2014] [Indexed: 12/11/2022]
Abstract
Many different types of cancer show a high incidence of TP53 mutations, leading to the expression of mutant p53 proteins. There is growing evidence that these mutant p53s have both lost wild-type p53 tumor suppressor activity and gained functions that help to contribute to malignant progression. Understanding the functions of mutant p53 will help in the development of new therapeutic approaches that may be useful in a broad range of cancer types.
Collapse
Affiliation(s)
- Patricia A J Muller
- Medical Research Council Toxicology Unit, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, UK.
| | - Karen H Vousden
- CR-UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK.
| |
Collapse
|
68
|
Li H, Li M, Xu D, Zhao C, Liu G, Wang F. Overexpression of Snail in retinal pigment epithelial triggered epithelial-mesenchymal transition. Biochem Biophys Res Commun 2014; 446:347-51. [PMID: 24607896 DOI: 10.1016/j.bbrc.2014.02.119] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 02/25/2014] [Indexed: 01/18/2023]
Abstract
Snail transcription factor has been implicated as an important regulator in epithelial-mesenchymal transition (EMT) during tumourigenesis and fibrogenesis. Our previous work showed that Snail transcription factor was activated in transforming growth factor β1 (TGF-β1) induced EMT in retinal pigment epithelial (RPE) cells and may contribute to the development of retinal fibrotic disease such as proliferative vitreoretinopathy (PVR). However, whether Snail alone has a direct role on retinal pigment epithelial-mesenchymal transition has not been investigated. Here, we analyzed the capacity of Snail to drive EMT in human RPE cells. A vector encoding Snail gene or an empty vector were transfected into human RPE cell lines ARPE-19 respectively. Snail overexpression in ARPE-19 cells resulted in EMT, which was characterized by the expected phenotypic transition from a typical epithelial morphology to mesenchymal spindle-shaped. The expression of epithelial markers E-cadherin and Zona occludin-1 (ZO-1) were down-regulated, whereas mesenchymal markers a-smooth muscle actin (a-SMA) and fibronectin were up-regulated in Snail expression vector transfected cells. In addition, ectopic expression of Snail significantly enhanced ARPE-19 cell motility and migration. The present data suggest that overexpression of Snail in ARPE-19 cells could directly trigger EMT. These results may provide novel insight into understanding the regulator role of Snail in the development of retinal pigment epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Hui Li
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Affiliate of Tongji University, Shanghai 200072, PR China
| | - Min Li
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Affiliate of Tongji University, Shanghai 200072, PR China
| | - Ding Xu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Affiliate of Tongji University, Shanghai 200072, PR China
| | - Chun Zhao
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Affiliate of Tongji University, Shanghai 200072, PR China
| | - Guodong Liu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Affiliate of Tongji University, Shanghai 200072, PR China
| | - Fang Wang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Affiliate of Tongji University, Shanghai 200072, PR China.
| |
Collapse
|
69
|
Arjonen A, Kaukonen R, Mattila E, Rouhi P, Högnäs G, Sihto H, Miller BW, Morton JP, Bucher E, Taimen P, Virtakoivu R, Cao Y, Sansom OJ, Joensuu H, Ivaska J. Mutant p53-associated myosin-X upregulation promotes breast cancer invasion and metastasis. J Clin Invest 2014; 124:1069-82. [PMID: 24487586 PMCID: PMC3934176 DOI: 10.1172/jci67280] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 11/14/2013] [Indexed: 02/04/2023] Open
Abstract
Mutations of the tumor suppressor TP53 are present in many forms of human cancer and are associated with increased tumor cell invasion and metastasis. Several mechanisms have been identified for promoting dissemination of cancer cells with TP53 mutations, including increased targeting of integrins to the plasma membrane. Here, we demonstrate a role for the filopodia-inducing motor protein Myosin-X (Myo10) in mutant p53-driven cancer invasion. Analysis of gene expression profiles from 2 breast cancer data sets revealed that MYO10 was highly expressed in aggressive cancer subtypes. Myo10 was required for breast cancer cell invasion and dissemination in multiple cancer cell lines and murine models of cancer metastasis. Evaluation of a Myo10 mutant without the integrin-binding domain revealed that the ability of Myo10 to transport β₁ integrins to the filopodia tip is required for invasion. Introduction of mutant p53 promoted Myo10 expression in cancer cells and pancreatic ductal adenocarcinoma in mice, whereas suppression of endogenous mutant p53 attenuated Myo10 levels and cell invasion. In clinical breast carcinomas, Myo10 was predominantly expressed at the invasive edges and correlated with the presence of TP53 mutations and poor prognosis. These data indicate that Myo10 upregulation in mutant p53-driven cancers is necessary for invasion and that plasma-membrane protrusions, such as filopodia, may serve as specialized metastatic engines.
Collapse
Affiliation(s)
- Antti Arjonen
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Riina Kaukonen
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Elina Mattila
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Pegah Rouhi
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Gunilla Högnäs
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Harri Sihto
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Bryan W. Miller
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Jennifer P. Morton
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Elmar Bucher
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Pekka Taimen
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Reetta Virtakoivu
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Yihai Cao
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Owen J. Sansom
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Heikki Joensuu
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| | - Johanna Ivaska
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.
Turku Centre for Biotechnology, University of Turku, Turku, Finland.
Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden.
Laboratory of Molecular Oncology, University of Helsinki, Biomedicum, Helsinki, Finland.
CR-UK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom.
Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.
Department of Medicine and Health Sciences, Linköping University, Linköping, Sweden.
Department of Cardiovascular Sciences, University of Leicester, and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester, United Kingdom.
Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
Department of Biochemistry and Food Chemistry, University of Turku, Turku, Finland
| |
Collapse
|
70
|
Imai H, Kato S, Sakamoto Y, Kakudo Y, Shimodaira H, Ishioka C. High throughput RNAi screening identifies ID1 as a synthetic sick/lethal gene interacting with the common TP53 mutation R175H. Oncol Rep 2013; 31:1043-50. [PMID: 24378760 PMCID: PMC3926671 DOI: 10.3892/or.2013.2953] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 07/22/2013] [Indexed: 11/12/2022] Open
Abstract
The TP53 mutation (R175H) is one of the most common mutations in human cancer. It is a highly attractive strategy for cancer therapy to find the genes that lead the R175H-expressing cancer cells. The aim of this study was to identify the synthetic sick/lethal gene interacting with R175H. Using lentiviral bar-coded comprehensive shRNA library and a tetracycline-inducible R175H expressed in the SF126 human glioblastoma cell line (SF126-tet-R175H), we conducted high-throughput screening to identify the candidate genes that induce synthetic sickness/lethality in R175H-expressing cells. We identified 906 candidate gene suppressions that may lead to accelerated cell growth inhibition in the presence of R175H. Inhibitor of differentiation 1 (ID1) was one of the candidate genes, and its suppression by siRNA resulted in the acceleration of growth inhibition in cell lines both transiently and endogenously expressing R175H but not in TP53-null cell lines or other common p53 mutants (such as R273H). Flow cytometry analysis showed that ID1 suppression resulted in G1 arrest, and the arrest was accelerated by the expression of R175H. ID1 is a synthetic sick/lethal gene that interacts with R175H and is considered to be a novel molecular target for cancer therapy in R175H-expressing cells.
Collapse
Affiliation(s)
- Hiroo Imai
- Department of Clinical Oncology, IDAC, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Shunsuke Kato
- Department of Clinical Oncology, IDAC, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Yasuhiro Sakamoto
- Department of Clinical Oncology, IDAC, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Yuichi Kakudo
- Department of Clinical Oncology, IDAC, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Hideki Shimodaira
- Department of Clinical Oncology, IDAC, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Chikashi Ishioka
- Department of Clinical Oncology, IDAC, Tohoku University, Sendai, Miyagi 980-8575, Japan
| |
Collapse
|
71
|
Son H, Moon A. Epithelial-mesenchymal Transition and Cell Invasion. Toxicol Res 2013; 26:245-52. [PMID: 24278531 PMCID: PMC3834497 DOI: 10.5487/tr.2010.26.4.245] [Citation(s) in RCA: 245] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 11/02/2010] [Accepted: 11/14/2010] [Indexed: 12/31/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a complex process in which epithelial cells acquire the characteristics of invasive mesenchymal cells. EMT has been implicated in cancer progression and metastasis as well as the formation of many tissues and organs during development. Epithelial cells undergoing EMT lose cell-cell adhesion structures and polarity, and rearrange their cytoskeletons. Several oncogenic pathways such as transforming growth factor (TGF) -β, Wnt, and Notch signaling pathways, have been shown to induce EMT. These pathways have activated transcription factors including Snail, Slug, and the ZEB family which work as transcriptional repressors of E-cadherin, thereby making epithelial cells motile and resistant to apoptosis. Mounting evidence shows that EMT is associated with cell invasion and tumor progression.In this review, we summarize the characteristic features of EMT, pathways leading to EMT, and the role of EMT in cell invasion. Three topics are addressed in this review: (1) Definition of EMT, (2) Signaling pathways leading to EMT, (3) Role of EMT in cell invasion. Understanding the role of EMT in cell invasion will provide valuable information for establishing strategies to develop anti-metastatic therapeutics which modulate malignant cellular processes mediated by EMT.
Collapse
Affiliation(s)
- Hwajin Son
- College of Pharmacy, Duksung Women's University, Seoul 132-714, Korea
| | | |
Collapse
|
72
|
Perez CJ, Rundhaug JE, Johnson DG, Oberyszyn TM, Tober KL, Kusewitt DF. Slug expression in mouse skin and skin tumors is not regulated by p53. J Invest Dermatol 2013; 134:566-568. [PMID: 24008423 PMCID: PMC3947144 DOI: 10.1038/jid.2013.363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Carlos J Perez
- Department of Molecular Carcinogenesis, Science Park, University of Texas M.D. Anderson Cancer Center, Smithville, Texas, USA
| | - Joyce E Rundhaug
- Department of Molecular Carcinogenesis, Science Park, University of Texas M.D. Anderson Cancer Center, Smithville, Texas, USA
| | - David G Johnson
- Department of Molecular Carcinogenesis, Science Park, University of Texas M.D. Anderson Cancer Center, Smithville, Texas, USA
| | - Tatiana M Oberyszyn
- Department of Pathology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Kathleen L Tober
- Department of Pathology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Donna F Kusewitt
- Department of Molecular Carcinogenesis, Science Park, University of Texas M.D. Anderson Cancer Center, Smithville, Texas, USA.
| |
Collapse
|
73
|
Lafitte M, Moranvillier I, Garcia S, Peuchant E, Iovanna J, Rousseau B, Dubus P, Guyonnet-Dupérat V, Belleannée G, Ramos J, Bedel A, de Verneuil H, Moreau-Gaudry F, Dabernat S. FGFR3 has tumor suppressor properties in cells with epithelial phenotype. Mol Cancer 2013; 12:83. [PMID: 23902722 PMCID: PMC3750311 DOI: 10.1186/1476-4598-12-83] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 07/19/2013] [Indexed: 02/07/2023] Open
Abstract
Background Due to frequent mutations in certain cancers, FGFR3 gene is considered as an oncogene. However, in some normal tissues, FGFR3 can limit cell growth and promote cell differentiation. Thus, FGFR3 action appears paradoxical. Results FGFR3 expression was forced in pancreatic cell lines. The receptor exerted dual effects: it suppressed tumor growth in pancreatic epithelial-like cells and had oncogenic properties in pancreatic mesenchymal-like cells. Distinct exclusive pathways were activated, STATs in epithelial-like cells and MAP Kinases in mesenchymal-like cells. Both FGFR3 splice variants had similar effects and used the same intracellular signaling. In human pancreatic carcinoma tissues, levels of FGFR3 dropped in tumors. Conclusion In tumors from epithelial origin, FGFR3 signal can limit tumor growth, explaining why the 4p16.3 locus bearing FGFR3 is frequently lost and why activating mutations of FGFR3 in benign or low grade tumors of epithelial origin are associated with good prognosis. The new hypothesis that FGFR3 can harbor both tumor suppressive and oncogenic properties is crucial in the context of targeted therapies involving specific tyrosine kinase inhibitors (TKIs). TKIs against FGFR3 might result in adverse effects if used in the wrong cell context.
Collapse
Affiliation(s)
- Marie Lafitte
- INSERM U1035, Université Bordeaux Segalen, 146 rue Léo Saignat, Bordeaux 33076, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Li Z, Li N, Wu M, Li X, Luo Z, Wang X. Expression of miR-126 suppresses migration and invasion of colon cancer cells by targeting CXCR4. Mol Cell Biochem 2013; 381:233-42. [PMID: 23744532 DOI: 10.1007/s11010-013-1707-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 05/24/2013] [Indexed: 01/03/2023]
Abstract
A previous study demonstrated that miR-126 expression was significantly downregulated in highly metastatic colon cancer cells. This study was to investigate the biological function of miR-126 and its regulation of target genes in colon cancer cells. Quantitative PCR was used to detect miR-126 expression in colon cancer SW480 and SW620 cells. MTT assay was to measure the changed cell viability after miR-126 mimics transfection. Wound healing and Transwell migration and invasion assays measured capacity of tumor cell migration and invasion of SW480 and SW620 cells after miR-126 transfection. Luciferase reporter assay and Western blot were used to assess both transcriptional and expression levels of one of the miR-126 target genes (i.e., CXCR4). Levels of miR-126 expression were lower in colon cancer SW480 and SW620 cells than in the adjacent normal epithelial tissues (P < 0.05). Transfection of miR-126 mimics significantly reduced colon cancer cell viability compared to NC cells (P < 0.05). The wound healing and Transwell migration and invasion assays showed that miR-126 mimics inhibited SW480 and SW620 cell migration and invasion capacity. Bioinformatics predicted that CXCR4 is one of the miR-126 target genes. Indeed, luciferase reporter assay and Western blot confirmed that CXCR4 is a miR-126 target gene. Expression of miR-126 inhibited colon cancer cell viability and reduced tumor cell migration and invasion capacity by its negative regulation of CXCR4 expression.
Collapse
Affiliation(s)
- Zeng Li
- Department of Gastroenterology, The Xiangtan Central Hospital, Xiangtan, China
| | | | | | | | | | | |
Collapse
|
75
|
Abstract
Pro-oncogenic properties of mutant p53 were investigated with the aid of migration assays, adhesion assays, and soft agar growth assays using cells stably expressing gain-of-function p53 mutants. To determine cell migration, "wound-healing" (scratch) assays and haptotactic (chamber) assays were used. H1299 cells expressing mutant p53 were found to migrate more rapidly than cells transfected with empty vector alone. Results from both types of migration assay were broadly similar. Migratory ability differed for different p53 mutants, suggesting allele-specific effects. Cells expressing p53 mutants also showed enhanced adhesion to extracellular matrix compare to controls. Furthermore, stable transfection of mutant p53-H179L into NIH3T3 fibroblasts was sufficient to allow anchorage-independent growth in soft agar.
Collapse
|
76
|
Cong NN, Zhang QY. Role of EMT in gastrointestinal tract tumors. Shijie Huaren Xiaohua Zazhi 2013; 21:205-210. [DOI: 10.11569/wcjd.v21.i3.205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [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
Epithelial-mesenchymal transition (EMT) refers to the process by which cells transit from epithelial phenotype to mesenchymal phenotype. EMT is important for embryonic development, wound healing, and invasion of carcinomas. The molecular mechanisms of EMT are a hot topic of research in invasion and migration of malignant tumors, especially digestive carcinomas. Since malignant epithelial tumors account for a large proportion of tumors and are associated with very poor outcome and prognosis, exploration of the process of epithelial cell migration and invasion is of great significance for the prevention and treatment of tumors. The investigation of EMT provides a basis for understanding the pathogenesis of tumors and their prognosis and resistance to antitumor drugs. This review focuses on the molecular mechanisms and role of EMT in gastrointestinal tract tumors.
Collapse
|
77
|
Termén S, Tan EJ, Heldin CH, Moustakas A. p53 regulates epithelial-mesenchymal transition induced by transforming growth factor β. J Cell Physiol 2012; 228:801-13. [DOI: 10.1002/jcp.24229] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2012] [Accepted: 09/18/2012] [Indexed: 12/30/2022]
|
78
|
D'Costa ZJ, Jolly C, Androphy EJ, Mercer A, Matthews CM, Hibma MH. Transcriptional repression of E-cadherin by human papillomavirus type 16 E6. PLoS One 2012; 7:e48954. [PMID: 23189137 PMCID: PMC3506579 DOI: 10.1371/journal.pone.0048954] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 10/03/2012] [Indexed: 02/01/2023] Open
Abstract
There is increasing evidence supporting DNA virus regulation of the cell adhesion and tumour suppressor protein, E-cadherin. We previously reported that loss of E-cadherin in human papillomavirus (HPV) type 16-infected epidermis is contributed to by the major viral proto-oncogene E6 and is associated with reduced Langerhans cells density, potentially regulating the immune response. The focus of this study is determining how the HPV16 E6 protein mediates E-cadherin repression. We found that the E-cadherin promoter is repressed in cells expressing E6, resulting in fewer E-cadherin transcripts. On exploring the mechanism for this, repression by increased histone deacetylase activity or by increased binding of trans-repressors to the E-cadherin promoter Epal element was discounted. In contrast, DNA methyltransferase (DNMT) activity was increased in E6 expressing cells. Upon inhibiting DNMT activity using 5-Aza-2'-deoxycytidine, E-cadherin transcription was restored in the presence of HPV16 E6. The E-cadherin promoter was not directly methylated, however a mutational analysis showed general promoter repression and reduced binding of the transactivators Sp1 and AML1 and the repressor Slug. Expression of E7 with E6 resulted in a further reduction in surface E-cadherin levels. This is the first report of HPV16 E6-mediated transcriptional repression of this adhesion molecule and tumour suppressor protein.
Collapse
Affiliation(s)
- Zarina J. D'Costa
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Carol Jolly
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Elliot J. Androphy
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Andrew Mercer
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Charles M. Matthews
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Merilyn H. Hibma
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
79
|
Xu HX, Ma WQ, Yang RJ, Wang YM, Wang LJ, Zang LQ, He XX. Small interfering RNA-mediated MIF knockdown reduces cell invasion in murine colorectal cancer cell line CT-26. Shijie Huaren Xiaohua Zazhi 2012; 20:3000-3004. [DOI: 10.11569/wcjd.v20.i31.3000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [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 investigate the effect of small interfering RNA (siRNA)-mediated knockdown of the macrophage migration-inhibitory factor (MIF) gene on cell invasion in murine colorectal cancer cell line CT-26 and to explore possible mechanisms involved.
METHODS: CT-26 cells were divided into three groups: experimental group, control group, and blank group. The experimental group and control group were treated with a siRNA specific for the MIF gene (MIF siRNA) and a nonspecific siRNA, respectively, while the blank group was not treated with any agent. Transwell assay was used to determine cell invasion. ELISA was used to determine the level of MIF protein in cell supernatants, and the expression of MIF, CD74, tiam1 and E-cadherin mRNAs was detected by RT-PCR.
RESULTS: Twenty-four hours after treatment, cell invasion was significantly inhibited and the level of MIF protein in supernatants significantly declined in the experimental group compared to the control and blank groups (P = 0.012, 0.020). Compared to the control and blank groups, the expression of MIF, CD74 and tiam1 mRNAs decreased significantly and that of E-cadherin mRNA increased significantly (PE-Cadherin = 0.001) in the experimental group. In addition, the levels of MIF and CD74 proteins significantly declined in the experimental group compared to the control and the blank groups (PMIF = 0.006; PCD74 = 0.016).
CONCLUSION: MIF siRNA inhibits the invasion of CT-26 cells possibly by down-regulating the expression of MIF, CD74 and tiam1 and up-regulating the expression of E-cadherin.
Collapse
|
80
|
Jing Y, Han Z, Liu Y, Sun K, Zhang S, Jiang G, Li R, Gao L, Zhao X, Wu D, Cai X, Wu M, Wei L. Mesenchymal stem cells in inflammation microenvironment accelerates hepatocellular carcinoma metastasis by inducing epithelial-mesenchymal transition. PLoS One 2012; 7:e43272. [PMID: 22952657 PMCID: PMC3429457 DOI: 10.1371/journal.pone.0043272] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Accepted: 07/18/2012] [Indexed: 12/12/2022] Open
Abstract
In response to inflammation, mesenchymal stem cells (MSCs) are known to migrate to tissue injury sites to participate in immune modulation, tissue remodeling and wound healing. Tumors apply persistent mechanical and pathological stress to tissues and causes continual infiltration of MSCs. Here, we demonstrate that MSCs promote human hepatocellular carcinoma (HCC) metastasis under the influence of inflammation. The metastasis promoting effect could be imitated with the supernatant of MSCs pretreated with IFNγ and TNFα. Interestingly, treatment of HCC cells with the supernatant leads to epithelial-mesenchymal transition (EMT), an effect related to the production of TGFβ by cytokines stimulated MSCs. Importantly, the levels of MSCs expressing SSEA4 in clinical HCC samples significantly correlated with poor prognosis of HCC, and EMT of HCC was strongly associated with a shorter cancer-free interval (CFI) and a worse overall survival (OS). Therefore, our results suggest that MSCs in tumor inflammatory microenvironment could promote tumor metastasis through TGFβ-induced EMT.
Collapse
Affiliation(s)
- Yingying Jing
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Zhipeng Han
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Yan Liu
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Kai Sun
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Shanshan Zhang
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Guocheng Jiang
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Rong Li
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Lu Gao
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Xue Zhao
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Dong Wu
- Department of Combined Treatment, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Xiong Cai
- Department of Combined Treatment, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Mengchao Wu
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, People’s Republic of China
| | - Lixin Wei
- Tumor Immunology and Gene Therapy Center, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, People’s Republic of China
- * E-mail:
| |
Collapse
|
81
|
Walerych D, Napoli M, Collavin L, Del Sal G. The rebel angel: mutant p53 as the driving oncogene in breast cancer. Carcinogenesis 2012; 33:2007-17. [PMID: 22822097 PMCID: PMC3483014 DOI: 10.1093/carcin/bgs232] [Citation(s) in RCA: 216] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Breast cancer is the most frequent invasive tumor diagnosed in women, causing over 400 000 deaths yearly worldwide. Like other tumors, it is a disease with a complex, heterogeneous genetic and biochemical background. No single genomic or metabolic condition can be regarded as decisive for its formation and progression. However, a few key players can be pointed out and among them is the TP53 tumor suppressor gene, commonly mutated in breast cancer. In particular, TP53 mutations are exceptionally frequent and apparently among the key driving factors in triple negative breast cancer -the most aggressive breast cancer subgroup-whose management still represents a clinical challenge. The majority of TP53 mutations result in the substitution of single aminoacids in the central region of the p53 protein, generating a spectrum of variants ('mutant p53s', for short). These mutants lose the normal p53 oncosuppressive functions to various extents but can also acquire oncogenic properties by gain-of-function mechanisms. This review discusses the molecular processes translating gene mutations to the pathologic consequences of mutant p53 tumorigenic activity, reconciling cell and animal models with clinical outcomes in breast cancer. Existing and speculative therapeutic methods targeting mutant p53 are also discussed, taking into account the overlap of mutant and wild-type p53 regulatory mechanisms and the crosstalk between mutant p53 and other oncogenic pathways in breast cancer. The studies described here concern breast cancer models and patients-unless it is indicated otherwise and justified by the importance of data obtained in other models.
Collapse
Affiliation(s)
- Dawid Walerych
- Laboratorio Nazionale CIB (LNCIB), Area Science Park, 34149 Trieste, Italy
| | | | | | | |
Collapse
|
82
|
Khapare N, Kundu ST, Sehgal L, Sawant M, Priya R, Gosavi P, Gupta N, Alam H, Karkhanis M, Naik N, Vaidya MM, Dalal SN. Plakophilin3 loss leads to an increase in PRL3 levels promoting K8 dephosphorylation, which is required for transformation and metastasis. PLoS One 2012; 7:e38561. [PMID: 22701666 PMCID: PMC3368841 DOI: 10.1371/journal.pone.0038561] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Accepted: 05/08/2012] [Indexed: 12/13/2022] Open
Abstract
The desmosome anchors keratin filaments in epithelial cells leading to the formation of a tissue wide IF network. Loss of the desmosomal plaque protein plakophilin3 (PKP3) in HCT116 cells, leads to an increase in neoplastic progression and metastasis, which was accompanied by an increase in K8 levels. The increase in levels was due to an increase in the protein levels of the Phosphatase of Regenerating Liver 3 (PRL3), which results in a decrease in phosphorylation on K8. The increase in PRL3 and K8 protein levels could be reversed by introduction of an shRNA resistant PKP3 cDNA. Inhibition of K8 expression in the PKP3 knockdown clone S10, led to a decrease in cell migration and lamellipodia formation. Further, the K8 PKP3 double knockdown clones showed a decrease in colony formation in soft agar and decreased tumorigenesis and metastasis in nude mice. These results suggest that a stabilisation of K8 filaments leading to an increase in migration and transformation may be one mechanism by which PKP3 loss leads to tumor progression and metastasis.
Collapse
Affiliation(s)
- Nileema Khapare
- KS215, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Samrat T. Kundu
- KS215, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Lalit Sehgal
- KS215, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Mugdha Sawant
- KS215, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Rashmi Priya
- KS215, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Prajakta Gosavi
- KS215, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Neha Gupta
- KS215, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Hunain Alam
- KS215, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Madhura Karkhanis
- Pharmacology Department, Piramal Life Sciences Ltd., Mumbai, Maharashtra, India
| | - Nishigandha Naik
- Pharmacology Department, Piramal Life Sciences Ltd., Mumbai, Maharashtra, India
| | - Milind M. Vaidya
- KS215, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| | - Sorab N. Dalal
- KS215, Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar Node, Navi Mumbai, Maharashtra, India
| |
Collapse
|
83
|
Fujita T, Gotohda N, Kato Y, Kinoshita T, Takahashi S, Konishi M, Daiko H, Nishimura M, Kuwata T, Ochiai A, Kinoshita T. Clinicopathological features of stomach cancer with invasive micropapillary component. Gastric Cancer 2012; 15:179-87. [PMID: 21987353 DOI: 10.1007/s10120-011-0094-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 08/28/2011] [Indexed: 02/07/2023]
Abstract
BACKGROUND Invasive micropapillary carcinoma has been recognized as a rare disease entity with aggressive tumor behavior. However, few reports have described invasive micropapillary carcinoma in the gastrointestinal tract, particularly its involvement in gastric cancer. METHODS We retrospectively analyzed 930 patients diagnosed with gastric cancer who underwent gastrectomy, and we then histopathologically evaluated the existence of a regional invasive micropapillary component. Clinicopathological features were investigated in patients with an invasive micropapillary component and compared with such features in 100 patients with gastric adenocarcinoma, selected as stage-matched controls, who underwent gastrectomy during the same period. RESULTS Of the 930 patients, 14 were histopathologically diagnosed with gastric cancer with a regional invasive micropapillary component. There were no significant differences in age, gender, tumor location, macroscopic type, or type of surgery between patients with an invasive micropapillary component and the pT-matched controls. Histopathologically, significant differences were observed in lymphatic infiltration, venous invasion, the percentage of cases with lymph node metastasis, and the median number of metastatic lymph nodes. The three-year disease-free and overall survival rates of patients with an invasive micropapillary component were 40.5 and 59.3%, respectively, compared with those for the stage-matched controls, which were 72.6 and 80.6%, respectively (p = 0.02 and 0.07). CONCLUSIONS Patients with gastric cancer with a regional invasive micropapillary component showed marked cancer infiltration in the lymphatic pathway and poor prognosis after gastrectomy.
Collapse
Affiliation(s)
- Takeo Fujita
- Division of Digestive Surgery, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba 277-8577, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Screening of drugs to counteract human papillomavirus 16 E6 repression of E-cadherin expression. Invest New Drugs 2012; 30:2236-51. [PMID: 22359217 DOI: 10.1007/s10637-012-9803-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 02/10/2012] [Indexed: 12/11/2022]
Abstract
Persistent infections with certain high-risk human papillomavirus (HPV) types such as 16 and 18 can result in the development of cervical cancer. Neither of the two prophylactic vaccines against HPV16 and 18 that are in current use have any therapeutic efficacy for prevalent HPV infections. Ablative therapy is widely used for the treatment of HPV cervical dysplasia however disease recurrence is a widely recognized problem. Thus there is a continuing need for therapeutic approaches for the treatment of HPV infections. The HPV16 E6 viral oncoprotein represses surface expression of the cellular adhesion molecule, E-cadherin. Reduced E-cadherin expression on HPV-infected keratinocytes is associated with lowered numbers of antigen-presenting Langerhans cells in the infected epidermis, potentially reducing immune surveillance for HPV. Four chemicals reported to up-regulate E-cadherin were screened for their ability to counteract E6 repression of surface E-cadherin. 5-Aza-2'-deoxycytidine (AzaDC), a DNA methyltransferase inhibitor, and Indole-3-carbinol (I3C), reported to increase E-cadherin through a p21(Waf1/Cip1)-dependent mechanism, had low cytotoxicity and increased or restored E-cadherin expression and adhesive function in HPV16 E6 expressing HCT116 cells. Doxorubicin, also known to induce p21(Waf1/Cip1), increased E-cadherin in E6 expressing cells but had some associated cytotoxicity. Tamoxifen, which can restore adhesive function of surface E-cadherin, was ineffective in counteracting E6 repression of E-cadherin. AzaDC and I3C both show potential to restore antigen-presenting cells to HPV infected skin by antagonizing E6 repression of E-cadherin, thereby counteracting an important immune evasion mechanism of HPV16 and reinstating immune function at the infected site.
Collapse
|
85
|
Tang FY, Pai MH, Chiang EPI. Consumption of high-fat diet induces tumor progression and epithelial-mesenchymal transition of colorectal cancer in a mouse xenograft model. J Nutr Biochem 2012; 23:1302-13. [PMID: 22221675 DOI: 10.1016/j.jnutbio.2011.07.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Revised: 06/09/2011] [Accepted: 07/27/2011] [Indexed: 12/22/2022]
Abstract
Epidemiologic studies suggest that intake of high-fat diet (HFD) promotes colon carcinogenesis. Epithelial-mesenchymal transition (EMT) and inflammation play important roles during tumor progression of colorectal cancer (CRC). Oncogenic pathways such as phosphatidylinositol-3-kinase (PI3K)/Akt/mTOR and mitogen-activated protein kinase (MAPK)/ERK signaling cascades induce EMT and inflammation in cancer. No experimental evidence has been demonstrated regarding HFD-mediated tumor progression including EMT in CRC so far. Our results demonstrated that HFD consumption could induce tumor growth and progression, including EMT and inflammation, in a mouse xenograft tumor model. The molecular mechanisms were through activation of MAPK/ERK and PI3K/Akt/mTOR signaling pathways. HFD induced up-regulation of cyclooxygenase-2, cyclin D1 and proliferating cell nuclear antigen proteins concomitant with increases in expression of nuclear factor-κB p65 (RelA) and β-catenin proteins. Surprisingly, HFD consumption could suppress p21(CIP1/WAF1) expression through increases in nuclear histone deacetylase complex (HDAC). Moreover, HFD could mediate the disassembly of E-cadherin adherent complex and the up-regulation of Vimentin and N-cadherin proteins in tumor tissues. Taken together, our novel findings support evidence for HFD-mediated modulation of HDAC activity and activation of oncogenic cascades, which involve EMT and inflammation in CRC, playing important roles in tumor growth and progression in a mouse xenograft model.
Collapse
Affiliation(s)
- Feng-Yao Tang
- Department of Nutrition, Biomedical Science Laboratory, China Medical University, Taichung 40402, Taiwan.
| | | | | |
Collapse
|
86
|
Yi Q, Zhao X, Huang Y, Ma T, Zhang Y, Hou H, Cooke HJ, Yang DQ, Wu M, Shi Q. p53 dependent centrosome clustering prevents multipolar mitosis in tetraploid cells. PLoS One 2011; 6:e27304. [PMID: 22076149 PMCID: PMC3208627 DOI: 10.1371/journal.pone.0027304] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 10/13/2011] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND p53 abnormality and aneuploidy often coexist in human tumors, and tetraploidy is considered as an intermediate between normal diploidy and aneuploidy. The purpose of this study was to investigate whether and how p53 influences the transformation from tetraploidy to aneuploidy. PRINCIPAL FINDINGS Live cell imaging was performed to determine the fates and mitotic behaviors of several human and mouse tetraploid cells with different p53 status, and centrosome and spindle immunostaining was used to investigate centrosome behaviors. We found that p53 dominant-negative mutation, point mutation, or knockout led to a 2∼ 33-fold increase of multipolar mitosis in N/TERT1, 3T3 and mouse embryonic fibroblasts (MEFs), while mitotic entry and cell death were not significantly affected. In p53-/- tetraploid MEFs, the ability of centrosome clustering was compromised, while centrosome inactivation was not affected. Suppression of RhoA/ROCK activity by specific inhibitors in p53-/- tetraploid MEFs enhanced centrosome clustering, decreased multipolar mitosis from 38% to 20% and 16% for RhoA and ROCK, respectively, while expression of constitutively active RhoA in p53+/+ tetraploid 3T3 cells increased the frequency of multipolar mitosis from 15% to 35%. CONCLUSIONS p53 could not prevent tetraploid cells entering mitosis or induce tetraploid cell death. However, p53 abnormality impaired centrosome clustering and lead to multipolar mitosis in tetraploid cells by modulating the RhoA/ROCK signaling pathway.
Collapse
Affiliation(s)
- Qiyi Yi
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaoyu Zhao
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Yun Huang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Tieliang Ma
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Yingyin Zhang
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Heli Hou
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Howard J. Cooke
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
- MRC Human Genetics Unit and Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Da-Qing Yang
- Sanford Research/University of South Dakota, Sanford Health, Sioux Falls, South Dakota, United States of America
| | - Mian Wu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
| | - Qinghua Shi
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, China
- * E-mail:
| |
Collapse
|
87
|
Zlobec I, Lugli A. Epithelial mesenchymal transition and tumor budding in aggressive colorectal cancer: tumor budding as oncotarget. Oncotarget 2011; 1:651-61. [PMID: 21317460 PMCID: PMC3248128 DOI: 10.18632/oncotarget.199] [Citation(s) in RCA: 253] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Epithelial mesenchymal transition (EMT) is proposed as a critical mechanism for the acquisition of malignant phenotypes by epithelial cells. In colorectal cancer, tumor cells having undergone EMT are histologically represented by the presence of tumor buds defined as single cells or small clusters of de-differentiated tumor cells at the invasive front. Tumor budding is not a static, histological feature rather it represents a snap-shot of a dynamic process undertaken by an aggressive tumor with the potential to disseminate and metastasize. Strong, consistent evidence shows that tumor budding is a predictor of lymph node metastasis, distant metastatic disease, local recurrence, worse overall and disease-free survival time and an independent prognostic factor. Moreover, the International Union against Cancer (UICC) recognizes tumor budding as a highly relevant, additional prognostic parameter. The aim of this review is to summarize the evidence supporting the implementation of tumor budding into diagnostic pathology and patient management and additionally to illustrate its worthiness as a potential therapeutic target.
Collapse
Affiliation(s)
- Inti Zlobec
- Institute for Pathology, University Hospital Basel, Schoenbeinstrasse 40, Basel, Switzerland
| | | |
Collapse
|
88
|
Chang HY, Kao MC, Way TD, Ho CT, Fu E. Diosgenin suppresses hepatocyte growth factor (HGF)-induced epithelial-mesenchymal transition by down-regulation of Mdm2 and vimentin. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:5357-5363. [PMID: 21504235 DOI: 10.1021/jf200598w] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Substantial activation of the hepatocyte growth factor (HGF)/c-Met pathway leads to cancer cell scattering and invasion and has been observed in several types of cancers, including prostate and colorectal cancers. The phosphorylation cascade downstream of HGF, particularly PI3K/Akt signaling, regulates epithelial-to-mesenchymal transition (EMT). How this signaling governs EMT and whether specific kinases respond to particular EMT effectors remain unclear. This study found specific increases in Mdm2 and vimentin rather than the coregulation of an array of EMT marker proteins in response to HGF-induced EMT in DU145 prostate cancer cells. Importantly, it was further found that diosgenin abrogated HGF-induced DU145 cell scattering and invasion. Moreover, diosgenin effectively inhibited the HGF-induced increases in Mdm2 and vimentin by down-regulating phosphorylated Akt and mTOR. In summary, the results suggest that diosgenin may be a potential compound for use in prostate cancer therapy to target the major HGF-induced EMT pathway.
Collapse
Affiliation(s)
- Hsiang-Yu Chang
- Graduate Institute of Life Sciences, National Defense Medical Center and Academia Sinica, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
89
|
Jiang Y, Xie X, Li Z, Wang Z, Zhang Y, Ling ZQ, Ling Z, Pan Y, Wang Z, Chen Y. Functional cooperation of RKTG with p53 in tumorigenesis and epithelial-mesenchymal transition. Cancer Res 2011; 71:2959-68. [PMID: 21385899 DOI: 10.1158/0008-5472.can-10-4077] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Raf kinase trapping to Golgi (RKTG) is a potential tumor suppressor gene due to its negative roles in regulating Ras/Raf/MEK/ERK (extracellular signal-regulated kinase) pathway and GPCR (G protein-coupled receptor) Gβγ subunit signaling. Interestingly, RKTG-deficient mice are free of tumors, although they are prone to form skin cancer on carcinogen administration. On the other hand, p53 is a well-characterized tumor suppressor gene and p53 heterozygous mice develop sarcoma and other tumors starting from 12 months of age. In RKTG-null mouse embryonic fibroblasts, lypophosphatidic acid (LPA), but not EGF (epidermal growth factor), could stimulate hyperphosphorylation of AKT and GSK3β, accompanied by increases in phosphorylation of p53 at Ser15 and accumulation of p53, as well as its target genes p21 and p16. Spontaneous skin cancer-like tumors were detected in about 25% of RKTG nullizygous and p53 heterozygous mice within 7 months of age. Hyperplasia and epithelial-mesenchymal transition (EMT) were observed in the tumor-overlying epidermis, in which LOH of p53 occurred and EMT features emerged. In p53-mutated A431 epithelial carcinoma cells, knockdown of RKTG led to enhancement of LPA-stimulated AKT and GSK3β phosphorylation, together with increased accumulation of β-catenin and appearance of EMT features that were antagonized by p53 overexpression. In HepG2 epithelial cells, LPA-stimulated AKT phosphorylation and EMT features reached maximum when both RKTG and p53 were simultaneously silenced. In summary, these results not only indicate that RKTG has an in vivo tumor suppressor function to cooperate with p53 in tumorigenesis but also suggest that p53 has an EMT checkpoint function and the loss of this function can combine with loss of RKTG to drive EMT and tumor progression.
Collapse
Affiliation(s)
- Yuhui Jiang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Emami S. Interplay between p53-family, their regulators, and PARPs in DNA repair. Clin Res Hepatol Gastroenterol 2011; 35:98-104. [PMID: 21177056 DOI: 10.1016/j.gcb.2010.10.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2010] [Revised: 09/28/2010] [Accepted: 10/04/2010] [Indexed: 02/07/2023]
Abstract
Abnormalities of the p53 tumor suppressor gene are among the most frequent molecular events in human neoplasia. p53 is consequently one of the most studied proteins, and is the subject of over 55,500 scientific papers. In this review, attention is focused on the functions of p53 in DNA repair. We highlight the recent progress in the analysis of protein signals to p53, including PARPs, and ubiquitination cascade proteins MDM2, CRM1, USP10 and 14-3-3σ.
Collapse
Affiliation(s)
- S Emami
- Inserm UMR S938, centre de recherche Saint-Antoine, université Pierre-et-Marie-Curie (université Paris-6), 184, rue du faubourg-Saint-Antoine, 75571 Paris cedex 12, France.
| |
Collapse
|
91
|
Puppin C, Passon N, Frasca F, Vigneri R, Tomay F, Tomaciello S, Damante G. In thyroid cancer cell lines expression of periostin gene is controlled by p73 and is not related to epigenetic marks of active transcription. Cell Oncol (Dordr) 2011; 34:131-40. [DOI: 10.1007/s13402-011-0009-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2010] [Indexed: 12/26/2022] Open
|
92
|
Cheng JC, Auersperg N, Leung PCK. Inhibition of p53 induces invasion of serous borderline ovarian tumor cells by accentuating PI3K/Akt-mediated suppression of E-cadherin. Oncogene 2010; 30:1020-31. [PMID: 20972462 DOI: 10.1038/onc.2010.486] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Serous borderline ovarian tumors (SBOTs) are slow-growing, non-invasive ovarian epithelial neoplasms. SBOTs are considered to be distinct entities that give rise to invasive low-grade serous carcinomas (LGCs), which have a relatively poor prognosis and are unrelated to high-grade serous carcinomas (HGCs). The mechanisms underlying the progression of non-invasive SBOTs to invasive epithelial ovarian carcinomas are not understood. We recently established short-term cultures of SBOT cells from tumor biopsies and showed that inactivation of p53, retinoblastoma (Rb) and/or PP2A by the simian virus 40 (SV40) large (LT) and small T antigens extends the life span of the cells and endows them with the ability to invade Matrigel-coated transwells. In this study, we show that concurrent inhibition of p53 and Rb by the SV40 LT produces cells (referred to as SBOT4-LT) with increased life span and cell invasion. To distinguish the roles of p53 and Rb in the progression from SBOTs to invasive ovarian carcinomas, we performed small interfering RNA-mediated knockdown of endogenous p53 in a spontaneously immortalized SBOT cell line, SBOT3.1, which increased cell invasion. This increased invasive activity was associated with the transcriptional downregulation of E-cadherin, correlated with an increase in PIK3CA levels and the increased activation of Akt. Conversely, in invasive LGC-derived MPSC1 cells, enhancing the levels of p53 decreased cell invasion and diminished the phosphatidylinositol 3-kinase (PI3K)/Akt-mediated downregulation of E-cadherin. Inhibition of Rb also enhanced invasiveness, but did not affect the levels of PIK3CA and E-cadherin in SBOT3.1 cells, suggesting that it functions by a different pathway. To our knowledge, this study is the first to show that p53 has an important role in the progression from SBOTs to invasive carcinomas. In addition, our findings suggest that downregulation of E-cadherin by the PI3K/Akt pathway contributes to this progression.
Collapse
Affiliation(s)
- J-C Cheng
- Department of Obstetrics and Gynecology, Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | |
Collapse
|