1
|
Governa P, Romagnoli G, Albanese P, Rossi F, Manetti F, Biagi M. Effect of in vitro simulated digestion on the anti- Helicobacter Pylori activity of different Propolis extracts. J Enzyme Inhib Med Chem 2023; 38:2183810. [PMID: 36916299 PMCID: PMC10026752 DOI: 10.1080/14756366.2023.2183810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023] Open
Abstract
Helicobacter pylori (HP) is among the most common pathogens causing infection in humans worldwide. Oxidative stress and gastric inflammation are involved in the progression of HP-related gastric diseases, and they can be targeted by integrating conventional antibiotic treatment with polyphenol-enriched natural products. In this work, we characterised three different propolis extracts and evaluated their stability under in vitro simulated gastric digestion, compared to their main constituents alone. The extract with the highest stability to digestion (namely, the dark propolis extract, DPE) showed a minimum bactericidal concentration (MBC) lower than 1 mg/mL on HP strains with different virulence factors. Finally, since urease is one of the virulence factors contributing to the establishment of a microenvironment that promotes HP infection, we evaluated the possible inhibition of this enzyme by using molecular docking simulations and in vitro colorimetric assay, showing that galangin and pinocembrin may be involved in this activity.
Collapse
Affiliation(s)
- Paolo Governa
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Giulia Romagnoli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Paola Albanese
- Department of Physical Sciences, Earth and Environment, University of Siena, Siena, Italy
| | - Federico Rossi
- Department of Physical Sciences, Earth and Environment, University of Siena, Siena, Italy
| | - Fabrizio Manetti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Marco Biagi
- Department of Physical Sciences, Earth and Environment, University of Siena, Siena, Italy
| |
Collapse
|
2
|
Zhao YX, Ma LB, Yang Z, Wang F, Wang HY, Dang JY. Cancerous inhibitor of protein phosphatase 2A enhances chemoresistance of gastric cancer cells to oxaliplatin. World J Gastrointest Oncol 2023; 15:286-302. [PMID: 36908323 PMCID: PMC9994047 DOI: 10.4251/wjgo.v15.i2.286] [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] [Received: 09/08/2022] [Revised: 11/23/2022] [Accepted: 01/05/2023] [Indexed: 02/14/2023] Open
Abstract
BACKGROUND Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a newly discovered oncogene. It is an active cell proliferation regulatory factor that inhibits tumor apoptosis in gastric cancer (GC) cells. CIP2A is functionally related to chemoresistance in various types of tumors according to recent studies. The underlying mechanism, however, is unknown. Further, the primary treatment regimen for GC is oxaliplatin-based chemotherapy. Nonetheless, it often fails due to chemoresistance of GC cells to oxaliplatin.
AIM The goal of this study was to examine CIP2A expression and its association with oxaliplatin resistance in human GC cells.
METHODS Immunohistochemistry was used to examine CIP2A expression in GC tissues and adjacent normal tissues. CIP2A expression in GC cell lines was reduced using small interfering RNA. After confirming the silencing efficiency, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide tetrazolium and flow cytometry assays were used to evaluate cell proliferation and apoptosis caused by oxaliplatin treatment. Further, the key genes and protein changes were verified using real-time quantitative reverse transcription PCR and Western blotting, respectively, before and after intervention. For bioinformatics analysis, we used the R software and Bioconductor project. For statistical analysis, we used GraphPad Prism 6.0 and the Statistical Package for the Social Sciences software version 20.0 (IBM, Armonk, United States).
RESULTS A high level of CIP2A expression was associated with tumor size, T stage, lymph node metastasis, Tumor Node Metastasis stage, and a poor prognosis. Further, CIP2A expression was higher in GC cells than in normal human gastric epithelial cells. Using small interfering RNA against CIP2A, we discovered that CIP2A knockdown inhibited cell proliferation and significantly increased GC cell sensitivity to oxaliplatin. Moreover, CIP2A knockdown enhanced oxaliplatin-induced apoptosis in GC cells. Hence, high CIP2A levels in GC may be a factor in chemoresistance to oxaliplatin. In human GC cells, CIP2A regulated protein kinase B phosphorylation, and chemical inhibition of the protein kinase B signaling pathway was significantly associated with increased sensitivity to oxaliplatin. Therefore, the protein kinase B signaling pathway was correlated with CIP2A-enhanced chemoresistance of human GC cells to oxaliplatin.
CONCLUSION CIP2A expression could be a novel therapeutic strategy for chemoresistance in GC.
Collapse
Affiliation(s)
- Yong-Xun Zhao
- The Seventh Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Li-Bin Ma
- The Seventh Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Ze Yang
- The Seventh Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Fang Wang
- Department of Pathology, The First Hospital of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Hui-Ying Wang
- The First Clinical Medical School, Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Jia-Yao Dang
- The First Clinical Medical School, Lanzhou University, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
3
|
Kashani E, Vassella E. Pleiotropy of PP2A Phosphatases in Cancer with a Focus on Glioblastoma IDH Wildtype. Cancers (Basel) 2022; 14:5227. [PMID: 36358647 PMCID: PMC9654311 DOI: 10.3390/cancers14215227] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/13/2022] [Accepted: 10/20/2022] [Indexed: 07/29/2023] Open
Abstract
Serine/Threonine protein phosphatase 2A (PP2A) is a heterotrimeric (or occasionally, heterodimeric) phosphatase with pleiotropic functions and ubiquitous expression. Despite the fact that they all contribute to protein dephosphorylation, multiple PP2A complexes exist which differ considerably by their subcellular localization and their substrate specificity, suggesting diverse PP2A functions. PP2A complex formation is tightly regulated by means of gene expression regulation by transcription factors, microRNAs, and post-translational modifications. Furthermore, a constant competition between PP2A regulatory subunits is taking place dynamically and depending on the spatiotemporal circumstance; many of the integral subunits can outcompete the rest, subjecting them to proteolysis. PP2A modulation is especially important in the context of brain tumors due to its ability to modulate distinct glioma-promoting signal transduction pathways, such as PI3K/Akt, Wnt, Ras, NF-κb, etc. Furthermore, PP2A is also implicated in DNA repair and survival pathways that are activated upon treatment of glioma cells with chemo-radiation. Depending on the cancer cell type, preclinical studies have shown some promise in utilising PP2A activator or PP2A inhibitors to overcome therapy resistance. This review has a special focus on "glioblastoma, IDH wild-type" (GBM) tumors, for which the therapy options have limited efficacy, and tumor relapse is inevitable.
Collapse
Affiliation(s)
- Elham Kashani
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Erik Vassella
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
4
|
Sharafutdinov I, Ekici A, Vieth M, Backert S, Linz B. Early and late genome-wide gastric epithelial transcriptome response during infection with the human carcinogen Helicobacterpylori. CELL INSIGHT 2022; 1:100032. [PMID: 37193047 PMCID: PMC10120309 DOI: 10.1016/j.cellin.2022.100032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 05/18/2023]
Abstract
Infection of the stomach by Helicobacter pylori is a major risk factor for the development of gastric cancer. Colonization of the gastric epithelium leads to the activation of multiple disease-related signaling pathways. Serine protease HtrA represents an important secreted virulence factor that mediates cleavage of cellular junctions. However, its potential role in nuclear responses is unknown. Here, we performed a genome-wide RNA-seq analysis of polarized gastric epithelial cells infected by wild-type (wt) and ΔhtrA mutant bacteria. Fluorescence microscopy showed that H. pylori wt, but not ΔhtrA bacteria, preferably localized at cellular junctions. Our results pinpointed early (2 h) and late (6 h) transcriptional responses, with most differentially expressed genes at 6 h post infection. The transcriptomes revealed HtrA-dependent targeting of genes associated with inflammation and apoptosis (e.g. IL8, ZFP36, TNF). Accordingly, infection with the ΔhtrA mutant induced increased apoptosis rates in host cells, which was associated with reduced H. pylori CagA expression. In contrast, transcription of various carcinogenesis-associated genes (e.g. DKK1, DOCK8) was affected by H. pylori independent of HtrA. These findings suggest that H. pylori disturbs previously unknown molecular pathways in an HtrA-dependent and HtrA-independent manner, and provide valuable new insights of this significant pathogen in humans and thus potential targets for better controlling the risk of malignant transformation.
Collapse
Affiliation(s)
- Irshad Sharafutdinov
- Department of Biology, Division of Microbiology, Friedrich Alexander Universität Erlangen-Nürnberg, Staudtstr. 5, D-91058, Erlangen, Germany
| | - Arif Ekici
- Institute of Human Genetics, University Hospital, Friedrich Alexander Universität Erlangen-Nürnberg, Schwabachanlage 10, D-91054, Erlangen, Germany
| | - Michael Vieth
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Klinikum Bayreuth, Preuschwitzer Str 101, D-95445, Bayreuth, Germany
| | - Steffen Backert
- Department of Biology, Division of Microbiology, Friedrich Alexander Universität Erlangen-Nürnberg, Staudtstr. 5, D-91058, Erlangen, Germany
| | - Bodo Linz
- Department of Biology, Division of Microbiology, Friedrich Alexander Universität Erlangen-Nürnberg, Staudtstr. 5, D-91058, Erlangen, Germany
| |
Collapse
|
5
|
Son HE, Jang WG. Cip2A modulates osteogenic differentiation via the ERK-Runx2 pathway in MG63 cells. Biofactors 2021; 47:658-664. [PMID: 34077593 DOI: 10.1002/biof.1760] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/08/2021] [Indexed: 01/25/2023]
Abstract
Cancerous inhibitor of protein phosphatase 2A (Cip2A) is an oncoprotein that promotes the development of several types of cancer. However, its molecular function in osteoblast differentiation remains unclear. In this study, we found that Cip2A was upregulated under osteogenic conditions in MG63 cells. Besides, overexpression of Cip2A significantly increased the expression of Runt-related transcription factor 2 (Runx2) and alkaline phosphatase (ALP). Inversely, the knockdown of Cip2A in MG63 cells suppressed osteoblast differentiation. Cip2A expression during osteogenic differentiation was mediated by extracellular signal-regulated kinase (ERK) activation. Taken together, our results suggest that Cip2A plays important role in regulating osteoblast differentiation by inducing ERK phosphorylation in MG63 cells.
Collapse
Affiliation(s)
- Hyo-Eun Son
- Department of Biotechnology, School of Engineering, Daegu University, Gyeongbuk, Republic of Korea
- Research Institute of Anti-Aging, Daegu University, Gyeongbuk, Republic of Korea
| | - Won-Gu Jang
- Department of Biotechnology, School of Engineering, Daegu University, Gyeongbuk, Republic of Korea
- Research Institute of Anti-Aging, Daegu University, Gyeongbuk, Republic of Korea
| |
Collapse
|
6
|
Mahoney KE, Shabanowitz J, Hunt DF. MHC Phosphopeptides: Promising Targets for Immunotherapy of Cancer and Other Chronic Diseases. Mol Cell Proteomics 2021; 20:100112. [PMID: 34129940 PMCID: PMC8724925 DOI: 10.1016/j.mcpro.2021.100112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 05/11/2021] [Accepted: 06/02/2021] [Indexed: 12/27/2022] Open
Abstract
Major histocompatibility complex-associated peptides have been considered as potential immunotherapeutic targets for many years. MHC class I phosphopeptides result from dysregulated cell signaling pathways that are common across cancers and both viral and bacterial infections. These antigens are recognized by central memory T cells from healthy donors, indicating that they are considered antigenic by the immune system and that they are presented across different individuals and diseases. Based on these responses and the similar dysregulation, phosphorylated antigens are promising candidates for prevention or treatment of different cancers as well as a number of other chronic diseases.
Collapse
Affiliation(s)
- Keira E Mahoney
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA
| | - Jeffrey Shabanowitz
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA.
| | - Donald F Hunt
- Department of Chemistry, University of Virginia, Charlottesville, Virginia, USA; Department of Pathology, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
7
|
Denk S, Schmidt S, Schurr Y, Schwarz G, Schote F, Diefenbacher M, Armendariz C, Dejure F, Eilers M, Wiegering A. CIP2A regulates MYC translation (via its 5'UTR) in colorectal cancer. Int J Colorectal Dis 2021; 36:911-918. [PMID: 33078202 PMCID: PMC8178152 DOI: 10.1007/s00384-020-03772-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/07/2020] [Indexed: 02/04/2023]
Abstract
BACKGROUND Deregulated expression of MYC is a driver of colorectal carcinogenesis, suggesting that decreasing MYC expression may have significant therapeutic value. CIP2A is an oncogenic factor that regulates MYC expression. CIP2A is overexpressed in colorectal cancer (CRC), and its expression levels are an independent marker for long-term outcome of CRC. Previous studies suggested that CIP2A controls MYC protein expression on a post-transcriptional level. METHODS To determine the mechanism by which CIP2A regulates MYC in CRC, we dissected MYC translation and stability dependent on CIP2A in CRC cell lines. RESULTS Knockdown of CIP2A reduced MYC protein levels without influencing MYC stability in CRC cell lines. Interfering with proteasomal degradation of MYC by usage of FBXW7-deficient cells or treatment with the proteasome inhibitor MG132 did not rescue the effect of CIP2A depletion on MYC protein levels. Whereas CIP2A knockdown had marginal influence on global protein synthesis, we could demonstrate that, by using different reporter constructs and cells expressing MYC mRNA with or without flanking UTR, CIP2A regulates MYC translation. This interaction is mainly conducted by the MYC 5'UTR. CONCLUSIONS Thus, instead of targeting MYC protein stability as reported for other tissue types before, CIP2A specifically regulates MYC mRNA translation in CRC but has only slight effects on global mRNA translation. In conclusion, we propose as novel mechanism that CIP2A regulates MYC on a translational level rather than affecting MYC protein stability in CRC.
Collapse
Affiliation(s)
- S. Denk
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany ,Department of General, Visceral, Transplant, Vascular and Pediatric Surgery (Department of Surgery I), University Hospital Würzburg, Oberduerrbacherstr. 6, 97080 Würzburg, Germany
| | - S. Schmidt
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany ,Department of General, Visceral, Transplant, Vascular and Pediatric Surgery (Department of Surgery I), University Hospital Würzburg, Oberduerrbacherstr. 6, 97080 Würzburg, Germany
| | - Y. Schurr
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - G. Schwarz
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - F. Schote
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - M. Diefenbacher
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - C. Armendariz
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - F. Dejure
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - M. Eilers
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany ,Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | - Armin Wiegering
- Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany ,Department of General, Visceral, Transplant, Vascular and Pediatric Surgery (Department of Surgery I), University Hospital Würzburg, Oberduerrbacherstr. 6, 97080 Würzburg, Germany ,Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| |
Collapse
|
8
|
Zendehdel A, Roham M. Biological evidence of the relationship between
Helicobacter pylori
and associated extragastric diseases. J Cell Biochem 2019; 120:12128-12140. [PMID: 30977160 DOI: 10.1002/jcb.28681] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/07/2019] [Accepted: 02/14/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Abolfazl Zendehdel
- Department of Geriatric Medicine, Ziaeian Hospital Tehran University of Medical Sciences Tehran Iran
| | - Maryam Roham
- Antimicrobial‐Resistant Research Center Iran University of Medical Sciences Tehran Iran
| |
Collapse
|
9
|
Nath S, Ohlmeyer M, Salathe MA, Poon J, Baumlin N, Foronjy RF, Geraghty P. Chronic Cigarette Smoke Exposure Subdues PP2A Activity by Enhancing Expression of the Oncogene CIP2A. Am J Respir Cell Mol Biol 2018; 59:695-705. [PMID: 30011381 PMCID: PMC12057641 DOI: 10.1165/rcmb.2018-0173oc] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 06/25/2018] [Indexed: 12/16/2022] Open
Abstract
Phosphatase activity of the major serine threonine phosphatase, protein phosphatase 2A (PP2A), is blunted in the airways of individuals with chronic obstructive pulmonary disease (COPD), which results in heightened inflammation and proteolytic responses. The objective of this study was to investigate how PP2A activity is modulated in COPD airways. PP2A activity and endogenous inhibitors of PP2A were investigated in animal and cell models of COPD. In primary human bronchial epithelial (HBE) cells isolated from smokers and donors with COPD, we observed enhanced expression of cancerous inhibitor of PP2A (CIP2A), an oncoprotein encoded by the KIAA1524 gene, compared with cells from nonsmokers. CIP2A expression was induced by chronic cigarette smoke exposure in mice that coincided with a reduction in PP2A activity, airspace enlargements, and loss of lung function, as determined by PP2A phosphatase activity, mean linear intercept analysis, and forced expiratory volume in 0.05 second/forced vital capacity. Modulating CIP2A expression in HBE cells by silencing RNA or chemically with erlotinib enhanced PP2A activity, reduced extracellular-signal-regulated kinase phosphorylation, and reduced the responses of matrix metalloproteinases 1 and 9 in HBE cells isolated from subjects with COPD. Enhanced epithelial growth factor receptor responses in cells from subjects with COPD were observed to modulate CIP2A expression levels. Our study indicates that chronic cigarette smoke induction of epithelial growth factor receptor signaling and CIP2A expression can impair PP2A responses that are associated with loss of lung function and enhancement of proteolytic responses. Augmenting PP2A activity by manipulating CIP2A expression may represent a feasible therapeutic approach to counter smoke-induced lung disease.
Collapse
Affiliation(s)
- Sridesh Nath
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | | | - Matthias A Salathe
- 3 Division of Pulmonary, Critical Care, and Sleep Medicine, University of Miami, Miami, Florida; and
- 4 Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Justin Poon
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - Nathalie Baumlin
- 3 Division of Pulmonary, Critical Care, and Sleep Medicine, University of Miami, Miami, Florida; and
- 4 Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Robert F Foronjy
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
- 5 Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, New York
| | - Patrick Geraghty
- 1 Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
- 5 Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, New York
| |
Collapse
|
10
|
Park B, Lim JW, Kim H. Lycopene treatment inhibits activation of Jak1/Stat3 and Wnt/β-catenin signaling and attenuates hyperproliferation in gastric epithelial cells. Nutr Res 2018; 70:70-81. [PMID: 30098838 DOI: 10.1016/j.nutres.2018.07.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 07/05/2018] [Accepted: 07/19/2018] [Indexed: 12/19/2022]
Abstract
Helicobacter pylori (H pylori) colonizes the human stomach and increases the risk of gastric diseases including gastric cancer. H pylori increases reactive oxygen species (ROS), which activate Janus-activator kinase 1 (Jak1)/signal transducers and activators of transcription 3 (Stat3) in gastric epithelial cells. ROS mediate hyperproliferation, a hallmark of carcinogenesis, by activating Wnt/β-catenin signaling in various cells. Lycopene is a potent antioxidant exhibiting anticancer effects. We hypothesized that lycopene may inhibit H pylori-induced hyperproliferation by suppressing ROS-mediated activation of Jak1/Stat3 and Wnt/β-catenin signaling, and β-catenin target gene expression in gastric epithelial cells. We determined cell viability, ROS levels, and the protein levels of phospho- and total Jak1/Stat3, Wnt/β-catenin signaling molecules, Wnt-1, lipoprotein-related protein 5, and β-catenin target oncogenes (c-Myc and cyclin E) in H pylori-infected gastric epithelial AGS cells. The Jak1/Stat3 inhibitor AG490 served as the control treatment. The significance of the differences among groups was calculated using the 1-way analysis of variance followed by Newman-Keuls post hoc tests. The results show that lycopene reduced ROS levels and inhibited Jak1/Stat3 activation, alteration of Wnt/β-catenin multiprotein complex molecules, expression of c-Myc and cyclin E, and cell proliferation in H pylori-infected AGS cells. AG490 similarly inhibited H pylori-induced cell proliferation, alteration of Wnt/β-catenin multiprotein complex molecules, and oncogene expression. H pylori increased the levels of Wnt-1 and its receptor lipoprotein-related protein 5; this increase was inhibited by either lycopene or AG490 in AGS cells. In conclusion, lycopene inhibits ROS-mediated activation of Jak1/Stat3 and Wnt/β-catenin signaling and, thus, oncogene expression in relation to hyperproliferation in H pylori-infected gastric epithelial cells. Lycopene might be a potential and promising nutrient for preventing H pylori-associated gastric diseases including gastric cancer.
Collapse
Affiliation(s)
- Bohye Park
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul 03722, Korea.
| | - Joo Weon Lim
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul 03722, Korea.
| | - Hyeyoung Kim
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul 03722, Korea.
| |
Collapse
|
11
|
Gagnaire A, Nadel B, Raoult D, Neefjes J, Gorvel JP. Collateral damage: insights into bacterial mechanisms that predispose host cells to cancer. Nat Rev Microbiol 2017; 15:109-128. [DOI: 10.1038/nrmicro.2016.171] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
12
|
Yu XJ, Zhao Q, Wang XB, Zhang JX, Wang XB. Gambogenic acid induces proteasomal degradation of CIP2A and sensitizes hepatocellular carcinoma to anticancer agents. Oncol Rep 2016; 36:3611-3618. [PMID: 27779687 DOI: 10.3892/or.2016.5188] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/11/2016] [Indexed: 11/06/2022] Open
Abstract
Cancerous inhibitor of protein phosphatase 2A (CIP2A) is an oncoprotein that is overexpressed in many human malignancies. It regulates phosphorylated AKT and stabilizes c‑Myc in cell proliferation and tumor formation, suggesting that CIP2A plays an essential role in the development of cancer. In the present study, we report that a natural compound, gambogenic acid (GEA), induced the degradation of CIP2A via the ubiquitin‑proteasome pathway. Interestingly, the combination of GEA and proteasome inhibitors potentiated the accumulation of ubiquitinated CIP2A and aggresome formation. In addition, GEA exhibited an inhibitory effect on cell proliferation and CIP2A‑downstream signaling molecules (c‑Myc and pAKT). Furthermore, GEA and CIP2A silencing enhanced the chemosensitivity of hepatocellular carcinoma cells to anticancer agents, suggesting that a combination of a CIP2A inhibitor and anticancer agents could be a valuable clinical therapeutic strategy. These results indicate that GEA is a CIP2A inhibitor that interferes with the ubiquitination and destabilization of CIP2A, providing a promising strategy to enhance the combinational therapy for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xian-Jun Yu
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital and School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Qun Zhao
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Graduate School of the Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, P.R. China
| | - Xuan-Bin Wang
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital and School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Jing-Xuan Zhang
- Laboratory of Chinese Herbal Pharmacology, Oncology Center, Renmin Hospital and School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, P.R. China
| | - Xiao-Bo Wang
- Center for Translational Medicine, Suizhou Hospital, Hubei University of Medicine, Suizhou, Hubei 441300, P.R. China
| |
Collapse
|
13
|
Subhash VV, Ho B. Inflammation and proliferation - a causal event of host response to Helicobacter pylori infection. MICROBIOLOGY (READING, ENGLAND) 2015; 161:1150-1160. [PMID: 25721850 DOI: 10.1099/mic.0.000066] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Helicobacter pylori is a major aetiological agent in the development of various gastroduodenal diseases. Its persistence in gastric mucosa is determined by the interaction between various host, microbial and environmental factors. The bacterium colonizes the gastric epithelium and induces activation of various chemokine mediators, including NFκB, the master regulator of inflammation. H. pylori infection is also associated with an increase in expression of cell cycle regulators, thereby leading to mucosal cell hyper-proliferation. Thus, H. pylori-associated infections manifest activation of key host response events, which inadvertently could lead to the establishment of chronic infection and neoplastic progression. This article reviews and elaborates the current knowledge in H. pylori-induced activation of various host signalling pathways that could promote cancer development. Special focus is placed on the inflammatory and proliferative responses that could serve as suitable biomarkers of infection, since a sustained cell proliferation in an environment rich in inflammatory cells is characteristic in H. pylori-associated gastric malignancies. Here, the role of ERK and WNT signalling in H. pylori-induced activation of inflammatory and proliferative responses respectively is discussed in detail. An in depth analysis of the underlying signalling pathways and interacting partners causing alterations in these crucial host responses could contribute to the development of successful therapeutic strategies for the prevention, management and treatment of H. pylori infection.
Collapse
Affiliation(s)
- Vinod Vijay Subhash
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, 117545, Singapore
| | - Bow Ho
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, 117545, Singapore
| |
Collapse
|
14
|
Cao L, Yu J. Effect of Helicobacter pylori Infection on the Composition of Gastric Microbiota in the Development of Gastric Cancer. Gastrointest Tumors 2015; 2:14-25. [PMID: 26673084 DOI: 10.1159/000380893] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Gastric cancer is one of the most common cancer types worldwide. In China, gastric cancer has become one of the major threats for public health, ranking second on incidence and third on cause of cancer death. Despite the common risk factors that promote the development of gastric cancer, the huge quantity of microorganism colonies within the gastrointestinal tract, particularly Helicobacter pylori infection, demonstrates a correlation with chronic inflammation and gastric carcinogenesis, as epidemiological studies have determined that H. pylori infection confers approximately 75% of the attributable risk for gastric cancer. SUMMARY The current article draws an overview on the correlation between the microbiota, inflammation and gastric tumorigenesis. H. pylori infection has been identified as the main risk factor as it triggers epithelial barrier disruption, survival signaling as well as genetic/epigenetic modulation. Apart from H. pylori, the existence of a diverse and complex composition of microbiota in the stomach has been identified, which supports a role of microbiota in the development of gastric cancer. Moreover, metagenomics studies focused on the composition and function of the microbiota have associated microbiota with gastric metabolic diseases and even tumorigenesis. Apart from the gastric microbiota, inflammation is another identified contributor to cancer development as well. KEY MESSAGE Though H. pylori infection and the non-H. pylori microbiota play a role in gastric cancer, the properties of gastric microbiota and mechanisms by which they participate in the genesis of gastric cancer are still not clearly depicted. Moreover, it remains to be understood how the presence of microbiota along with H. pylori infection affects the progress from gastric disease to cancer. PRACTICAL IMPLICATIONS This article summarized a clue of the current studies on microbiota, H. pylori infection and the progression from gastric disease to cancer.
Collapse
Affiliation(s)
- Le Cao
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, China
| | - Ju Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, The Chinese University of Hong Kong, Shatin, Hong Kong, SAR, China
| |
Collapse
|
15
|
Chen KF, Yen CC, Lin JK, Chen WS, Yang SH, Jiang JK, Lan YT, Lin CC, Yu HC, Hsu HM, Lin WL, Teng HW. Cancerous inhibitor of protein phosphatase 2A (CIP2A) is an independent prognostic marker in wild-type KRAS metastatic colorectal cancer after colorectal liver metastasectomy. BMC Cancer 2015; 15:301. [PMID: 25896895 PMCID: PMC4404594 DOI: 10.1186/s12885-015-1300-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 03/31/2015] [Indexed: 01/07/2023] Open
Abstract
Background The impact of KRAS signaling on cancerous inhibitor of protein phosphatase 2A (CIP2A) expression has not yet been explored. We investigated the impact of KRAS on CIP2A expression in colorectal cancer patients after colorectal liver metastasectomy. Methods We examined CIP2A expression by immunohistochemistry (IHC) and used direct sequencing to identify the mutational status of KRAS exon 2 (codon 12 and 13). The association between CIP2A expression, KRAS genotype, clinicopathological parameters and survival were examined by the Kaplan–Meier method and the Cox proportional hazards model. A combination of immunoblotting and proliferation assays were employed to elucidate the role of CIP2A in signal transduction pathways in wild-type KRAS Caco-2 cells. Results A total of 220 colorectal cancer patients who had undergone colorectal liver metastasectomy were included in the study. The mutant KRAS genotype was associated with CIP2A overexpression. CIP2A expression was an independent prognostic marker in patients with wild-type KRAS metastatic colorectal cancer after colorectal liver metastasectomy (relative risk = 1.873, P = 0.019). Targeted silencing of CIP2A in Caco-2 cells (wild-type KRAS) led to decreased expression of pERK/ERK and decreased cell proliferation. Overexpression of mutant KRAS G12D in Caco-2 cells led to an increase in CIP2A expression and cell proliferation. In Caco-2 cells with the KRAS G12D, KRAS overexpression preserved the regulation effect of CIP2A in KRAS and abrogated the impact of CIP2A regulation on pERK/ERK and cell proliferation. CIP2A inhibition also increased the efficacy of cetuximab in Caco-2 cells. Conclusions CIP2A is an independent prognostic marker in patients with wild-type KRAS metastatic colorectal cancer after colorectal liver metastasectomy.
Collapse
Affiliation(s)
- Kuen-Feng Chen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan. .,National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan.
| | - Chueh-Chuan Yen
- Division of Hematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, No. 201, Section 2, Shih-Pai Road, Taipei, Taiwan. .,National Yang-Ming University School of Medicine, Taipei, Taiwan.
| | - Jen-Kou Lin
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan. .,National Yang-Ming University School of Medicine, Taipei, Taiwan.
| | - Wei-Shone Chen
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan. .,National Yang-Ming University School of Medicine, Taipei, Taiwan.
| | - Shung-Haur Yang
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan. .,National Yang-Ming University School of Medicine, Taipei, Taiwan.
| | - Jeng-Kai Jiang
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan. .,National Yang-Ming University School of Medicine, Taipei, Taiwan.
| | - Yuan-Tzu Lan
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan. .,National Yang-Ming University School of Medicine, Taipei, Taiwan.
| | - Chun-Chi Lin
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan. .,National Yang-Ming University School of Medicine, Taipei, Taiwan.
| | - Hui-Chuan Yu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan. .,National Center of Excellence for Clinical Trial and Research, National Taiwan University Hospital, Taipei, Taiwan.
| | - Hui-Mei Hsu
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan. .,National Yang-Ming University School of Medicine, Taipei, Taiwan.
| | - Wen-Ling Lin
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan. .,National Yang-Ming University School of Medicine, Taipei, Taiwan.
| | - Hao-Wei Teng
- Division of Hematology and Oncology, Department of Medicine, Taipei Veterans General Hospital, No. 201, Section 2, Shih-Pai Road, Taipei, Taiwan. .,Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan. .,National Yang-Ming University School of Medicine, Taipei, Taiwan.
| |
Collapse
|
16
|
Chao TT, Wang CY, Lai CC, Chen YL, Tsai YT, Chen PT, Lin HI, Huang YCT, Shiau CW, Yu CJ, Chen KF. TD-19, an erlotinib derivative, induces epidermal growth factor receptor wild-type nonsmall-cell lung cancer apoptosis through CIP2A-mediated pathway. J Pharmacol Exp Ther 2014; 351:352-8. [PMID: 25187431 DOI: 10.1124/jpet.114.215418] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Some patients with nonsmall-cell lung cancer (NSCLC) without epidermal growth factor receptor (EGFR) mutations still respond to gefitinib and erlotinib, suggesting that there may be a mechanism(s) other than the EGFR pathway that mediates the tumoricidal effects. In the current study, we tested the efficacy of TD-19, a novel compound chemically modified from erlotinib, which has more potent apoptotic effects than erlotinib in EGFR wild-type NSCLC cell lines. TD-19 induced significant cell death and apoptosis in H358, H441, H460, and A549 cells, as evidenced by increased caspase-3 activity and cleavage of procaspase-9 and poly (ADP-ribose) polymerase. The apoptotic effect of TD-19 in H460 cells, which were resistant to erlotinib, was associated with downregulation of cancerous inhibitor of protein phosphatase 2A (CIP2A), increased protein phosphatase 2A (PP2A) activity, and decreased AKT phosphorylation, but minimal effects on EGFR phosphorylation. Overexpression of CIP2A partially protected the H460 cells from TD-19-induced apoptosis. Okadaic acid, a known PP2A inhibitor, significantly reduced TD-19-induced apoptosis, while forskolin, which increased PP2A activity, increased the apoptotic effect of TD-19. TD-19 inhibited the growth of H460 xenograft tumors by ∼80%. We conclude that TD-19 exerted tumoricidal effects on NSCLC cells. TD-19 provides proof that the CIP2A pathway may be a novel target for the treatment of EGFR wild-type NSCLC.
Collapse
Affiliation(s)
- Ting-Ting Chao
- Medical Research Center (T.-T.C., C.-Y.W., Y.-T.T., P.-T.C.), Department of Internal Medicine (C.-Y.W., H.-I.L.), and Department of Pathology (Y.-L.C.), Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan (C.-Y.W.); Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan (C.-C.L.); Department of Medicine, Duke University Medical Center, Durham, North Carolina (Y.-C.T.H.); Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan (C.-W.S.); and Department of Internal Medicine (C.-J.Y.) and Department of Medical Research and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital and National Taiwan University, Taipei, Taiwan
| | - Cheng-Yi Wang
- Medical Research Center (T.-T.C., C.-Y.W., Y.-T.T., P.-T.C.), Department of Internal Medicine (C.-Y.W., H.-I.L.), and Department of Pathology (Y.-L.C.), Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan (C.-Y.W.); Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan (C.-C.L.); Department of Medicine, Duke University Medical Center, Durham, North Carolina (Y.-C.T.H.); Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan (C.-W.S.); and Department of Internal Medicine (C.-J.Y.) and Department of Medical Research and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital and National Taiwan University, Taipei, Taiwan
| | - Chih-Cheng Lai
- Medical Research Center (T.-T.C., C.-Y.W., Y.-T.T., P.-T.C.), Department of Internal Medicine (C.-Y.W., H.-I.L.), and Department of Pathology (Y.-L.C.), Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan (C.-Y.W.); Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan (C.-C.L.); Department of Medicine, Duke University Medical Center, Durham, North Carolina (Y.-C.T.H.); Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan (C.-W.S.); and Department of Internal Medicine (C.-J.Y.) and Department of Medical Research and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital and National Taiwan University, Taipei, Taiwan
| | - Yen-Lin Chen
- Medical Research Center (T.-T.C., C.-Y.W., Y.-T.T., P.-T.C.), Department of Internal Medicine (C.-Y.W., H.-I.L.), and Department of Pathology (Y.-L.C.), Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan (C.-Y.W.); Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan (C.-C.L.); Department of Medicine, Duke University Medical Center, Durham, North Carolina (Y.-C.T.H.); Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan (C.-W.S.); and Department of Internal Medicine (C.-J.Y.) and Department of Medical Research and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital and National Taiwan University, Taipei, Taiwan
| | - Yi-Ting Tsai
- Medical Research Center (T.-T.C., C.-Y.W., Y.-T.T., P.-T.C.), Department of Internal Medicine (C.-Y.W., H.-I.L.), and Department of Pathology (Y.-L.C.), Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan (C.-Y.W.); Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan (C.-C.L.); Department of Medicine, Duke University Medical Center, Durham, North Carolina (Y.-C.T.H.); Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan (C.-W.S.); and Department of Internal Medicine (C.-J.Y.) and Department of Medical Research and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital and National Taiwan University, Taipei, Taiwan
| | - Pao-Tzu Chen
- Medical Research Center (T.-T.C., C.-Y.W., Y.-T.T., P.-T.C.), Department of Internal Medicine (C.-Y.W., H.-I.L.), and Department of Pathology (Y.-L.C.), Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan (C.-Y.W.); Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan (C.-C.L.); Department of Medicine, Duke University Medical Center, Durham, North Carolina (Y.-C.T.H.); Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan (C.-W.S.); and Department of Internal Medicine (C.-J.Y.) and Department of Medical Research and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital and National Taiwan University, Taipei, Taiwan
| | - Hen-I Lin
- Medical Research Center (T.-T.C., C.-Y.W., Y.-T.T., P.-T.C.), Department of Internal Medicine (C.-Y.W., H.-I.L.), and Department of Pathology (Y.-L.C.), Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan (C.-Y.W.); Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan (C.-C.L.); Department of Medicine, Duke University Medical Center, Durham, North Carolina (Y.-C.T.H.); Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan (C.-W.S.); and Department of Internal Medicine (C.-J.Y.) and Department of Medical Research and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital and National Taiwan University, Taipei, Taiwan
| | - Yuh-Chin T Huang
- Medical Research Center (T.-T.C., C.-Y.W., Y.-T.T., P.-T.C.), Department of Internal Medicine (C.-Y.W., H.-I.L.), and Department of Pathology (Y.-L.C.), Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan (C.-Y.W.); Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan (C.-C.L.); Department of Medicine, Duke University Medical Center, Durham, North Carolina (Y.-C.T.H.); Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan (C.-W.S.); and Department of Internal Medicine (C.-J.Y.) and Department of Medical Research and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital and National Taiwan University, Taipei, Taiwan
| | - Chung-Wai Shiau
- Medical Research Center (T.-T.C., C.-Y.W., Y.-T.T., P.-T.C.), Department of Internal Medicine (C.-Y.W., H.-I.L.), and Department of Pathology (Y.-L.C.), Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan (C.-Y.W.); Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan (C.-C.L.); Department of Medicine, Duke University Medical Center, Durham, North Carolina (Y.-C.T.H.); Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan (C.-W.S.); and Department of Internal Medicine (C.-J.Y.) and Department of Medical Research and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital and National Taiwan University, Taipei, Taiwan
| | - Chong-Jen Yu
- Medical Research Center (T.-T.C., C.-Y.W., Y.-T.T., P.-T.C.), Department of Internal Medicine (C.-Y.W., H.-I.L.), and Department of Pathology (Y.-L.C.), Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan (C.-Y.W.); Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan (C.-C.L.); Department of Medicine, Duke University Medical Center, Durham, North Carolina (Y.-C.T.H.); Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan (C.-W.S.); and Department of Internal Medicine (C.-J.Y.) and Department of Medical Research and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital and National Taiwan University, Taipei, Taiwan
| | - Kuen-Feng Chen
- Medical Research Center (T.-T.C., C.-Y.W., Y.-T.T., P.-T.C.), Department of Internal Medicine (C.-Y.W., H.-I.L.), and Department of Pathology (Y.-L.C.), Cardinal Tien Hospital, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan (C.-Y.W.); Department of Intensive Care Medicine, Chi Mei Medical Center, Liouying, Tainan, Taiwan (C.-C.L.); Department of Medicine, Duke University Medical Center, Durham, North Carolina (Y.-C.T.H.); Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan (C.-W.S.); and Department of Internal Medicine (C.-J.Y.) and Department of Medical Research and National Center of Excellence for Clinical Trial and Research (K.-F.C.), National Taiwan University Hospital and National Taiwan University, Taipei, Taiwan
| |
Collapse
|
17
|
Chen S, Duan G, Zhang R, Fan Q. Helicobacter pylori cytotoxin-associated gene A protein upregulates α-enolase expression via Src/MEK/ERK pathway: implication for progression of gastric cancer. Int J Oncol 2014; 45:764-770. [PMID: 24841372 DOI: 10.3892/ijo.2014.2444] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/29/2014] [Indexed: 12/13/2022] Open
Abstract
Persistent infection with Helicobacter pylori confers an increased risk for the development of gastric cancer. In our previous investigations, we found that ENO1 was overexpression in cagA-positive H. pylori-infected gastric epithelial AGS cells by proteomic method, in contrast to the isogenic cagA knock out mutant H. pylori-infected cells. ENO1 is a newly identified oncoprotein overexpressed in some cancer. However, the relationship between H. pylori infection and ENO1 expression still remains undefined. The AGS gastric cancer cells were transfected with WT-cagA plasmid and PR-cagA plasmids. Expression of ENO1 mRNA and protein were measured by real-time quantitative PCR and western blot analysis. Signal protein inhibitor treatment was used to investigate the signal pathways. It was found that the ENO1 mRNA and protein overexpression levels were dependent on cagA gene expression and CagA protein phosphorylation. Further analysis revealed that the Src, MEK and ERK pathway was involved in this upregulation effect. Our data suggest that ENO1 was upregulated by CagA protein through activating the Src and MEK/ERK signal pathways, thereby providing a novel mechanism underlying H. pylori-mediated gastric diseases.
Collapse
Affiliation(s)
- Shuaiyin Chen
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Guangcai Duan
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Rongguang Zhang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Qingtang Fan
- Henan Key Laboratory of Molecular Medicine, Zhengzhou, Henan, P.R. China
| |
Collapse
|
18
|
CIP2A is overexpressed and involved in the pathogenesis of chronic myelocytic leukemia by interacting with breakpoint cluster region-Abelson leukemia virus. Med Oncol 2014; 31:112. [DOI: 10.1007/s12032-014-0112-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 06/30/2014] [Indexed: 01/26/2023]
|
19
|
De P, Carlson J, Leyland-Jones B, Dey N. Oncogenic nexus of cancerous inhibitor of protein phosphatase 2A (CIP2A): an oncoprotein with many hands. Oncotarget 2014; 5:4581-602. [PMID: 25015035 PMCID: PMC4148086 DOI: 10.18632/oncotarget.2127] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/20/2014] [Indexed: 12/23/2022] Open
Abstract
Oncoprotein CIP2A a Cancerous Inhibitor of PP2A forms an "oncogenic nexus" by virtue of its control on PP2A and MYC stabilization in cancer cells. The expression and prognostic function of CIP2A in different solid tumors including colorectal carcinoma, head and neck cancers, gastric cancers, lung carcinoma, cholangiocarcinoma, esophageal cancers, pancreatic carcinoma, brain cancers, breast carcinoma, bladder cancers, ovarian carcinoma, renal cell carcinomas, tongue cancers, cervical carcinoma, prostate cancers, and oral carcinoma as well as a number of hematological malignancies are just beginning to emerge. Herein, we reviewed the recent progress in our understanding of (1) how an "oncogenic nexus" of CIP2A participates in the tumorigenic transformation of cells and (2) how we can prospect/view the clinical relevance of CIP2A in the context of cancer therapy. The review will try to understand the role of CIP2A (a) as a biomarker in cancers and evaluate the prognostic value of CIP2A in different cancers (b) as a therapeutic target in cancers and (c) in drug response and developing chemo-resistance in cancers.
Collapse
Affiliation(s)
- Pradip De
- Department of Molecular & Experimental Medicine, Avera Research Institute, Sioux Falls, SD
- Department of Internal Medicine, SSOM, University of South Dakota, Sioux Falls, SD
| | - Jennifer Carlson
- Department of Molecular & Experimental Medicine, Avera Research Institute, Sioux Falls, SD
| | - Brian Leyland-Jones
- Department of Molecular & Experimental Medicine, Avera Research Institute, Sioux Falls, SD
- Department of Internal Medicine, SSOM, University of South Dakota, Sioux Falls, SD
| | - Nandini Dey
- Department of Molecular & Experimental Medicine, Avera Research Institute, Sioux Falls, SD
- Department of Internal Medicine, SSOM, University of South Dakota, Sioux Falls, SD
| |
Collapse
|
20
|
Choi YA, Koo JS, Park JS, Park MY, Jeong AL, Oh KS, Yang Y. Estradiol enhances CIP2A expression by the activation of p70 S6 kinase. Endocr Relat Cancer 2014; 21:189-202. [PMID: 24280132 DOI: 10.1530/erc-13-0453] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cancerous inhibitor of PP2A (CIP2A) stimulates the proliferation of various cancer cells, and 17β-estradiol (E₂) enhances the proliferation of breast cancer cells. E₂ activates epidermal growth factor receptor (EGFR), stimulating the MEK1/2 and PI3K pathways, and CIP2A expression is increased by the MEK1/2-induced transcription factor ETS1. It is possible for E₂ to increase CIP2A expression. This study examined whether E₂ could increase CIP2A expression and whether CIP2A is highly expressed in estrogen receptor (ER)-positive breast cancer tissues. E₂ increased CIP2A expression at the translational level in a c-MYC-independent manner in MCF-7 cells. E₂-enhanced proliferation was impaired without CIP2A expression. E₂-stimulated EGFR activated the MAPK and PI3K pathways, which converged to activate p70 S6 kinase (S6K). Phosphorylation at all the three phosphorylation sites (S424/T421, T229, and T389) on S6K was required for the phosphorylation of eukaryotic initiation factor 4B (eIF4B), which was responsible for the increase in CIP2A translation. Furthermore, CIP2A expression was higher in ER-positive tissues than in ER-negative tissues. This is the first study, to our knowledge, to demonstrate that CIP2A is a key factor in E₂-enhanced proliferation and that estrogen regulates CIP2A expression by non-genomic action through EGFR.
Collapse
Affiliation(s)
- Yeon A Choi
- Department of Life Science, Research Center for Women's Disease, Sookmyung Women's University, Seoul 140-742, Republic of Korea Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | | | | | | | | | | | | |
Collapse
|
21
|
Wang HW, Yang SH, Huang GD, Lin JK, Chen WS, Jiang JK, Lan YT, Lin CC, Hwang WL, Tzeng CH, Li AFY, Yen CC, Teng HW. Temsirolimus enhances the efficacy of cetuximab in colon cancer through a CIP2A-dependent mechanism. J Cancer Res Clin Oncol 2014; 140:561-71. [PMID: 24493623 DOI: 10.1007/s00432-014-1596-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 01/20/2014] [Indexed: 12/19/2022]
Abstract
PURPOSE A dozen clinical trials examining a combination of temsirolimus and cetuximab in treating metastatic colon cancer are currently underway. We investigated the role of cancerous inhibitor of protein phosphatase 2A (CIP2A) in the synergism between temsirolimus and cetuximab in colon cancer. METHODS Five colon cancer cell lines were used for in vitro studies. Signal transduction pathways were assessed by immunoblotting. The synergism between studied drugs was analyzed with combination indexes. Gene silencing was performed using small interfering RNAs. The efficacies of temsirolimus and cetuximab were tested in nude mice with colon cancer xenografts. Transcriptional activity was assessed using a reporter assay. The inhibitors leupeptin, chloroquine, and MG132 were used to assess protein degradation. The association between CIP2A, clinicopathological parameters, and survival was examined by immunohistochemical staining using a tumor tissue microarray. RESULTS Temsirolimus decreased the resistance of cells to cetuximab by both inhibiting transcription of CIP2A and increasing degradation of CIP2A through the lysosomal-autophagy pathway. The mammalian target of rapamycin (mTOR) protein immunoprecipitated along with CIP2A. Temsirolimus decreased expression of phosphorylated extracellular regulated protein kinase (pErk) and phosphorylated v-akt murine thymoma viral oncogene (pAKT) and decreased the interaction of CIP2A and mTOR in cell lines without the K-ras codon 12 mutation. CIP2A was a prognostic marker only in colon cancer patients with weak expression of pErk or pAKT. CONCLUSIONS Temsirolimus decreases cellular resistance to cetuximab by regulating CIP2A expression in colon cancer cells. Potential biomarkers for CIP2A inhibitors include pErk and pAKT.
Collapse
Affiliation(s)
- Hsei-Wei Wang
- National Yang-Ming University School of Medicine, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Liu Z, Zhang X, Xu X, Chen L, Li W, Yu H, Sun Y, Zeng J, Jia J. RUNX3 inhibits survivin expression and induces cell apoptosis in gastric cancer. Eur J Cell Biol 2014; 93:118-26. [PMID: 24636883 DOI: 10.1016/j.ejcb.2014.02.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Revised: 02/13/2014] [Accepted: 02/14/2014] [Indexed: 01/18/2023] Open
Abstract
Transcription factor RUNX3 is associated with gastric tumorigenesis and progression through regulating the expression of its target genes. Survivin is a member of the inhibitor of apoptosis (IAP) family and has been shown to inhibit cell apoptosis and promote cell proliferation. Increased survivin expression has been found in various cancer types, including gastric cancer. In this study, we found that restoration of RUNX3 promotes cell apoptosis through inhibiting the survivin expression, while RUNX3 inhibition increases the expression of survivin in gastric cancer cell lines. Moreover, RUNX3 over-expression inhibits,whereas its inhibition increases, the promoter activity of survivin gene, respectively. RUNX3-R122C, a mutation located in the conserved Runt domain, has no effect on the promoter activity of survivin gene. We further identified a RUNX3-binding site in the promoter of survivin gene and the binding of RUNX3 on survivin promoter leads to significantly inhibition of survivin expression. Finally, we confirmed the negative correlation of RUNX3 and survivin expression in clinical specimens of gastric cancer. These findings reveal a novel mechanism of RUNX3 for the induction of cell apoptosis in human gastric cancer.
Collapse
Affiliation(s)
- Zhifang Liu
- Department of Biochemistry and molecular biology, School of Medicine, Shandong University, Jinan, PR China
| | - Xinchao Zhang
- Department of Biochemistry and molecular biology, School of Medicine, Shandong University, Jinan, PR China
| | - Xia Xu
- Department of Biochemistry and molecular biology, School of Medicine, Shandong University, Jinan, PR China
| | - Long Chen
- Department of Biochemistry and molecular biology, School of Medicine, Shandong University, Jinan, PR China
| | - Wenjuan Li
- Department of Microbiology and Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Medicine, Shandong University, Jinan, PR China
| | - Han Yu
- Department of Microbiology and Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Medicine, Shandong University, Jinan, PR China
| | - Yundong Sun
- Department of Microbiology and Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Medicine, Shandong University, Jinan, PR China
| | - Jiping Zeng
- Department of Biochemistry and molecular biology, School of Medicine, Shandong University, Jinan, PR China
| | - Jihui Jia
- Department of Microbiology and Key Laboratory for Experimental Teratology of Chinese Ministry of Education, School of Medicine, Shandong University, Jinan, PR China.
| |
Collapse
|
23
|
Zhao Y, Li Y, Han J, Liu T, Guan Q, Zhao P, Guo L, Liu K, He D. Helicobacter pylori enhances CIP2A expression and cell proliferation via JNK2/ATF2 signaling in human gastric cancer cells. Int J Mol Med 2014; 33:703-10. [PMID: 24398514 DOI: 10.3892/ijmm.2014.1615] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 12/24/2013] [Indexed: 11/06/2022] Open
Abstract
Helicobacter pylori (H. pylori) infection plays an important role in the development of gastric carcinomas. Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a novel human oncoprotein that functions as an important regulator of cell growth and malignant transformation. In the present study, we aimed to investigate the potential mechanisms by which H. pylori upregulates the expression of CIP2A and the functional impact of H. pylori-induced CIP2A in gastric cancer cells. We demonstrated that infection of MKN-45 cells with H. pylori led to a marked increase in the expression of CIP2A at the mRNA and protein levels. H. pylori-induced CIP2A was associated with increased cell proliferation. In addition, H. pylori was found to activate the JNK2 pathway. Importantly, both H. pylori-induced CIP2A production and cell proliferation were partially reversed by inhibition of JNK2 signaling. Similarly, the blockade of H. pylori-induced CIP2A expression by siRNA against CIP2A also inhibited cell proliferation. Thus, H. pylori appears to stimulate the expression of CIP2A and proliferation of gastric cancer cells via JNK2 signaling. These findings suggest that H. pylori-induced upregulation of CIP2A contributes to the development and progression of gastric cancer. Further in vivo studies are warranted to explore the biological role of CIP2A and its interaction with JNK2 signaling in gastric cancer.
Collapse
Affiliation(s)
- Yongxun Zhao
- The Second Clinical Medical School of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Yumin Li
- Department of General Surgery, The Second Hospital of Lanzhou University, Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou, Gansu 730030, P.R. China
| | - Jian Han
- Institute of Pathogenic Biology, School of Basic Medical Science, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Tao Liu
- Department of General Surgery, The Second Hospital of Lanzhou University, Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou, Gansu 730030, P.R. China
| | - Quanlin Guan
- Department of Surgical Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Peng Zhao
- Department of Surgical Oncology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Lingyun Guo
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Kaikun Liu
- The Second Clinical Medical School of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Dongqiang He
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
24
|
Xing JM, Huang ZG. Association between CagA/ERK signaling pathway and gastric cancer. Shijie Huaren Xiaohua Zazhi 2013; 21:3363-3368. [DOI: 10.11569/wcjd.v21.i31.3363] [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
Cytotoxin-associated gene A (CagA) of Helicobacter pylori is the first identified bacterial oncoprotein that plays a critical role in gastric carcinogenesis. Upon delivery into gastric epithelial cells via type IV secretion, CagA can interfere with a number of host signaling pathways. Extracellular signal-regulated kinase (ERK) signaling pathway is a hub in cellular signal transduction, through which CagA elicits a series of cellular events including cell proliferation, apoptosis, scatter and metastasis, all of which are associated with gastric carcinogenesis. Here we perform a review of the association between CagA/ERK signaling pathway and gastric cancer.
Collapse
|
25
|
CIP2A influences survival in colon cancer and is critical for maintaining Myc expression. PLoS One 2013; 8:e75292. [PMID: 24098375 PMCID: PMC3788051 DOI: 10.1371/journal.pone.0075292] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 08/12/2013] [Indexed: 11/19/2022] Open
Abstract
The cancerous inhibitor of protein phosphatase 2A (CIP2A) is an oncogenic factor that stabilises the c-Myc protein. CIP2A is overexpressed in several tumours, and expression levels are an independent marker for long-term outcome. To determine whether CIP2A expression is elevated in colon cancer and whether it might serve as a prognostic marker for survival, we analysed CIP2A mRNA expression by real-time PCR in 104 colon cancer samples. CIP2A mRNA was overexpressed in colon cancer samples and CIP2A expression levels correlated significantly with tumour stage. We found that CIP2A serves as an independent prognostic marker for disease-free and overall survival. Further, we investigated CIP2A-dependent effects on levels of c-Myc, Akt and on cell proliferation in three colon cancer cell lines by silencing CIP2A using small interfering (si) and short hairpin (sh) RNAs. Depletion of CIP2A substantially inhibited growth of colon cell lines and reduced c-Myc levels without affecting expression or function of the upstream regulatory kinase, Akt. Expression of CIP2A was found to be dependent on MAPK activity, linking elevated c-Myc expression to deregulated signal transduction in colon cancer.
Collapse
|
26
|
Deng B, Li Y, Zhang Y, Bai L, Yang P. Helicobacter pylori infection and lung cancer: a review of an emerging hypothesis. Carcinogenesis 2013; 34:1189-95. [PMID: 23568955 DOI: 10.1093/carcin/bgt114] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Helicobacter pylori (Hp) is one of the most common bacteria infecting humans. Recently, certain extragastric manifestations, linked to Hp infection, have been widely investigated, suggesting that Hp infection might be a 'systemic' disease. Accumulating, yet limited, evidence points to a potential association between Hp infection and lung cancer risk. Epidemiologic studies have shown that odds ratios (estimated relative risks) of lung cancer with Hp infection range from 1.24 to 17.78 compared with the controls, suggesting an increased lung cancer risk in the population exposed to Hp infection although far from supporting a causal relationship between Hp and lung cancer. Many studies have demonstrated the existence of Hp in the mucosa of the upper respiratory tract with no direct evidence of Hp-localization in lung tissue in the published literatures, rendering the possible functional mechanism underlying the association an open question. We followed the classic hypothesis-generating path, where we have thoroughly reviewed the publications on lung cancer and Hp infection from serological association to possible mechanisms as: (i) p130cas activated by Src kinase following Hp-host communication and p130cas-related carcinogenesis as in various malignancies; and (ii) gastroesophageal reflux and inhalation of urease or gastrin, which are Hp-related carcinogenic factors and present in lung tissues. We propose rigorous investigations regarding the Hp-lung cancer association and, if confirmed, the mechanisms of Hp infection leading to lung cancer development and progression. Clarification on Hp-lung cancer association is important for the understanding of lung cancer beyond tobacco-smoking-related carcinogenesis.
Collapse
Affiliation(s)
- Bo Deng
- Department of Health Sciences Research, Mayo Clinic, College of Medicine, Rochester, MN 55905, USA
| | | | | | | | | |
Collapse
|
27
|
Wang G, Gao F, Zhang W, Chen J, Wang T, Zhang G, Shen L. Involvement of Aquaporin 3 in Helicobacter pylori-related gastric diseases. PLoS One 2012; 7:e49104. [PMID: 23152856 PMCID: PMC3494660 DOI: 10.1371/journal.pone.0049104] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 10/04/2012] [Indexed: 01/26/2023] Open
Abstract
Background Gastric cancer is one of the most common and lethal malignant cancers worldwide, and numerous epidemiological studies have demonstrated that Helicobacter pylori (H. pylori) infection plays a key role in the development of gastric carcinomas. Our previous studies showed that aquaporin 3 (AQP3) is overexpressed in gastric carcinoma and promotes the migration and proliferation of human gastric carcinoma cells, suggesting that AQP3 may be a potentially important determinant of gastric carcinoma. However, the role of AQP3 in H. pylori carcinogenesis is unknown. Methods The AQP3 protein and H. pylori were detected in human gastric tissues by immunohistochemistry and modified Giemsa staining respectively. AQP3 knockdown was obtained by small interfering (si) RNA. Western blot assays and RT-PCR were used to evaluate the change of AQP3 in the human gastric cancer AGS and SGC7901 cell lines after co-culture with H. pylori. Sprague Dawley rats were orally inoculated with H. pylori to establish a rat model colonized by H. pylori. Results The present study found that AQP3 expression correlated with H. pylori infection status in gastric cancer tissues and corresponding normal mucosa, and H. pylori co-culture upregulated AQP3 expression in human gastric adenocarcinoma cells in vitro via the extracellular signal-regulated kinase signaling pathway. H. pylori infection also increased AQP3 expression in gastric mucosa colonized by H. pylori in a Sprague Dawley rat model. Conclusions These findings provide further information to understand the mechanism of H. pylori carcinogenesis and a potential strategy for the treatment of H. pylori-associated gastric carcinoma.
Collapse
Affiliation(s)
- Gang Wang
- Division of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fei Gao
- Division of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weiming Zhang
- Department of Pathology, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jia Chen
- Division of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tao Wang
- Division of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guoxin Zhang
- Department of Gastroenterology, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lizong Shen
- Division of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
- * E-mail:
| |
Collapse
|
28
|
Huang LP, Savoly D, Sidi AA, Adelson ME, Mordechai E, Trama JP. CIP2A protein expression in high-grade, high-stage bladder cancer. Cancer Med 2012; 1:76-81. [PMID: 23342256 PMCID: PMC3544439 DOI: 10.1002/cam4.15] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 06/04/2012] [Accepted: 06/06/2012] [Indexed: 12/22/2022] Open
Abstract
Bladder cancer is one of the most common cancers in the United States. Numerous markers have been evaluated for suitability of bladder cancer detection and surveillance. However, few of them are acceptable as a routine tool. Therefore, there exists a continuing need for an assay that detects the presence of bladder cancer in humans. It would be advantageous to develop an assay with a protein that is associated with the development of bladder cancer. We have identified the cancerous inhibitor of PP2A (CIP2A) protein as a novel bladder cancer biomarker. In this study, Western blot analysis was used to assess the expression level of CIP2A protein in bladder cancer cell lines and bladder cancer patient tissues (n = 43). Our studies indicated CIP2A protein was abundantly expressed in bladder cancer cell lines but not in nontumor epithelial cell lines. Furthermore, CIP2A was specifically expressed in transitional cell carcinoma (TCC) of the bladder tumor tissues but not in adjacent nontumor bladder tissue. Our data showed that CIP2A protein detection in high-grade TCC tissues had a sensitivity of 65%, which is 3.4-fold higher than that seen in low-grade TCC tissues (19%). The level of CIP2A protein expression increased with the stage of disease (12%, 27%, 67%, and 100% for pTa, pT1, pT2, and pT3 tumor, respectively). In conclusion, our studies suggest that CIP2A protein is specifically expressed in human bladder tumors. CIP2A is preferentially expressed in high-grade and high-stage TCC tumors, which are high-risk and invasive tumors. Our studies reported here support the role of CIP2A in bladder cancer progression and its usefulness for the surveillance of recurrence or progression of human bladder cancer.
Collapse
Affiliation(s)
- Lisa P Huang
- Oncoveda, Cancer Research Center, Medical Diagnostic Laboratories, L.L.C., Hamilton, NJ, 08690, USA.
| | | | | | | | | | | |
Collapse
|
29
|
CIP2A is a predictor of poor prognosis in colon cancer. J Gastrointest Surg 2012; 16:1037-47. [PMID: 22328001 DOI: 10.1007/s11605-012-1828-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 01/19/2012] [Indexed: 01/31/2023]
Abstract
PURPOSE The cancerous inhibitor of protein phosphatase 2A (CIP2A) oncoprotein is overexpressed in colon cancer tissue compared to normal colon mucosa. We investigated the impact of CIP2A on colon cancer. METHODS A tissue microarray consisting of 167 colon cancer specimens was investigated. The association between CIP2A and clinicopathological parameters was analyzed using the χ(2) test. Survival was analyzed using the Kaplan-Meier method. The impact of CIP2A on proliferation and drug resistance was evaluated using the 3-(4, 5-dimethylthiazolyl-2)-2, 5-diphenyltetrazolium bromide test. An anchorage-independent colony formation assay was also performed. RESULTS CIP2A was an independent prognostic factor in colon cancer after controlling for other clinical confounding factors, such as stage and lymphovascular invasion, particularly in stages III and IV (hazard ratio = 2.974, P < 0.001). The knockdown of CIP2A reduced the proliferation and anchorage-independent colony formation of colon cancer cells. Knockdown of CIP2A decreased the resistance of the cells to 5-fluorouracil, oxaliplatin, and SN38 (an active metabolite of irinotecan). Treatment with 5-fluorouracil, oxaliplatin, and SN38 decreased CIP2A expression. CONCLUSIONS CIP2A is a prognostic factor in colon cancer. The knockdown of CIP2A reduced proliferation and anchorage-independent colony formation and increased 5-fluorouracil, oxaliplatin, and SN38 efficacy in colon cancer cell lines.
Collapse
|
30
|
Liu Z, Xu X, Chen L, Li W, Sun Y, Zeng J, Yu H, Chen C, Jia J. Helicobacter pylori CagA inhibits the expression of Runx3 via Src/MEK/ERK and p38 MAPK pathways in gastric epithelial cell. J Cell Biochem 2012; 113:1080-6. [DOI: 10.1002/jcb.23440] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
Godet AN, Guergnon J, Galioot A, Falanga PB, Colle JH, Cayla X, Garcia A. [Targeting of PP2A enzymes by viral proteins and cancer signalling]. Med Sci (Paris) 2011; 27:1106-11. [PMID: 22192750 DOI: 10.1051/medsci/20112712017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Protein phosphatase 2A (PP2A) is a large family of holoenzymes that comprises 1% of total cellular proteins and accounts for the majority of Ser/Thr phosphatase activity in eukaryotic cells. PP2A proteins are made of a core dimer, composed of a catalytic (C) subunit and a structural (A) subunit, in association with a third variable -regulatory (B) subunit. Although initially considered as a constitutive housekeeping enzyme, PP2A is indeed highly regulated by post-translational modifications of its catalytic subunit or by the identity of a regulatory type B subunit, which determines substrate specificity, subcellular localization and enzymatic activity of a defined holoenzyme. During the two last decades, multiple studies of structural and functional regulation of PP2A holoenzymes by viral proteins led to the identification of critical pathways for both viral biology and tumorigenesis. To date a dozen of different viruses (ADN/ARN or retrovirus) have been identified that encode viral proteins associated to PP2A. In this review, we analyze a biological strategy, used by various viruses based on the targeting of PP2A enzymes by viral proteins, in order to specifically deregulate cellular pathways of their hosts. The impact of such PP2A targeting for biomedical search, and in further therapeutic developments against cancer, will also be discussed.
Collapse
Affiliation(s)
- Angélique N Godet
- Laboratoire E3 Phosphatases, Unité Signalisation Moléculaire et Activation Cellulaire (SMAC), Institut Pasteur, 25, rue du Docteur Roux, 75015 Paris, France
| | | | | | | | | | | | | |
Collapse
|
32
|
Expression of CIP2A in renal cell carcinomas correlates with tumour invasion, metastasis and patients' survival. Br J Cancer 2011; 105:1905-11. [PMID: 22075943 PMCID: PMC3251889 DOI: 10.1038/bjc.2011.492] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background: Cancerous inhibitor of protein phosphatase 2A (CIP2A) drives cellular transformation. The objective of this study was to detect the potential effects of CIP2A in renal cell carcinomas (RCCs). Methods: A total of 107 RCC patients were involved in the study. Cancerous inhibitor of protein phosphatase 2A expression was investigated by real-time PCR and immunohistochemistry. In vitro, we examined the expression of CIP2A and c-Myc and tested the migration and invasion capability of A498 and KRC/Y cells with scratch migration assay and Matrigel invasion assay after down-regulating CIP2A expression using siRNA. Results: Cancerous inhibitor of protein phosphatase 2A was over-expressed in RCC tissues. Clear cell RCC showed an even higher-CIP2A expression level than papillary or chromophobe RCC did. The CIP2A immunostaining level was positively correlated with primary tumour stage, lymph node metastasis, distant metastasis, TNM stage and histological grade (all P<0.05). High-CIP2A expression implied poor survival for patients (P<0.05). Cancerous inhibitor of protein phosphatase 2A depletion by siRNA down-regulated c-Myc expression and attenuated the migration and invasion of RCC cells. Conclusion: Higher-CIP2A expression positively correlates with the aggressive phenotype of RCCs, and predicts poor prognosis for patients. Cancerous inhibitor of protein phosphatase 2A may be a novel target for prevention and treatment of RCC metastasis and recurrence.
Collapse
|
33
|
Böckelman C, Lassus H, Hemmes A, Leminen A, Westermarck J, Haglund C, Bützow R, Ristimäki A. Prognostic role of CIP2A expression in serous ovarian cancer. Br J Cancer 2011; 105:989-95. [PMID: 21897396 PMCID: PMC3185957 DOI: 10.1038/bjc.2011.346] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Cancerous inhibitor of protein phosphatase 2A (CIP2A) is an oncoprotein expressed in several solid cancers. Our purpose was to study its role in serous ovarian cancer patients, and the association to clinicopathological variables and molecular markers. Methods: We collected retrospectively 562 consecutive serous ovarian cancer patients treated at the Helsinki University Central Hospital. We stained tumour tissue microarrays for CIP2A by immunohistochemistry and constructed survival curves according to the Kaplan–Meier method. Associations to clinicopathological and molecular markers were assessed by the χ2-test. Results: We found strong cytoplasmic CIP2A immunoreactivity in 212 (40.4%) specimens, weak positivity in 222 (42.4%) specimens, and negative in 90 (17.2%). Immunopositive CIP2A expression was associated with high grade (P<0.0001), advanced stage (P=0.0005), and aneuploidy (P=0.001, χ2-test). Cancerous inhibitor of protein phosphatase 2A overexpression was also associated with EGFR protein expression (P=0.006) and EGFR amplification (P=0.043). Strong cytoplasmic CIP2A immunopositivity predicted poor outcome in ovarian cancer patients (P<0.0001, log-rank test). Conclusion: Our results show that CIP2A associates with reduced survival and parameters associated with high grade in ovarian cancer patients, and may thus be one of the factors that identify aggressive subtype (type II) of this disease.
Collapse
Affiliation(s)
- C Böckelman
- Department of Pathology, HUSLAB and Haartman Institute, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Guergnon J, Godet AN, Galioot A, Falanga PB, Colle JH, Cayla X, Garcia A. PP2A targeting by viral proteins: a widespread biological strategy from DNA/RNA tumor viruses to HIV-1. Biochim Biophys Acta Mol Basis Dis 2011; 1812:1498-507. [PMID: 21856415 DOI: 10.1016/j.bbadis.2011.07.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2011] [Revised: 06/22/2011] [Accepted: 07/05/2011] [Indexed: 12/27/2022]
Abstract
Protein phosphatase 2A (PP2A) is a large family of holoenzymes that comprises 1% of total cellular proteins and accounts for the majority of Ser/Thr phosphatase activity in eukaryotic cells. Although initially viewed as constitutive housekeeping enzymes, it is now well established that PP2A proteins represent a family of highly and sophistically regulated phosphatases. The past decade, multiple complementary studies have improved our knowledge about structural and functional regulation of PP2A holoenzymes. In this regard, after summarizing major cellular regulation, this review will mainly focus on discussing a particulate biological strategy, used by various viruses, which is based on the targeting of PP2A enzymes by viral proteins in order to specifically deregulate, for their own benefit, cellular pathways of their hosts. The impact of such PP2A targeting for research in human diseases, and in further therapeutic developments, is also discussed.
Collapse
Affiliation(s)
- Julien Guergnon
- Laboratoire E3 Phosphatases-Unité Signalisation Moléculaire et Activation Cellulaire, Institut Pasteur 25, rue du Dr Roux, 75015 Paris, France
| | | | | | | | | | | | | |
Collapse
|
35
|
Liu J, Wang X, Zhou G, Wang H, Xiang L, Cheng Y, Liu W, Wang Y, Jia J, Zhao W. Cancerous inhibitor of protein phosphatase 2A is overexpressed in cervical cancer and upregulated by human papillomavirus 16 E7 oncoprotein. Gynecol Oncol 2011; 122:430-6. [DOI: 10.1016/j.ygyno.2011.04.031] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2011] [Revised: 04/19/2011] [Accepted: 04/20/2011] [Indexed: 12/31/2022]
|
36
|
Overexpression and small molecule-triggered downregulation of CIP2A in lung cancer. PLoS One 2011; 6:e20159. [PMID: 21655278 PMCID: PMC3105001 DOI: 10.1371/journal.pone.0020159] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 04/12/2011] [Indexed: 11/19/2022] Open
Abstract
Background Lung cancer is the leading cause of cancer deaths worldwide, with a five-year overall survival rate of only 15%. Cancerous inhibitor of PP2A (CIP2A) is a human oncoprotein inhibiting PP2A in many human malignancies. However, whether CIP2A can be a new drug target for lung cancer is largely unclear. Methodology/Principal Findings Normal and malignant lung tissues were derived from 60 lung cancer patients from southern China. RT-PCR, Western blotting and immunohistochemistry were used to evaluate the expression of CIP2A. We found that among the 60 patients, CIP2A was undetectable or very low in paratumor normal tissues, but was dramatically elevated in tumor samples in 38 (63.3%) patients. CIP2A overexpression was associated with cigarette smoking. Silencing CIP2A by siRNA inhibited the proliferation and clonogenic activity of lung cancer cells. Intriguingly, we found a natural compound, rabdocoetsin B which is extracted from a Traditional Chinese Medicinal herb Rabdosia coetsa, could induce down-regulation of CIP2A and inactivation of Akt pathway, and inhibit proliferation and induce apoptosis in a variety of lung cancer cells. Conclusions/Significance Our findings strongly indicate that CIP2A could be an effective target for lung cancer drug development, and the therapeutic potentials of CIP2A-targeting agents warrant further investigation.
Collapse
|
37
|
Zhao Y, Zhou Y, Sun Y, Yu A, Yu H, Li W, Liu Z, Zeng J, Li X, Chen C, Jia J. Virulence factor cytotoxin-associated gene A in Helicobacter pylori is downregulated by interferon-γin vitro. ACTA ACUST UNITED AC 2010; 61:76-83. [DOI: 10.1111/j.1574-695x.2010.00750.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
38
|
Vaarala MH, Väisänen MR, Ristimäki A. CIP2A expression is increased in prostate cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2010; 29:136. [PMID: 20964854 PMCID: PMC2984408 DOI: 10.1186/1756-9966-29-136] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Accepted: 10/21/2010] [Indexed: 01/17/2023]
Abstract
BACKGROUND The CIP2A protein is a recently characterized oncoprotein which inhibits protein phosphatase 2A activity. Expression of CIP2A has been detected in several carcinomas, but its expression and significance in prostate cancer has not been examined so far. METHODS Expression of the CIP2A protein was studied using immunohistochemistry in prostate cancer (n = 59) and in benign prostatic hyperplasia (n = 20) specimens. The CIP2A staining scores were compared with several clinicopathological parameters. RESULTS Expression of CIP2A was increased in prostate cancer epithelium as compared with the benign hyperplastic epithelium (p < 0.001). The expression of CIP2A was associated with high Gleason scores (p < 0.001) and among patients treated with radical prostatectomy, CIP2A expression was associated with pre-treatment risk stratification (p = 0.011) and pathological T-class (p = 0.031). No statistically significant association was detected between CIP2A expression and prostate specific antigen concentrations. CONCLUSIONS Expression of the CIP2A protein is increased in prostate cancer specimens and its expression is associated with poorly differentiated and high-risk tumors.
Collapse
Affiliation(s)
- Markku H Vaarala
- Department of Surgery, Oulu University Hospital, PO Box 21, 90029 OYS, Finland.
| | | | | |
Collapse
|