1
|
Targeting Transcription Factors ATF5, CEBPB and CEBPD with Cell-Penetrating Peptides to Treat Brain and Other Cancers. Cells 2023; 12:cells12040581. [PMID: 36831248 PMCID: PMC9954556 DOI: 10.3390/cells12040581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Developing novel therapeutics often follows three steps: target identification, design of strategies to suppress target activity and drug development to implement the strategies. In this review, we recount the evidence identifying the basic leucine zipper transcription factors ATF5, CEBPB, and CEBPD as targets for brain and other malignancies. We describe strategies that exploit the structures of the three factors to create inhibitory dominant-negative (DN) mutant forms that selectively suppress growth and survival of cancer cells. We then discuss and compare four peptides (CP-DN-ATF5, Dpep, Bpep and ST101) in which DN sequences are joined with cell-penetrating domains to create drugs that pass through tissue barriers and into cells. The peptide drugs show both efficacy and safety in suppressing growth and in the survival of brain and other cancers in vivo, and ST101 is currently in clinical trials for solid tumors, including GBM. We further consider known mechanisms by which the peptides act and how these have been exploited in rationally designed combination therapies. We additionally discuss lacunae in our knowledge about the peptides that merit further research. Finally, we suggest both short- and long-term directions for creating new generations of drugs targeting ATF5, CEBPB, CEBPD, and other transcription factors for treating brain and other malignancies.
Collapse
|
2
|
Klempnauer KH. C/EBPβ sustains the oncogenic program of AML cells by cooperating with MYB and co-activator p300 in a transcriptional module. Exp Hematol 2022; 108:8-15. [PMID: 35032593 DOI: 10.1016/j.exphem.2022.01.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/02/2022] [Accepted: 01/07/2022] [Indexed: 11/17/2022]
Abstract
Transcription factor MYB is a key regulator of gene expression in hematopoietic cells and has emerged as a novel drug target for acute myeloid leukemia (AML). Studies aiming to identify potential MYB inhibitors have shown that the natural compound helenalin acetate (HA) inhibits viability and induces cell death and differentiation of AML cells by disrupting the MYB-induced gene expression program. Interestingly, CCAAT-box/enhancer binding protein beta (C/EBPβ), a transcription factor known to cooperate with MYB and the co-activator p300 in myeloid cells, rather than MYB itself, was identified as the primary target of HA. This supports a model in which MYB, C/EBPβ and p300 form the core of a transcriptional module that is essential for the maintenance of proliferative potential of AML cells, highlighting a novel role of C/EBPβ as a pro-leukemogenic factor.
Collapse
Affiliation(s)
- Karl-Heinz Klempnauer
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, D-48149 Münster, Germany.
| |
Collapse
|
3
|
Ghani LA, Yusenko MV, Frank D, Moorthy R, Widen JC, Dörner W, Khandanpour C, Harki DA, Klempnauer KH. A synthetic covalent ligand of the C/EBPβ transactivation domain inhibits acute myeloid leukemia cells. Cancer Lett 2022; 530:170-180. [DOI: 10.1016/j.canlet.2022.01.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/18/2022] [Accepted: 01/18/2022] [Indexed: 12/20/2022]
|
4
|
Yusenko MV, Trentmann A, Casolari DA, Abdel Ghani L, Lenz M, Horn M, Dörner W, Klempnauer S, Mootz HD, Arteaga MF, Mikesch JH, D'Andrea RJ, Gonda TJ, Müller-Tidow C, Schmidt TJ, Klempnauer KH. C/EBPβ is a MYB- and p300-cooperating pro-leukemogenic factor and promising drug target in acute myeloid leukemia. Oncogene 2021; 40:4746-4758. [PMID: 33958723 PMCID: PMC8298201 DOI: 10.1038/s41388-021-01800-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/29/2021] [Accepted: 04/14/2021] [Indexed: 02/05/2023]
Abstract
Transcription factor MYB has recently emerged as a promising drug target for the treatment of acute myeloid leukemia (AML). Here, we have characterized a group of natural sesquiterpene lactones (STLs), previously shown to suppress MYB activity, for their potential to decrease AML cell proliferation. Unlike what was initially thought, these compounds inhibit MYB indirectly via its cooperation partner C/EBPβ. C/EBPβ-inhibitory STLs affect the expression of a large number of MYB-regulated genes, suggesting that the cooperation of MYB and C/EBPβ broadly shapes the transcriptional program of AML cells. We show that expression of GFI1, a direct MYB target gene, is controlled cooperatively by MYB, C/EBPβ, and co-activator p300, and is down-regulated by C/EBPβ-inhibitory STLs, exemplifying that they target the activity of composite MYB-C/EBPβ-p300 transcriptional modules. Ectopic expression of GFI1, a zinc-finger protein that is required for the maintenance of hematopoietic stem and progenitor cells, partially abrogated STL-induced myelomonocytic differentiation, implicating GFI1 as a relevant target of C/EBPβ-inhibitory STLs. Overall, our data identify C/EBPβ as a pro-leukemogenic factor in AML and suggest that targeting of C/EBPβ may have therapeutic potential against AML.
Collapse
MESH Headings
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Humans
- CCAAT-Enhancer-Binding Protein-beta/metabolism
- CCAAT-Enhancer-Binding Protein-beta/genetics
- Proto-Oncogene Proteins c-myb/metabolism
- Proto-Oncogene Proteins c-myb/genetics
- Transcription Factors/metabolism
- Transcription Factors/genetics
- DNA-Binding Proteins/metabolism
- DNA-Binding Proteins/genetics
- Cell Proliferation
- E1A-Associated p300 Protein/metabolism
- E1A-Associated p300 Protein/genetics
- Cell Line, Tumor
- Lactones/pharmacology
- Gene Expression Regulation, Leukemic/drug effects
- Sesquiterpenes/pharmacology
Collapse
Affiliation(s)
- Maria V Yusenko
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, Münster, Germany
| | - Amke Trentmann
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, Münster, Germany
| | - Debora A Casolari
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Luca Abdel Ghani
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, Münster, Germany
| | - Mairin Lenz
- Institute for Pharmaceutical Biology and Phytochemistry, Westfälische-Wilhelms-Universität, Münster, Germany
| | - Melanie Horn
- Department of Medicine V, Hematology, Oncology, Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Wolfgang Dörner
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, Münster, Germany
| | | | - Henning D Mootz
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, Münster, Germany
| | - Maria Francisca Arteaga
- Department of Medicine A, Hematology and Oncology, University Hospital, Westfälische-Wilhelms-Universität, Münster, Germany
| | - Jan-Henrik Mikesch
- Department of Medicine A, Hematology and Oncology, University Hospital, Westfälische-Wilhelms-Universität, Münster, Germany
| | - Richard J D'Andrea
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Thomas J Gonda
- Cancer Research Institute, University of South Australia, Adelaide, SA, Australia
| | - Carsten Müller-Tidow
- Department of Medicine V, Hematology, Oncology, Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas J Schmidt
- Institute for Pharmaceutical Biology and Phytochemistry, Westfälische-Wilhelms-Universität, Münster, Germany
| | | |
Collapse
|
5
|
Afrough H, Ghafouri-Fard S, Yousefi H, Pakzad P, Kholghi Oskooei V, Taheri M. DICER-AS1 lncRNA: A putative culprit in the pathogenesis of gastric cancer. Exp Mol Pathol 2020; 116:104490. [PMID: 32663487 DOI: 10.1016/j.yexmp.2020.104490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/15/2020] [Accepted: 07/07/2020] [Indexed: 12/24/2022]
Abstract
The nuclear factor-κB (NF-κB) has a pivotal role in the pathogenesis of several cancers including gastric cancer. We have recently reported dysregulation of a number of NF-κB-associated lncRNAs in a variety of human disorders including breast cancer and coronary artery disease. In the current study, we evaluated expression of five NF-κB-associated lncRNAs (CHAST, ADINR, DICER1-AS1, HNF1A-AS1 and NKILA) and two NF-κB-associated-mRNA coding genes (CEBPA and ATG5) in gastric cancer tissues and their paired non-cancerous tissues using real time PCR method. Expression of DICER-AS1 was significantly down-regulated in gastric cancer tissues compared with the corresponding non-cancerous tissues (Expression ratio = 0.23, P value = .01). Expressions of other genes were not significantly different between these two sets of samples. Relative expression of DICER1-AS1 in cancer tissues versus non-cancerous tissues tended to associated with histological grade (P = .05). Tumoral expression levels of NKILA, ADINR, CEBPA and HNF1A-AS1 were significantly higher in patients with positive family history of cancer compared with those without such history (P values = .03, 0.02, 0.02 1nd 0.03, respectively). Besides, expression levels of NKILA, ADINR, DICER1-AS1, CEBPA, CHAST, HNF1A-AS1 and ATG5 were lower in H. pylori-infected tissues (P values = .01, 0.02, 0.03, 0.01, 0.004, 0.004 and 0.04, respectively). The lowest tumoral expression of DICER1-AS1 was detected in stage II cancers, while the highest expression of this lncRNA was reported in a single stage I tumor tissue. Similar pattern of expression was detected for ATG5. Significant pairwise correlations were demonstrated between expression levels of NF-ƙB-associated genes in both gastric cancer tissues and non-cancerous tissues. Expression levels of DICER1-AS1 had sensitivity and specificity values of 63.3% and 63.3% in differentiating between tumoral and non-tumoral tissues (Estimate criterion>6.96, J = 0.27, P value = .01, AUC = 0.67). Although previous studies have reported involvement of NF-κB pathway in the pathogenesis of gastric cancer, among the reported lncRNAs associated with this pathway, we could only detect differential expression of DICER1-AS1 between tumoral and non-tumoral tissues. Thus, the mechanism underlying dysregulation of this pathway might be different among various cancers.
Collapse
Affiliation(s)
- Hanieh Afrough
- College of Biology Sciences, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC, School of Medicine, New Orleans, USA
| | - Parviz Pakzad
- Department of Microbiology, School of Biology Sciences, North Branch, Islamic Azad University, Tehran, Iran
| | - Vahid Kholghi Oskooei
- Department of Laboratory Sciences, School of Paramedical Sciences, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran; Neuroscience Research Center, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Yang Y, Fan J, Han S, Li E. TNIP1 Inhibits Proliferation And Promotes Apoptosis In Clear Cell Renal Carcinoma Through Targeting C/Ebpβ. Onco Targets Ther 2019; 12:9861-9871. [PMID: 31819484 PMCID: PMC6874165 DOI: 10.2147/ott.s216138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 10/31/2019] [Indexed: 12/25/2022] Open
Abstract
Background/Purpose TNF-α-induced protein 3-interacting protein 1 (TNIP1) is active in various cancers, but its expression and function in renal cell carcinoma (RCC) have not been described. This study investigated the role of TNIP1 in clear cell renal cell carcinomas (ccRCC), which accounts for 75–80% of RCC and has a poor prognosis. Methods The expression of TNIP1 in human ccRCC tissues and cells was detected by real-time quantitative reverse transcription–polymerase chain reaction (qRT-PCR), Western blot (WB), and immunohistochemical (IHC) staining. Cell proliferation was assayed by a cell counting kit (CCK)-8 assay; cell cycle analysis and apoptosis assay were done by flow cytometry. Results TNIP1 is downregulated in both ccRCC human tissues and cells. Besides, TNIP1 downregulation promoted cell proliferation with more cell cycle entry, and inhibited apoptosis. TNIP1 downregulation was associated with increased of expression of the Bcl-2 anti-apoptosis gene and decreased expression of the Bax apoptosis-promoting gene and cleaved-caspase-3 by negatively regulating C/EBPβ expression. Conclusion TNIP1 acted as a tumor-inhibitor in ccRCC by targeting C/EBPβ. The results warrant study of TNIP1 as a potential diagnostic marker and therapeutic target of ccRCC.
Collapse
Affiliation(s)
- Yong Yang
- Department of Urology, The Ninth Hospital of Xi'an, Xi'an, Shaanxi, People's Republic of China
| | - Jinhai Fan
- Department of Urology, The First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, Shaanxi, People's Republic of China
| | - Shenglu Han
- Department of Urology, The Ninth Hospital of Xi'an, Xi'an, Shaanxi, People's Republic of China
| | - Enyuan Li
- Department of Urology, The Ninth Hospital of Xi'an, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
7
|
Ren M, McGowan E, Li Y, Zhu X, Lu X, Zhu Z, Lin Y, He S. Saikosaponin-d Suppresses COX2 Through p-STAT3/C/EBPβ Signaling Pathway in Liver Cancer: A Novel Mechanism of Action. Front Pharmacol 2019; 10:623. [PMID: 31191326 PMCID: PMC6549044 DOI: 10.3389/fphar.2019.00623] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 05/15/2019] [Indexed: 01/30/2023] Open
Abstract
Saikosaponin-d (SSd) is an active extract from Radix Bupleuri, the dried root from the plant Bupleurum falcatum used in China for thousands of years to treat liver diseases. The SSd extract possesses valuable pharmacological activities including anti-cancer and anti-inflammatory effects; however, the mechanism underlying the anti-cancer activity of SSd is largely unknown. Here, we explored the mechanism of action of SSd as an anti-cancer agent for liver cancer in two human hepatocellular carcinoma cell lines. Using MTT and annexin-V-FITC/PI assays, Western blots, immunohistochemistry, qRT-PCR, luciferase reporter assay, and a JAK2-specific inhibitor (AG490), we demonstrated that the anti-tumorigenic effects of SSd act through the intermediatory p-STAT3/C/EBPβ signaling pathway to suppress cyclooxygenase (COX)-2. SSd effectively inhibited cell proliferation in a dose-dependent manner. Apoptosis was significantly increased in cells treated with SSd (2.5–15 µg/ml) with concurrent increase and decrease in pro- and anti-apoptosis proteins, respectively. COX-2, C/EBPβ, and p-STAT3 were significantly decreased, at both the translational and transcriptional levels, by SSd treatment. AG490 produced similar inhibitory effects on STAT3, p-STAT3, C/EBPβ, and COX-2. In conclusion, our data suggest that SSd controls liver cancer proliferation through suppression of the p-STAT3/C/EBPβ signaling pathway inhibiting COX2 expression. These findings further our understanding of the pharmacological action of SSd, providing new information on SSd mechanism of action and showing potential for SSd as a novel therapy for liver cancer.
Collapse
Affiliation(s)
- Mudan Ren
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiao tong University, Xi'an, China
| | - Eileen McGowan
- School of Life Sciences, University of Technology Sydney, Broadway, NSW, Australia
| | - Yarui Li
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiao tong University, Xi'an, China
| | - Xiaofeng Zhu
- Department of Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xinlan Lu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiao tong University, Xi'an, China
| | - Zhanfang Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiao tong University, Xi'an, China
| | - Yiguang Lin
- School of Life Sciences, University of Technology Sydney, Broadway, NSW, Australia
| | - Shuixiang He
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiao tong University, Xi'an, China
| |
Collapse
|
8
|
Gil-Martín E, Egea J, Reiter RJ, Romero A. The emergence of melatonin in oncology: Focus on colorectal cancer. Med Res Rev 2019; 39:2239-2285. [PMID: 30950095 DOI: 10.1002/med.21582] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/04/2019] [Accepted: 03/16/2019] [Indexed: 12/17/2022]
Abstract
Within the last few decades, melatonin has increasingly emerged in clinical oncology as a naturally occurring bioactive molecule with substantial anticancer properties and a pharmacological profile optimal for joining the currently available pharmacopeia. In addition, extensive experimental data shows that this chronobiotic agent exerts oncostatic effects throughout all stages of tumor growth, from initial cell transformation to mitigation of malignant progression and metastasis; additionally, melatonin alleviates the side effects and improves the welfare of radio/chemotherapy-treated patients. Thus, the support of clinicians and oncologists for the use of melatonin in both the treatment and proactive prevention of cancer is gaining strength. Because of its epidemiological importance and symptomatic debut in advanced stages of difficult clinical management, colorectal cancer (CRC) is a preferential target for testing new therapies. In this regard, the development of effective forms of clinical intervention for the improvement of CRC outcome, specifically metastatic CRC, is urgent. At the same time, the need to reduce the costs of conventional anti-CRC therapy results is also imperative. In light of this status quo, the therapeutic potential of melatonin, and the direct and indirect critical processes of CRC malignancy it modulates, have aroused much interest. To illuminate the imminent future on CRC research, we focused our attention on the molecular mechanisms underlying the multiple oncostatic actions displayed by melatonin in the onset and evolution of CRC and summarized epidemiological evidence, as well as in vitro, in vivo and clinical findings that support the broadly protective potential demonstrated by melatonin.
Collapse
Affiliation(s)
- Emilio Gil-Martín
- Department of Biochemistry, Genetics and Immunology, Biomedical Research Center (CINBIO, 'Centro Singular de Investigación de Galicia'), University of Vigo, Vigo, Spain
| | - Javier Egea
- Molecular Neuroinflammation and Neuronal Plasticity Laboratory, Research Unit, Hospital Universitario Santa Cristina, Madrid, Spain.,Servicio de Farmacología Clínica, Instituto de Investigación Sanitaria, Hospital Universitario de la Princesa, Madrid, Spain.,Departamento de Farmacología y Terapéutica, Instituto-Fundación Teófilo Hernando, Universidad Autónoma de Madrid, Madrid, Spain
| | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health Science Center, San Antonio, Texas, USA
| | - Alejandro Romero
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
9
|
C/EBPβ Is a Transcriptional Regulator of Wee1 at the G₂/M Phase of the Cell Cycle. Cells 2019; 8:cells8020145. [PMID: 30754676 PMCID: PMC6407104 DOI: 10.3390/cells8020145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 02/06/2019] [Accepted: 02/09/2019] [Indexed: 12/25/2022] Open
Abstract
The CCAAT/enhancer-binding protein β (C/EBPβ) is a transcription factor that regulates cellular proliferation, differentiation, apoptosis and tumorigenesis. Although the pro-oncogenic roles of C/EBPβ have been implicated in various human cancers, how it contributes to tumorigenesis or tumor progression has not been determined. Immunohistochemistry with human non-small cell lung cancer (NSCLC) tissues revealed that higher levels of C/EBPβ protein were expressed compared to normal lung tissues. Knockdown of C/EBPβ by siRNA reduced the proliferative capacity of NSCLC cells by delaying the G2/M transition in the cell cycle. In C/EBPβ-knockdown cells, a prolonged increase in phosphorylation of cyclin dependent kinase 1 at tyrosine 15 (Y15-pCDK1) was displayed with simultaneously increased Wee1 and decreased Cdc25B expression. Chromatin immunoprecipitation (ChIP) analysis showed that C/EBPβ bound to distal promoter regions of WEE1 and repressed WEE1 transcription through its interaction with histone deacetylase 2. Treatment of C/EBPβ-knockdown cells with a Wee1 inhibitor induced a decrease in Y15-pCDK1 and recovered cells from G2/M arrest. In the xenograft tumors, the depletion of C/EBPβ significantly reduced tumor growth. Taken together, these results indicate that Wee1 is a novel transcription target of C/EBPβ that is required for the G2/M phase of cell cycle progression, ultimately regulating proliferation of NSCLC cells.
Collapse
|
10
|
Bustos-Carpinteyro AR, Oliveira C, Sousa A, Oliveira P, Pinheiro H, Carvalho J, Magaña-Torres MT, Flores-Miramontes MG, Aguilar-Lemarroy A, Jave-Suárez LF, Peregrina-Sandoval J, Cruz-Ramos JA, Sánchez-López JY. CDH1 somatic alterations in Mexican patients with diffuse and mixed sporadic gastric cancer. BMC Cancer 2019; 19:69. [PMID: 30642281 PMCID: PMC6332846 DOI: 10.1186/s12885-019-5294-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 01/07/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Diffuse gastric cancer (DGC) is associated with the reduction or absence of the expression of the cell adhesion protein E-cadherin (encoded by the CDH1 gene). Molecular characteristics are less well described for mixed gastric cancer (MGC). The main somatic alterations that have been described in the CDH1 gene are mutations, loss of heterozygosity (LOH) and promoter methylation. The aim was to analyze CDH1 somatic alterations in Mexican patients with diffuse and mixed gastric cancer. METHODS We searched for mutations in the CDH1 gene in tumor DNA from DGC (n = 13) and MGC (n = 7) patients by next generation sequencing (NGS). Validation of findings was performed using Sanger sequencing. LOH was analyzed using dinucleotide repeat markers surrounding the CDH1 gene, and methylation was investigated by DNA bisulfite conversion and sequencing. E-cadherin protein deficiency was analyzed by immunohistochemistry. RESULTS Seventeen point variants were identified by NGS, 13 of them were validated by Sanger sequencing. Only 1/13 had not been previously reported (c.-137C > A), and 12/13 were already reported as polymorphisms. Two DGC cases presented LOH at the locus 16q22.1 (13.3%). CDH1 promoter methylation was positive in (7/11) 63.6% and (4/6) 66.6% of the cases with DGC and MGC, respectively. E-cadherin protein deficiency was observed in 58.3% of DGC cases while 100% in MGC cases. CONCLUSIONS While no pathogenic somatic mutations were found that could explain the diffuse histology of gastric cancer in DGC and MGC, methylation was the most common somatic inactivation event of the CDH1 gene, and LOH was rare. The previously unreported c.-137C > A variant modify the CDH1 gene expression since it alters the binding sites for transcription factors.
Collapse
Affiliation(s)
- Andrea Rebeca Bustos-Carpinteyro
- División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada N. 800, Col. Independencia, C. P. 44340, Guadalajara, Jalisco, México.,Doctorado en Genética Humana, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Carla Oliveira
- Expression Regulation in Cancer, IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S; University of Porto, Porto, Portugal.,Faculty of Medicine, University of Porto, Porto, Portugal
| | - Abel Sousa
- Expression Regulation in Cancer, IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S; University of Porto, Porto, Portugal
| | - Patricia Oliveira
- Expression Regulation in Cancer, IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S; University of Porto, Porto, Portugal
| | - Hugo Pinheiro
- Expression Regulation in Cancer, IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S; University of Porto, Porto, Portugal.,Department of Internal Medicine, Centro Hospitalar Tâmega e Sousa Avenida do Hospital Padre Américo, N° 210 4564-007, Guilhufe - Penafiel, Portugal
| | - Joana Carvalho
- Expression Regulation in Cancer, IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S; University of Porto, Porto, Portugal
| | - María Teresa Magaña-Torres
- División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada N. 800, Col. Independencia, C. P. 44340, Guadalajara, Jalisco, México
| | - María Guadalupe Flores-Miramontes
- División de Inmunología, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México
| | - Adriana Aguilar-Lemarroy
- División de Inmunología, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México
| | - Luis Felipe Jave-Suárez
- División de Inmunología, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México
| | - Jorge Peregrina-Sandoval
- Laboratorio de Inmunobiología, Departamento de Biología Celular y Molecular. Centro Universitario de Ciencias Biológicas y Agropecuarias, Universidad de Guadalajara, CP 45510, Nextipac, Jalisco, México
| | | | - Josefina Yoaly Sánchez-López
- División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada N. 800, Col. Independencia, C. P. 44340, Guadalajara, Jalisco, México.
| |
Collapse
|
11
|
do Rosário Pinheiro D, Harada ML, Rodriguez Burbano RM, do Nascimento Borges B. COX-2 gene expression and methylation profile in Sapajus apella as an experimental model for gastric adenocarcinoma. Genet Mol Biol 2018; 41:496-501. [PMID: 29767663 PMCID: PMC6082229 DOI: 10.1590/1678-4685-gmb-2016-0329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 10/19/2017] [Indexed: 11/27/2022] Open
Abstract
Gastric cancer (GC) remains one of the main causes of cancer-related death worldwide. There are two distinct histological types of GC: diffuse and intestinal. The latter is characterized by the presence of pre-neoplastic lesions. One of the most frequently altered enzymes in intestinal GC is COX-2, an important lesion marker. This work aimed to study COX-2 methylation and expression in N-methyl-N-Nitrosurea (MNU)-induced intestinal GC in six Sapajus apella animals. The partial promoter sequence of S. apella COX-2 gene was obtained and used to identify transcription factors and cis-regulatory element binding sites. The COX-2 methylation pattern was assessed using Methylation-Specific PCR (MSP), and expression was analyzed by immunohistochemistry (IHQ). A total of 20 samples were obtained. A 675 bp fragment of the S. apella COX-2 promoter region was obtained, and it was 99.2% and 68.2% similar to H. sapiens and S. boliviensis, respectively. Similar to humans, several transcription factors and cis-regulatory element binding sites were identified in the S. apella sequence. MSP revealed that all samples were methylated. However, IHQ results demonstrated positive COX-2 expression in all pre-neoplastic and tumoral samples. The results suggest that the analyzed fragment is not crucial in COX-2 regulation of GC in S. apella.
Collapse
Affiliation(s)
- Danilo do Rosário Pinheiro
- Universidade Federal do
ParáUniversidade Federal do ParáInstituto de Ciências
BiológicasMolecular Biology LaboratoryBelémPABrazilMolecular Biology Laboratory, Instituto de
Ciências Biológicas. Universidade Federal do Pará, Belém, PA,
Brazil
| | - Maria Lucia Harada
- Universidade Federal do
ParáUniversidade Federal do ParáInstituto de Ciências
BiológicasMolecular Biology LaboratoryBelémPABrazilMolecular Biology Laboratory, Instituto de
Ciências Biológicas. Universidade Federal do Pará, Belém, PA,
Brazil
| | - Rommel Mario Rodriguez Burbano
- Universidade Federal do
ParáUniversidade Federal do ParáInstituto de Ciências
BiológicasHuman Cytogenetics LaboratoryBelémPABrazilHuman Cytogenetics Laboratory, Instituto de
Ciências Biológicas. Universidade Federal do Pará, Belém, PA,
Brazil
| | - Barbara do Nascimento Borges
- Universidade Federal do
ParáUniversidade Federal do ParáInstituto de Ciências
BiológicasMolecular Biology LaboratoryBelémPABrazilMolecular Biology Laboratory, Instituto de
Ciências Biológicas. Universidade Federal do Pará, Belém, PA,
Brazil
| |
Collapse
|
12
|
Tomizawa M, Shinozaki F, Motoyoshi Y, Sugiyama T, Yamamoto S, Ishige N. CCAAT/enhancer-binding protein α decreases the viability of gastric cancer cells. Oncol Lett 2017; 13:4322-4326. [PMID: 28599433 DOI: 10.3892/ol.2017.5987] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 02/23/2017] [Indexed: 01/13/2023] Open
Abstract
CCAAT/enhancer-binding protein (C/EBP) α, C/EBPβ and C/EBPδ are involved in inflammation and cell differentiation. In the present study, their roles in human gastric cancer cells were investigated. The human gastric cancer cell lines MKN45 and MKN74 were subjected to the reverse transcription-quantitative polymerase chain reaction (RT-qPCR) to analyze the expression levels of C/EBPα, C/EBPβ and C/EBPδ. The cells were transfected with expression plasmids for either C/EBPα or C/EBPδ, and subjected to a 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium inner salt (MTS) assay and RT-qPCR for analysis of cyclin D1 expression. Expression levels of C/EBPα and C/EBPδ were decreased in MKN45 and MKN74 cells compared with in normal gastric tissue. Expression levels of C/EBPβ were decreased in MKN45 cells and increased in MKN74 cells. Viability of MKN45 cells was decreased by C/EBPα and C/EBPδ. Viability of MKN74 cells was decreased by C/EBPα, but increased by C/EBPδ. Expression levels of cyclin D1 were decreased in association with C/EBPα and C/EBPδ overexpression in MKN45 cells. Expression levels of cyclin D1 were decreased in association with C/EBPα overexpression, but increased in association with C/EBPδ overexpression, in MKN74 cells. The results of the present study indicate that C/EBPα is potentially useful for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Minoru Tomizawa
- Department of Gastroenterology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Fuminobu Shinozaki
- Department of Radiology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Yasufumi Motoyoshi
- Department of Neurology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Takao Sugiyama
- Department of Rheumatology, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Shigenori Yamamoto
- Department of Pediatrics, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| | - Naoki Ishige
- Department of Neurosurgery, National Hospital Organization, Shimoshizu Hospital, Yotsukaido, Chiba 284-0003, Japan
| |
Collapse
|
13
|
Falkenberg KD, Jakobs A, Matern JC, Dörner W, Uttarkar S, Trentmann A, Steinmann S, Coulibaly A, Schomburg C, Mootz HD, Schmidt TJ, Klempnauer KH. Withaferin A, a natural compound with anti-tumor activity, is a potent inhibitor of transcription factor C/EBPβ. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1349-1358. [PMID: 28476645 DOI: 10.1016/j.bbamcr.2017.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 04/27/2017] [Accepted: 05/01/2017] [Indexed: 02/07/2023]
Abstract
Recent work has shown that deregulation of the transcription factor Myb contributes to the development of leukemia and several other human cancers, making Myb and its cooperation partners attractive targets for drug development. By employing a myeloid Myb-reporter cell line we have identified Withaferin A (WFA), a natural compound that exhibits anti-tumor activities, as an inhibitor of Myb-dependent transcription. Analysis of the inhibitory mechanism of WFA showed that WFA is a significantly more potent inhibitor of C/EBPβ, a transcription factor cooperating with Myb in myeloid cells, than of Myb itself. We show that WFA covalently modifies specific cysteine residues of C/EBPβ, resulting in the disruption of the interaction of C/EBPβ with the co-activator p300. Our work identifies C/EBPβ as a novel direct target of WFA and highlights the role of p300 as a crucial co-activator of C/EBPβ. The finding that WFA is a potent inhibitor of C/EBPβ suggests that inhibition of C/EBPβ might contribute to the biological activities of WFA.
Collapse
Affiliation(s)
- Kim D Falkenberg
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, D-48149 Münster, Germany
| | - Anke Jakobs
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, D-48149 Münster, Germany
| | - Julian C Matern
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, D-48149 Münster, Germany
| | - Wolfgang Dörner
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, D-48149 Münster, Germany
| | - Sagar Uttarkar
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, D-48149 Münster, Germany
| | - Amke Trentmann
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, D-48149 Münster, Germany
| | - Simone Steinmann
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, D-48149 Münster, Germany
| | - Anna Coulibaly
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, D-48149 Münster, Germany
| | - Caroline Schomburg
- Institute for Pharmaceutical Biology and Phytochemistry, Westfälische-Wilhelms-Universität, D-48149 Münster, Germany
| | - Henning D Mootz
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, D-48149 Münster, Germany
| | - Thomas J Schmidt
- Institute for Pharmaceutical Biology and Phytochemistry, Westfälische-Wilhelms-Universität, D-48149 Münster, Germany
| | - Karl-Heinz Klempnauer
- Institute for Biochemistry, Westfälische-Wilhelms-Universität, D-48149 Münster, Germany.
| |
Collapse
|
14
|
Regalo G, Förster S, Resende C, Bauer B, Fleige B, Kemmner W, Schlag PM, Meyer TF, Machado JC, Leutz A. C/EBPβ regulates homeostatic and oncogenic gastric cell proliferation. J Mol Med (Berl) 2016; 94:1385-1395. [PMID: 27522676 PMCID: PMC5143359 DOI: 10.1007/s00109-016-1447-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 06/24/2016] [Accepted: 07/17/2016] [Indexed: 12/26/2022]
Abstract
Abstract Cancer of the stomach is among the leading causes of death from cancer worldwide. The transcription factor C/EBPβ is frequently overexpressed in gastric cancer and associated with the suppression of the differentiation marker TFF1. We show that the murine C/EBPβ knockout stomach displays unbalanced homeostasis and reduced cell proliferation and that tumorigenesis of human gastric cancer xenograft is inhibited by knockdown of C/EBPβ. Cross-species comparison of gene expression profiles between C/EBPβ-deficient murine stomach and human gastric cancer revealed a subset of tumors with a C/EBPβ signature. Within this signature, the RUNX1t1 tumor suppressor transcript was down-regulated in 38 % of gastric tumor samples. The RUNX1t1 promoter was frequently hypermethylated and ectopic expression of RUNX1t1 in gastric cancer cells inhibited proliferation and enhanced TFF1 expression. These data suggest that the tumor suppressor activity of both RUNX1t1 and TFF1 are mechanistically connected to C/EBPβ and that cross-regulation between C/EBPβ-RUNX1t1-TFF1 plays an important role in gastric carcinogenesis. Key message C/EBPβ controls proliferation and differentiation balance in the stomach. Homeostatic differentiation/proliferation balance is altered in gastric cancer. RUNX1t1 is a C/EBPβ-associated tumor suppressor. RUNX1t1 negatively regulates C/EBPβ pro-oncogenic functions.
Electronic supplementary material The online version of this article (doi:10.1007/s00109-016-1447-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Goncalo Regalo
- Max-Delbrueck-Center for Molecular Medicine, Robert-Roessle-Str. 10, 13125, Berlin, Germany.
| | - Susann Förster
- Max-Delbrueck-Center for Molecular Medicine, Robert-Roessle-Str. 10, 13125, Berlin, Germany
| | - Carlos Resende
- Institute of Pathology and Molecular Immunology of the University of Porto, 4200-465, Porto, Portugal
| | - Bianca Bauer
- Max-Planck Institute for Infection Biology, 10117, Berlin, Germany
| | - Barbara Fleige
- Institut für Gewebediagnostik Berlin am MVZ des HELIOS Klinikum, 13125, Berlin, Germany
| | - Wolfgang Kemmner
- Max-Delbrueck-Center for Molecular Medicine, Robert-Roessle-Str. 10, 13125, Berlin, Germany
| | - Peter M Schlag
- Charité Comprehensive Cancer Centers, Charité-Universitätsmedizin, 10117, Berlin, Germany
| | - Thomas F Meyer
- Max-Planck Institute for Infection Biology, 10117, Berlin, Germany
| | - José C Machado
- Institute of Pathology and Molecular Immunology of the University of Porto, 4200-465, Porto, Portugal
| | - Achim Leutz
- Max-Delbrueck-Center for Molecular Medicine, Robert-Roessle-Str. 10, 13125, Berlin, Germany.
- Humboldt-University of Berlin, Institute of Biology, 10115, Berlin, Germany.
| |
Collapse
|
15
|
Wu SM, Lin WY, Shen CC, Pan HC, Keh-Bin W, Chen YC, Jan YJ, Lai DW, Tang SC, Tien HR, Chiu CS, Tsai TC, Lai YL, Sheu ML. Melatonin set out to ER stress signaling thwarts epithelial mesenchymal transition and peritoneal dissemination via calpain-mediated C/EBPβ and NFκB cleavage. J Pineal Res 2016; 60:142-54. [PMID: 26514342 DOI: 10.1111/jpi.12295] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 10/26/2015] [Indexed: 12/11/2022]
Abstract
Peritoneal dissemination of tumor has high mortality and is associated with the loss of epithelial features, acquisition of motile mesenchymal morphology characteristics, and invasive properties by tumor cells. Melatonin is an endogenously produced molecule in all plant species that is known to exert antitumor activity, but to date, its underlying mechanisms and antiperitoneal metastasis efficacy is not well defined. This study determined the antiperitoneal dissemination potential of melatonin in vivo and assessed its association with the inhibition of epithelial-to-mesenchymal transition (EMT) signaling mechanism by endoplasmic reticulum (ER) stress, which may be a major molecular mechanism of melatonin against cancer. The results demonstrate that melatonin inhibited peritoneal metastasis in vivo and activated ER stress in Cignal ERSE Reporter Assay, organelle structure in transmission electron microscopy images, calpain activity, and protein biomarkers like p-elf2α. Moreover, the overexpression of transcription factor C/EBPβ in gastric cancer interacted with NFκB and further regulates COX-2 expression. These were dissociated and downregulated by melatonin, as proven by immunofluorescence imaging, immunoprecipitation, EMSA, and ChIP assay. Melatonin or gene silencing of C/EBPβ decreased the EMT protein markers (E-cadherin, Snail, and Slug) and Wnt/beta-catenin activity by Topflash activity, and increased ER stress markers. In an animal study, the results of melatonin therapy were consistent with those of in vitro findings and attenuated systemic proangiogenesis factor production. In conclusion, C/EBPβ and NFκB inhibition by melatonin may impede both gastric tumor growth and peritoneal dissemination by inducing ER stress and inhibiting EMT.
Collapse
Affiliation(s)
- Sheng-Mao Wu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Wan-Yu Lin
- Department of Nuclear Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chin-Chang Shen
- Institute of Nuclear Energy Research, Atomic Energy Council, Taoyuan, Taiwan
| | - Hung-Chuan Pan
- Faculty of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Wang Keh-Bin
- Department of Nuclear Medicine, Kuang Tien General Hospital, Taichung, Taiwan
| | - Yi-Ching Chen
- Department of Nuclear Medicine, Kuang Tien General Hospital, Taichung, Taiwan
| | - Yee-Jee Jan
- Department of Pathology and Laboratory Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - De-Wei Lai
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Shu-Ching Tang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Hsing-Ru Tien
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Chien-Shan Chiu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Tsung-Chih Tsai
- Department of life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Liang Lai
- Division of Gastroenterology, Department of Internal Medicine, Armed Forces Taichung General Hospital, Taiping, Taichung, Taiwan
| | - Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan
- Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
16
|
Skierucha M, Milne ANA, Offerhaus GJA, Polkowski WP, Maciejewski R, Sitarz R. Molecular alterations in gastric cancer with special reference to the early-onset subtype. World J Gastroenterol 2016; 22:2460-74. [PMID: 26937134 PMCID: PMC4768192 DOI: 10.3748/wjg.v22.i8.2460] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 11/06/2015] [Accepted: 12/30/2015] [Indexed: 02/06/2023] Open
Abstract
Currently, gastric cancer (GC) is one of the most frequently diagnosed neoplasms, with a global burden of 723000 deaths in 2012. It is the third leading cause of cancer-related death worldwide. There are numerous possible factors that stimulate the pro-carcinogenic activity of important genes. These factors include genetic susceptibility expressed in a single-nucleotide polymorphism, various acquired mutations (chromosomal instability, microsatellite instability, somatic gene mutations, epigenetic alterations) and environmental circumstances (e.g., Helicobcter pylori infection, EBV infection, diet, and smoking). Most of the aforementioned pathways overlap, and authors agree that a clear-cut pathway for GC may not exist. Thus, the categorization of carcinogenic events is complicated. Lately, it has been claimed that research on early-onset gastric carcinoma (EOGC) and hereditary GC may contribute towards unravelling some part of the mystery of the GC molecular pattern because young patients are less exposed to environmental carcinogens and because carcinogenesis in this setting may be more dependent on genetic factors. The comparison of various aspects that differ and coexist in EOGCs and conventional GCs might enable scientists to: distinguish which features in the pathway of gastric carcinogenesis are modifiable, discover specific GC markers and identify a specific target. This review provides a summary of the data published thus far concerning the molecular characteristics of GC and highlights the outstanding features of EOGC.
Collapse
|
17
|
Interleukin-1B signalling leads to increased survival of gastric carcinoma cells through a CREB-C/EBPβ-associated mechanism. Gastric Cancer 2016; 19:74-84. [PMID: 25740226 DOI: 10.1007/s10120-014-0448-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 11/17/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND Polymorphisms in inflammation-related genes have been associated with a risk of gastric carcinoma (GC). However, the biological mechanisms underlying these associations are still elusive. Our objective was to determine whether chronic inflammation-associated IL1Β signalling, as seen in the context of Helicobacter pylori infection, could be linked to gastric carcinogenesis by modulating the behaviour of gastric epithelial cells. METHODS The effect of IL1B was assessed by studying the expression and activation status of the IL1Β-activated transcription factors C/EBPβ and CREB in GC cell lines. Interaction between CREB and C/EBPβ was explored through interference RNA, chromatin immunoprecipitation and chemical inhibition. CREB and C/EBPβ expression was analysed in 66 samples of primary GC and in normal gastric mucosa. GC cell growth was analysed in vitro by BrdU incorporation and in vivo employing a chicken embryo chorioallantoic membrane model. RESULTS We found that IL1B regulates the expression/activation status of both C/EBPβ and CREB in GC cells through an ERK1/2-dependent mechanism. Our results show that CREB is a direct transactivator of CEBPB, acting as an upstream effector in this regulatory mechanism. Furthermore, we found CREB to be overexpressed in 94 % of GC samples and significantly associated with C/EBPβ expression (P < 0.05). Finally, we demonstrated both in vitro and in vivo that CREB can mediate IL1B-induced GC cell proliferation. CONCLUSIONS Our results support the hypothesis that the effect of chronic inflammation on gastric carcinogenesis, as seen in the context of genetically susceptible individuals infected with Helicobacter pylori, includes the modulation of signalling pathways that regulate survival mechanisms in epithelial cells. IL1B is able to increase the expression/activation status of CREB and its target gene C/EBPβ, which are mandatory for GC cell survival. Our results may help inform new strategies for the prevention and treatment of GC, including the control of chronic inflammation.
Collapse
|
18
|
Hu TZ, Huang LH, Xu CX, Liu XM, Wang Y, Xiao J, Zhou L, Luo L, Jiang XX. Expressional profiles of transcription factors in the progression of Helicobacter pylori-associated gastric carcinoma based on protein/DNA array analysis. Med Oncol 2015; 32:265. [PMID: 26563475 DOI: 10.1007/s12032-015-0711-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/02/2015] [Indexed: 12/17/2022]
Abstract
Transcription factors (TFs) are crucial modulators of gene expression during the development and progression of gastric carcinoma. Helicobacter pylori (H. pylori) is one of the most significant risk factors of gastric carcinoma, and it is widely known that chronic inflammation with H. pylori infection triggers gastric carcinogenesis through inflammation-carcinoma chain [gastric carcinogenesis stages: non-atrophic gastritis, chronic atrophic gastritis, intestinal metaplasia, dysplasia and gastric carcinoma (GC)], but its mechanism regarding changed TFs remains unknown. In this study, we investigated the expressional profiles of 345 transcription factors in gastric mucosa of healthy volunteers and patients at different gastric carcinogenesis stages using protein/DNA array-based approach. The data demonstrated the up-regulated TFs such as GATA-3, AP4, c-Myc and Pbx1 in the gastric mucosa of GC patients compared with the healthy volunteers, while other TFs, particularly CCAAT and CACC, showed the consistently decreasing trend along the gastric carcinogenesis. The increased expressions of AP4, Pbx1 and C/EBPα were further validated by quantitative real-time PCR and Western blot in various H. pylori-infected models such as clinical gastric tissues, gastric epithelial cell lines and Mongolian gerbils. This study provides insights into and potential laws for gene transcriptional regulation by identifying potential TFs targets against the development of H. pylori-associated gastric carcinoma.
Collapse
Affiliation(s)
- Ting-Zi Hu
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, 138 Tongzipo Street, Changsha, 410013, Hunan Province, China
| | - Li-Hua Huang
- Center for Medical Experiment, Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan Province, China
| | - Can-Xia Xu
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, 138 Tongzipo Street, Changsha, 410013, Hunan Province, China.
- Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Changsha, 410013, China.
| | - Xiao-Ming Liu
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, 138 Tongzipo Street, Changsha, 410013, Hunan Province, China
| | - Yu Wang
- Department of Internal Medicine, The Third People's Hospital of Huaihua, Huaihua, 418000, Hunan Province, China
| | - Jing Xiao
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, 138 Tongzipo Street, Changsha, 410013, Hunan Province, China
| | - Li Zhou
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, 138 Tongzipo Street, Changsha, 410013, Hunan Province, China
| | - Ling Luo
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, 138 Tongzipo Street, Changsha, 410013, Hunan Province, China
| | - Xiao-Xia Jiang
- Department of Gastroenterology, Third Xiangya Hospital of Central South University, 138 Tongzipo Street, Changsha, 410013, Hunan Province, China
| |
Collapse
|
19
|
C/EBP-β-activated microRNA-223 promotes tumour growth through targeting RASA1 in human colorectal cancer. Br J Cancer 2015; 112:1491-500. [PMID: 25867276 PMCID: PMC4453668 DOI: 10.1038/bjc.2015.107] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 02/19/2015] [Indexed: 01/02/2023] Open
Abstract
Background: Evidences have shown that the RAS signalling pathway plays an important role in colorectal cancer (CRC). Moreover, RAS-GTPase-activating proteins (RASGAPs) as RAS signalling terminators are associated with tumourigenicity and tumour progression. In this study, we used bioinformatics analysis to predict and study important miRNAs that could target RAS p21 GTPase-activating protein 1 (RASA1), an important member of RASGAPs. Methods: The levels of RASA1 and miR-223 were analysed by real-time PCR, western blotting or in situ immunofluorescence analyses. The functional effects of miR-223 and the effects of miR-223-targeted inhibitors were examined in vivo using established assays. Results: Upregulation of miR-223 was detected in CRC tissues (P<0.01) and was involved in downregulation of RASA1 in CRC tissues. Furthermore, the direct inhibition of RASA1 translation by miR-223 and the activation of miR-223 by CCAAT/enhancer binding protein-β (C/EBP-β) were evaluated in CRC cells. An in vivo xenograft model of CRC suggested that the upregulation of miR-223 could promote tumour growth and that the inhibition of miR-223 might prevent solid tumour growth. Conclusions: These results identify that C/EBP-β-activated miR-223 contributes to tumour growth by targeting RASA1 in CRC and miR-223-targeted inhibitors may have clinical promise for CRC treatment via suppression of miR-223.
Collapse
|
20
|
Telomerase reverse transcriptase inhibition stimulates cyclooxygenase 2 expression in cancer cells and synergizes with celecoxib to exert anti-cancer effects. Br J Cancer 2013; 108:2272-80. [PMID: 23681187 PMCID: PMC3681031 DOI: 10.1038/bjc.2013.208] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background: Telomerase and telomerase reverse transcriptase (hTERT) confer cancer cells sustained proliferation and survival potentials. Targeting telomerase or hTERT is a novel anti-cancer strategy. However, telomerase/hTERT inhibition alone has minimal clinical efficacy. We explored the relationship between hTERT and cyclooxygenase 2 (COX2) and evaluated synergistic anti-cancer effects of targeting both hTERT and COX2. Methods: hTERT was depleted in gastric and cervical cancer cells using small interfering RNA (siRNA) and analysed for COX2 expression using quantitative PCR and immunoblotting. Viable cells and apoptotic cells in gastric cancer cells treated with hTERT siRNA or/and the COX2 inhibitor celecoxib were measured using Trypen blue exclusion and flow cytometry. The in vivo anti-cancer effect of hTERT depletion or/and celecoxib was evaluated using mouse xenograft models. Results: Knocking down hTERT expression in cancer cells led to robust increases in mRNA and protein levels of COX2. The COX2 promoter activity increased substantially in hTERT-depleted cells. hTERT depletion led to the activation of p38 mitogen-activated protein kinase responsible for the stimulation of COX2 gene transcription. hTERT depletion or celecoxib alone did not affect cancer cell survival, whereas their combination synergistically killed them both in vitro and in vivo. Conclusion: hTERT induces COX2 expression and simultaneously targeting hTERT and COX2 synergistically kills cancer cells.
Collapse
|
21
|
Abstract
OBJECTIVE Several gastrointestinal peptides are now recognized to have target functions beyond the intestinal wall, including effects on adipocytes. Secretin (SEC), one of the first identified, has not been evaluated in this context. METHODS Using cultured 3T3-L1 preadipocytes, adipocytes and primary rat adipocytes we evaluated the effect of SEC on cell proliferation, mitochondrial activity, differentiation, triglyceride (TG) synthesis, lipolysis as well expression of the SEC receptor (SCTR) in rodent and human adipose tissues. RESULTS In preadipocytes, SEC significantly increased mitochondrial activity (115%; P<0.01), thymidine incorporation (149.7%; P<0.05) and C/EBPβ expression (123.4%; P<0.05). During standard differentiation, SCTR mRNA increased up to a maximum of ninefold (P<0.001). In human adipose tissue, SCTR correlated with body mass index and plasma insulin, and SCTR mRNA expression was also detected in rat adipose tissues. SEC supplementation during differentiation enhanced TG accumulation (+138%; P<0.01). In mature adipocytes, SEC increased fatty acid (FA) uptake (186%; P<0.01), adiponectin and monocyte chemotactic protein-1 secretion (+142% and +149%, respectively; P<0.05) and mRNA expression of PPARγ (+206%; P<0.01), FABP4 (+164%; P<0.001), DGAT-1 (+144%; P<0.01), adiponectin (+138%; P<0.001) and CD36 (+149%; P<0.05). In primary rat adipocytes, SEC also increased FA uptake (137%; P<0.05). Pretreatment with a SEC antagonist impaired SEC-induced FA uptake and cAMP accumulation. SEC treatment simultaneously stimulated lipolysis measured as glycerol release in 3T3-L1 adipocytes and rat adipose tissue. CONCLUSION The present results suggest that SEC is a potent modulator of adipocyte functions, demonstrating overall a role in enhanced substrate cycling.
Collapse
|
22
|
Du ZP, Yuan HM, Wu BL, Chang JX, Lv Z, Shen J, Wu JY, Chen HB, Li EM, Xu LY. Neutrophil gelatinase-associated lipocalin in gastric carcinoma cells and its induction by TPA are controlled by C/EBPβ. Biochem Cell Biol 2011; 89:314-24. [PMID: 21612443 DOI: 10.1139/o11-002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL) expression has been found to be upregulated in a variety of tumors, but the mechanism of NGAL elevation in gastric carcinoma remains unknown. Here, immunohistochemistry was applied to analyze NGAL expression in gastric carcinoma patients. Reverse transcription PCR, Western blot, and enzyme-linked immunosorbent assay (ELISA) were performed to evaluate NGAL mRNA and protein levels before and after 12-O-tetradecanoylphorbol-13-acetate (TPA) induction. Luciferase reporter assay was carried out to identify the core cis element in NGAL promoter. The binding ability and specificity of transcription factors were analyzed by electrophoretic mobility-shift assay (EMSA) and chromatin immunoprecipitation (ChIP), respectively. Results showed that NGAL was overexpressed in gastric tumor tissues. Gastric cancer cells treated with TPA resulted in the transactivation of NGAL promoter and the upregulation of its mRNA and protein levels. We identified the -110 to -79 sequence segment upstream from the transcription initiation site of NGAL as a TPA responsive element (TRE) and confirmed that C/EBPβ was able to bind to the -87 to -79 segment. Forced expression of C/EBPβ significantly increased the promoter activity of NGAL as well as its mRNA level. These results suggest that NGAL is overexpressed in gastric cancer, the binding of C/EBPβ to the TRE of its gene promoter mediates its TPA-induced overexpression in gastric carcinoma cells.
Collapse
Affiliation(s)
- Ze-Peng Du
- Institute of Oncologic Pathology, Medical College of Shantou University, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Fujimori K, Amano F. Niacin promotes adipogenesis by reducing production of anti-adipogenic PGF2α through suppression of C/EBPβ-activated COX-2 expression. Prostaglandins Other Lipid Mediat 2011; 94:96-103. [DOI: 10.1016/j.prostaglandins.2011.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 12/22/2010] [Accepted: 01/04/2011] [Indexed: 01/21/2023]
|
24
|
C/EBP alpha expression is associated with homeostasis of the gastric epithelium and with gastric carcinogenesis. J Transl Med 2010; 90:1132-9. [PMID: 20386538 DOI: 10.1038/labinvest.2010.79] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Transcription factors from the CCAAT/enhancer-binding protein (C/EBP) family are fundamental for the control of differentiation and proliferation of many adult tissues. C/EBP alpha has a crucial role in inducing terminal differentiation and is an established tumor suppressor gene in several cancer models. The objective of this study was to analyze the putative role of C/EBP alpha in gastric carcinoma (GC). We analyzed the expression of C/EBP alpha in normal and neoplastic gastric tissues, and assessed the role of C/EBP alpha on proliferation and differentiation of GC cells. In normal gastric mucosa, C/EBP alpha is expressed in the foveolar epithelium and co-localizes with the gastric differentiation marker trefoil factor 1 (TFF1). The expression of C/EBP alpha was found to be lost in 30% of GC cases. To evaluate the role of C/EBP alpha in cell proliferation and differentiation, we transfected GC cells with a full-length C/EBP alpha protein. We observed a significant decrease in proliferation in C/EBP alpha-transfected cells. This was accompanied by a decrease in Cyclin D1, an increase in P27 expression, and an increased expression of TFF1. Finally, we showed that inhibition of the Ras/MAPK pathway leads to increased C/EBP alpha and TFF1 expression, and decreased cell proliferation and cyclin D1 expression in GC cells. Our results suggest that C/EBP alpha (together with other members of the C/EBP family) has an active role in the control of differentiation and proliferation in normal gastric mucosa. In GC, loss of C/EBP alpha may be associated with the switch from a cellular differentiation to a cellular proliferation program, presumably as a consequence of Ras/MAPK pathway activation.
Collapse
|
25
|
Meir O, Dvash E, Werman A, Rubinstein M. C/EBP-beta regulates endoplasmic reticulum stress-triggered cell death in mouse and human models. PLoS One 2010; 5:e9516. [PMID: 20209087 PMCID: PMC2831074 DOI: 10.1371/journal.pone.0009516] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2009] [Accepted: 02/11/2010] [Indexed: 11/18/2022] Open
Abstract
Endoplasmic reticulum (ER) stress elicits the unfolded protein response (UPR), initially aimed at coping with the stress, but triggering cell death upon further stress. ER stress induces the C/EBP-beta variant Liver-enriched Activating Protein (LAP), followed by the dominant-negative variant, Liver Inhibitory Protein (LIP). However, the distinct role of LAP and LIP in ER stress is unknown. We found that the kinetics of the ER stress-induced expression of LIP overlapped with that of the cell death in mouse B16 melanoma cells. Furthermore, inducible over-expression of LIP augmented ER stress-triggered cell death whereas over-expression of LAP attenuated cell death. Similar results were obtained in human 293T cells. Limited vasculature in tumors triggers hypoxia, nutrient shortage and accumulation of toxic metabolites, all of which eliciting continuous ER stress. We found that LAP promoted and LIP inhibited B16 melanoma tumor progression without affecting angiogenesis or accelerating the cell cycle. Rather, LAP attenuated, whereas LIP augmented tumor ER stress. We therefore suggest that C/EBP-beta regulates the transition from the protective to the death-promoting phase of the UPR. We further suggest that the over-expression of LAP observed in many solid tumors promotes tumor progression by attenuating ER stress-triggered tumor cell death [corrected].
Collapse
Affiliation(s)
- Ofir Meir
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Efrat Dvash
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Ariel Werman
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
| | - Menachem Rubinstein
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|
26
|
Abstract
The immensity of genes and molecules implicated in gastric carcinogenesis is overwhelming and the relevant importance of some of these molecules is too often unclear. This review serves to bring us up-to-date with the latest findings as well as to look at the larger picture in terms of how to tackle the problem of solving this multi-piece puzzle. In this review, the environmental nurturing of intestinal cancer is discussed, beginning with epidemiology (known causative factors for inducing molecular change), an update of H. pylori research, including the role of inflammation and stem cells in premalignant lesions. The role of E-cadherin in the nature (genotype) of diffuse gastric cancer is highlighted, and finally the ever growing discipline of SNP analysis (including IL1B) is discussed.
Collapse
|
27
|
Abstract
The immensity of genes and molecules implicated in gastric carcinogenesis is overwhelming and the relevant importance of some of these molecules is too often unclear. This review serves to bring us up-to-date with the latest findings as well as to look at the larger picture in terms of how to tackle the problem of solving this multi-piece puzzle. In this review, the environmental nurturing of intestinal cancer is discussed, beginning with epidemiology (known causative factors for inducing molecular change), an update of H. pylori research, including the role of inflammation and stem cells in premalignant lesions. The role of E-cadherin in the nature (genotype) of diffuse gastric cancer is highlighted, and finally the ever growing discipline of SNP analysis (including IL1B) is discussed.
Collapse
|
28
|
Park MJ, Kim KH, Kim HY, Kim K, Cheong J. Bile acid induces expression of COX-2 through the homeodomain transcription factor CDX1 and orphan nuclear receptor SHP in human gastric cancer cells. Carcinogenesis 2008; 29:2385-93. [PMID: 18775915 DOI: 10.1093/carcin/bgn207] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The caudal-related homeobox gene, CDX1, encodes for an intestinal-specific transcription factor and is involved in the induction of intestinal metaplasia (IM) of the stomach in gastric cancer. Gastric IM induced by bile reflux is a precancerous gastric adenocarcinomal lesion and has been associated with the induction of cyclooxygenase-2 (COX-2). In this study, we demonstrate the molecular mechanisms underlying the transcriptional regulation of COX-2 by bile acid in gastric cells. We noted that the ectopic expression of CDX1 enhanced COX-2 gene expression and that bile acid was associated with the induction of CDX1 expression. Furthermore, the induction of CDX1 by bile acid was mediated by the orphan nuclear receptor, small heterodimer partner (SHP). Finally, it was verified that the expression of COX-2, CDX1, SHP and CCAAT element-binding protein beta messenger RNA in human IM lesions were significantly higher than in lesions associated with gastritis. Collectively, these results reveal that bile acid induces an increase in the gene expression of COX-2 via the sequential transcriptional induction of SHP and CDX1 in precancerous lesions of human gastric cancer.
Collapse
Affiliation(s)
- Min Jung Park
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan, Korea
| | | | | | | | | |
Collapse
|
29
|
de Maat MFG, van de Velde CJH, Umetani N, de Heer P, Putter H, van Hoesel AQ, Meijer GA, van Grieken NC, Kuppen PJK, Bilchik AJ, Tollenaar RAEM, Hoon DSB. Epigenetic silencing of cyclooxygenase-2 affects clinical outcome in gastric cancer. J Clin Oncol 2007; 25:4887-94. [PMID: 17971584 DOI: 10.1200/jco.2006.09.8921] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Overexpression of cyclooxygenase-2 (COX-2) in gastric cancer has been shown to enhance tumor progression. We investigated whether silencing by promoter region hypermethylation of the COX-2 gene contributes to disease outcome in gastric cancer. MATERIALS AND METHODS COX-2 methylation status was initially assessed by capillary array electrophoresis methylation-specific polymerase chain reaction (CAE-MSP) and COX-2 protein expression by immunohistochemistry (IHC) in 40 primary gastric cancer tissues in a pilot study. Prognostic end points of correlative studies of COX-2 methylation status were time to recurrence, overall survival, and standard clinicopathologic features. CAE-MSP analysis was then validated in a second independent gastric cancer population (n = 137). RESULTS COX-2 methylation was detected in 23% and 28% of the pilot and validation patient groups, respectively. COX-2 expression (IHC) in gastric tumors inversely correlated with COX-2 gene methylation status in the pilot study (P = .02). COX-2 methylation in tumors was significantly associated with lower T, N, and TNM stage in the validation patient group (P = .02, P = .006, and P = .008, respectively). Patients with COX-2 methylated tumors had significantly longer time to recurrence and improved overall survival in a multivariate analysis in the validation patient group (hazard ratio[HR], 0.49; 95% CI, 0.24% to 0.99%; HR, 0.62; 95% CI, 0.38% to 0.99%, respectively). CONCLUSION Hypermethylation of COX-2 gene promoter was identified as an independent prognostic factor in gastric cancer patients. The results suggest promoter hypermethylation to be an important regulatory mechanism of COX-2 expression in gastric cancer and an important prognostic biomarker.
Collapse
Affiliation(s)
- Michiel F G de Maat
- Department of Molecular Oncology and Division of Gastrointestinal Surgery, John Wayne Cancer Institute, Santa Monica, CA 90404, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Wang W, Bergh A, Damber JE. Increased expression of CCAAT/enhancer-binding protein beta in proliferative inflammatory atrophy of the prostate: relation with the expression of COX-2, the androgen receptor, and presence of focal chronic inflammation. Prostate 2007; 67:1238-46. [PMID: 17570496 DOI: 10.1002/pros.20595] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Proliferative inflammatory atrophy (PIA) in the prostate has been proposed to be a precursor to prostate cancer. CCAAT/enhancer-binding protein beta (C/EBPbeta) is an important transcription factor involved in cellular proliferation and differentiation. Activation of C/EBPbeta plays a crucial role during the initial stage of cyclo-oxygenase 2 (COX-2) induction by proinflammatory mediators. Overexpression of C/EBPbeta has been reported in several human tumors. Nevertheless, the C/EBPbeta expression and functions in human prostate tissue are basically unknown. METHODS C/EBPbeta immunohistochemical staining was performed on 45 benign prostate hyperplasia (BPH) samples. The expression of C/EBPbeta in PIA lesions and normal-appearing acini was analyzed. In addition, by using double-IHC staining, C/EBPbeta expression and the association with chronic inflammatory cell density, co-expression of COX-2 and androgen receptor (AR) were also investigated. RESULTS C/EBPbeta was occasionally observed in normal-appearing prostate acini (4.9% +/- 6.7%, Mean +/- SD) but was clearly overexpressed in PIA lesions (81.8% +/- 16.4%) (P < 0.0001). Atrophic glands with T-lymphocyte and macrophage inflammation expressed higher level of C/EBPbeta. Furthermore, C/EBPbeta correlated significantly with COX-2 expression. Downregulation of the AR was common in PIA and was also related to the C/EBPbeta overexpression. CONCLUSIONS The data demonstrated that chronic inflammation appeared to play roles in the induction of C/EBPbeta expression in prostate epithelium, which was in turn associated with increased COX-2 expression and AR downregulation. In combining with other molecular alteration in the epithelium of PIA, it is suggested that these cells might be a kind of intermediate cells and involved in the pathogenesis of prostate cancer.
Collapse
Affiliation(s)
- Wanzhong Wang
- Department of Urology, Lundberg Laboratory for Cancer Research, The Sahlgrenska Academy at Göteborg University, Göteborg, Sweden
| | | | | |
Collapse
|
31
|
Resende C, Regalo G, Durães C, Carneiro F, Machado JC. Genetic Changes of CEBPA in Cancer: Mutations or Polymorphisms? J Clin Oncol 2007; 25:2493-4; author reply 2494-5. [PMID: 17557966 DOI: 10.1200/jco.2007.10.7227] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|