201
|
Jeon HM, Lee SY, Ju MK, Kim CH, Park HG, Kang HS. Early growth response 1 regulates glucose deprivation-induced necrosis. Oncol Rep 2012; 29:669-75. [PMID: 23152075 PMCID: PMC3583586 DOI: 10.3892/or.2012.2134] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/23/2012] [Accepted: 11/07/2012] [Indexed: 01/22/2023] Open
Abstract
Necrosis is commonly found in the core region of solid tumours due to metabolic stress such as hypoxia and glucose deprivation (GD) resulting from insufficient vascularization. Necrosis promotes tumour growth and development by releasing the tumour-promoting cytokine high mobility group box 1 (HMGB1); however, the molecular mechanism underlying necrotic cell death remains largely unknown. In this study, we show that early growth response 1 (Egr-1) is induced in a reactive oxygen species (ROS)-dependent manner by GD in several cell lines such as A549, MDA-MB-231 and HepG2 cells that exhibit necrosis upon GD. We found that Egr-1 short hairpin RNA (shRNA) prevented GD-induced necrosis and HMGB1 release. Necrosis-inhibiting activity of Egr-1 shRNA was also seen in multicellular tumour spheroids (MTSs), an in vitro tumour model system. In contrast, Egr-1 overexpression appeared to make tumour cells more susceptible to GD-induced necrosis. Finally, Egr-1 shRNA suppressed the growth of MTSs. These findings demonstrate that Egr-1 is implicated in GD-induced necrosis and tumour progression.
Collapse
Affiliation(s)
- Hyun Min Jeon
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Pusan 609-735, Republic of Korea
| | | | | | | | | | | |
Collapse
|
202
|
Abstract
When cellular reducing enzymes fail to shield the cell from increased amounts of reactive oxygen species (ROS), oxidative stress arises. The redox state is misbalanced, DNA and proteins are damaged and cellular transcription networks are activated. This condition can lead to the initiation and/or to the progression of atherosclerosis, tumors or pulmonary hypertension; diseases that are decisively furthered by the presence of oxidizing agents. Redox sensitive genes, like the zinc finger transcription factor early growth response 1 (Egr-1), play a pivotal role in the pathophysiology of these diseases. Apart from inducing apoptosis, signaling partners like the MEK/ERK pathway or the protein kinase C (PKC) can activate salvage programs such as cell proliferation that do not ameliorate, but rather worsen their outcome. Here, we review the currently available data on Egr-1 related signal transduction cascades in response to oxidative stress in the progression of epidemiologically significant diseases. Knowing the molecular pathways behind the pathology will greatly enhance our ability to identify possible targets for the development of new therapeutic strategies.
Collapse
|
203
|
Banning A, Ockenga W, Finger F, Siebrasse P, Tikkanen R. Transcriptional regulation of flotillins by the extracellularly regulated kinases and retinoid X receptor complexes. PLoS One 2012; 7:e45514. [PMID: 23029064 PMCID: PMC3445523 DOI: 10.1371/journal.pone.0045514] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/12/2012] [Accepted: 08/21/2012] [Indexed: 11/25/2022] Open
Abstract
Flotillin-1 and flotillin-2 are important regulators of signal transduction pathways such as growth factor signaling. Flotillin expression is increased under pathological conditions such as neurodegenerative disorders and cancer. Despite their importance for signal transduction, very little is known about the transcriptional regulation of flotillins. Here, we analyzed the expression of flotillins at transcriptional level and identified flotillins as downstream targets of the mitogen activated kinases ERK1/2. The promoter activity of flotillins was increased upon growth factor stimulation in a MAPK dependent manner. Overexpression of serum response factor or early growth response gene 1 resulted in increased flotillin mRNA and protein expression. Furthermore, both promoter activity and expression of endogenous flotillins were increased upon treatment with retinoic acid or by overexpression of the retinoid X receptor and its binding partners RARα and PPARγ. Our data indicate that the expression of flotillins, which can be detected in all cultured cells, is fine-tuned in response to various external stimuli. This regulation may be critical for the outcome of signaling cascades in which flotillins are known to be involved.
Collapse
Affiliation(s)
- Antje Banning
- Institute of Biochemistry, Medical Faculty, University of Giessen, Giessen, Germany
| | - Wymke Ockenga
- Institute of Biochemistry, Medical Faculty, University of Giessen, Giessen, Germany
| | - Fabian Finger
- Institute of Biochemistry, Medical Faculty, University of Giessen, Giessen, Germany
| | - Philipp Siebrasse
- Institute of Biochemistry, Medical Faculty, University of Giessen, Giessen, Germany
| | - Ritva Tikkanen
- Institute of Biochemistry, Medical Faculty, University of Giessen, Giessen, Germany
- * E-mail:
| |
Collapse
|
204
|
Balzarolo M, Karrich JJ, Engels S, Blom B, Medema JP, Wolkers MC. The transcriptional regulator NAB2 reveals a two-step induction of TRAIL in activated plasmacytoid DCs. Eur J Immunol 2012; 42:3019-27. [PMID: 22806638 DOI: 10.1002/eji.201242385] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/12/2012] [Revised: 06/11/2012] [Accepted: 07/11/2012] [Indexed: 01/27/2023]
Abstract
Plasmacytoid dendritic cells (pDCs) are key players in antiviral immunity. In addition to massive type I interferon production, activated pDCs express the apoptosis-inducing molecule TRAIL, which enables them to clear infected cells that express the TRAIL receptors TRAIL-R1 and TRAIL-R2. In this study, we examined the molecular mechanisms that govern TRAIL expression in human pDCs. We identify NGFI-A-binding protein 2 (NAB2) as a novel transcriptional regulator that governs TRAIL induction in stimulated pDCs. We show with the pDC-like cell line CAL-1 that NAB2 is exclusively induced downstream of TLR7 and TLR9 signaling, and not upon type I IFN-R signaling. Furthermore, PI3K signaling is required for NAB2-mediated TRAIL expression. Finally, we show that TRAIL induction in CpG-activated human pDCs occurs through two independent signaling pathways: the first is initiated through TLR9 signaling upon recognition of nucleic acids, followed by type I IFN-R-mediated signaling. In conclusion, our data suggest that these two pathways are downstream of different activation signals, but act in concert to allow for full TRAIL expression in pDCs.
Collapse
Affiliation(s)
- Melania Balzarolo
- Laboratory of Experimental Oncology and Radiobiology, Center for Experimental Molecular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | | | | | | | | | | |
Collapse
|
205
|
Ozen E, Gozukizil A, Erdal E, Uren A, Bottaro DP, Atabey N. Heparin inhibits Hepatocyte Growth Factor induced motility and invasion of hepatocellular carcinoma cells through early growth response protein 1. PLoS One 2012; 7:e42717. [PMID: 22912725 PMCID: PMC3418296 DOI: 10.1371/journal.pone.0042717] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/21/2012] [Accepted: 07/11/2012] [Indexed: 12/16/2022] Open
Abstract
The Hepatocyte Growth Factor (HGF)/c-Met signaling pathway regulates hepatocyte proliferation, and pathway aberrations are implicated in the invasive and metastatic behaviors of hepatocellular carcinoma (HCC). In addition to c-Met, heparin acts as a co-receptor to modulate pathway activity. Recently, anti-metastatic and anti-cancer effects of heparin have been reported. However, the role of heparin in the regulation of HGF signaling remains controversial and the effects of heparin on HGF-induced biological responses during hepatocarcinogenesis is not yet defined. In this study we determined the effects of heparin on HGF-induced activities of HCC cells and the underlying molecular mechanisms. Here, we report for the first time that heparin inhibits HGF-induced adhesion, motility and invasion of HCC cells. In addition, heparin reduced HGF-induced activation of c-Met and MAPK in a dose-dependent manner, as well as decreased transcriptional activation and expression of Early growth response factor 1 (Egr1). HGF-induced MMP-2 and MMP-9 activation, and MT1-MMP expression, also were inhibited by heparin. Stable knockdown of Egr1 caused a significant decrease in HGF-induced invasion, as well as the activation and expression of MMPs. Parallel to these findings, the overexpression of Egr1 increased the invasiveness of HCC cells. Our results suggest that Egr1 activates HGF-induced cell invasion through the regulation of MMPs in HCC cells and heparin inhibits HGF-induced cellular invasion via the downregulation of Egr1. Therefore, heparin treatment might be a therapeutic approach to inhibit invasion and metastasis of HCC, especially for patients with active HGF/c-Met signaling.
Collapse
Affiliation(s)
- Evin Ozen
- Department of Medical Biology and Genetics, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Aysim Gozukizil
- Department of Medical Biology and Genetics, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Esra Erdal
- Department of Medical Biology and Genetics, School of Medicine, Dokuz Eylul University, Izmir, Turkey
| | - Aykut Uren
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Donald P. Bottaro
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National, Institutes of Health, Bethesda, Maryland, United States of America
| | - Nese Atabey
- Department of Medical Biology and Genetics, School of Medicine, Dokuz Eylul University, Izmir, Turkey
- * E-mail:
| |
Collapse
|
206
|
Polk WW. FH535 potentiation of cigarette smoke condensate cytotoxicity is associated with changes in β-catenin and EGR-1 signaling. Int J Toxicol 2012; 31:380-9. [PMID: 22713211 DOI: 10.1177/1091581812447956] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/16/2023]
Abstract
Cigarette smoke condensate (CSC) has been reported to elicit morphological and transcriptional changes that suggest epithelial-to-mesenchymal transition (EMT) in cultured bronchial epithelial cells. The transdifferentiation potential of acute and prolonged CSC exposure alone or in combination with the β-catenin inhibitor, FH535, was investigated in the bronchial epithelial cell line, BEAS-2B, through assessment of cell morphology, transcript expression, protein expression, and protein localization. Changes in morphology, β-catenin translocation, E-cadherin expression, metalloproteinase expression, and fibronectin could be demonstrated independent of molecular or physiological evidence of EMT. FH535 was shown to increase CSC-induced cytotoxicity and depress β-catenin expression. However, FH535 effects were not limited to the β-catenin pathway as it also blocked the expression of early growth responsive protein 1 (EGR-1) target genes, fibronectin and phosphatase and tensin homologue, without affecting EGR-1 nuclear accumulation.
Collapse
Affiliation(s)
- William W Polk
- A.W. Spears Research Center, Lorillard Tobacco Company, Greensboro, NC 27420, USA.
| |
Collapse
|
207
|
Fan Q, Barathi VA, Cheng CY, Zhou X, Meguro A, Nakata I, Khor CC, Goh LK, Li YJ, Lim W, Ho CEH, Hawthorne F, Zheng Y, Chua D, Inoko H, Yamashiro K, Ohno-Matsui K, Matsuo K, Matsuda F, Vithana E, Seielstad M, Mizuki N, Beuerman RW, Tai ES, Yoshimura N, Aung T, Young TL, Wong TY, Teo YY, Saw SM. Genetic variants on chromosome 1q41 influence ocular axial length and high myopia. PLoS Genet 2012; 8:e1002753. [PMID: 22685421 PMCID: PMC3369958 DOI: 10.1371/journal.pgen.1002753] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/10/2012] [Accepted: 04/20/2012] [Indexed: 12/14/2022] Open
Abstract
As one of the leading causes of visual impairment and blindness, myopia poses a significant public health burden in Asia. The primary determinant of myopia is an elongated ocular axial length (AL). Here we report a meta-analysis of three genome-wide association studies on AL conducted in 1,860 Chinese adults, 929 Chinese children, and 2,155 Malay adults. We identified a genetic locus on chromosome 1q41 harboring the zinc-finger 11B pseudogene ZC3H11B showing genome-wide significant association with AL variation (rs4373767, β = -0.16 mm per minor allele, P(meta) =2.69 × 10(-10)). The minor C allele of rs4373767 was also observed to significantly associate with decreased susceptibility to high myopia (per-allele odds ratio (OR) =0.75, 95% CI: 0.68-0.84, P(meta) =4.38 × 10(-7)) in 1,118 highly myopic cases and 5,433 controls. ZC3H11B and two neighboring genes SLC30A10 and LYPLAL1 were expressed in the human neural retina, retinal pigment epithelium, and sclera. In an experimental myopia mouse model, we observed significant alterations to gene and protein expression in the retina and sclera of the unilateral induced myopic eyes for the murine genes ZC3H11A, SLC30A10, and LYPLAL1. This supports the likely role of genetic variants at chromosome 1q41 in influencing AL variation and high myopia.
Collapse
Affiliation(s)
- Qiao Fan
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Veluchamy A. Barathi
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, National University of Singapore, Singapore, Singapore
| | - Ching-Yu Cheng
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, National University of Singapore, Singapore, Singapore
| | - Xin Zhou
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Akira Meguro
- Department of Ophthalmology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Isao Nakata
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Center for Genomic Medicine and Inserm U.852, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Chiea-Chuen Khor
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology, and Research, Singapore, Singapore
- Centre for Molecular Epidemiology, National University of Singapore, Singapore, Singapore
- Department of Pediatrics, National University of Singapore, Singapore, Singapore
| | - Liang-Kee Goh
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
- Duke–National University of Singapore Graduate Medical School, Singapore, Singapore
- Department of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Yi-Ju Li
- Department of Biostatistics and Bioinformatics, Duke University Medical School, Durham, North Carolina, United States of America
- Center for Human Genetics, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Wan'e Lim
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Candice E. H. Ho
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Felicia Hawthorne
- Center for Human Genetics, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Yingfeng Zheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Daniel Chua
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
| | - Hidetoshi Inoko
- Department of Molecular Life Science, Division of Molecular Medical Science and Molecular Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Kenji Yamashiro
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kyoko Ohno-Matsui
- Department of Ophthalmology and Visual Science, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keitaro Matsuo
- Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine and Inserm U.852, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Eranga Vithana
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, National University of Singapore, Singapore, Singapore
| | - Mark Seielstad
- Institute for Human Genetics and Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Nobuhisa Mizuki
- Department of Ophthalmology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Roger W. Beuerman
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, National University of Singapore, Singapore, Singapore
- Duke–National University of Singapore Graduate Medical School, Singapore, Singapore
| | - E.-Shyong Tai
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
- Department of Medicine, National University of Singapore, Singapore, Singapore
| | - Nagahisa Yoshimura
- Department of Ophthalmology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tin Aung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, National University of Singapore, Singapore, Singapore
| | - Terri L. Young
- Duke–National University of Singapore Graduate Medical School, Singapore, Singapore
- Center for Human Genetics, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Tien-Yin Wong
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, National University of Singapore, Singapore, Singapore
- Centre for Eye Research Australia, University of Melbourne, Melbourne, Australia
| | - Yik-Ying Teo
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science, Technology, and Research, Singapore, Singapore
- Graduate School for Integrative Science and Engineering, National University of Singapore, Singapore, Singapore
- Department of Statistics and Applied Probability, National University of Singapore, Singapore, Singapore
- * E-mail: (S-MS); (Y-YT)
| | - Seang-Mei Saw
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, National University of Singapore, Singapore, Singapore
- Duke–National University of Singapore Graduate Medical School, Singapore, Singapore
- Graduate School for Integrative Science and Engineering, National University of Singapore, Singapore, Singapore
- * E-mail: (S-MS); (Y-YT)
| |
Collapse
|
208
|
Zhang P, Chen JH, Guo XL. New insights into PTEN regulation mechanisms and its potential function in targeted therapies. Biomed Pharmacother 2012; 66:485-90. [PMID: 22902055 DOI: 10.1016/j.biopha.2012.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/02/2012] [Accepted: 04/15/2012] [Indexed: 12/12/2022] Open
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a tumor suppressor gene of phosphatased activity. Its low or lacking expression closely relates with tumor progress and poor prognosis. The regulation and function ascribed to PTEN have become more diverse since its discovery as a putative phosphatase mutated in many human tumors. PTEN function is positively and negatively regulated at the transcriptional level, as well as post-translationally by phosphorylation, oxidation and acetylation. Deregulation of PTEN is implicated in other human diseases in addition to cancers, including diabetes and obesity, modulation of PTEN level has widespread therapeutic applications to those tumorigenesis and non-tumor diseases. This review will summarize the new points on the regulation of PTEN and briefly discuss the potential therapeutic role of PTEN in some diseases.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | | | | |
Collapse
|
209
|
Derdak Z, Villegas KA, Wands JR. Early growth response-1 transcription factor promotes hepatic fibrosis and steatosis in long-term ethanol-fed Long-Evans rats. Liver Int 2012; 32:761-70. [PMID: 22292946 PMCID: PMC10026596 DOI: 10.1111/j.1478-3231.2012.02752.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/01/2011] [Accepted: 12/28/2011] [Indexed: 12/30/2022]
Abstract
BACKGROUND Previous studies demonstrated that the Long-Evans (LE) rats exhibited liver injury and lipid metabolic abnormalities after 8 weeks of ethanol feeding. AIMS The goal of this study was to investigate if the LE rats develop more advanced hepatic abnormalities (e.g., fibrosis) after long-term feeding with an ethanol-containing Lieber-DeCarli diet. In addition, the contribution of early growth response-1 (EGR1) transcription factor to these pathological changes was assessed. METHODS Long-Evans rats were fed an ethanol-containing or isocaloric control liquid diet for 18 months. Livers were processed for histological analyses, studies of fibrosis-related gene expression, cell fractionation and triglyceride measurement. Serum alanine aminotransferase (ALT) levels were assessed. DNA binding activities of p53 and the sterol regulatory element-binding protein-1c (SREBP1c) were analysed. The abundance of EGR1 and enzymes involved in fatty acid synthesis were determined. Chromatin immunoprecipitation was employed to study EGR1 binding to the SREBP1c promoter region. RESULTS Ethanol feeding generated steatosis, chicken wire fibrosis and ALT elevations in the LE rats. Fibrosis was associated with the upregulation of EGR1 and its downstream target genes. EGR1 upregulation was associated with enhanced p53 activity and an increase in the cellular p66(shc) abundance. Steatosis was linked to the activation of SREBP1c. Importantly, EGR1 upregulation paralleled the expression and transcriptional activity of SREBP1c. Finally, EGR1 was shown to bind to the SREBP1c promoter region. CONCLUSIONS Long-term ethanol feeding promoted steatosis and fibrosis in LE rats via EGR1 activation. The highly abundant EGR1 bound to the SREBP1c promoter and contributed to the steatosis observed in the LE rat model.
Collapse
Affiliation(s)
- Zoltan Derdak
- Division of Gastroenterology & Liver Research Center, Warren Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA.
| | | | | |
Collapse
|
210
|
Abstract
Inflammation is an important contributor to the development and progression of human cancers. Inflammatory lipid metabolites, prostaglandins, formed from arachidonic acid by prostaglandin H synthases commonly called cyclooxygenases (COXs) bind to specific receptors that activate signaling pathways driving the development and progression of tumors. Inhibitors of prostaglandin formation, COX inhibitors, or nonsteroidal anti-inflammatory drugs (NSAIDs) are well documented as agents that inhibit tumor growth and with long-term use prevent tumor development. NSAIDs also alter gene expression independent of COX inhibition and these changes in gene expression also appear to contribute to the anti-tumorigenic activity of these drugs. Many NSAIDs, as illustrated by sulindac sulfide, alter gene expressions by altering the expression or phosphorylation status of the transcription factors specificity protein 1 and early growth response-1 with the balance between these two events resulting in increases or decreases in specific target genes. In this review, we have summarized and discussed the various genes altered by this mechanism after NSAID treatment and how these changes in expression relate to the anti-tumorigenic activity. A major focus of the review is on NSAID-activated gene (NAG-1) or growth differentiation factor 15. This unique member of the TGF-β superfamily is highly induced by NSAIDs and numerous drugs and chemicals with anti-tumorigenic activities. Investigations with a transgenic mouse expressing the human NAG-1 suggest it acts to suppress tumor development in several mouse models of cancer. The biochemistry and biology of NAG-1 were discussed as potential contributor to cancer prevention by COX inhibitors.
Collapse
|
211
|
Li J, Wang H, Ma Z, Fan W, Li Y, Han B, Zhang Z, Wang J. TAT-Apoptin induces apoptosis in the human bladder cancer EJ cell line and regulates Bax, Bcl-2, caspase-3 and survivin expression. Exp Ther Med 2012; 3:1033-1038. [PMID: 22970013 DOI: 10.3892/etm.2012.520] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/08/2011] [Accepted: 03/14/2012] [Indexed: 11/06/2022] Open
Abstract
In order to identify the antitumor effect of TAT-Apoptin on the human bladder cancer EJ cell line and study its impact on the expression of the apoptosis-related genes bax, bcl-2, caspase-3 and survivin, the MTT assay, real-time quantitative PCR and western blot analysis were used in this study. The results of the MTT assay indicated that TAT-Apoptin was able to inhibit the proliferation of EJ cells in a dose-dependent manner. The expression of Bax, Bcl-2, Caspase-3 and Survivin mRNA and protein following the treatment of the EJ cells with TAT-Apoptin (0.1, 0.5, 1, 10, 50 and 100 μg/ml) for 24, 48 and 72 h was analyzed. Cell proliferation was significantly different after treatment with the various concentrations of TAT-Apoptin and the durations of treatment. The level of expression of Bcl-2 and Survivin in EJ cells decreased significantly, while that of Bax and Caspase-3 increased significantly at the mRNA and protein levels.
Collapse
Affiliation(s)
- Jun Li
- Department of Nephrology, The First Affiliated Hospital, Kunming Medical University, Kunming 650031
| | | | | | | | | | | | | | | |
Collapse
|
212
|
van Loo KMJ, Schaub C, Pernhorst K, Yaari Y, Beck H, Schoch S, Becker AJ. Transcriptional regulation of T-type calcium channel CaV3.2: bi-directionality by early growth response 1 (Egr1) and repressor element 1 (RE-1) protein-silencing transcription factor (REST). J Biol Chem 2012; 287:15489-501. [PMID: 22431737 DOI: 10.1074/jbc.m111.310763] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/06/2022] Open
Abstract
The pore-forming Ca(2+) channel subunit Ca(V)3.2 mediates a low voltage-activated (T-type) Ca(2+) current (I(CaT)) that contributes pivotally to neuronal and cardiac pacemaker activity. Despite the importance of tightly regulated Ca(V)3.2 levels, the mechanisms regulating its transcriptional dynamics are not well understood. Here, we have identified two key factors that up- and down-regulate the expression of the gene encoding Ca(V)3.2 (Cacna1h). First, we determined the promoter region and observed several stimulatory and inhibitory clusters. Furthermore, we found binding sites for the transcription factor early growth response 1 (Egr1/Zif268/Krox-24) to be highly overrepresented within the Ca(V)3.2 promoter region. mRNA expression analyses and dual-luciferase promoter assays revealed that the Ca(V)3.2 promoter was strongly activated by Egr1 overexpression in vitro and in vivo. Subsequent chromatin immunoprecipitation assays in NG108-15 cells and mouse hippocampi confirmed specific Egr1 binding to the Ca(V)3.2 promoter. Congruently, whole-cell I(CaT) values were significantly larger after Egr1 overexpression. Intriguingly, Egr1-induced activation of the Ca(V)3.2 promoter was effectively counteracted by the repressor element 1-silencing transcription factor (REST). Thus, Egr1 and REST can bi-directionally regulate Ca(V)3.2 promoter activity and mRNA expression and, hence, the size of I(CaT). This mechanism has critical implications for the regulation of neuronal and cardiac Ca(2+) homeostasis under physiological conditions and in episodic disorders such as arrhythmias and epilepsy.
Collapse
Affiliation(s)
- Karen M J van Loo
- Department of Neuropathology, University of Bonn Medical Center, D-53105 Bonn, Germany.
| | | | | | | | | | | | | |
Collapse
|
213
|
PI3K/AKT/PTEN Signaling as a Molecular Target in Leukemia Angiogenesis. Adv Hematol 2012; 2012:843085. [PMID: 22505939 PMCID: PMC3299269 DOI: 10.1155/2012/843085] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/05/2011] [Accepted: 12/11/2011] [Indexed: 11/17/2022] Open
Abstract
PI3K/AKT/PTEN pathway is important in the regulation of angiogenesis mediated by vascular endothelial growth factor in many tumors including leukemia. The signaling pathway is activated in leukemia patients as well as leukemia cell lines together with a decrease in the expression of PTEN gene. The mechanism by which the signaling pathway regulates angiogenesis remains to be further elucidated. However, it has become an attractive target for drug therapy against leukemia, because angiogenesis is a key process in malignant cell growth. In this paper, we will focus on the roles and mechanisms of PI3K/AKT/PTEN pathway in regulating angiogenesis.
Collapse
|
214
|
Song LJ, Liu RJ, Zeng Z, Alper SL, Cui HJ, Lu Y, Zheng L, Yan ZW, Fu GH. Gastrin inhibits a novel, pathological colon cancer signaling pathway involving EGR1, AE2, and P-ERK. J Mol Med (Berl) 2012; 90:707-18. [PMID: 22228178 DOI: 10.1007/s00109-011-0851-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/18/2011] [Revised: 12/13/2011] [Accepted: 12/23/2011] [Indexed: 12/22/2022]
Abstract
Human anion exchanger 2 (AE2) is a plasma membrane protein that regulates intracellular pH and cell volume. AE2 contributes to transepithelial transport of chloride and bicarbonate in normal colon and other epithelial tissues. We now report that AE2 overexpression in colon cancer cells is correlated with expression of the nuclear proliferation marker, Ki67. Survival analysis of 24 patients with colon cancer in early stage or 33 patients with tubular adenocarcinoma demonstrated that expression of AE2 is correlated with poor prognosis. Cellular and molecular experiments indicated that AE2 expression promoted proliferation of colon cancer cells. In addition, we found that transcription factor EGR1 underlies AE2 upregulation and the AE2 sequester p16INK4a (P16) in the cytoplasm of colon cancer cells. Cytoplasmic P16 enhanced ERK phosphorylation and promoted proliferation of colon cancer cells. Gastrin inhibited proliferation of colon cancer cells by suppressing expression of EGR1 and AE2 and by blocking ERK phosphorylation. Taken together, our data describe a novel EGR1/AE2/P16/P-ERK signaling pathway in colon carcinogenesis, with implications for pathologic prognosis and for novel therapeutic approaches.
Collapse
Affiliation(s)
- Ling-Jun Song
- Department of Pathology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
215
|
Tarcic G, Avraham R, Pines G, Amit I, Shay T, Lu Y, Zwang Y, Katz M, Ben-Chetrit N, Jacob-Hirsch J, Virgilio L, Rechavi G, Mavrothalassitis G, Mills GB, Domany E, Yarden Y. EGR1 and the ERK-ERF axis drive mammary cell migration in response to EGF. FASEB J 2011; 26:1582-92. [PMID: 22198386 DOI: 10.1096/fj.11-194654] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/23/2022]
Abstract
The signaling pathways that commit cells to migration are incompletely understood. We employed human mammary cells and two stimuli: epidermal growth factor (EGF), which induced cellular migration, and serum factors, which stimulated cell growth. In addition to strong activation of ERK by EGF, and AKT by serum, early transcription remarkably differed: while EGF induced early growth response-1 (EGR1), and this was required for migration, serum induced c-Fos and FosB to enhance proliferation. We demonstrate that induction of EGR1 involves ERK-mediated down-regulation of microRNA-191 and phosphorylation of the ETS2 repressor factor (ERF) repressor, which subsequently leaves the nucleus. Unexpectedly, knockdown of ERF inhibited migration, which implies migratory roles for exported ERF molecules. On the other hand, chromatin immunoprecipitation identified a subset of direct EGR1 targets, including EGR1 autostimulation and SERPINB2, whose transcription is essential for EGF-induced cell migration. In summary, EGR1 and the EGF-ERK-ERF axis emerge from our study as major drivers of growth factor-induced mammary cell migration.
Collapse
Affiliation(s)
- Gabi Tarcic
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
216
|
Abstract
The myelodysplastic syndromes are a diverse group of clonal stem cell disorders characterized by ineffective hematopoiesis, peripheral cytopenias, and an increased propensity to evolve to acute myeloid leukemia. The molecular pathogenesis of these disorders is poorly understood, but recurring chromosomal abnormalities occur in approximately 50% of cases and are the focus of much investigation. The availability of newer molecular techniques has allowed the identification of additional genetic aberrations, including mutations and epigenetic changes of prognostic and potential therapeutic importance. This review focuses on the key role of cytogenetic analysis in myelodysplastic syndromes in the context of the diagnosis, prognosis, and pathogenesis of these disorders.
Collapse
Affiliation(s)
- Olatoyosi Odenike
- Assistant Professor of Medicine, Section of Hematology/Oncology, and the Comprehensive Cancer Center, University of Chicago
| | - John Anastasi
- Associate Professor of Pathology, Hematopathology and Clinical Hematology Laboratory, and the Comprehensive Cancer Center, University of Chicago
| | - Michelle M. Le Beau
- Professor of Medicine, Section of Hematology/Oncology, and the Comprehensive Cancer Center, University of Chicago
| |
Collapse
|
217
|
Umeda JE, Demuth DR, Ando ES, Faveri M, Mayer MPA. Signaling transduction analysis in gingival epithelial cells after infection with Aggregatibacter actinomycetemcomitans. Mol Oral Microbiol 2011; 27:23-33. [PMID: 22230463 DOI: 10.1111/j.2041-1014.2011.00629.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/28/2022]
Abstract
Periodontal diseases result from the interaction of bacterial pathogens with the host's gingival tissue. Gingival epithelial cells are constantly challenged by microbial cells and respond by altering their transcription profiles, inducing the production of inflammatory mediators. Different transcription profiles are induced by oral bacteria and little is known about how the gingival epithelium responds after interaction with the periodontopathogenic organism Aggregatibacter actinomycetemcomitans. In the present study, we examined the transcription of genes involved in signaling transduction pathways in gingival epithelial cells exposed to viable A. actinomycetemcomitans. Immortalized gingival epithelial cells (OBA-9) were infected with A. actinomycetemcomitans JP2 for 24 h and the transcription profile of genes encoding human signal transduction pathways was determined. Functional analysis of inflammatory mediators positively transcribed was performed by ELISA in culture supernatant and in gingival tissues. Fifteen of 84 genes on the array were over-expressed (P < 0.01) after 24 h of infection with viable A. actinomycetemcomitans. Over-expressed genes included those implicated in tissue remodeling and bone resorption, such as CSF2, genes encoding components of the LDL pathway, nuclear factor-κB-dependent genes and other cytokines. The ELISA data confirmed that granulocyte-macrophage colony-stimulating factor/colony-stimulating factor 2, tumor necrosis factor-α and intercellular adhesion molecule-1 were highly expressed by infected gingival cells when compared with control non-infected cells, and presented higher concentrations in tissues from patients with aggressive and chronic periodontitis than in tissues from healthy controls. The induction in epithelial cells of factors such as the pro-inflammatory cytokine CSF2, which is involved in osteoclastogenesis, may help to explain the outcomes of A. actinomycetemcomitans infection.
Collapse
Affiliation(s)
- J E Umeda
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
218
|
DIF-1 inhibits the Wnt/β-catenin signaling pathway by inhibiting TCF7L2 expression in colon cancer cell lines. Biochem Pharmacol 2011; 83:47-56. [PMID: 22005519 DOI: 10.1016/j.bcp.2011.10.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/11/2011] [Revised: 09/29/2011] [Accepted: 10/03/2011] [Indexed: 12/18/2022]
Abstract
We previously reported that differentiation-inducing factor-1 (DIF-1), a morphogen in Dictyostelium discoideum, inhibits the proliferation of human cancer cell lines by inducing β-catenin degradation and suppressing the Wnt/β-catenin signaling pathway. To determine whether β-catenin degradation is essential for the effect of DIF-1, we examined the effect of DIF-1 on human colon cancer cell lines (HCT-116, SW-620 and DLD-1), in which the Wnt/β-catenin signaling pathway is constitutively active. DIF-1 strongly inhibited cell proliferation and arrested the cell cycle in the G(0)/G(1) phase via the suppression of cyclin D1 expression at mRNA and protein levels without reducing β-catenin protein. TCF-dependent transcriptional activity and cyclin D1 promoter activity were revealed to be inhibited via suppression of transcription factor 7-like 2 (TCF7L2) expression. Luciferase reporter assays and EMSAs using the TCF7L2 promoter fragments indicated that the binding site for the transcription factor early growth response-1 (Egr-1), which is located in the -609 to -601 bp region relative to the start codon in the TCF7L2 promoter, was involved in DIF-1 activity. Moreover, RNAi-mediated depletion of endogenous TCF7L2 resulted in reduced cyclin D1 promoter activity and protein expression, and the overexpression of TCF7L2 overrode the inhibition of the TCF-dependent transcriptional activity and cyclin D1 promoter activity induced by DIF-1. Therefore, DIF-1 seemed to inhibit the Wnt/β-catenin signaling pathway by suppressing TCF7L2 expression via reduced Egr-1-dependent transcriptional activity in these colon cancer cell lines. Our results provide a novel insight into the mechanisms by which DIF-1 inhibits the Wnt/β-catenin signaling pathway.
Collapse
|
219
|
Manente AG, Pinton G, Tavian D, Lopez-Rodas G, Brunelli E, Moro L. Coordinated sumoylation and ubiquitination modulate EGF induced EGR1 expression and stability. PLoS One 2011; 6:e25676. [PMID: 21998680 PMCID: PMC3187784 DOI: 10.1371/journal.pone.0025676] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/12/2011] [Accepted: 09/08/2011] [Indexed: 11/18/2022] Open
Abstract
Background Human early growth response-1 (EGR1) is a member of the zing-finger family of transcription factors induced by a range of molecular and environmental stimuli including epidermal growth factor (EGF). In a recently published paper we demonstrated that integrin/EGFR cross-talk was required for Egr1 expression through activation of the Erk1/2 and PI3K/Akt/Forkhead pathways. EGR1 activity and stability can be influenced by many different post-translational modifications such as acetylation, phosphorylation, ubiquitination and the recently discovered sumoylation. The aim of this work was to assess the influence of sumoylation on EGF induced Egr1 expression and/or stability. Methods We modulated the expression of proteins involved in the sumoylation process in ECV304 cells by transient transfection and evaluated Egr1 expression in response to EGF treatment at mRNA and protein levels. Results We demonstrated that in ECV304 cells Egr1 was transiently induced upon EGF treatment and a fraction of the endogenous protein was sumoylated. Moreover, SUMO-1/Ubc9 over-expression stabilized EGF induced ERK1/2 phosphorylation and increased Egr1 gene transcription. Conversely, in SUMO-1/Ubc9 transfected cells, EGR1 protein levels were strongly reduced. Data obtained from protein expression and ubiquitination analysis, in the presence of the proteasome inhibitor MG132, suggested that upon EGF stimuli EGR1 sumoylation enhanced its turnover, increasing ubiquitination and proteasome mediated degradation. Conclusions Here we demonstrate that SUMO-1 modification improving EGR1 ubiquitination is involved in the modulation of its stability upon EGF mediated induction.
Collapse
Affiliation(s)
- Arcangela Gabriella Manente
- Dipartimento di Scienze Chimiche, Alimentari, Farmaceutiche e Farmacologiche, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Giulia Pinton
- Dipartimento di Scienze Chimiche, Alimentari, Farmaceutiche e Farmacologiche, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Daniela Tavian
- Department of Psychology, Catholic University of the Sacred Heart, Milan, Italy
| | | | - Elisa Brunelli
- Dipartimento di Scienze Chimiche, Alimentari, Farmaceutiche e Farmacologiche, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Laura Moro
- Dipartimento di Scienze Chimiche, Alimentari, Farmaceutiche e Farmacologiche, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
- * E-mail:
| |
Collapse
|
220
|
Seki T, Kokuryo T, Yokoyama Y, Suzuki H, Itatsu K, Nakagawa A, Mizutani T, Miyake T, Uno M, Yamauchi K, Nagino M. Antitumor effects of α-bisabolol against pancreatic cancer. Cancer Sci 2011; 102:2199-205. [PMID: 21883695 DOI: 10.1111/j.1349-7006.2011.02082.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/25/2022] Open
Abstract
In the present study, we investigated whether α-bisabolol, a sesquiterpene alcohol present in essential oils derived from a variety of plants, has antitumor effects against pancreatic cancer. α-Bisabolol induced a decrease in cell proliferation and viability in pancreatic cancer cell lines (KLM1, KP4, Panc1, MIA Paca2), but not in pancreatic epithelial cells (ACBRI515). α-Bisabolol treatment induced apoptosis and suppressed Akt activation in pancreatic cancer cell lines. Furthermore, α-bisabolol treatment induced the overexpression of early growth response-1 (EGR1), whereas EGR1 siRNA decreased the α-bisabolol-induced cell death of KLM1 cells. Tumor growth in both subcutaneous and peritoneal xenograft nude mouse models was significantly inhibited by intragastric administration of 1000 mg/kg of α-bisabolol, once a week for three weeks. The results indicate that α-bisabolol could be a novel therapeutic option for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Takashi Seki
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
221
|
Sarma SN, Kim YJ, Ryu JC. Differential gene expression profiles of human leukemia cell lines exposed to benzene and its metabolites. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2011; 32:285-295. [PMID: 21843810 DOI: 10.1016/j.etap.2011.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 10/22/2010] [Revised: 04/08/2011] [Accepted: 06/08/2011] [Indexed: 05/31/2023]
Abstract
Benzene is a well-known environmental pollutant that can induce hematotoxicity, aplastic anemia, acute myelogenous leukemia, and lymphoma. Benzene toxicity is likely mediated through metabolites induced by means of multiple pathways. Although benzene metabolites are known to induce oxidative stress and disrupt the cell cycle, the mechanism underlying leukemogenesis is not fully understood. The aim of this study was to analyze the genome-wide expression profiles of human promyelocytic leukemia HL-60 cells that had been exposed to benzene and its metabolites. This was carried out using whole human genome oligonucleotide microarrays to ascertain potential biomarkers. Genes that were differentially expressed (>1.5-fold and p-values <0.05) after exposure to benzene (BZ), hydroquinone (HQ), and 1,4-benzoquinone (BQ) were then classified with GO, KEGG and GSEA pathway annotation. All genes that were identified were then functionally categorized as being involved in the cell cycle, the p53 signaling pathway, apoptosis, the MAPK signaling pathway, or the T cell receptor signaling pathway. Functionally important genes were further validated by means of real-time RT-PCR. The results showed that EGR1, PMAIP1, AR, CCL2, CD69, HSPA8, SLC7A11, HERPUD1, ELK1, and MKI57 genes altered their expression profiles. Similar expression profiles were also found in human erythromyeloblastoid leukemia K562 cells and in human leukemic monocyte lymphoma U937 cells. In conclusion, gene expression profiles along with GO, KEGG and GSEA pathway annotation analysis have provided an insight into the leukemogenesis as well as highlighted potential gene-based biomarkers of human leukemia cell lines when they are exposed to benzene and its metabolites.
Collapse
Affiliation(s)
- Sailendra Nath Sarma
- Cellular and Molecular Toxicology Laboratory, Korea Institute of Science & Technology, P.O. Box 131, Cheongryang, Seoul 130-650, Republic of Korea.
| | | | | |
Collapse
|
222
|
Lee HA, Park I, Byun HJ, Jeoung D, Kim YM, Lee H. Metastasis suppressor KAI1/CD82 attenuates the matrix adhesion of human prostate cancer cells by suppressing fibronectin expression and β1 integrin activation. Cell Physiol Biochem 2011; 27:575-86. [PMID: 21691075 DOI: 10.1159/000329979] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 04/27/2011] [Indexed: 11/19/2022] Open
Abstract
KAI1/CD82, a tetraspanin membrane protein functions as a metastasis suppressor in many types of human cancers and has been shown to regulate cell adhesion properties. In the present study, we investigated the underlying mechanism of KAI1/CD82-mediated changes in cell adhesion to the extracellular matrix using human prostate cancer cells. We found that high KAI1/CD82 expression attenuated short-term cell adhesion to uncoated- or fibronectin-coated plates. Moreover, high KAI1/CD82 expression generated an extracellular environment unfavorable for cell adhesion as compared to low KAI1/CD82 expression, suggesting KAI1/CD82-dependent regulation of extracellular matrix (ECM) molecule(s) expression and/or secretion. Among ECM components examined, fibronectin exhibited decreased expression and secretion in high KAI1/CD82-expressing cells. Furthermore, high KAI1/CD82 expression interfered with the activation of β (1) integrin at the cell surface while total β (1) integrin levels remained unchanged, concomitant with reduced formation of focal adhesion complex and decreased bundling of actin filaments. Finally, high KAI1/CD82 expression significantly retarded cell motility in a scratch wound assay. Taken together, our results strongly suggest that KAI1/CD82 attenuates the activation of β (1) integrin, and thereby down-regulates outside-in signaling of β (1) integrin, leading to the reduction of focal adhesion formation and fibronectin expression/secretion, which subsequently interferes with cell adhesion properties and motility.
Collapse
Affiliation(s)
- Hyun-Ah Lee
- Medical & Bio-Material Research Center, College of Natural Sciences, School of Medicine, Kangwon National University, Chuncheon, Kangwon-do, Republic of Korea
| | | | | | | | | | | |
Collapse
|
223
|
Ritchie MF, Zhou Y, Soboloff J. WT1/EGR1-mediated control of STIM1 expression and function in cancer cells. Front Biosci (Landmark Ed) 2011; 16:2402-15. [PMID: 21622185 DOI: 10.2741/3862] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/09/2023]
Abstract
There have been numerous publications linking Ca(2+) signaling and cancer, however, a clear explanation for this link has remained elusive. We recently identified the oncogenes/tumor suppressors Wilms Tumor Suppressor 1 (WT1) and Early Growth Response 1 (EGR1) as regulators of the expression of STIM1, an essential regulator of Ca(2+) entry in non-excitable cells. The current review focuses on the literature defining both differential Ca(2+) signaling and WT1/EGR1 expression patterns in 6 specific cancer subtypes: Acute Myeloid Leukemia, Wilms Tumor, breast cancer, ovarian cancer, glioblastoma and prostate cancer. For each tumor-type, we have assessed how specific changes in WT1 and EGR1 expression might contribute to aberrant Ca(2+) homeostasis as well as the therapeutic potential of these observations.
Collapse
Affiliation(s)
- Michael F Ritchie
- Department of Biochemistry, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | |
Collapse
|
224
|
Chu JH, Lazarus R, Carey VJ, Raby BA. Quantifying differential gene connectivity between disease states for objective identification of disease-relevant genes. BMC SYSTEMS BIOLOGY 2011; 5:89. [PMID: 21627793 PMCID: PMC3128864 DOI: 10.1186/1752-0509-5-89] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 11/24/2010] [Accepted: 05/31/2011] [Indexed: 02/16/2023]
Abstract
Background Network modeling of whole transcriptome expression data enables characterization of complex epistatic (gene-gene) interactions that underlie cellular functions. Though numerous methods have been proposed and successfully implemented to develop these networks, there are no formal methods for comparing differences in network connectivity patterns as a function of phenotypic trait. Results Here we describe a novel approach for quantifying the differences in gene-gene connectivity patterns across disease states based on Graphical Gaussian Models (GGMs). We compare the posterior probabilities of connectivity for each gene pair across two disease states, expressed as a posterior odds-ratio (postOR) for each pair, which can be used to identify network components most relevant to disease status. The method can also be generalized to model differential gene connectivity patterns within previously defined gene sets, gene networks and pathways. We demonstrate that the GGM method reliably detects differences in network connectivity patterns in datasets of varying sample size. Applying this method to two independent breast cancer expression data sets, we identified numerous reproducible differences in network connectivity across histological grades of breast cancer, including several published gene sets and pathways. Most notably, our model identified two gene hubs (MMP12 and CXCL13) that each exhibited differential connectivity to more than 30 transcripts in both datasets. Both genes have been previously implicated in breast cancer pathobiology, but themselves are not differentially expressed by histologic grade in either dataset, and would thus have not been identified using traditional differential gene expression testing approaches. In addition, 16 curated gene sets demonstrated significant differential connectivity in both data sets, including the matrix metalloproteinases, PPAR alpha sequence targets, and the PUFA synthesis pathway. Conclusions Our results suggest that GGM can be used to formally evaluate differences in global interactome connectivity across disease states, and can serve as a powerful tool for exploring the molecular events that contribute to disease at a systems level.
Collapse
Affiliation(s)
- Jen-hwa Chu
- Channing Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston MA 02115, USA.
| | | | | | | |
Collapse
|
225
|
Stoddart A, McNerney ME, Bartom E, Bergerson R, Young DJ, Qian Z, Wang J, Fernald AA, Davis EM, Larson RA, White KP, Le Beau MM. Genetic pathways leading to therapy-related myeloid neoplasms. Mediterr J Hematol Infect Dis 2011; 3:e2011019. [PMID: 21713073 PMCID: PMC3113274 DOI: 10.4084/mjhid.2011.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/18/2011] [Accepted: 04/21/2011] [Indexed: 01/03/2023] Open
Abstract
Therapy-related myeloid neoplasm (t-MN) is a distinctive clinical syndrome occurring after exposure to chemotherapy or radiotherapy. t-MN arises in most cases from a multipotential hematopoietic stem cell or, less commonly, in a lineage committed progenitor cell. The prognosis for patients with t-MN is poor, as current forms of therapy are largely ineffective. Cytogenetic analysis, molecular analysis and gene expression profiling analysis of t-MN has revealed that there are distinct subtypes of the disease; however, our understanding of the genetic basis of t-MN is incomplete. Elucidating the genetic pathways and molecular networks that are perturbed in t-MNs, may facilitate the identification of therapeutic targets that can be exploited for the development of urgently-needed targeted therapies.
Collapse
|
226
|
Sill H, Olipitz W, Zebisch A, Schulz E, Wölfler A. Therapy-related myeloid neoplasms: pathobiology and clinical characteristics. Br J Pharmacol 2011; 162:792-805. [PMID: 21039422 DOI: 10.1111/j.1476-5381.2010.01100.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/21/2022] Open
Abstract
Therapy-related myeloid neoplasms (t-MNs) are serious long-term consequences of cytotoxic treatments for an antecedent disorder. t-MNs are observed after ionizing radiation as well as conventional chemotherapy including alkylating agents, topoisomerase-II-inhibitors and antimetabolites. In addition, adjuvant use of recombinant human granulocyte-colony stimulating factor may also increase the risk of t-MNs. There is clinical and biological overlap between t-MNs and high-risk de novo myelodysplastic syndromes and acute myeloid leukaemia suggesting similar mechanisms of leukaemogenesis. Human studies and animal models point to a prominent role of genetic susceptibilty in the pathogenesis of t-MNs. Common genetic variants have been identified that modulate t-MN risk, and t-MNs have been observed in some cancer predisposition syndromes. In either case, establishing a leukaemic phenotype requires acquisition of somatic mutations - most likely induced by the cytotoxic treatment. Knowledge of the specific nature of the initiating exposure has allowed the identification of crucial pathogenetic mechanisms and for these to be modelled in vitro and in vivo. Prognosis of patients with t-MNs is dismal and at present, the only curative approach for the majority of these individuals is haematopoietic stem cell transplantation, which is characterized by high transplant-related mortality rates. Novel transplantation strategies using reduced intensity conditioning regimens as well as novel drugs - demethylating agents and targeted therapies - await clinical testing and may improve outcome. Ultimately, individual assessment of genetic risk factors may translate into tailored therapies and establish a strategy for reducing t-MN incidences without jeopardizing therapeutic success rates for the primary disorders.
Collapse
Affiliation(s)
- H Sill
- Department of Internal Medicine, Division of Haematology, Medical University of Graz, Graz, Austria.
| | | | | | | | | |
Collapse
|
227
|
Bhattacharyya S, Wu M, Fang F, Tourtellotte W, Feghali-Bostwick C, Varga J. Early growth response transcription factors: key mediators of fibrosis and novel targets for anti-fibrotic therapy. Matrix Biol 2011; 30:235-42. [PMID: 21511034 DOI: 10.1016/j.matbio.2011.03.005] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/11/2011] [Revised: 03/11/2011] [Accepted: 03/15/2011] [Indexed: 02/06/2023]
Abstract
Fibrosis is a deregulated and ultimately defective form of tissue repair that underlies a large number of chronic human diseases, as well as obesity and aging. The pathogenesis of fibrosis involves multiple cell types and extracellular signals, of which transforming growth factor-ß (TGF-ß) is pre-eminent. The prevalence of fibrosis is rising worldwide, and to date no agents has shown clinical efficacy in the attenuating or reversing the process. Recent studies implicate the immediate-early response transcription factor Egr-1 in the pathogenesis of fibrosis. Egr-1 couples acute changes in the cellular environment to sustained alterations in gene expression, and mediates a broad spectrum of biological responses to injury and stress. In contrast to other ligand-activated transcription factors such as NF-κB, c-jun and Smad2/3 that undergo post-translational modification such as phosphorylation and nuclear translocation, Egr-1 activity is regulated via its biosynthesis. Aberrant Egr-1 expression or activity is implicated in cancer, inflammation, atherosclerosis, and ischemic injury and recent studies now indicate an important role for Egr-1 in TGF-ß-dependent profibrotic responses. Fibrosis in various animal models and human diseases such as scleroderma (SSc) and idiopathic pulmonary fibrosis (IPF) is accompanied by aberrant Egr-1 expression. Moreover Egr-1 appears to be required for physiologic and pathological connective tissue remodeling, and Egr-1-null mice are protected from fibrosis. As a novel profibrotic mediator, Egr-1 thus appears to be a promising potential target for the development of anti-fibrotic therapies.
Collapse
Affiliation(s)
- Swati Bhattacharyya
- Division of Rheumatology, Northwestern University Feinberg School of Medicine, Chicago, USA
| | | | | | | | | | | |
Collapse
|
228
|
Lu Y, Li T, Qureshi HY, Han D, Paudel HK. Early growth response 1 (Egr-1) regulates phosphorylation of microtubule-associated protein tau in mammalian brain. J Biol Chem 2011; 286:20569-81. [PMID: 21489990 DOI: 10.1074/jbc.m111.220962] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/06/2022] Open
Abstract
In the normal brain, tau protein is phosphorylated at a number of proline- and non-proline directed sites, which reduce tau microtubule binding and thus regulate microtubule dynamics. In Alzheimer disease (AD), tau is abnormally hyperphosphorylated, leading to neurofibrillary tangle formation and microtubule disruption, suggesting a loss of regulatory mechanisms controlling tau phosphorylation. Early growth response 1 (Egr-1) is a transcription factor that is significantly up-regulated in AD brain. The pathological significance of this up-regulation is not known. In this study, we found that lentivirus-mediated overexpression of Egr-1 in rat brain hippocampus and primary neurons in culture activates proline-directed kinase Cdk5, inactivates PP1, promotes tau phosphorylation at both proline-directed Ser(396/404) and non-proline-directed Ser(262) sites, and destabilizes microtubules. Furthermore, in Egr-1(-/-) mouse brain, Cdk5 activity was decreased, PP1 activity was increased, and tau phosphorylation was reduced at both proline-directed and non-proline-directed sites. By using nerve growth factor-exposed PC12 cells, we determined that Egr-1 activates Cdk5 to promote phosphorylation of tau and inactivates PP1 via phosphorylation. When Cdk5 was inhibited, tau phosphorylation at both proline- and non-proline directed sites and PP1 phosphorylation were blocked, indicating that Egr-1 acts through Cdk5. By using an in vitro kinase assay and HEK-293 cells transfected with tau, PP1, and Cdk5, we found that Cdk5 phosphorylates Ser(396/404) directly. In addition, by phosphorylating and inactivating PP1, Cdk5 promotes tau phosphorylation at Ser(262) indirectly. Our results indicate that Egr-1 is an in vivo regulator of tau phosphorylation and suggest that in AD brain increased levels of Egr-1 aberrantly activate an Egr-1/Cdk5/PP1 pathway, leading to accumulation of hyperphosphorylated tau, thus destabilizing the microtubule cytoskeleton.
Collapse
Affiliation(s)
- Yifan Lu
- The Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec H3T 1E2, Canada
| | | | | | | | | |
Collapse
|
229
|
Hannafon BN, Sebastiani P, de las Morenas A, Lu J, Rosenberg CL. Expression of microRNA and their gene targets are dysregulated in preinvasive breast cancer. Breast Cancer Res 2011; 13:R24. [PMID: 21375733 PMCID: PMC3219184 DOI: 10.1186/bcr2839] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/21/2010] [Revised: 01/24/2011] [Accepted: 03/04/2011] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION microRNA (miRNA) are short, noncoding RNA that negatively regulate gene expression and may play a causal role in invasive breast cancer. Since many genetic aberrations of invasive disease are detectable in early stages, we hypothesized that miRNA expression dysregulation and the predicted changes in gene expression might also be found in early breast neoplasias. METHODS Expression profiling of 365 miRNA by real-time quantitative polymerase chain reaction assay was combined with laser capture microdissection to obtain an epithelium-specific miRNA expression signature of normal breast epithelium from reduction mammoplasty (RM) (n = 9) and of paired samples of histologically normal epithelium (HN) and ductal carcinoma in situ (DCIS) (n = 16). To determine how miRNA may control the expression of codysregulated mRNA, we also performed gene expression microarray analysis in the same paired HN and DCIS samples and integrated this with miRNA target prediction. We further validated several target pairs by modulating the expression levels of miRNA in MCF7 cells and measured the expression of target mRNA and proteins. RESULTS Thirty-five miRNA were aberrantly expressed between RM, HN and DCIS. Twenty-nine miRNA and 420 mRNA were aberrantly expressed between HN and DCIS. Combining these two data sets with miRNA target prediction, we identified two established target pairs (miR-195:CCND1 and miR-21:NFIB) and tested several novel miRNA:mRNA target pairs. Overexpression of the putative tumor suppressor miR-125b, which is underexpressed in DCIS, repressed the expression of MEMO1, which is required for ErbB2-driven cell motility (also a target of miR-125b), and NRIP1/RIP140, which modulates the transcriptional activity of the estrogen receptor. Knockdown of the putative oncogenic miRNA miR-182 and miR-183, both highly overexpressed in DCIS, increased the expression of chromobox homolog 7 (CBX7) (which regulates E-cadherin expression), DOK4, NMT2 and EGR1. Augmentation of CBX7 by knockdown of miR-182 expression, in turn, positively regulated the expression of E-cadherin, a key protein involved in maintaining normal epithelial cell morphology, which is commonly lost during neoplastic progression. CONCLUSIONS These data provide the first miRNA expression profile of normal breast epithelium and of preinvasive breast carcinoma. Further, we demonstrate that altered miRNA expression can modulate gene expression changes that characterize these early cancers. We conclude that miRNA dysregulation likely plays a substantial role in early breast cancer development.
Collapse
Affiliation(s)
- Bethany N Hannafon
- Department of Medicine, Boston Medical Center and Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | | | | | | | | |
Collapse
|
230
|
Calogero A, Porcellini A, Lombari V, Fabbiano C, Arcella A, Miscusi M, Ponti D, Ragona G. Sensitivity to cisplatin in primary cell lines derived from human glioma correlates with levels of EGR-1 expression. Cancer Cell Int 2011; 11:5. [PMID: 21366897 PMCID: PMC3059282 DOI: 10.1186/1475-2867-11-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/14/2010] [Accepted: 03/02/2011] [Indexed: 11/20/2022] Open
Abstract
Background Less than 30% of malignant gliomas respond to adjuvant chemotherapy. Here, we have asked whether variations in the constitutive expression of early-growth response factor 1 (EGR-1) predicted acute cytotoxicity and clonogenic cell death in vitro, induced by six different chemotherapics. Materials and methods Cytotoxicity assays were performed on cells derived from fresh tumor explants of 18 human cases of malignant glioma. In addition to EGR-1, tumor cultures were investigated for genetic alterations and the expression of cancer regulating factors, related to the p53 pathway. Results We found that sensitivity to cisplatin correlates significantly with levels of EGR-1 expression in tumors with wild-type p53/INK4a/p16 status. Conclusion Increased knowledge of the mechanisms regulating EGR-1 expression in wild-type p53/INK4a/p16 cases of glioma may help in the design of new chemotherapeutic strategies for these tumors.
Collapse
Affiliation(s)
- Antonella Calogero
- Department of Medical-surgical Science and Biotechnologies, University of Rome "Sapienza", Corso della Repubblica 79, 04100 Latina, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
231
|
Suppression of osteosarcoma cell invasion by chemotherapy is mediated by urokinase plasminogen activator activity via up-regulation of EGR1. PLoS One 2011; 6:e16234. [PMID: 21283769 PMCID: PMC3024416 DOI: 10.1371/journal.pone.0016234] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/07/2010] [Accepted: 12/17/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The cellular and molecular mechanisms of tumour response following chemotherapy are largely unknown. We found that low dose anti-tumour agents up-regulate early growth response 1 (EGR1) expression. EGR1 is a member of the immediate-early gene group of transcription factors which modulate transcription of multiple genes involved in cell proliferation, differentiation, and development. It has been reported that EGR1 act as either tumour promoting factor or suppressor. We therefore examined the expression and function of EGR1 in osteosarcoma. METHODS We investigated the expression of EGR1 in human osteosarcoma cell lines and biopsy specimens. We next examined the expression of EGR1 following anti-tumour agents treatment. To examine the function of EGR1 in osteosarcoma, we assessed the tumour growth and invasion in vitro and in vivo. RESULTS Real-time PCR revealed that EGR1 was down-regulated both in osteosarcoma cell lines and osteosarcoma patients' biopsy specimens. In addition, EGR1 was up-regulated both in osteosarcoma patient' specimens and osteosarcoma cell lines following anti-tumour agent treatment. Although forced expression of EGR1 did not prevent osteosarcoma growth, forced expression of EGR1 prevented osteosarcoma cell invasion in vitro. In addition, forced expression of EGR1 promoted down-regulation of urokinase plasminogen activator, urokinase receptor, and urokinase plasminogen activity. Xenograft mice models showed that forced expression of EGR1 prevents osteosarcoma cell migration into blood vessels. CONCLUSIONS These findings suggest that although chemotherapy could not prevent osteosarcoma growth in chemotherapy-resistant patients, it did prevent osteosarcoma cell invasion by down-regulation of urokinase plasminogen activity via up-regulation of EGR1 during chemotherapy periods.
Collapse
|
232
|
Lu J, Li XP, Dong Q, Kung HF, He ML. TBX2 and TBX3: the special value for anticancer drug targets. Biochim Biophys Acta Rev Cancer 2010; 1806:268-74. [PMID: 20624445 PMCID: PMC7127380 DOI: 10.1016/j.bbcan.2010.07.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/09/2010] [Revised: 06/29/2010] [Accepted: 07/02/2010] [Indexed: 01/04/2023]
Abstract
TBX2 and TBX3 are members of the T-box family of transcription factors, which are implicated in embryonic development. Unlike most members of the T-box family, TBX2 and TBX3 are the only mammalian T-box factors which function as transcriptional repressors, mediated by the repression domain in the C-terminal. In addition to a role in development, recent evidence suggests that TBX2 and TBX3 are overexpressed in a number of cancers, including melanoma, breast, liver, lung, pancreas, ovarian, and cervical cancers. However, there is little information about the mechanisms for how these T-box genes contribute to tumorigenesis. Upregulation of TBX2 and TBX3 suppresses the expression of p14(ARF) and p21(CIP1) and promotes bypass of senescence through inactivation of p53 pathway. TBX2 functionally interacts with pRb, and pRb modulates TBX2 functional specificity. In addition, TBX2 is a player of Wnt signaling while TBX3 is a downstream target of the Wnt/beta-catenin pathway, and overexpression of TBX2 and TBX3 represses the expression of E-cadherin, which is demonstrated to be a prerequisite for epithelial tumor cell invasion. Moreover, TBX2 is shown to interact with EGR1 to block multiple downstream tumor suppressors. Here, we review the current knowledge on TBX2 and TBX3 in tumorigenesis and prospect their special value for development of target-based anticancer drugs.
Collapse
Key Words
- cdks, cyclin-dependent kinases
- egr1, early growth response 1
- fgf, fibroblast growth factor
- mefs, mouse embryonic fibroblasts
- rd, repression domain
- rnai, rna interference
- sirna, small interfering rna
- tgfβ, transforming growth factor β
- ums, ulnar-mammary syndrome
- ctcl, cutaneous t-cell lymphoma
- tbx2
- tbx3
- tumorigenesis
Collapse
Affiliation(s)
- Juan Lu
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | | | | | | | | |
Collapse
|
233
|
Sarver AL, Li L, Subramanian S. MicroRNA miR-183 functions as an oncogene by targeting the transcription factor EGR1 and promoting tumor cell migration. Cancer Res 2010; 70:9570-80. [PMID: 21118966 DOI: 10.1158/0008-5472.can-10-2074] [Citation(s) in RCA: 236] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/16/2022]
Abstract
The transcription factor EGR1 is a tumor suppressor gene that is downregulated in many cancer types. Clinically, loss of EGR1 translates to increased tumor transformation and subsequent patient morbidity and mortality. In synovial sarcoma, the SS18-SSX fusion protein represses EGR1 expression through a direct association with the EGR1 promoter. However, the mechanism through which EGR1 becomes downregulated in other tumor types is unclear. Here, we report that EGR1 is regulated by microRNA (miR)-183 in multiple tumor types including synovial sarcoma, rhabdomyosarcoma (RMS), and colon cancer. Using an integrative network analysis, we identified that miR-183 is significantly overexpressed in these tumor types as well as in corresponding tumor cell lines. Bioinformatic analyses suggested that miR-183 could target EGR1 mRNA and this specific interaction was validated in vitro. miR-183 knockdown in synovial sarcoma, RMS, and colon cancer cell lines revealed deregulation of a miRNA network composed of miR-183-EGR1-PTEN in these tumors. Integrated miRNA- and mRNA-based genomic analyses indicated that miR-183 is an important contributor to cell migration in these tumor types and this result was functionally validated to be occurring via an EGR1-based mechanism. In conclusion, our findings have significant implications in the mechanisms underlying EGR1 regulation in cancers. miR-183 has a potential oncogenic role through the regulation of 2 tumor suppressor genes, EGR1 and PTEN, and the deregulation of this fundamental miRNA regulatory network may be central to many tumor types.
Collapse
Affiliation(s)
- Aaron L Sarver
- Biostatistics and Bioinformatics, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | |
Collapse
|
234
|
Cabello CM, Lamore SD, Bair WB, Davis AL, Azimian SM, Wondrak GT. DCPIP (2,6-dichlorophenolindophenol) as a genotype-directed redox chemotherapeutic targeting NQO1*2 breast carcinoma. Free Radic Res 2010; 45:276-92. [PMID: 21034357 DOI: 10.3109/10715762.2010.526766] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022]
Abstract
Accumulative experimental evidence suggests feasibility of chemotherapeutic intervention targeting human cancer cells by pharmacological modulation of cellular oxidative stress. Current efforts aim at personalization of redox chemotherapy through identification of predictive tumour genotypes and redox biomarkers. Based on earlier research demonstrating that anti-melanoma activity of the pro-oxidant 2,6-dichlorophenolindophenol (DCPIP) is antagonized by cellular NAD(P)H:quinone oxidoreductase (NQO1) expression, this study tested DCPIP as a genotype-directed redox chemotherapeutic targeting homozygous NQO1*2 breast carcinoma, a common missense genotype [rs1800566 polymorphism; NP_000894.1:p.Pro187Ser] encoding a functionally impaired NQO1 protein. In a panel of cultured breast carcinoma cell lines and NQO1-transfectants with differential NQO1 expression levels, homozygous NQO1*2 MDA-MB231 cells were hypersensitive to DCPIP-induced caspase-independent cell death that occurred after early onset of oxidative stress with glutathione depletion and loss of genomic integrity. Array analysis revealed upregulated expression of oxidative (GSTM3, HMOX1, EGR1), heat shock (HSPA6, HSPA1A, CRYAB) and genotoxic stress response (GADD45A, CDKN1A) genes confirmed by immunoblot detection of HO-1, Hsp70, Hsp70B', p21 and phospho-p53 (Ser15). In a murine xenograft model of human homozygous NQO1*2-breast carcinoma, systemic administration of DCPIP displayed significant anti-tumour activity, suggesting feasibility of redox chemotherapeutic intervention targeting the NQO1*2 genotype.
Collapse
Affiliation(s)
- Christopher M Cabello
- Department of Pharmacology and Toxicology, College of Pharmacy, Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | | | | | | | | | | |
Collapse
|
235
|
Chromik AM, Hahn SA, Daigeler A, Flier A, Bulut D, May C, Harati K, Roschinsky J, Sülberg D, Weyhe D, Mittelkötter U, Uhl W. Gene expression analysis of cell death induction by taurolidine in different malignant cell lines. BMC Cancer 2010; 10:595. [PMID: 21034493 PMCID: PMC2988031 DOI: 10.1186/1471-2407-10-595] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/19/2010] [Accepted: 10/30/2010] [Indexed: 11/24/2022] Open
Abstract
Background The anti-infective agent Taurolidine (TRD) has been shown to have cell death inducing properties, but the mechanism of its action is largely unknown. The aim of this study was to identify potential common target genes modulated at the transcriptional level following TRD treatment in tumour cell lines originating from different cancer types. Methods Five different malignant cell lines (HT29, Chang Liver, HT1080, AsPC-1 and BxPC-3) were incubated with TRD (100 μM, 250 μM and 1000 μM). Proliferation after 8 h and cell viability after 24 h were analyzed by BrdU assay and FACS analysis, respectively. Gene expression analyses were carried out using the Agilent -microarray platform to indentify genes which displayed conjoint regulation following the addition of TRD in all cell lines. Candidate genes were subjected to Ingenuity Pathways Analysis and selected genes were validated by qRT-PCR and Western Blot. Results TRD 250 μM caused a significant inhibition of proliferation as well as apoptotic cell death in all cell lines. Among cell death associated genes with the strongest regulation in gene expression, we identified pro-apoptotic transcription factors (EGR1, ATF3) as well as genes involved in the ER stress response (PPP1R15A), in ubiquitination (TRAF6) and mitochondrial apoptotic pathways (PMAIP1). Conclusions This is the first conjoint analysis of potential target genes of TRD which was performed simultaneously in different malignant cell lines. The results indicate that TRD might be involved in different signal transduction pathways leading to apoptosis.
Collapse
Affiliation(s)
- Ansgar M Chromik
- Department of Visceral and General Surgery, St. Josef Hospital, Ruhr-University Bochum, Gudrunstrasse 56, D-44791 Bochum, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
236
|
Kim N, Kukkonen S, Gupta S, Aldovini A. Association of Tat with promoters of PTEN and PP2A subunits is key to transcriptional activation of apoptotic pathways in HIV-infected CD4+ T cells. PLoS Pathog 2010; 6:e1001103. [PMID: 20862322 PMCID: PMC2940756 DOI: 10.1371/journal.ppat.1001103] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/03/2009] [Accepted: 08/13/2010] [Indexed: 11/25/2022] Open
Abstract
Apoptosis in HIV-1-infected CD4+ primary T cells is triggered by the alteration of the PI3K and p53 pathways, which converge on the FOXO3a transcriptional activator. Tat alone can cause activation of FOXO3a and of its proapoptotic target genes. To understand how Tat affects this pathway, we carried out ChIP-Chip experiments with Tat. Tat associates with the promoters of PTEN and two PP2A subunit genes, but not with the FOXO3a promoter. PTEN and PP2A encode phosphatases, whose levels and activity are increased when Tat is expressed. They counteract phosphorylation of Akt1 and FOXO3a, and so activate transcriptional activity of FOXO3a. FOXO3a promotes increased transcription of Egr-1, which can further stimulate the transcription of PTEN, thereby reinforcing the pathway that leads to FOXO3a transcriptional activation. RNAi experiments support the role of PTEN and PP2A in the initiation of the Tat-mediated cascade, which is critical to apoptosis. The increased accumulation of PTEN and PP2A subunit mRNAs during Tat expression is more likely to be the result of increased transcription initiation and not relief of promoter-proximal pausing of RNAPII. The Tat-PTEN and -PP2A promoter interactions provide a mechanistic explanation of Tat-mediated apoptosis in CD4+ T cells. HIV infection leads to the depletion of CD4+ T cells, the major viral cell target. The destruction of these cells can occur because of cytopathic effect or apoptosis. HIV Tat is one of the proteins that can contribute to the apoptotic process of both infected and uninfected cells, as it is released in the plasma and enter uninfected cells. Tat expression in CD4+ T-cells is linked to increased transcriptional activity of FOXO3a, a factor that targets the transcription of pro-apoptotic genes. The mechanism by which Tat leads to activation apoptotic pathways is by associating with the promoters of the phospatases PTEN and PP2A and by increasing their levels. The increased amount of these proteins leads to a decreased amount of pAKt1 and increased amount of non-phosphorylated FOXO3a, which migrates from the cytoplasm to the nucleus and increases the transcription of its proapoptotic target genes. These results, together with experiments that silence PTEN and PP2A and measure their activities, identify the association of Tat with PTEN and PP2A promoters as the initiating event of Tat-mediated apoptosis.
Collapse
Affiliation(s)
- Nayoung Kim
- Children's Hospital Boston, Department of Medicine, and Harvard Medical School, Department of Pediatrics, Boston, Massachusetts, United States of America
| | - Sami Kukkonen
- Children's Hospital Boston, Department of Medicine, and Harvard Medical School, Department of Pediatrics, Boston, Massachusetts, United States of America
| | - Sumeet Gupta
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, United States of America
| | - Anna Aldovini
- Children's Hospital Boston, Department of Medicine, and Harvard Medical School, Department of Pediatrics, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
237
|
Zheng L, Pu J, Jiang G, Weng M, He J, Mei H, Hou X, Tong Q. Abnormal expression of early growth response 1 in gastric cancer: association with tumor invasion, metastasis and heparanase transcription. Pathol Int 2010; 60:268-77. [PMID: 20403028 DOI: 10.1111/j.1440-1827.2010.02512.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/29/2023]
Abstract
Given that previous studies indicated that early growth response 1 (EGR1) exerts pro-tumorigenic effects through regulating heparanase (HPA) transcription, it was hypothesized that EGR1 may correlate with the progression of gastric cancer. One hundred and fifteen patients with gastric cancer were evaluated for the protein and transcript expression of EGR1 and HPA on immunohistochemistry and real-time quantitative polymerase chain reaction (PCR). In normal gastric mucosa, EGR1 protein expression was absent or weak, whereas gastric cancer was positive for EGR1. Seventy gastric cancer patients (60.9%) were positive for cytoplasmic EGR1 expression, and 26 (22.6%) had nuclear expression of EGR1. In the gastric cancer examined, the transcripts of EGR1 were enhanced compared to that of normal gastric mucosa, and positively correlated with EGR1 protein expression. The cytoplasmic or nuclear expression of EGR1 and its transcripts in gastric cancer was positively correlated with tumor infiltration (P < 0.05), lymph node and distant metastasis (P < 0.05), tumor node metastasis (TNM) stages (P < 0.05), but not with age, gender, tumor location and size, histological types or differentiation. Moreover, the protein and transcript expression of EGR1 was correlated with that of HPA in gastric cancer. These results indicate that aberrant expression of EGR1 in gastric cancer is associated with tumor invasion and metastasis, and HPA transcription.
Collapse
Affiliation(s)
- Liduan Zheng
- Department of Pathology, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | | | | | | | | | | | | | | |
Collapse
|
238
|
Avraham R, Sas-Chen A, Manor O, Steinfeld I, Shalgi R, Tarcic G, Bossel N, Zeisel A, Amit I, Zwang Y, Enerly E, Russnes HG, Biagioni F, Mottolese M, Strano S, Blandino G, Børresen-Dale AL, Pilpel Y, Yakhini Z, Segal E, Yarden Y. EGF decreases the abundance of microRNAs that restrain oncogenic transcription factors. Sci Signal 2010; 3:ra43. [PMID: 20516477 DOI: 10.1126/scisignal.2000876] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/02/2022]
Abstract
Epidermal growth factor (EGF) stimulates cells by launching gene expression programs that are frequently deregulated in cancer. MicroRNAs, which attenuate gene expression by binding complementary regions in messenger RNAs, are broadly implicated in cancer. Using genome-wide approaches, we showed that EGF stimulation initiates a coordinated transcriptional program of microRNAs and transcription factors. The earliest event involved a decrease in the abundance of a subset of 23 microRNAs. This step permitted rapid induction of oncogenic transcription factors, such as c-FOS, encoded by immediate early genes. In line with roles as suppressors of EGF receptor (EGFR) signaling, we report that the abundance of this early subset of microRNAs is decreased in breast and in brain tumors driven by the EGFR or the closely related HER2. These findings identify specific microRNAs as attenuators of growth factor signaling and oncogenesis.
Collapse
Affiliation(s)
- Roi Avraham
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
239
|
Zhang J, Valianou M, Cheng JD. Identification and characterization of the promoter of fibroblast activation protein. Front Biosci (Elite Ed) 2010; 2:1154-63. [PMID: 20515787 DOI: 10.2741/e175] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/07/2023]
Abstract
Fibroblast activation protein (FAP) is a type II integral membrane glycoprotein belonging to the serine protease family. It is selectively expressed by tumor stromal fibroblasts and transiently in the fibroblasts of healing wounds. FAP has been shown to modulate growth, differentiation, adhesion, and metastasis of tumor cells. Despite the importance of FAP in cancer, the mechanisms that govern its expression have not been defined. In this study, we determined the transcription start site of the FAP gene and identified a 2-kb segment with promoter activity in cells expressing FAP. Truncation of this fragment revealed that the core promoter activity resided in a 245-bp fragment surrounding the transcription start site. Electrophoretic mobility shift assay showed that EGR1 binds to the FAP promoter. Mutation of the EGR1 site within this fragment significantly decreased the promoter activity of FAP and eliminated EGR1 binding. Down-regulation of EGR1 resulted in a significant reduction in endogenous FAP mRNA expression. These findings identify the basal transcriptional requirements of FAP gene expression and show EGR1 is an important regulator of FAP expression.
Collapse
Affiliation(s)
- Jiping Zhang
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | | | | |
Collapse
|
240
|
Andréasson U, Edén P, Peterson C, Högerkorp CM, Jerkeman M, Andersen N, Berglund M, Sundström C, Rosenquist R, Borrebaeck CAK, Ek S. Identification of uniquely expressed transcription factors in highly purified B-cell lymphoma samples. Am J Hematol 2010; 85:418-25. [PMID: 20513119 DOI: 10.1002/ajh.21701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/23/2023]
Abstract
Transcription factors (TFs) are critical for B-cell differentiation, affecting gene expression both by repression and transcriptional activation. Still, this information is not used for classification of B-cell lymphomas (BCLs). Traditionally, BCLs are diagnosed based on a phenotypic resemblance to normal B-cells; assessed by immunohistochemistry or flow cytometry, by using a handful of phenotypic markers. In the last decade, diagnostic and prognostic evaluation has been facilitated by global gene expression profiling (GEP), providing a new powerful means for the classification, prediction of survival, and response to treatment of lymphomas. However, most GEP studies have typically been performed on whole tissue samples, containing varying degrees of tumor cell content, which results in uncertainties in data analysis. In this study, global GEP analyses were performed on highly purified, flow-cytometry sorted tumor-cells from eight subgroups of BCLs. This enabled identification of TFs that can be uniquely associated to the tumor cells of chronic lymphocytic leukemia (CLL), diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL), hairy cell leukemia (HCL), and mantle cell lymphoma (MCL). The identified transcription factors influence both the global and specific gene expression of the BCLs and have possible implications for diagnosis and treatment.
Collapse
|
241
|
Kuo YS, Tang YB, Lu TY, Wu HC, Lin CT. IGFBP-6 plays a role as an oncosuppressor gene in NPC pathogenesis through regulating EGR-1 expression. J Pathol 2010; 222:299-309. [DOI: 10.1002/path.2735] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/05/2023]
|
242
|
Gregg JL, Brown KE, Mintz EM, Piontkivska H, Fraizer GC. Analysis of gene expression in prostate cancer epithelial and interstitial stromal cells using laser capture microdissection. BMC Cancer 2010; 10:165. [PMID: 20426842 PMCID: PMC2876079 DOI: 10.1186/1471-2407-10-165] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/13/2009] [Accepted: 04/28/2010] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The prostate gland represents a multifaceted system in which prostate epithelia and stroma have distinct physiological roles. To understand the interaction between stroma and glandular epithelia, it is essential to delineate the gene expression profiles of these two tissue types in prostate cancer. Most studies have compared tumor and normal samples by performing global expression analysis using a mixture of cell populations. This report presents the first study of prostate tumor tissue that examines patterns of differential expression between specific cell types using laser capture microdissection (LCM). METHODS LCM was used to isolate distinct cell-type populations and identify their gene expression differences using oligonucleotide microarrays. Ten differentially expressed genes were then analyzed in paired tumor and non-neoplastic prostate tissues by quantitative real-time PCR. Expression patterns of the transcription factors, WT1 and EGR1, were further compared in established prostate cell lines. WT1 protein expression was also examined in prostate tissue microarrays using immunohistochemistry. RESULTS The two-step method of laser capture and microarray analysis identified nearly 500 genes whose expression levels were significantly different in prostate epithelial versus stromal tissues. Several genes expressed in epithelial cells (WT1, GATA2, and FGFR-3) were more highly expressed in neoplastic than in non-neoplastic tissues; conversely several genes expressed in stromal cells (CCL5, CXCL13, IGF-1, FGF-2, and IGFBP3) were more highly expressed in non-neoplastic than in neoplastic tissues. Notably, EGR1 was also differentially expressed between epithelial and stromal tissues. Expression of WT1 and EGR1 in cell lines was consistent with these patterns of differential expression. Importantly, WT1 protein expression was demonstrated in tumor tissues and was absent in normal and benign tissues. CONCLUSIONS The prostate represents a complex mix of cell types and there is a need to analyze distinct cell populations to better understand their potential interactions. In the present study, LCM and microarray analysis were used to identify novel gene expression patterns in prostate cell populations, including identification of WT1 expression in epithelial cells. The relevance of WT1 expression in prostate cancer was confirmed by analysis of tumor tissue and cell lines, suggesting a potential role for WT1 in prostate tumorigenesis.
Collapse
Affiliation(s)
- Jennifer L Gregg
- School of Biomedical Sciences, Kent State University, Kent, OH, USA
| | | | | | | | | |
Collapse
|
243
|
Saletta F, Suryo Rahmanto Y, Noulsri E, Richardson DR. Iron chelator-mediated alterations in gene expression: identification of novel iron-regulated molecules that are molecular targets of hypoxia-inducible factor-1 alpha and p53. Mol Pharmacol 2010; 77:443-58. [PMID: 20023006 DOI: 10.1124/mol.109.061028] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/11/2023] Open
Abstract
Iron deficiency affects 500 million people, yet the molecular role of iron in gene expression remains poorly characterized. In addition, the alterations in global gene expression after iron chelation remain unclear and are important to assess for understanding the molecular pathology of iron deficiency and the biological effects of chelators. Considering this, we assessed the effect on whole genome gene expression of two iron chelators (desferrioxamine and 2-hydroxy-1-napthylaldehyde isonicotinoyl hydrazone) that have markedly different permeability properties. Sixteen genes were significantly regulated by both ligands, whereas a further 50 genes were significantly regulated by either compound. Apart from iron-mediated regulation of expression via hypoxia inducible factor-1 alpha, it was noteworthy that the transcription factor p53 was also involved in iron-regulated gene expression. Examining 16 genes regulated by both chelators in normal and neoplastic cells, five genes (APP, GDF15, CITED2, EGR1, and PNRC1) were significantly differentially expressed between the cell types. In view of their functions in tumor suppression, proliferation, and apoptosis, these findings are important for understanding the selective antiproliferative effects of chelators against neoplastic cells. Most of the genes identified have not been described previously to be iron-regulated and are important for understanding the molecular and cellular effects of iron depletion.
Collapse
Affiliation(s)
- Federica Saletta
- Iron Metabolism and Chelation Program, Department of Pathology, Bosch Institute, University of Sydney, Sydney, New South Wales, 2006, Australia
| | | | | | | |
Collapse
|
244
|
Mallakin A, Sugiyama T, Kai F, Taneja P, Kendig RD, Frazier DP, Maglic D, Matise LA, Willingham MC, Inoue K. The Arf-inducing transcription factor Dmp1 encodes a transcriptional activator of amphiregulin, thrombospondin-1, JunB and Egr1. Int J Cancer 2010; 126:1403-16. [PMID: 19816943 DOI: 10.1002/ijc.24938] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/01/2023]
Abstract
Dmp1 (Dmtf1) encodes a Myb-like transcription factor implicated in tumor suppression through direct activation of the Arf-p53 pathway. The human DMP1 gene is frequently deleted in non-small cell lung cancers, especially those that retain wild-type INK4a/ARF and/or p53. To identify novel genes that are regulated by Dmp1, transcriptional profiles of lung tissue from Dmp1-null and wild-type mice were generated using the GeneChip Microarray. Comparative analysis of gene expression changes between the two groups resulted in identification of numerous genes that may be regulated by Dmp1. Notably, amphiregulin (Areg), thrombospondin-1 (Tsp-1), JunB, Egr1, adrenomedullin (Adm), Bcl-3 and methyl-CpG binding domain protein 1 (Mbd1) were downregulated in the lungs from Dmp1-null mice while Gas1 and Ect2 genes were upregulated. These target genes were chosen for further analyses since they are involved in cell proliferation, transcription, angiogenesis/metastasis, apoptosis, or DNA methylation, and thus could account for the tumor suppressor phenotype of Dmp1. Dmp1 directly bound to the genomic loci of Areg, Tsp-1, JunB and Egr1. Significant upregulation or downregulation of the novel Dmp1 target genes was observed upon transient expression of Dmp1 in alveolar epithelial cells, an effect which was nullified by the inhibition of de novo mRNA synthesis. Interestingly, these genes and their protein products were significantly downregulated or upregulated in the lungs from Dmp1-heterozygous mice as well. Identification of novel Dmp1 target genes not only provides insights into the effects of Dmp1 on global gene expression, but also sheds light on the mechanism of haploid insufficiency of Dmp1 in tumor suppression.
Collapse
Affiliation(s)
- Ali Mallakin
- Department of Pathology, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
245
|
TAR1, a human anti-p53 single-chain antibody, restores tumor suppressor function to mutant p53 variants. J Immunother 2010; 33:146-54. [PMID: 20139776 DOI: 10.1097/cji.0b013e3181be14dc] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/26/2022]
Abstract
The tumor suppressor gene p53 is mutated in more than half of human tumors. One important characteristic of p53 mutants is their accumulation in the nucleus of cancer cells. Thus, reactivation of mutant p53 proteins may trigger massive apoptosis in tumor cells. Pharmacologic methods are currently under development to induce mutant p53 proteins to resume their wild-type function. We have identified a human single-chain Fv fragment, designated as transcriptional transactivation and apoptosis restoring (TAR1), which specifically and with high affinity binds to mutant p53 and restores its wild-type active conformation. Binding of TAR1 to mutant p53 induced transcriptional transactivation of p53 target genes and down-regulation of mutant p53 transcriptional target genes. TAR1 treatment induced apoptosis in a variety of cell lines endogenously expressing p53 carrying different point mutations DNA contact or structural p53 mutants. Moreover, in an animal model of mice carrying human xenografts, TAR1 induced tumor regression with no apparent deleterious side effects. Thus, it may be considered as a potential candidate for anticancer treatment, targeting tumors with mutant p53.
Collapse
|
246
|
Shin SY, Kim CG, Kim SH, Kim YS, Lim Y, Lee YH. Chlorpromazine activates p21Waf1/Cip1gene transcription via early growth response-1 (Egr-1) in C6 glioma cells. Exp Mol Med 2010; 42:395-405. [DOI: 10.3858/emm.2010.42.5.041] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Soon Young Shin
- Institute of Biomedical Science and Technology, Konkuk University Hospital, Seoul 143-729, Korea
- Department of Biomedical Science and Technology, Research Center for Transcription Control, Konkuk University, Seoul 143-701, Korea
| | - Chang Gun Kim
- Institute of Biomedical Science and Technology, Konkuk University Hospital, Seoul 143-729, Korea
- Department of Biomedical Science and Technology, Research Center for Transcription Control, Konkuk University, Seoul 143-701, Korea
| | - Se Hyun Kim
- Department of Psychiatry, Clinical Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Yong Sik Kim
- Department of Psychiatry, Clinical Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 110-744, Korea
| | - Yoongho Lim
- Division of Bioscience and Biotechnology, BMIC, Konkuk University, Seoul 143-701, Korea
| | - Young Han Lee
- Institute of Biomedical Science and Technology, Konkuk University Hospital, Seoul 143-729, Korea
- Department of Biomedical Science and Technology, Research Center for Transcription Control, Konkuk University, Seoul 143-701, Korea
| |
Collapse
|
247
|
Choi BH, Kim CG, Lim Y, Lee YH, Shin SY. Transcriptional activation of the human Klotho gene by epidermal growth factor in HEK293 cells; role of Egr-1. Gene 2010; 450:121-7. [DOI: 10.1016/j.gene.2009.11.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/28/2009] [Revised: 11/05/2009] [Accepted: 11/06/2009] [Indexed: 11/25/2022]
|
248
|
Abstract
Prostate cancer is a major cause of cancer-related death in American men, for which finding new therapeutic strategies remains a challenge. Early growth response-1 (EGR1) is a transcription factor involved in cell proliferation and in the regulation of apoptosis. Although it has long been considered a tumor suppressor, a wealth of new evidence shows that EGR1 promotes the progression of prostate cancer. This review addresses the paradoxes of EGR1 function. While EGR1 mediates apoptosis in response to stress and DNA damage by regulating a tumor suppressor network, it also promotes the proliferation of prostate cancer cells by a mechanism that is not fully understood. Thus, EGR1 might be targeted for prostate cancer therapy either by ectopic expression in combination with radiotherapy or chemotherapy, or by direct inhibition for systemic treatment. Possible strategies to antagonize EGR1 function in a therapeutic setting are discussed.
Collapse
Affiliation(s)
- Delphine Gitenay
- The Vaccine Research Institute of San Diego, San Diego, CA 92121, USA. 19429708
| | | |
Collapse
|
249
|
Moulos P, Papadodima O, Chatziioannou A, Loutrari H, Roussos C, Kolisis FN. A transcriptomic computational analysis of mastic oil-treated Lewis lung carcinomas reveals molecular mechanisms targeting tumor cell growth and survival. BMC Med Genomics 2009; 2:68. [PMID: 20003503 PMCID: PMC2801511 DOI: 10.1186/1755-8794-2-68] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/04/2009] [Accepted: 12/15/2009] [Indexed: 12/17/2022] Open
Abstract
Background Mastic oil from Pistacia lentiscus variation chia, a blend of bioactive terpenes with recognized medicinal properties, has been recently shown to exert anti-tumor growth activity through inhibition of cancer cell proliferation, survival, angiogenesis and inflammatory response. However, no studies have addressed its mechanisms of action at genome-wide gene expression level. Methods To investigate molecular mechanisms triggered by mastic oil, Lewis Lung Carcinoma cells were treated with mastic oil or DMSO and RNA was collected at five distinct time points (3-48 h). Microarray expression profiling was performed using Illumina mouse-6 v1 beadchips, followed by computational analysis. For a number of selected genes, RT-PCR validation was performed in LLC cells as well as in three human cancer cell lines of different origin (A549, HCT116, K562). PTEN specific inhibition by a bisperovanadium compound was applied to validate its contribution to mastic oil-mediated anti-tumor growth effects. Results In this work we demonstrated that exposure of Lewis lung carcinomas to mastic oil caused a time-dependent alteration in the expression of 925 genes. GO analysis associated expression profiles with several biological processes and functions. Among them, modifications on cell cycle/proliferation, survival and NF-κB cascade in conjunction with concomitant regulation of genes encoding for PTEN, E2F7, HMOX1 (up-regulation) and NOD1 (down-regulation) indicated some important mechanistic links underlying the anti-proliferative, pro-apoptotic and anti-inflammatory effects of mastic oil. The expression profiles of Hmox1, Pten and E2f7 genes were similarly altered by mastic oil in the majority of test cancer cell lines. Inhibition of PTEN partially reversed mastic oil effects on tumor cell growth, indicating a multi-target mechanism of action. Finally, k-means clustering, organized the significant gene list in eight clusters demonstrating a similar expression profile. Promoter analysis in a representative cluster revealed shared putative cis-elements suggesting a common regulatory transcription mechanism. Conclusions Present results provide novel evidence on the molecular basis of tumor growth inhibition mediated by mastic oil and set a rational basis for application of genomics and bioinformatic methodologies in the screening of natural compounds with potential cancer chemopreventive activities.
Collapse
Affiliation(s)
- Panagiotis Moulos
- Institute of Biological Research and Biotechnology, National Hellenic Research Foundation, Athens, Greece.
| | | | | | | | | | | |
Collapse
|
250
|
Schippert R, Schaeffel F, Feldkaemper MP. Microarray analysis of retinal gene expression in Egr-1 knockout mice. Mol Vis 2009; 15:2720-39. [PMID: 20019881 PMCID: PMC2793901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/09/2009] [Accepted: 12/04/2009] [Indexed: 10/25/2022] Open
Abstract
PURPOSE We found earlier that 42 day-old Egr-1 knockout mice had longer eyes and a more myopic refractive error compared to their wild-types. To identify genes that could be responsible for the temporarily enhanced axial eye growth, a microarray analysis was performed in knockout and wild-type mice at the postnatal ages of 30 and 42 days. METHODS The retinas of homozygous and wild-type Egr-1 knockout mice (Taconic, Ry, Denmark) were prepared for RNA isolation (RNeasy Mini Kit, Qiagen) at the age of 30 or 42 days, respectively (n=12 each). Three retinas were pooled and labeled cRNA was made. The samples were hybridized to Affymetrix GeneChip Mouse Genome 430 2.0 Arrays. Hybridization signals were calculated using GC-RMA normalization. Genes were identified as differentially expressed if they showed a fold-change (FC) of at least 1.5 and a p-value <0.05. A false-discovery rate of 5% was applied. Ten genes with potential biologic relevance were examined further with semiquantitative real-time RT-PCR. RESULTS Comparing mRNA expression levels between wild-type and homozygous Egr-1 knockout mice, we found 73 differentially expressed genes at the age of 30 days and 135 genes at the age of 42 days. Testing for differences in gene expression between the two ages (30 versus 42 days), 54 genes were differently expressed in wild-type mice and 215 genes in homozygous animals. Based on three networks proposed by Ingenuity pathway analysis software, nine differently expressed genes in the homozygous Egr-1 knockout mice were chosen for further validation by real-time RT-PCR, three genes in each network. In addition, the gene that was most prominently regulated in the knockout mice, compared to wild-type, at both 30 days and 42 days of age (protocadherin beta-9 [Pcdhb9]), was tested with real-time RT-PCR. Changes in four of the ten genes could be confirmed by real-time RT-PCR: nuclear prelamin A recognition factor (Narf), oxoglutarate dehydrogenase (Ogdh), selenium binding protein 1 (Selenbp1), and Pcdhb9. Except for Pcdhb9, the genes whose mRNA expression levels were validated were listed in one of the networks proposed by Ingenuity pathway analysis software. In addition to these genes, the software proposed several key-regulators which did not change in our study: retinoic acid, vascular endothelial growth factor A (VEGF-A), FBJ murine osteosarcoma viral oncogene homolog (cFos), and others. CONCLUSIONS Identification of genes that are differentially regulated during the development period between postnatal day 30 (when both homozygous and wild-type mice still have the same axial length) and day 42 (where the difference in eye length is apparent) could improve the understanding of mechanisms for the control of axial eye growth and may lead to potential targets for pharmacological intervention. With the aid of pathway-analysis software, a coarse picture of possible biochemical pathways could be generated. Although the mRNA expression levels of proteins proposed by the software, like VEGF, FOS, retinoic acid (RA) receptors, or cellular RA binding protein, did not show any changes in our experiment, these molecules have previously been implicated in the signaling cascades controlling axial eye growth. According to the pathway-analysis software, they represent links between several proteins whose mRNA expression was changed in our study.
Collapse
|