1
|
Gabbianelli R, Shahar E, de Simone G, Rucci C, Bordoni L, Feliziani G, Zhao F, Ferrati M, Maggi F, Spinozzi E, Mahajna J. Plant-Derived Epi-Nutraceuticals as Potential Broad-Spectrum Anti-Viral Agents. Nutrients 2023; 15:4719. [PMID: 38004113 PMCID: PMC10675658 DOI: 10.3390/nu15224719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Although the COVID-19 pandemic appears to be diminishing, the emergence of SARS-CoV-2 variants represents a threat to humans due to their inherent transmissibility, immunological evasion, virulence, and invulnerability to existing therapies. The COVID-19 pandemic affected more than 500 million people and caused over 6 million deaths. Vaccines are essential, but in circumstances in which vaccination is not accessible or in individuals with compromised immune systems, drugs can provide additional protection. Targeting host signaling pathways is recommended due to their genomic stability and resistance barriers. Moreover, targeting host factors allows us to develop compounds that are effective against different viral variants as well as against newly emerging virus strains. In recent years, the globe has experienced climate change, which may contribute to the emergence and spread of infectious diseases through a variety of factors. Warmer temperatures and changing precipitation patterns can increase the geographic range of disease-carrying vectors, increasing the risk of diseases spreading to new areas. Climate change may also affect vector behavior, leading to a longer breeding season and more breeding sites for disease vectors. Climate change may also disrupt ecosystems, bringing humans closer to wildlife that transmits zoonotic diseases. All the above factors may accelerate the emergence of new viral epidemics. Plant-derived products, which have been used in traditional medicine for treating pathological conditions, offer structurally novel therapeutic compounds, including those with anti-viral activity. In addition, plant-derived bioactive substances might serve as the ideal basis for developing sustainable/efficient/cost-effective anti-viral alternatives. Interest in herbal antiviral products has increased. More than 50% of approved drugs originate from herbal sources. Plant-derived compounds offer diverse structures and bioactive molecules that are candidates for new drug development. Combining these therapies with conventional drugs could improve patient outcomes. Epigenetics modifications in the genome can affect gene expression without altering DNA sequences. Host cells can use epigenetic gene regulation as a mechanism to silence incoming viral DNA molecules, while viruses recruit cellular epitranscriptomic (covalent modifications of RNAs) modifiers to increase the translational efficiency and transcript stability of viral transcripts to enhance viral gene expression and replication. Moreover, viruses manipulate host cells' epigenetic machinery to ensure productive viral infections. Environmental factors, such as natural products, may influence epigenetic modifications. In this review, we explore the potential of plant-derived substances as epigenetic modifiers for broad-spectrum anti-viral activity, reviewing their modulation processes and anti-viral effects on DNA and RNA viruses, as well as addressing future research objectives in this rapidly emerging field.
Collapse
Affiliation(s)
- Rosita Gabbianelli
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Ehud Shahar
- Department of Nutrition and Natural Products, Migal—Galilee Research Institute, Kiryat Shmona 11016, Israel;
- Department of Biotechnology, Tel-Hai College, Kiryat Shmona 1220800, Israel
| | - Gaia de Simone
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Chiara Rucci
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Laura Bordoni
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Giulia Feliziani
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Fanrui Zhao
- Unit of Molecular Biology and Nutrigenomics, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (R.G.); (G.d.S.); (L.B.); (G.F.); (F.Z.)
| | - Marta Ferrati
- Chemistry Interdisciplinary Project (ChIP) Research Centre, School of Pharmacy, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (M.F.); (F.M.); (E.S.)
| | - Filippo Maggi
- Chemistry Interdisciplinary Project (ChIP) Research Centre, School of Pharmacy, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (M.F.); (F.M.); (E.S.)
| | - Eleonora Spinozzi
- Chemistry Interdisciplinary Project (ChIP) Research Centre, School of Pharmacy, University of Camerino, Via Madonna delle Carceri, 62032 Camerino, Italy; (M.F.); (F.M.); (E.S.)
| | - Jamal Mahajna
- Department of Nutrition and Natural Products, Migal—Galilee Research Institute, Kiryat Shmona 11016, Israel;
- Department of Biotechnology, Tel-Hai College, Kiryat Shmona 1220800, Israel
| |
Collapse
|
2
|
Transposon-triggered innate immune response confers cancer resistance to the blind mole rat. Nat Immunol 2021; 22:1219-1230. [PMID: 34556881 PMCID: PMC8488014 DOI: 10.1038/s41590-021-01027-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/11/2021] [Indexed: 02/05/2023]
Abstract
Blind mole rats (BMRs) are small rodents, characterized by exceptionally long lifespan (> 21 years) and resistance to both spontaneous and induced tumorigenesis. Here we report that cancer resistance in the BMR is mediated by retrotransposable elements (RTEs). BMR cells and tissues express very low levels of DNA methyltransferase 1 (DNMT1). Upon cell hyperplasia, the BMR genome DNA loses methylation, resulting in activation of RTEs. Up-regulated RTEs form cytoplasmic RNA/DNA hybrids, which activate cGAS-STING pathway to induce cell death. Although this mechanism is enhanced in the BMR, we show that it functions in mice and human. We propose that RTEs were coopted to serve as tumor suppressors that monitor cell proliferation and are activated in premalignant cells to trigger cell death via activation of innate immune response. RTEs activation is a double-edged sword, serving as a tumor suppressor but in late life contributing to aging via induction of sterile inflammation.
Collapse
|
3
|
Karimi L, Mansoori B, shanebandi D, Mohammadi A, Aghapour M, Baradaran B. Function of microRNA-143 in different signal pathways in cancer: New insights into cancer therapy. Biomed Pharmacother 2017; 91:121-131. [DOI: 10.1016/j.biopha.2017.04.060] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/05/2017] [Accepted: 04/13/2017] [Indexed: 01/05/2023] Open
|
4
|
Liu Y, Zhi L, Shen J, Li S, Yao J, Yang X. Effect of in ovo folic acid injection on hepatic IGF2 expression and embryo growth of broilers. J Anim Sci Biotechnol 2016; 7:40. [PMID: 27453780 PMCID: PMC4957392 DOI: 10.1186/s40104-016-0099-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 06/28/2016] [Indexed: 11/12/2022] Open
Abstract
Background Insulin-like factor 2 (IGF2) plays an important role in embryonic growth process by modulating intermediary metabolism and cell proliferation. Folic acid is involved in one carbon metabolism and contributes to DNA methylation which is related to gene expression. The purpose of this study was to explore whether folic acid could regulate IGF2 expression via epigenetic mechanism and further promote embryonic growth of new-hatched broilers. Methods In the present study, 360 fertile eggs were selected and randomly assigned to four treatments. On 11 embryonic day of incubation (E11), 0, 50, 100 and 150 μg folic acid were injected into eggs respectively. After hatched, growth performance of broilers were calculated. Hepatic IGF2 expression, methylation level and chromatin structure of promoter region were analyzed. Results Results have showed that IGF2 expression was up-regulated in 150 μg folic acid group (P < 0.05) and other two dose of folic acid did not affect gene expression (P > 0.05). Meanwhile, methylation level of IGF2 promoter were lower in 100 and 150 μg groups, which was consistent with lower expression of DNA methyltransferase 1 (DNMT1) (P < 0.05). What’s more, chromatin looseness of IGF2 promoter was higher in 150 μg group than control group (P < 0.05). Further, birth weight (BW), liver and bursa index of new-hatched chickens in 150 μg folic acid group were higher than the other groups (P < 0.05). There were positive correlations between hepatic IGF2 expression and BW and organs index (P < 0.05). Conclusion In conclusion, our data have demonstrated that 150 μg folic acid injection on E11 could up-regulate IGF2 expression by modulating DNA hypomethylation and improving chromatin accessibility in the gene promoter region, and ulteriorly facilitate embryonic growth and organ development of broilers. Electronic supplementary material The online version of this article (doi:10.1186/s40104-016-0099-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 China
| | - Lihui Zhi
- School of Mathematics and Computer Science, ShanXi Normal University, Linfen, 041000 China
| | - Jing Shen
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 China
| | - Shizhao Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 China
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100 China
| |
Collapse
|
5
|
Boosani CS, Agrawal DK. Epigenetic Regulation of Innate Immunity by microRNAs. Antibodies (Basel) 2016; 5:E8. [PMID: 31557989 PMCID: PMC6698855 DOI: 10.3390/antib5020008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/16/2016] [Accepted: 03/16/2016] [Indexed: 12/20/2022] Open
Abstract
The innate immune response, which is usually referred to as the first line of defense, protects the hosts against pathogenic micro-organisms. Some of the biomolecules released from the pathogens, such as proteins, lipoproteins and nucleic acids, which are collectively termed as pathogen-associated molecular patterns (PAMPs), elicit signaling mechanisms that trigger immune responses in the hosts. Pathogen recognition receptors (PRRs) on the host cells recognize these PAMPs and initiate intracellular signaling through toll-like receptors (TLRs), RIG-I-like receptors (RLRs), and other pathways which induce production of pro-inflammatory cytokines and type I interferons. Recently, different members of tripartite motif containing proteins (TRIM) family of proteins were identified to intercept and regulate these cellular pathways. Specific targets of TRIM proteins have been identified and their molecular mechanisms were unraveled and identified unique domains involved in protein-protein interactions. Though innate immunity represents a tight and well conserved immune system in the host, gene expression in innate immunity was identified to be influenced by several epigenetic mechanisms including regulation by microRNAs (miRNAs). In this review, we present critical analysis of the findings on the identification of specific miRNAs that modulate expression of target genes involved in the regulation of innate immunity.
Collapse
Affiliation(s)
- Chandra S Boosani
- Department of Clinical & Translational Science, Creighton University School of Medicine, Omaha, NE 68178, USA.
| | - Devendra K Agrawal
- Department of Clinical & Translational Science, Creighton University School of Medicine, Omaha, NE 68178, USA.
| |
Collapse
|
6
|
Cheishvili D, Boureau L, Szyf M. DNA demethylation and invasive cancer: implications for therapeutics. Br J Pharmacol 2015; 172:2705-15. [PMID: 25134627 DOI: 10.1111/bph.12885] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 08/01/2014] [Accepted: 08/13/2014] [Indexed: 02/06/2023] Open
Abstract
One of the hallmarks of cancer is aberrant DNA methylation, which is associated with abnormal gene expression. Both hypermethylation and silencing of tumour suppressor genes as well as hypomethylation and activation of prometastatic genes are characteristic of cancer cells. As DNA methylation is reversible, DNA methylation inhibitors were tested as anticancer drugs with the idea that such agents would demethylate and reactivate tumour suppressor genes. Two cytosine analogues, 5-azacytidine (Vidaza) and 5-aza-2'-deoxycytidine, were approved by the Food and Drug Administration as antitumour agents in 2004 and 2006 respectively. However, these agents might cause activation of a panel of prometastatic genes in addition to activating tumour suppressor genes, which might lead to increased metastasis. This poses the challenge of how to target tumour suppressor genes and block cancer growth with DNA-demethylating drugs while avoiding the activation of prometastatic genes and precluding the morbidity of cancer metastasis. This paper reviews current progress in using DNA methylation inhibitors in cancer therapy and the potential promise and challenges ahead.
Collapse
Affiliation(s)
- David Cheishvili
- Department of Pharmacology and Therapeutics, McGill University Medical School, Montreal, QC, Canada
| | - Lisa Boureau
- Department of Pharmacology and Therapeutics, McGill University Medical School, Montreal, QC, Canada.,Department of Physiology Medical Sciences, University of Toronto 1 King's College Circle Toronto, ON, Canada
| | - Moshe Szyf
- Department of Pharmacology and Therapeutics, McGill University Medical School, Montreal, QC, Canada.,Sackler Program for Epigenetics and Developmental Psychobiology, McGill University Medical School, Montreal, QC, Canada.,Canadian Institute for Advanced Research, Faculty of Medicine, University of Toronto 1 King's College Circle Toronto, ON, Canada
| |
Collapse
|
7
|
Wellhauser L, Gojska NM, Belsham DD. Delineating the regulation of energy homeostasis using hypothalamic cell models. Front Neuroendocrinol 2015; 36:130-49. [PMID: 25223866 DOI: 10.1016/j.yfrne.2014.09.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Revised: 08/28/2014] [Accepted: 09/02/2014] [Indexed: 12/27/2022]
Abstract
Attesting to its intimate peripheral connections, hypothalamic neurons integrate nutritional and hormonal cues to effectively manage energy homeostasis according to the overall status of the system. Extensive progress in the identification of essential transcriptional and post-translational mechanisms regulating the controlled expression and actions of hypothalamic neuropeptides has been identified through the use of animal and cell models. This review will introduce the basic techniques of hypothalamic investigation both in vivo and in vitro and will briefly highlight the key advantages and challenges of their use. Further emphasis will be place on the use of immortalized models of hypothalamic neurons for in vitro study of feeding regulation, with a particular focus on cell lines proving themselves most fruitful in deciphering fundamental basics of NPY/AgRP, Proglucagon, and POMC neuropeptide function.
Collapse
Affiliation(s)
- Leigh Wellhauser
- Department of Physiology, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Nicole M Gojska
- Department of Physiology, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Denise D Belsham
- Departments of Physiology, Medicine and OB/GYN, University of Toronto, Toronto, Ontario M5G 1A8, Canada; Division of Cellular and Molecular Biology, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5S 1A8, Canada.
| |
Collapse
|
8
|
Alvarado S, Fernald RD, Storey KB, Szyf M. The dynamic nature of DNA methylation: a role in response to social and seasonal variation. Integr Comp Biol 2014; 54:68-76. [PMID: 24813708 DOI: 10.1093/icb/icu034] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
An organism's ability to adapt to its environment depends on its ability to regulate and maintain tissue specific, temporal patterns of gene transcription in response to specific environmental cues. Epigenetic mechanisms are responsible for many of the intricacies of a gene's regulation that alter expression patterns without affecting the genetic sequence. In particular, DNA methylation has been shown to have an important role in regulating early development and in some human diseases. Within these domains, DNA methylation has been extensively characterized over the past 60 years, but the discovery of its role in regulating behavioral outcomes has led to renewed interest in its potential roles in animal behavior and phenotypic plasticity. The conservation of DNA methylation across the animal kingdom suggests a possible role in the plasticity of genomic responses to environmental cues in natural environments. Here, we review the historical context for the study of DNA methylation, its function and mechanisms, and provide examples of gene/environment interactions in response to social and seasonal cues. Finally, we discuss useful tools to interrogate and dissect the function of DNA methylation in non-model organisms.
Collapse
Affiliation(s)
- Sebastian Alvarado
- *Stanford University, Gilbert Biology #314, 371 Serra Mall, Palo Alto CA 940305; Carleton University, Steacie Building #507, 1125 Colonel By Ottawa Ontario, K1S5B6; McGill University, McIntyre Medical Building #1309, 3655 Sir William Osler,Montreal, Quebec H3G1Y6
| | - Russell D Fernald
- *Stanford University, Gilbert Biology #314, 371 Serra Mall, Palo Alto CA 940305; Carleton University, Steacie Building #507, 1125 Colonel By Ottawa Ontario, K1S5B6; McGill University, McIntyre Medical Building #1309, 3655 Sir William Osler,Montreal, Quebec H3G1Y6
| | - Kenneth B Storey
- *Stanford University, Gilbert Biology #314, 371 Serra Mall, Palo Alto CA 940305; Carleton University, Steacie Building #507, 1125 Colonel By Ottawa Ontario, K1S5B6; McGill University, McIntyre Medical Building #1309, 3655 Sir William Osler,Montreal, Quebec H3G1Y6
| | - Moshe Szyf
- *Stanford University, Gilbert Biology #314, 371 Serra Mall, Palo Alto CA 940305; Carleton University, Steacie Building #507, 1125 Colonel By Ottawa Ontario, K1S5B6; McGill University, McIntyre Medical Building #1309, 3655 Sir William Osler,Montreal, Quebec H3G1Y6*Stanford University, Gilbert Biology #314, 371 Serra Mall, Palo Alto CA 940305; Carleton University, Steacie Building #507, 1125 Colonel By Ottawa Ontario, K1S5B6; McGill University, McIntyre Medical Building #1309, 3655 Sir William Osler,Montreal, Quebec H3G1Y6
| |
Collapse
|
9
|
Stefanska B, Cheishvili D, Suderman M, Arakelian A, Huang J, Hallett M, Han ZG, Al-Mahtab M, Akbar SMF, Khan WA, Raqib R, Tanvir I, Khan HA, Rabbani SA, Szyf M. Genome-wide study of hypomethylated and induced genes in patients with liver cancer unravels novel anticancer targets. Clin Cancer Res 2014; 20:3118-32. [PMID: 24763612 DOI: 10.1158/1078-0432.ccr-13-0283] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We utilized whole-genome mapping of promoters that are activated by DNA hypomethylation in hepatocellular carcinoma (HCC) clinical samples to shortlist novel targets for anticancer therapeutics. We provide a proof of principle of this approach by testing six genes short-listed in our screen for their essential role in cancer growth and invasiveness. EXPERIMENTAL DESIGN We used siRNA- or shRNA-mediated depletion to determine whether inhibition of these genes would reduce human tumor xenograft growth in mice as well as cell viability, anchorage-independent growth, invasive capacities, and state of activity of nodal signaling pathways in liver, breast, and bladder cancer cell lines. RESULTS Depletion of EXOSC4, RNMT, SENP6, WBSCR22, RASAL2, and NENF effectively and specifically inhibits cancer cell growth and cell invasive capacities in different types of cancer, but, remarkably, there is no effect on normal cell growth, suggesting a ubiquitous causal role for these genes in driving cancer growth and metastasis. Depletion of RASAL2 and NENF in vitro reduces their growth as explants in vivo in mice. RASAL2 and NENF depletion interferes with AKT, WNT, and MAPK signaling pathways as well as regulation of epigenetic proteins that were previously demonstrated to drive cancer growth and metastasis. CONCLUSION Our results prove that genes that are hypomethylated and induced in tumors are candidate targets for anticancer therapeutics in multiple cancer cell types. Because these genes are particularly activated in cancer, they constitute a group of targets for specific pharmacologic inhibitors of cancer and cancer metastasis. Clin Cancer Res; 20(12); 3118-32. ©2014 AACR.
Collapse
Affiliation(s)
- Barbara Stefanska
- Authors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, PakistanAuthors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, Pakistan
| | - David Cheishvili
- Authors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, Pakistan
| | - Matthew Suderman
- Authors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, Pakistan
| | - Ani Arakelian
- Authors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, Pakistan
| | - Jian Huang
- Authors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, Pakistan
| | - Michael Hallett
- Authors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, Pakistan
| | - Ze-Guang Han
- Authors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, Pakistan
| | - Mamun Al-Mahtab
- Authors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, Pakistan
| | - Sheikh Mohammad Fazle Akbar
- Authors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, Pakistan
| | - Wasif Ali Khan
- Authors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, Pakistan
| | - Rubhana Raqib
- Authors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, Pakistan
| | - Imrana Tanvir
- Authors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, Pakistan
| | - Haseeb Ahmed Khan
- Authors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, Pakistan
| | - Shafaat A Rabbani
- Authors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, Pakistan
| | - Moshe Szyf
- Authors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, PakistanAuthors' Affiliations: Departments of Pharmacology and Therapeutics and Medicine, McGill University Health Centre, Montreal; McGill Centre for Bioinformatics; and Sackler program for Psychobiology and Epigenetics at McGill University, Montreal, Quebec, Canada; Department of Nutrition Science, Purdue University, West Lafayette, Indiana; Shanghai-MOST Key Laboratory for Disease and Health Genomics, Chinese National Human Genome Center at Shanghai, Shanghai, China; Department of Hepatology, Bangabandhu Sheikh Mujib Medical University; International Centre for Diarrhoeal Diseases Research, Bangladesh (icddr,b), Dhaka, Dhaka District, Bangladesh; Department of Medical Sciences, Toshiba General Hospital, Tokyo, Kanto, Japan; and Department of Pathology, Fatima Memorial Hospital College of Medicine and Dentistry Lahore, Pakistan
| |
Collapse
|
10
|
Fagan RL, Wu M, Chédin F, Brenner C. An ultrasensitive high throughput screen for DNA methyltransferase 1-targeted molecular probes. PLoS One 2013; 8:e78752. [PMID: 24236046 PMCID: PMC3827244 DOI: 10.1371/journal.pone.0078752] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 09/20/2013] [Indexed: 12/20/2022] Open
Abstract
DNA methyltransferase 1 (DNMT1) is the enzyme most responsible for epigenetic modification of human DNA and the intended target of approved cancer drugs such as 5-aza-cytidine and 5-aza-2'-deoxycytidine. 5-aza nucleosides have complex mechanisms of action that require incorporation into DNA, and covalent trapping and proteolysis of DNMT isozymes. Direct DNMT inhibitors are needed to refine understanding of the role of specific DNMT isozymes in cancer etiology and, potentially, to improve cancer prevention and treatment. Here, we developed a high throughput pipeline for identification of direct DNMT1 inhibitors. The components of this screen include an activated form of DNMT1, a restriction enzyme-coupled fluorigenic assay performed in 384 well plates with a z-factor of 0.66, a counter screen against the restriction enzyme, a screen to eliminate DNA intercalators, and a differential scanning fluorimetry assay to validate direct binders. Using the Microsource Spectrum collection of 2320 compounds, this screen identified nine compounds with dose responses ranging from 300 nM to 11 µM, representing at least two different pharmacophores with DNMT1 inhibitory activity. Seven of nine inhibitors identified exhibited two to four-fold selectivity for DNMT1 versus DNMT3A.
Collapse
Affiliation(s)
- Rebecca L. Fagan
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Meng Wu
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Frédéric Chédin
- Department of Molecular and Cellular Biology and Genome Center, University of California Davis, Davis, California, United States of America
| | - Charles Brenner
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
11
|
Zhou R, Chen F, Chang F, Bai Y, Chen L. Persistent overexpression of DNA methyltransferase 1 attenuating GABAergic inhibition in basolateral amygdala accounts for anxiety in rat offspring exposed perinatally to low-dose bisphenol A. J Psychiatr Res 2013; 47:1535-44. [PMID: 23791455 DOI: 10.1016/j.jpsychires.2013.05.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 05/03/2013] [Accepted: 05/15/2013] [Indexed: 12/18/2022]
Abstract
Substantial evidence indicates that predisposition to diseases can be acquired during early stages of development and interactions between environmental and genetic factors may be implicated in the onset of many pathological conditions. We have shown that perinatal exposure to bisphenol A (BPA) at environmental dose level causes long-term anxiety-like behaviors in rats. The aim of this study was to examine epigenetic reprogramming effect of BPA on anxiety-related neurobehavior in the rat offspring. The results of real-time RT-PCR displayed that the overexpression of DNA methyltransferase 1 (DNMT1) mRNA was accompanied by the reduction of glutamic acid decarboxylase 67 (GAD67) mRNA level in the basolateral amygdala (BLA) of postnatal day 45 BPA-exposed female rats. Chronic intro-BLA injection with 5-ada-CdR could rectify the GAD67 mRNA expression. Behavioral data showed that the anxiety-like behaviors in BPA-exposed rats were reversed by intro-BLA treatment with 5-ada-CdR which could be further blocked by PTX. Electrophysiological study revealed behavioral alterations were associated with the increase of postsynaptic neuronal excitability in the cortical-BLA pathway which appeared as multispike responses, paired-pulse facilitation instead of paired-pulse inhibition and long-term potentiation and 5-aza-CdR treatment restored the increased synaptic transmission in the BLA via improving GABAergic system. The above results suggest that the overexpression of DNMT1 in the BLA is responsible for the etiology of anxiety associated with BPA exposure via GABAergic disinhibition. In addition, we also find these long-term neurobehavioral effects of developmental BPA exposure are reversible in adolescent period.
Collapse
Affiliation(s)
- Rong Zhou
- Department of Physiology, Nanjing Medical University, Hanzhong Road 140, Nanjing, Jiangsu, China
| | | | | | | | | |
Collapse
|
12
|
Stefanska B, Suderman M, Machnes Z, Bhattacharyya B, Hallett M, Szyf M. Transcription onset of genes critical in liver carcinogenesis is epigenetically regulated by methylated DNA-binding protein MBD2. Carcinogenesis 2013; 34:2738-49. [PMID: 23955541 DOI: 10.1093/carcin/bgt273] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We previously delineated genes whose promoters are hypomethylated and induced in hepatocellular carcinoma (HCC) patients. The purpose of this study was to establish the players that regulate these genes in liver cancer cells. We performed chromatin immunoprecipitation with methyl-CpG-binding domain protein 2 (MBD2), RNA polymerase II (RNA pol II), CCAAT/enhancer-binding protein alpha (CEBPA) antibodies and methylated DNA immunoprecipitation in HepG2 liver cancer cells treated with scrambled small interfering RNA (siRNA) and siRNA to MBD2 or CEBPA. We then hybridized DNA to microarrays spanning the entire coding sequences, introns and regulatory regions of several hundred HCC-hypomethylated genes. These analyses reveal that MBD2 binds a significant fraction of the hypomethylated genes, determines RNA pol II binding and DNA methylation state. MBD2 binding can result in promoter activation and hypomethylation or in repression. In activated target genes, MBD2 colocalizes with the transcription factor CEBPA, and MBD2 binding at these positions is reduced upon CEBPA depletion. Significant fraction of MBD2 effects on DNA methylation and transcription appears to be indirect since changes occur upon MBD2 depletion in genes where no MBD2 binding was detected. Our study delineates the rules governing the interaction of MBD2 with its targets and the consequences to RNA pol II binding and DNA methylation states. This has important implications for understanding the role of DNA methylation in cancer and targeting DNA methylation proteins in cancer therapy.
Collapse
Affiliation(s)
- Barbara Stefanska
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | | | | | | | | | | |
Collapse
|
13
|
Abstract
SLC5A8 is a putative tumor suppressor that is inactivated in more than 10 different types of cancer, but neither the oncogenic signaling responsible for SLC5A8 inactivation nor the functional relevance of SLC5A8 loss to tumor growth has been elucidated. Here, we identify oncogenic HRAS (HRAS(G12V)) as a potent mediator of SLC5A8 silencing in human nontransformed normal mammary epithelial cell lines and in mouse mammary tumors through DNMT1. Further, we demonstrate that loss of Slc5a8 increases cancer-initiating stem cell formation and promotes mammary tumorigenesis and lung metastasis in an HRAS-driven murine model of mammary tumors. Mammary-gland-specific overexpression of Slc5a8 (mouse mammary tumor virus-Slc5a8 transgenic mice), as well as induction of endogenous Slc5a8 in mice with inhibitors of DNA methylation, protects against HRAS-driven mammary tumors. Collectively, our results provide the tumor-suppressive role of SLC5A8 and identify the oncogenic HRAS as a mediator of tumor-associated silencing of this tumor suppressor in mammary glands. These findings suggest that pharmacological approaches to reactivate SLC5A8 expression in tumor cells have potential as a novel therapeutic strategy for breast cancer treatment.
Collapse
|
14
|
Fagan RL, Cryderman DE, Kopelovich L, Wallrath LL, Brenner C. Laccaic acid A is a direct, DNA-competitive inhibitor of DNA methyltransferase 1. J Biol Chem 2013; 288:23858-67. [PMID: 23839987 DOI: 10.1074/jbc.m113.480517] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Methylation of cytosines in CpG dinucleotides is the predominant epigenetic mark on vertebrate DNA. DNA methylation is associated with transcriptional repression. The pattern of DNA methylation changes during development and with disease. Human DNA methyltransferase 1 (Dnmt1), a 1616-amino acid multidomain enzyme, is essential for maintenance of DNA methylation in proliferating cells and is considered an important cancer drug target. Using a fluorogenic, endonuclease-coupled DNA methylation assay with an activated form of Dnmt1 engineered to lack the replication foci targeting sequence domain, we discovered that laccaic acid A (LCA), a highly substituted anthraquinone natural product, is a direct inhibitor with a 310 nm Ki. LCA is competitive with the DNA substrate in in vitro methylation assays and alters the expression of methylated genes in MCF-7 breast cancer cells synergistically with 5-aza-2'-deoxycytidine. LCA represents a novel class of Dnmt-targeted molecular probes, with biochemical properties that allow it to distinguish between non DNA-bound and DNA-bound Dnmt1.
Collapse
Affiliation(s)
- Rebecca L Fagan
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | |
Collapse
|
15
|
Amodio N, Leotta M, Bellizzi D, Di Martino MT, D'Aquila P, Lionetti M, Fabiani F, Leone E, Gullà AM, Passarino G, Caraglia M, Negrini M, Neri A, Giordano A, Tagliaferri P, Tassone P. DNA-demethylating and anti-tumor activity of synthetic miR-29b mimics in multiple myeloma. Oncotarget 2012; 3:1246-58. [PMID: 23100393 PMCID: PMC3717964 DOI: 10.18632/oncotarget.675] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Accepted: 10/19/2012] [Indexed: 12/31/2022] Open
Abstract
Aberrant DNA methylation plays a relevant role in multiple myeloma (MM) pathogenesis. MicroRNAs (miRNAs) are a class of small non-coding RNAs that recently emerged as master regulator of gene expression by targeting protein-coding mRNAs. However, miRNAs involvement in the regulation of the epigenetic machinery and their potential use as therapeutics in MM remain to be investigated. Here, we provide evidence that the expression of de novo DNA methyltransferases (DNMTs) is deregulated in MM cells. Moreover, we show that miR-29b targets DNMT3A and DNMT3B mRNAs and reduces global DNA methylation in MM cells. In vitro transfection of MM cells with synthetic miR-29b mimics significantly impairs cell cycle progression and also potentiates the growth-inhibitory effects induced by the demethylating agent 5-azacitidine. Most importantly, in vivo intratumor or systemic delivery of synthetic miR-29b mimics, in two clinically relevant murine models of human MM, including the SCID-synth-hu system, induces significant anti-tumor effects. All together, our findings demonstrate that aberrant DNMTs expression is efficiently modulated by tumor suppressive synthetic miR-29b mimics, indicating that methyloma modulation is a novel matter of investigation in miRNA-based therapy of MM.
Collapse
MESH Headings
- Animals
- Antimetabolites, Antineoplastic/pharmacology
- Azacitidine/pharmacology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Biomimetics
- Blotting, Western
- Bone Marrow/metabolism
- Bone Marrow/pathology
- Case-Control Studies
- Cell Cycle
- Cell Proliferation
- Cellular Microenvironment/drug effects
- DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors
- DNA (Cytosine-5-)-Methyltransferases/genetics
- DNA (Cytosine-5-)-Methyltransferases/metabolism
- DNA Methylation
- DNA Methyltransferase 3A
- Gene Expression Profiling
- Humans
- Immunoenzyme Techniques
- Leukemia, Plasma Cell/genetics
- Leukemia, Plasma Cell/pathology
- Leukemia, Plasma Cell/prevention & control
- Male
- Mice
- Mice, SCID
- MicroRNAs/chemical synthesis
- MicroRNAs/genetics
- Multiple Myeloma/genetics
- Multiple Myeloma/pathology
- Multiple Myeloma/prevention & control
- Oligonucleotide Array Sequence Analysis
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Cells, Cultured
- DNA Methyltransferase 3B
Collapse
Affiliation(s)
- Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University
and Medical Oncology Unit, T. Campanella Cancer Center, Salvatore Venuta University Campus,
Catanzaro, Italy
| | - Marzia Leotta
- Department of Experimental and Clinical Medicine, Magna Graecia University
and Medical Oncology Unit, T. Campanella Cancer Center, Salvatore Venuta University Campus,
Catanzaro, Italy
| | - Dina Bellizzi
- Department of Cell Biology, University of Calabria, Cosenza
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University
and Medical Oncology Unit, T. Campanella Cancer Center, Salvatore Venuta University Campus,
Catanzaro, Italy
| | | | - Marta Lionetti
- Department of Medical Sciences University of Milan, Hematology 1, IRCCS
Policlinico Foundation, Milan, Italy
| | - Fernanda Fabiani
- Department of Experimental and Clinical Medicine, Magna Graecia University
and Medical Oncology Unit, T. Campanella Cancer Center, Salvatore Venuta University Campus,
Catanzaro, Italy
| | - Emanuela Leone
- Department of Experimental and Clinical Medicine, Magna Graecia University
and Medical Oncology Unit, T. Campanella Cancer Center, Salvatore Venuta University Campus,
Catanzaro, Italy
| | - Anna Maria Gullà
- Department of Experimental and Clinical Medicine, Magna Graecia University
and Medical Oncology Unit, T. Campanella Cancer Center, Salvatore Venuta University Campus,
Catanzaro, Italy
| | | | - Michele Caraglia
- Department of Biochemistry and Biophysics, Second University of Naples,
Naples, Italy
| | - Massimo Negrini
- Department of Experimental Medicine and Diagnostics, University of
Ferrara
| | - Antonino Neri
- Department of Medical Sciences University of Milan, Hematology 1, IRCCS
Policlinico Foundation, Milan, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for
Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University
and Medical Oncology Unit, T. Campanella Cancer Center, Salvatore Venuta University Campus,
Catanzaro, Italy
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University
and Medical Oncology Unit, T. Campanella Cancer Center, Salvatore Venuta University Campus,
Catanzaro, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for
Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| |
Collapse
|
16
|
Lafontaine J, Rodier F, Ouellet V, Mes-Masson AM. Necdin, a p53-target gene, is an inhibitor of p53-mediated growth arrest. PLoS One 2012; 7:e31916. [PMID: 22355404 PMCID: PMC3280226 DOI: 10.1371/journal.pone.0031916] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 01/20/2012] [Indexed: 01/09/2023] Open
Abstract
In vitro, cellular immortalization and transformation define a model for multistep carcinogenesis and current ongoing challenges include the identification of specific molecular events associated with steps along this oncogenic pathway. Here, using NIH3T3 cells, we identified transcriptionally related events associated with the expression of Polyomavirus Large-T antigen (PyLT), a potent viral oncogene. We propose that a subset of these alterations in gene expression may be related to the early events that contribute to carcinogenesis. The proposed tumor suppressor Necdin, known to be regulated by p53, was within a group of genes that was consistently upregulated in the presence of PyLT. While Necdin is induced following p53 activation with different genotoxic stresses, Necdin induction by PyLT did not involve p53 activation or the Rb-binding site of PyLT. Necdin depletion by shRNA conferred a proliferative advantage to NIH3T3 and PyLT-expressing NIH3T3 (NIHLT) cells. In contrast, our results demonstrate that although overexpression of Necdin induced a growth arrest in NIH3T3 and NIHLT cells, a growing population rapidly emerged from these arrested cells. This population no longer showed significant proliferation defects despite high Necdin expression. Moreover, we established that Necdin is a negative regulator of p53-mediated growth arrest induced by nutlin-3, suggesting that Necdin upregulation could contribute to the bypass of a p53-response in p53 wild type tumors. To support this, we characterized Necdin expression in low malignant potential ovarian cancer (LMP) where p53 mutations rarely occur. Elevated levels of Necdin expression were observed in LMP when compared to aggressive serous ovarian cancers. We propose that in some contexts, the constitutive expression of Necdin could contribute to cancer promotion by delaying appropriate p53 responses and potentially promote genomic instability.
Collapse
Affiliation(s)
- Julie Lafontaine
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montréal, Québec, Canada
| | - Francis Rodier
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montréal, Québec, Canada
- Département de radiologie, radio-oncologie et médecine nucléaire, Université de Montréal, Montréal, Québec, Canada
| | - Véronique Ouellet
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montréal, Québec, Canada
| | - Anne-Marie Mes-Masson
- Centre de recherche du Centre hospitalier de l'Université de Montréal and Institut du cancer de Montréal, Montréal, Québec, Canada
- Département de médecine, Université de Montréal, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
17
|
He S, Wang F, Yang L, Guo C, Wan R, Ke A, Xu L, Hu G, Xu X, Shen J, Wang X. Expression of DNMT1 and DNMT3a are regulated by GLI1 in human pancreatic cancer. PLoS One 2011; 6:e27684. [PMID: 22110720 PMCID: PMC3215738 DOI: 10.1371/journal.pone.0027684] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 10/21/2011] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND AIMS GLI1, as an indispensable transcriptional factor of Hedgehog signaling pathway, plays an important role in the development of pancreatic cancer (PC). DNA methyltransferases (DNMTs) mediate the methylation of quantity of tumor-related genes. Our study aimed to explore the relationship between GLI1 and DNMTs. METHODS Expressions of GLI1 and DNMTs were detected in tumor and adjacent normal tissues of PC patients by immunohistochemistry (IHC). PANC-1 cells were treated by cyclopamine and GLI1-siRNA, while BxPC-3 cells were transfected with overexpression-GLI1 lentiviral vector. Then GLI1 and DNMTs expression were analyzed by qRT-PCR and western blot (WB). Then we took chromatin immunoprecipitation (ChIP) to demonstrate GLI1 bind to DNMT1. Finally, nested MSP was taken to valuate the methylation levels of APC and hMLH1, when GLI1 expression altered. RESULTS IHC result suggested the expressions of GLI1, DNMT1 and DNMT3a in PC tissues were all higher than those in adjacent normal tissues (p<0.05). After GLI1 expression repressed by cyclopamine in mRNA and protein level (down-regulation 88.1±2.2%, 86.4±2.2%, respectively), DNMT1 and DNMT3a mRNA and protein level decreased by 91.6%±2.2% and 83.8±4.8%, 87.4±2.7% and 84.4±1.3%, respectively. When further knocked down the expression of GLI1 by siRNA (mRNA decreased by 88.6±2.1%, protein decreased by 63.5±4.5%), DNMT1 and DNMT3a mRNA decreased by 80.9±2.3% and 78.6±3.8% and protein decreased by 64.8±2.8% and 67.5±5.6%, respectively. Over-expression of GLI1 by GLI1 gene transfection (mRNA increased by 655.5±85.9%, and protein increased by 272.3±14.4%.), DNMT1 and DNMT3a mRNA and protein increased by 293.0±14.8% and 578.3±58.5%, 143.5±17.4% and 214.0±18.9%, respectively. ChIP assays showed GLI1 protein bound to DNMT1 but not to DNMT3a. Results of nested MSP demonstrated GLI1 expression affected the DNA methylation level of APC but not hMLH1 in PC. CONCLUSION DNMT1 and DNMT3a are regulated by GLI1 in PC, and DNMT1 is its direct target gene.
Collapse
Affiliation(s)
- ShanShan He
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Feng Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - LiJuan Yang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, Shanghai, People's Republic of China
- Department of Gastroenterology, The First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - ChuanYong Guo
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Rong Wan
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - AiWu Ke
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Ling Xu
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - GuoYong Hu
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - XuanFu Xu
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - Jie Shen
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, Shanghai, People's Republic of China
| | - XingPeng Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University, Shanghai, People's Republic of China
- Department of Gastroenterology, The First People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
18
|
Chik F, Szyf M. Effects of specific DNMT gene depletion on cancer cell transformation and breast cancer cell invasion; toward selective DNMT inhibitors. Carcinogenesis 2011; 32:224-32. [PMID: 20980350 DOI: 10.1093/carcin/bgq221] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023] Open
Abstract
A hallmark of cancer is aberrant DNA methylation, consisting of global hypomethylation and regional hypermethylation of tumor suppressor genes. DNA methyltransferase inhibitors have been recognized as promising candidate anticancer drugs. Drug development has focused on DNA methylation inhibitors with the goal of activating tumor suppressor genes silenced by DNA methylation. 5-azacytidine (5-AC; Vidaza), a global DNA methyltransferase inhibitor, was Food and Drug Administration approved to treat myelodysplastic syndromes and is clinically tested for solid tumors. In this paper, it was demonstrated that 5'-aza-2'-deoxycytidine (5-azaCdR) activated both silenced tumor suppressor genes and pro-metastatic genes by demethylation, raising the concern that it would promote metastasis. 5-AzaCdR treatment increased the invasiveness of non-invasive breast cancer cell lines MCF-7 cells and ZR-75-1 and dramatically induced pro-metastatic genes; Urokinase plasminogen activator (uPA), matrix metalloproteinase 2 (MMP2), metastasis-associated gene (H-MTS1; S100A4) and C-X-C chemokine receptor 4 (CXCR4). The hypothesis that the blocking of cellular transformation activity of DNA methyltransferase inhibitor could be separated from the pro-metastatic activity was tested using short interfering RNA (siRNA) targeted to the different DNA methyltransferase (DNMT) genes. Although depletion of DNMT1 had the strongest effect on colony growth suppression in cellular transformation assays, it did not result in demethylation and activation of uPA, S100A4, MMP2 and CXCR4 in MCF-7 cells. Depletion of DNMT1 did not induce cellular invasion in MCF-7 and ZR-75-1 non-invasive breast cancer cell lines. These data have implications on the design of new DNA methyltransferase inhibitor and on the proper utilization of current inhibitors.
Collapse
Affiliation(s)
- Flora Chik
- Department of Pharmacology and Therapeutics, McGill University Medical School, Montreal, Quebec, Canada
| | | |
Collapse
|
19
|
Mansuy V, Geller S, Rey JP, Campagne C, Boccard J, Poulain P, Prevot V, Pralong FP. Phenotypic and molecular characterization of proliferating and differentiated GnRH-expressing GnV-3 cells. Mol Cell Endocrinol 2011; 332:97-105. [PMID: 20937356 DOI: 10.1016/j.mce.2010.10.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2010] [Revised: 09/14/2010] [Accepted: 10/02/2010] [Indexed: 12/27/2022]
Abstract
GnRH neurons provide the primary driving force upon the neuroendocrine reproductive axis. Here we used GnV-3 cells, a model of conditionally immortalized GnRH-expressing neurons, to perform an analysis of cell cycle and compare the gene expression profile of proliferating cells with differentiated cells. In the proliferation medium, 45 ± 1.5% of GnV-3 cells are in S-phase by FACS analysis. In the differentiation medium, only 9 ± 0.9% of them are in S-phase, and they acquire the characteristic bipolar shape displayed by preoptic GnRH neurons in vivo. In addition, GnV-3 cells in the differentiated state exhibit electrophysiological properties characteristic of neurons. Transcriptomic analysis identified up-regulation of 1931 genes and down-regulation of 1270 genes in cells grown in the differentiation medium compared to cells in the proliferation medium. Subsequent gene ontology study indicated that genes over-expressed in proliferating GnV-3 cells were mainly involved in cell cycle regulations, whereas genes over-expressed in differentiated cells were mainly involved in processes of differentiation, neurogenesis and neuronal morphogenesis. Taken together, these data demonstrate the occurrence of morphological and physiological changes in GnV-3 cells between the proliferating and the differentiated state. Moreover, the genes differentially regulated between these two different states are providing novel pathways potentially important for a better understanding of the physiology of mature GnRH neurons.
Collapse
Affiliation(s)
- Virginie Mansuy
- Service of Endocrinology, Diabetology and Metabolism, University Hospital and Faculty of Biology and Medicine, 1011 Lausanne, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Effect of maternal folic acid supplementation on hepatic one-carbon unit associated gene expressions in newborn piglets. Mol Biol Rep 2010; 38:3849-56. [DOI: 10.1007/s11033-010-0500-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2010] [Accepted: 11/13/2010] [Indexed: 01/21/2023]
|
21
|
Li W, Sun W, Liu L, Yang F, Li Y, Chen Y, Fang J, Zhang W, Wu J, Zhu Y. IL-32: a host proinflammatory factor against influenza viral replication is upregulated by aberrant epigenetic modifications during influenza A virus infection. THE JOURNAL OF IMMUNOLOGY 2010; 185:5056-65. [PMID: 20889550 DOI: 10.4049/jimmunol.0902667] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Our previous studies with clinical data analysis have shown that the proinflammatory factor IL-32 is activated in response to influenza virus infection. However, little is known about how influenza virus induces IL-32 production, and the role of IL-32 in the host immune responses during viral infection remains unclear. In this study, we show that IL-32 production is stimulated by influenza A virus or dsRNA in human PBMCs from healthy volunteers. We demonstrate that the NF-κB and CREB pathways play key roles in the activation of IL-32 production in response to influenza virus infection in A549 human lung epithelial cells. We then show that aberrant epigenetic modifications in the IL32 promoter are important in the transcriptional regulation of IL-32 expression. Interestingly, one CpG demethylation within the CREB binding site increases the binding of CREB to the promoter, which is followed by IL32 transcriptional activation in influenza A virus-infected cells. Overexpression assays combined with RNA interference show that DNA methyltransferases DNMT1 and DNMT3b are critical for IL32 promoter methylation and gene silencing before viral infection. We have demonstrated the anti-influenza virus function of IL-32. Assays for each of the six IL-32 isoforms (α, β, γ, δ, ε, and ζ) during influenza virus infection indicated that all the isoforms have antiviral activity, with different inhibitory rates, and that the effect of IL-32γ is strongest. Our results indicate that the elevated IL-32 levels triggered by influenza virus infection in turn hamper viral replication.
Collapse
Affiliation(s)
- Wei Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Paschos K, Allday MJ. Epigenetic reprogramming of host genes in viral and microbial pathogenesis. Trends Microbiol 2010; 18:439-47. [PMID: 20724161 PMCID: PMC3089700 DOI: 10.1016/j.tim.2010.07.003] [Citation(s) in RCA: 158] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Revised: 07/12/2010] [Accepted: 07/14/2010] [Indexed: 12/12/2022]
Abstract
One of the key questions in the study of mammalian gene regulation is how epigenetic methylation patterns on histones and DNA are initiated and established. These stable, heritable, covalent modifications are largely associated with the repression or silencing of gene transcription, and when deregulated can be involved in the development of human diseases such as cancer. This article reviews examples of viruses and bacteria known or thought to induce epigenetic changes in host cells, and how this might contribute to disease. The heritable nature of these processes in gene regulation suggests that they could play important roles in chronic diseases associated with microbial persistence; they might also explain so-called ‘hit-and-run’ phenomena in infectious disease pathogenesis.
Collapse
|
23
|
Rabbani SA, Valentino ML, Arakelian A, Ali S, Boschelli F. SKI-606 (Bosutinib) blocks prostate cancer invasion, growth, and metastasis in vitro and in vivo through regulation of genes involved in cancer growth and skeletal metastasis. Mol Cancer Ther 2010; 9:1147-57. [PMID: 20423991 DOI: 10.1158/1535-7163.mct-09-0962] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the current study, we have examined the efficacy of a Src/Abl kinase inhibitor SKI-606 (Bosutinib) for its effect on prostate cancer growth and skeletal metastasis. Treatment of highly invasive human prostate cancer cells PC-3 and DU-145 with different doses of SKI-606 decreased Src activation, cell proliferation, migration, and invasion as determined by Matrigel Boyden chamber invasion assay. For in vivo studies, PC-3 cells were inoculated through s.c. or i.t. route into male BALB/c nu/nu or Fox Chase severe combined immunodeficient mice, respectively. Experimental animals treated with SKI-606 developed tumors of a significantly smaller volume and a significant decrease (50%) in experimental skeletal lesion area. A marked increase (32%) in bone volume to tumor volume ratio was also seen by micro-computed tomography analysis of tibias from control and experimental groups of animals. Western blot analysis showed the ability of SKI-606 to significantly decrease the phosphorylation of signaling molecules (AKT, mitogen-activated protein kinase, focal adhesion kinase) and the expression of tumor progression-associated genes uPAR, MMP-2, MMP-9, N-cadherin, fibronectin, BMP-2 (bone morphogenetic protein 2), BMP-6 (bone morphogenetic protein 6), IL-8 (interleukin 8), and TGF-beta (transforming growth factor beta) in prostate cancer cells. SKI-606 is currently in clinical trials for breast cancer and chronic myelogenous leukemia. Results from these studies provide convincing evidence for evaluating its efficacy in prostate cancer patients.
Collapse
Affiliation(s)
- Shafaat A Rabbani
- Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada.
| | | | | | | | | |
Collapse
|
24
|
Abstract
Evidence is emerging that several diseases and behavioral pathologies result from defects in gene function. The best-studied example is cancer, but other diseases such as autoimmune disease, asthma, type 2 diabetes, metabolic disorders, and autism display aberrant gene expression. Gene function may be altered by either a change in the sequence of the DNA or a change in epigenetic programming of a gene in the absence of a sequence change. With epigenetic drugs, it is possible to reverse aberrant gene expression profiles associated with different disease states. Several epigenetic drugs targeting DNA methylation and histone deacetylation enzymes have been tested in clinical trials. Understanding the epigenetic machinery and the differential roles of its components in specific disease states is essential for developing targeted epigenetic therapy.
Collapse
Affiliation(s)
- Moshe Szyf
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Quebec H3G 1Y6, Canada.
| |
Collapse
|
25
|
Zheng DL, Zhang L, Cheng N, Xu X, Deng Q, Teng XM, Wang KS, Zhang X, Huang J, Han ZG. Epigenetic modification induced by hepatitis B virus X protein via interaction with de novo DNA methyltransferase DNMT3A. J Hepatol 2009; 50:377-87. [PMID: 19070387 DOI: 10.1016/j.jhep.2008.10.019] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2008] [Revised: 09/20/2008] [Accepted: 10/09/2008] [Indexed: 02/07/2023]
Abstract
BACKGROUND/AIMS The hepatitis B virus X protein (HBx) has been implicated as a potential trigger of the epigenetic deregulation of some genes, but the underlying mechanisms remain unknown. The aim of this study was to identify underlying mechanisms involved in HBx-mediated epigenetic modification. METHODS Interactions between HBx and DNA methyltransferase (DNMT) or histone deacetylase-1 (HDAC1) were assessed by co-immunoprecipitation. DNA methylation of gene promoters was detected by bisulfite sequencing, and HBx-mediated protein binding to gene regulatory elements was evaluated by chromatin immunoprecipitation. Target gene transcriptional activity was measured by real-time polymerase chain reaction. RESULTS HBx can interact directly with DNMT3A and HDAC1. HBx recruited DNMT3A to the regulatory promoters of interleukin-4 receptor and metallothionein-1F and subsequently silenced their transcription via de novo DNA methylation. By contrast, the transcription of CDH6 and IGFBP3 was triggered by HBx through the deprivation of DNMT3A from their promoters. Transcriptional levels of target genes in hepatocellular carcinoma (HCC) specimens were strongly correlated with the occurrence of HBx. CONCLUSIONS The interaction of HBx and DNMT3A facilitates cellular epigenetic modification (via regional hypermethylation or hypomethylation) at distinct genomic loci, providing an alternative mechanism within HBx-mediated transcriptional regulation, and a profound understanding of hepatitis and HCC pathogenesis.
Collapse
Affiliation(s)
- Da-Li Zheng
- National Human Genome Center of Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Rui-Jin Road II, Shanghai 200025, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wolfe A, Ng Y, Divall SA, Singh SP, Radovick S. Development of an immortalised, post-pubertal gonadotrophin-releasing hormone neuronal cell line. J Neuroendocrinol 2008; 20:1029-37. [PMID: 18624926 PMCID: PMC4888592 DOI: 10.1111/j.1365-2826.2008.01760.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Gonadotrophin-releasing hormone (GnRH) is important in reproduction, although some of the mechanisms for its synthesis and release remain elusive. Progress in understanding the GnRH neurone has been hampered by the limited number and diffuse distribution of the neurone in the mammalian brain. Several stable GnRH-expressing cell lines have been developed using in vivo expression of the simian virus 40 T Antigen (TAg), and they have been helpful for the study of gene expression and neuronal function. However, expression of an immortalising gene may interfere with normal cellular function. We developed a novel GnRH-secreting cell line transgenic mouse model suitable for targeted transformation in post-pubertal mice using a tetracycline-regulated TAg transgene. This clonal cell line, GRT, expresses neuronal markers and GnRH. GRT cells grown in medium containing tetracycline-free serum express increasing mRNA levels of GnRH associated with declining levels of TAg expression. The novelty and ultimately the usefulness of this cell line is that TAg expression, which could affect the GnRH neuronal phenotype, can be regulated by tetracycline.
Collapse
Affiliation(s)
- A Wolfe
- Johns Hopkins University College of Medicine, Department of Pediatrics, Baltimore, MD 21287, USA.
| | | | | | | | | |
Collapse
|
27
|
Xiao Y, Word B, Starlard-Davenport A, Haefele A, Lyn-Cook BD, Hammons G. Age and gender affect DNMT3a and DNMT3b expression in human liver. Cell Biol Toxicol 2008; 24:265-72. [PMID: 17929180 DOI: 10.1007/s10565-007-9035-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2007] [Accepted: 09/03/2007] [Indexed: 12/31/2022]
Abstract
DNA methylation is catalyzed by a family of DNA methyltransferases (DNMTs) including the maintenance enzyme DNMT 1 and de novo methyltransferases DNMT 3a and DNMT 3b. Elevated levels of DNMTs have been found in cancer cells and in several types of human tumors. A polymorphism found in DNMT3b has been associated with increased risk for several cancers. The factors influencing DNMT expression in human tissues have not been clearly determined. he present study examined TDNMT3a and DNMT3b levels in human liver tissue samples and compared the effect of ageing, cigarette smoking, and gender. DNMT3a and DNMT3b expression levels in the samples from older individuals (56-78 years, n = 28) were both significantly higher than those of the younger group (16-48 years, n = 27) (73.2 +/- 3.4 vs 8.3 +/- 2.8 and 56.1 +/- 1.9 vs 17.5 +/- 5.7, respectively; p < 0.05). Levels of DNMT3b in females were significantly higher than those in males (75.4 +/- 2.2 vs 16.3 +/- 4.7; p < 0.05); however, DNMT3a levels were similar for females and males (52.7 +/- 2.7 vs 48.4 +/- 2.0). Expression levels of DNMT3a and DNMT3b were similar in smokers and nonsmokers (58.1 +/- 3.5 vs 60.8 +/- 3.1 and 54.5 +/- 2.3 vs 48.3 +/- 1.8, respectively). Genotyping for DNMT3b (C-->T) variant in this sample pool showed a frequency distribution of CC (41%), CT (50%), and TT (9%). The findings from this study suggest that ageing and gender may be important factors influencing DNA methylation status.
Collapse
Affiliation(s)
- Yongmei Xiao
- Division of Personalized Nutrition and Medicine, National Center for Toxicological Research, Jefferson, AR 72079, USA
| | | | | | | | | | | |
Collapse
|
28
|
CpG island methylator phenotype (CIMP) in cancer: causes and implications. Cancer Lett 2008; 268:177-86. [PMID: 18471961 DOI: 10.1016/j.canlet.2008.03.022] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Revised: 03/19/2008] [Accepted: 03/19/2008] [Indexed: 12/31/2022]
Abstract
Strong evidence exists for a subgroup of tumours, from a variety of tissue types, exhibiting concordant tumour specific DNA methylation: the "CpG island methylator phenotype" (CIMP). Occurrence of CIMP is associated with a range of genetic and environmental factors, although the molecular causes are not well-understood. Both increased expression and aberrant targeting of DNA methyltransferases (DNMTs) could contribute to the occurrence of CIMP. One under-explored area is the possibility that DNA damage may induce or select for CIMP during carcinogenesis or treatment of tumours with chemotherapy. DNA damaging agents can induce DNA damage at guanine rich regions throughout the genome, including CpG islands. This DNA damage can result in stalled DNA synthesis, which will lead to localised increased DNMT1 concentration and therefore potentially increased DNA methylation at these sites. Chemotherapy can select for cells which have increased tolerance to DNA damage due to increased lesion bypass, in some cases by mechanisms which involve inactivation of genes by CpG island methylation. CIMP has been associated with worse patient prognosis, probably due to increased epigenetic plasticity. Therefore, further clinical testing of the diagnostic and prognostic value of the current CIMP markers, as well as increasing our understanding of the molecular causes underlying CIMP are required.
Collapse
|
29
|
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer and the third leading cause of cancer death worldwide. Hepatocarcinogenesis is a multistep process evolving from normal through chronic hepatitis/cirrhosis and dysplastic nodules to HCC. With advances in molecular methods, there is a growing understanding of the molecular mechanisms in hepatocarcinogenesis. Hepatocarcinogenesis is strongly linked to increases in allelic losses, chromosomal changes, gene mutations, epigenetic alterations and alterations in molecular cellular pathways. Some of these alterations are accompanied by a stepwise increase in the different pathological disease stages in hepatocarcinogenesis. Overall, a detailed understanding of the underlying molecular mechanisms involved in the progression of HCC is of fundamental importance to the development of effective prevention and treatment regimes for HCC.
Collapse
Affiliation(s)
- Chun-Ming Wong
- SH Ho Foundation Research Laboratory, Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | | |
Collapse
|
30
|
Szyf M, McGowan P, Meaney MJ. The social environment and the epigenome. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2008; 49:46-60. [PMID: 18095330 DOI: 10.1002/em.20357] [Citation(s) in RCA: 256] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The genome is programmed by the epigenome. Two of the fundamental components of the epigenome are chromatin structure and covalent modification of the DNA molecule itself by methylation. DNA methylation patterns are sculpted during development and it has been a long held belief that they remain stable after birth in somatic tissues. Recent data suggest that DNA methylation is dynamic later in life in postmitotic cells such as neurons and thus potentially responsive to different environmental stimuli throughout life. We hypothesize a mechanism linking the social environment early in life and long-term epigenetic programming of behavior and responsiveness to stress and health status later in life. We will also discuss the prospect that the epigenetic equilibrium remains responsive throughout life and that therefore environmental triggers could play a role in generating interindividual differences in human behavior later in life. We speculate that exposures to different environmental toxins alters long-established epigenetic programs in the brain as well as other tissues leading to late-onset disease.
Collapse
Affiliation(s)
- Moshe Szyf
- Department of Pharmacology and Therapeutics, McGill University, and Department of Psychiatry, Douglas Hospital Research Center, Montréal, Québec, Canada.
| | | | | |
Collapse
|
31
|
Abstract
The epigenome serves as an interface between the dynamic environment and the inherited static genome. The epigenome is comprised of chromatin and a covalent modification of DNA by methylation. The epigenome is sculpted during development to shape the diversity of gene expression programs in the different cell types of the organism by a highly organized process. Epigenetic aberrations have similar consequences to genetic polymorphisms resulting in variations in gene function. Recent data suggest that the epigenome is dynamic and is therefore responsive to environmental signals not only during the critical periods in development but also later in life as well. It is postulated here that not only chemicals but also exposure to social behavior, such as maternal care, could affect the epigenome. It is proposed that exposures to different environmental agents could lead to interindividual phenotypic diversity as well as differential susceptibility to disease and behavioral pathologies. Interindividual differences in the epigenetic state could also affect susceptibility to xenobiotics. Although our current understanding of how epigenetic mechanisms impact on the toxic action of xenobiotics is very limited, it is anticipated that in the future, epigenetics will be incorporated in the assessment of the safety of chemicals.
Collapse
Affiliation(s)
- Moshe Szyf
- Department of Pharmacology and Therapeutics, McGill University, 3655 Sir William Osler Promenade, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
32
|
Park IY, Sohn BH, Yu E, Suh DJ, Chung YH, Lee JH, Surzycki SJ, Lee YI. Aberrant epigenetic modifications in hepatocarcinogenesis induced by hepatitis B virus X protein. Gastroenterology 2007; 132:1476-94. [PMID: 17408664 DOI: 10.1053/j.gastro.2007.01.034] [Citation(s) in RCA: 227] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Accepted: 12/14/2006] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS The involvement of the hepatitis B virus X (HBx) protein in epigenetic modifications during hepatocarcinogenesis has not been previously characterized. The aim of the present study was to identify the involvement of HBx in regional hypermethylation and global hypomethylation during the formation of hepatocellular carcinoma (HCC). METHODS Liver cell lines were transiently or stably transfected with an HBx-expressing vector. DNA methyltransferase (DNMT) promoter activity changes were examined by luciferase assay and chromatin immunoprecipitation. The methylation status of insulin-like growth factor binding protein-3 was examined by methyl-specific polymerase chain reaction and bisulfite sequencing. Global DNA methylation levels were examined using 5-methylcytosine dot blot and methylation-sensitive Southern blot analysis. HBx-mediated DNA methylation abnormalities were confirmed in patient HCC samples using methyl-specific polymerase chain reaction and 5-methylcytosine dot blot analysis. RESULTS HBx expression increased total DNMT activities by up-regulation of DNMT1, DNMT3A1, and DNMT3A2 and selectively promoted regional hypermethylation of specific tumor suppressor genes. HBx specifically repressed insulin-like growth factor-3 expression through de novo methylation via DNMT3A1 and DNMT3A2 and by inhibiting SP1 binding via recruiting methyl CpG binding protein 2 to the newly methylated SP1 binding element. HBx also induced global hypomethylation of satellite 2 repeat sequences by down-regulating DNMT3B. The prevalence of these specific methylation abnormalities by HBx was significantly correlated with HBx expression in HBV-infected HCC patients. CONCLUSIONS Targeted deregulation of DNMTs by HBx promotes both specific regional hypermethylation and global hypomethylation. These epigenetic modulations by HBx may suggest a mechanism for epigenetic tumorigenesis during HBV-mediated hepatocarcinogenesis.
Collapse
Affiliation(s)
- In Young Park
- Liver Cell Signal Transduction Laboratory, Molecular Cancer Research Center, KRIBB, Daejeon, Korea
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Atkinson SP, Keith WN. Epigenetic control of cellular senescence in disease: opportunities for therapeutic intervention. Expert Rev Mol Med 2007; 9:1-26. [PMID: 17352843 DOI: 10.1017/s1462399407000269] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Understanding how senescence is established and maintained is an important area of study both for normal cell physiology and in tumourigenesis. Modifications to N-terminal tails of histone proteins, which can lead to chromatin remodelling, appear to be key to the regulation of the senescence phenotype. Epigenetic mechanisms such as modification of histone proteins have been shown to be sufficient to regulate gene expression levels and specific gene promoters can become epigenetically altered at senescence. This suggests that epigenetic mechanisms are important in senescence and further suggests epigenetic deregulation could play an important role in the bypass of senescence and the acquisition of a tumourigenic phenotype. Tumour suppressor proteins and cellular senescence are intimately linked and such proteins are now known to regulate gene expression through chromatin remodelling, again suggesting a link between chromatin modification and cellular senescence. Telomere dynamics and the expression of the telomerase genes are also both implicitly linked to senescence and tumourigenesis, and epigenetic deregulation of the telomerase gene promoters has been identified as a possible mechanism for the activation of telomere maintenance mechanisms in cancer. Recent studies have also suggested that epigenetic deregulation in stem cells could play an important role in carcinogenesis, and new models have been suggested for the attainment of tumourigenesis and bypass of senescence. Overall, proper regulation of the chromatin environment is suggested to have an important role in the senescence pathway, such that its deregulation could lead to tumourigenesis.
Collapse
Affiliation(s)
- Stuart P Atkinson
- Centre for Oncology and Applied Pharmacology, University of Glasgow, Cancer Research UK Beatson Laboratories, Bearsden, Glasgow, G61 1BD, UK
| | | |
Collapse
|
34
|
Burgers WA, Blanchon L, Pradhan S, de Launoit Y, Kouzarides T, Fuks F. Viral oncoproteins target the DNA methyltransferases. Oncogene 2007; 26:1650-5. [PMID: 16983344 PMCID: PMC3350866 DOI: 10.1038/sj.onc.1209950] [Citation(s) in RCA: 204] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2006] [Revised: 07/04/2006] [Accepted: 07/05/2006] [Indexed: 12/31/2022]
Abstract
Small DNA tumour viruses have evolved a number of mechanisms to drive nondividing cells into S phase. Virally encoded oncoproteins such as adenovirus E1A and human papillomavirus (HPV) E7 can bind an array of cellular proteins to override proliferation arrest. The DNA methyltransferase Dnmt1 is the major mammalian enzyme responsible for maintaining CpG methylation patterns in the cell following replication. One of the hallmarks of tumour cells is disrupted DNA methylation patterns, highlighting the importance of the proper regulation of DNA methyltransferases in normal cell proliferation. Here, we show that adenovirus 5 E1A and HPV-16 E7 associate in vitro and in vivo with the DNA methyltransferase Dnmt1. Consistent with this interaction, we find that E1A and E7 can purify DNA methyltransferase activity from nuclear extracts. These associations are direct and mediated by the extreme N-terminus of E1A and the CR3 zinc-finger domain of E7. Furthermore, we find that a point mutant at leucine 20 of E1A, a residue known to be critical for its transformation functions, is unable to bind Dnmt1 and DNA methyltransferase activity. Finally, both E1A and E7 can stimulate the methyltransferase activity of Dnmt1 in vitro. Our results provide the first indication that viral oncoproteins bind and regulate Dnmt1 enzymatic activity. These observations open up the possibility that this association may be used to control cellular proliferation pathways and suggest a new mechanism by which small DNA tumour viruses can steer cells through the cell cycle.
Collapse
Affiliation(s)
- WA Burgers
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, UK
- Faculty of Health Sciences, Division of Medical Virology, University of Cape Town Medical School, Cape Town, South Africa
| | - L Blanchon
- Laboratory of Cancer Epigenetics, Faculty of Medicine, Free University of Brussels, Brussels, Belgium
| | | | - Y de Launoit
- UMR 8161, CNRS Institut Pasteur de Lille, Universités de Lille 1 et 2, Institut de Biologie de Lille, Lille, Cedex, France
| | - T Kouzarides
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, UK
| | - F Fuks
- Wellcome Trust/CRUK Gurdon Institute, University of Cambridge, Cambridge, UK
- Laboratory of Cancer Epigenetics, Faculty of Medicine, Free University of Brussels, Brussels, Belgium
| |
Collapse
|
35
|
Morey SR, Smiraglia DJ, James SR, Yu J, Moser MT, Foster BA, Karpf AR. DNA methylation pathway alterations in an autochthonous murine model of prostate cancer. Cancer Res 2007; 66:11659-67. [PMID: 17178860 DOI: 10.1158/0008-5472.can-06-1937] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We examined the DNA methylation pathway in an autochthonous murine prostate cancer model, transgenic adenocarcinoma of mouse prostate (TRAMP). We observed that, compared with strain-matched normal prostates, primary and metastatic TRAMP tumors display increased cytosine DNA methyltransferase (Dnmt) activity, Dnmt1 and Dnmt3b protein expression, and Dnmt1, Dnmt3a, and Dnmt3b mRNA expression. Increased expression of Dnmt genes correlates with increased expression of cyclin A and E2F target genes, implicating increased cell proliferation and Rb inactivation in Dnmt overexpression. We analyzed DNA methylation in TRAMP and found that global levels of 5-methyl-2'-deoxycytidine are unaltered, whereas specific tumors display centromeric repeat hypomethylation. To interrogate locus-specific methylation, we did restriction landmark genomic scanning (RLGS) on normal prostates and primary tumors. In primary tumors, 2.3% of approximately 1,200 analyzed loci display aberrant DNA hypermethylation, whereas a considerably smaller number of events show hypomethylation. The pattern of RLGS changes was nonrandom, indicating a coordinated methylation defect. Two specific genes identified by RLGS were studied in detail. Surprisingly, methylation of a downstream exon of p16(INK4a) (p16) was the highest frequency hypermethylation event identified in TRAMP, where it is associated with increased p16 mRNA and protein expression. In contrast, hypermethylation of the 5' CpG island region of the homeobox gene Irx3 in TRAMP is associated with reduced gene expression. In summary, our data reveal a systemic DNA methylation pathway defect in TRAMP reminiscent of human prostate cancer, supporting the use of this model to investigate the functional role of DNA methylation pathway alterations in prostate cancer development.
Collapse
Affiliation(s)
- Shannon R Morey
- Departments of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Kwon YM, Park JH, Kim H, Shim YM, Kim J, Han J, Park J, Kim DH. Different susceptibility of increased DNMT1 expression by exposure to tobacco smoke according to histology in primary non-small cell lung cancer. J Cancer Res Clin Oncol 2006; 133:219-26. [PMID: 17053888 DOI: 10.1007/s00432-006-0160-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2006] [Accepted: 09/18/2006] [Indexed: 10/24/2022]
Abstract
PURPOSE DNA methyltransferase 1 (DNMT1) is known to play an important role in the development of cancers. However, the underlying mechanisms responsible for the altered expression of DNMT1 in non-small cell lung cancers (NSCLCs) remain to be elucidated. METHODS We investigated the relationships of mRNA expression levels of DNMT1 to the altered expression of retinoblastoma (Rb) and p53 and to the clinicopathological variables in 153 NSCLCs. The expression of DNMT1 was determined by quantitative real-time PCR, and the altered expressions of p53 and Rb were assessed by immunohistochemistry. RESULTS The increased expression of DNMT1 was found in 47 (31%) of 153 NSCLC patients examined. The prevalence of increased DNMT1 expression was significantly different between adenocarcinoma and squamous cell carcinoma (42% vs. 19%, respectively; P = 0.004). Patients who had smoked more than 65 packyears showed a 4.17 times [95% confidence interval (CI) = 1.17-69.49; P = 0.007] higher risk of increased DNMT1 expression compared to those who had smoked less than 45 packyears in adenocarcinoma. The expressions of Rb and p53 proteins were not associated with the increased expression of DNMT1 in 153 NSCLCs (P = 0.18 and 0.54, respectively). CONCLUSIONS The present study suggests that the susceptibility of increased DNMT1 expression by exposure to tobacco smoke may be different according to histologic subtypes in NSCLC.
Collapse
Affiliation(s)
- Young-Mi Kwon
- Division of Pulmonary and Critical Care Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
37
|
McCabe MT, Low JA, Imperiale MJ, Day ML. Human polyomavirus BKV transcriptionally activates DNA methyltransferase 1 through the pRb/E2F pathway. Oncogene 2006; 25:2727-35. [PMID: 16547506 DOI: 10.1038/sj.onc.1209266] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Many DNA tumor virus oncogenes are capable of activating and highjacking the host cell's DNA replication machinery for its own reproduction purposes through targeting and inactivation of the retinoblastoma pocket protein family. Pocket proteins function to regulate cell cycle progression and DNA synthesis through inhibitory interactions with the E2F transcription factors. The interaction of viral oncogenes with the pocket proteins is crucial for their transforming activity. We recently demonstrated that the DNA methyltransferase 1 (DNMT1) gene is an E2F target gene that is transcriptionally activated in cells lacking the retinoblastoma gene (Rb-/-). Overexpression of DNMT1 is implicated in tumor suppressor gene hypermethylation which is associated with tumorigenesis. Given that viral oncogenes potently stimulate E2F activity, we hypothesized that viral infection might activate DNMT1 and thereby promote transformation. Herein, we demonstrate that DNMT1 is strongly activated by the human polyomavirus BKV large T antigen (TAg) and adenovirus E1a. Viral oncogene mutants incapable of binding the pocket proteins are ineffective at activating DNMT1 compared to their wild-type counterparts. Additionally, mutation of the E2F sites within the DNMT1 promoters dramatically abrogates transcriptional activation. These data suggest that viral induction of DNMT1 through modulation of the pRB/E2F pathway may be involved in viral transformation.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/virology
- Adenovirus E1A Proteins/metabolism
- Animals
- Antigens, Polyomavirus Transforming/genetics
- Antigens, Polyomavirus Transforming/metabolism
- BK Virus/physiology
- Cell Transformation, Viral
- Cells, Cultured
- DNA (Cytosine-5-)-Methyltransferase 1
- DNA (Cytosine-5-)-Methyltransferases/genetics
- DNA (Cytosine-5-)-Methyltransferases/metabolism
- E2F Transcription Factors/genetics
- E2F Transcription Factors/metabolism
- Embryo, Mammalian/cytology
- Embryo, Mammalian/metabolism
- Enzyme Activation
- Fibroblasts/cytology
- Fibroblasts/metabolism
- Humans
- Luciferases
- Male
- Mice
- Mice, Knockout
- Mutation
- NIH 3T3 Cells/metabolism
- NIH 3T3 Cells/virology
- Polyomavirus Infections/immunology
- Polyomavirus Infections/virology
- Promoter Regions, Genetic/genetics
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Prostatic Neoplasms/virology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Retinoblastoma Protein/genetics
- Retinoblastoma Protein/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Transcription, Genetic
- Transcriptional Activation
Collapse
Affiliation(s)
- M T McCabe
- Program in Cellular and Molecular Biology, Department of Urology, University of Michigan, Ann Arbor, MI 48109-0944, USA
| | | | | | | |
Collapse
|
38
|
McCabe MT, Low JA, Daignault S, Imperiale MJ, Wojno KJ, Day ML. Inhibition of DNA methyltransferase activity prevents tumorigenesis in a mouse model of prostate cancer. Cancer Res 2006; 66:385-92. [PMID: 16397253 DOI: 10.1158/0008-5472.can-05-2020] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Transcriptional silencing of tumor suppressor genes by DNA methylation plays an important role in tumorigenesis. These aberrant epigenetic modifications may be mediated in part by elevated DNA methyltransferase levels. DNA methyltransferase 1 (DNMT1), in particular, is overexpressed in many tumor types. Recently, we showed that Dnmt1 is transcriptionally regulated by E2F transcription factors and that retinoblastoma protein (pRb) inactivation induces Dnmt1. Based on these observations, we investigated regulation of Dnmt1 by polyomavirus oncogenes, which potently inhibit the pRb pocket protein family. Infection of primary human prostate epithelial cells with BK polyomavirus dramatically induced Dnmt1 transcription following large T antigen (TAg) translation and E2F activation. For in vivo study of Dnmt1 regulation, we used the transgenic adenocarcinoma of the mouse prostate (TRAMP) model, which expresses the SV40 polyomavirus early region, including TAg, under control of a prostate-specific promoter. Analysis of TRAMP prostate lesions revealed greatly elevated Dnmt1 mRNA and protein levels beginning in prostatic intraepithelial neoplasia and continuing through advanced prostate cancer and metastasis. Interestingly, when TRAMP mice were treated in a chemopreventive manner with the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (5-aza), 0 of 14 mice developed prostate cancer at 24 weeks of age, whereas 7 of 13 (54%) control-treated mice developed poorly differentiated prostate cancer. Treatment with 5-aza also prevented the development of lymph node metastases and dramatically extended survival compared with control-treated mice. Taken together, these data suggest that Dnmt1 is rapidly activated by pRb pathway inactivation, and that DNA methyltransferase activity is required for malignant transformation and tumorigenesis.
Collapse
Affiliation(s)
- Michael T McCabe
- Department of Urology, University of Michigan, Ann Arbor, Michigan 48109-0944, USA
| | | | | | | | | | | |
Collapse
|
39
|
Szyf M. DNA methylation and demethylation as targets for anticancer therapy. BIOCHEMISTRY (MOSCOW) 2005; 70:533-49. [PMID: 15948707 DOI: 10.1007/s10541-005-0147-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cancer growth and metastasis require the coordinate change in gene expression of different sets of genes. While genetic alterations can account for some of these changes, it is becoming evident that many of the changes in gene expression observed are caused by epigenetic modifications. The epigenome consists of the chromatin and its modifications, the "histone code" as well as the pattern of distribution of covalent modifications of cytosines residing in the dinucleotide sequence CG by methylation. Although hypermethylation of tumor suppressor genes has attracted a significant amount of attention and inhibitors of DNA methylation were shown to activate methylated tumor suppressor genes and inhibit tumor growth, demethylation of critical genes plays a critical role in cancer as well. This review discusses the emerging role of demethylation in activation of pro-metastatic genes and the potential therapeutic implications of the demethylation machinery in metastasis.
Collapse
Affiliation(s)
- M Szyf
- Department of Pharmacology and Therapeutics, McGill University, Montreal PQ H3G 1Y6, Canada.
| |
Collapse
|
40
|
Krepulat F, Löhler J, Heinlein C, Hermannstädter A, Tolstonog GV, Deppert W. Epigenetic mechanisms affect mutant p53 transgene expression in WAP-mutp53 transgenic mice. Oncogene 2005; 24:4645-59. [PMID: 15870706 DOI: 10.1038/sj.onc.1208557] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We describe the construction and phenotypic characterization of 23 whey acidic protein (WAP)-mutp53 transgenic mouse lines. The mutp53-expressing lines showed a mosaic expression pattern for the transgenes, leading to a heterogeneous yet mouse line-specific expression pattern for mutp53 upon induction. Only few lines were obtained, in which the majority of the induced mammary epithelial cells expressed the mutp53 transgene, most of the transgenic lines did not express mutp53, or expressed the transgene in less than 2% of the induced mammary epithelial cells. Hormone requirements for mutp53 transgene expression from the WAP-promoter differed in high and low expressing lines, being low in high expressing lines, and even lower in multiparous mutp53 mice, where persistent expression of the transgene occurred. Repeated induction of mutp53 expression through repeated parturition resulted in the formation of expanding mutp53-expressing foci within the mammary alveolar epithelium. The data suggest that epigenetic mechanisms play a role in modulating the expression of the mutp53 transgene. To support this idea, we crossed a nonexpressing WAP-mutp53 line with a strongly SV40 T-antigen-expressing WAP-T mouse line. In the bitransgenic mice, T-antigen-induced chromatin remodeling led to re-expression of epigenetically silenced mutp53 transgene(s). In these mice, mutp53 expression was much more variable compared to SV40 T-antigen expression, and seemed to depend on the coexpression of SV40 T-antigen. Mutp53 expression in this system thus resembles the situation in many human tumors, where one can observe a heterogeneous expression of mutp53, despite a homogeneous distribution of the p53 mutation in the tumor cells.
Collapse
Affiliation(s)
- Frauke Krepulat
- Heinrich-Pette-Institut für Experimentelle Virologie und Immunologie an der Universität Hamburg, Martinistrasse 52, D-20251 Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
Tumor suppressor gene silencing by DNA hypermethylation contributes to tumorigenesis in many tumor types. This aberrant methylation may be due to increased expression and activity of DNA methyltransferases, which catalyze the transfer of methyl groups from S-adenosylmethionine to cytosines in CpG dinucleotides. Elevated expression of the maintenance DNA methyltransferase, DNA methyltransferase 1 (DNMT-1), has been shown in carcinomas of the colon, lung, liver, and prostate. Based on the nearly ubiquitous alterations of both DNA methylation and the retinoblastoma protein (pRb) pathway found in human cancer, we investigated a potential regulatory pathway linking the two alterations in murine and human prostate epithelial cells. Analysis of DNA methyltransferase levels in Rb-/- murine prostate epithelial cell lines revealed elevated Dnmt-1 levels. Genomic DNA sequence analysis identified conserved E2F consensus binding sites in proximity to the transcription initiation points of murine and human Dnmt-1. Furthermore, the Dnmt-1 promoter was shown to be regulated by the pRb/E2F pathway in murine and human cell lines of epithelial and fibroblast origin. In the absence of pRb, Dnmt-1 transcripts exhibited aberrant cell cycle regulation and Rb-/- cells showed aberrant methylation of the paternally expressed gene 3 (Peg3) tumor suppressor gene. These findings show a link between inactivation of the pRb pathway and induction of DNA hypermethylation of CpG island-containing genes in tumorigenesis.
Collapse
Affiliation(s)
- Michael T McCabe
- Department of Urology, Michigan Urology Center, University of Michigan, Ann Arbor, Michigan 48109-0944, USA
| | | | | |
Collapse
|
42
|
Suzuki M, Toyooka S, Shivapurkar N, Shigematsu H, Miyajima K, Takahashi T, Stastny V, Zern AL, Fujisawa T, Pass HI, Carbone M, Gazdar AF. Aberrant methylation profile of human malignant mesotheliomas and its relationship to SV40 infection. Oncogene 2004; 24:1302-8. [PMID: 15592515 DOI: 10.1038/sj.onc.1208263] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Malignant mesothelioma (MM) is associated with asbestos exposure and the presence of SV40 viral sequences. Recently, we reported that SV40 infection of human mesothelial cells (HM) causes aberrant methylation of the tumor suppressor gene (TSG) RASSF1A. We investigated methylation of 12 genes by methylation-specific PCR in 63 MMs, six MM cell lines, and two foci of SV40-infected HM. Methylation percentages of the tested genes ranged from 3 to 65%. The frequencies of HPP1, RASSF1A, Cyclin D2, and RRAD methylation, and the value of the methylation index, were significantly higher in SV40 sequence-positive MMs than in SV40-negative MMs. Methylation of TMS1 and HIC-1 was associated with shortened survival. SV40-infected HM showed progressive aberrant methylation of seven genes (RASSF1A, HPP1, DcR1, TMS1, CRBP1, HIC-1, and RRAD) during serial passage. Our results demonstrate a relationship between SV40 and methylation of multiple genes in MM, indicating that the virus plays a role in the pathogenesis of MM.
Collapse
Affiliation(s)
- Makoto Suzuki
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
May T, Hauser H, Wirth D. Transcriptional control of SV40 T-antigen expression allows a complete reversion of immortalization. Nucleic Acids Res 2004; 32:5529-38. [PMID: 15486202 PMCID: PMC524297 DOI: 10.1093/nar/gkh887] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Conditional proliferation of mouse embryo fibroblasts was achieved with a novel autoregulatory vector for Tet-dependent expression of the SV40 T-antigen. The majority of cell clones that were isolated under induced conditions showed strict regulation of cell growth. Status switches were found to be fully reversible and highly reproducible with respect to gene expression characteristics. A consequence of T-antigen expression is a significant deregulation of >400 genes. Deinduced cells turn to rest in G0/G1 phase and exhibit a senescent phenotype. The cells are not oncogenic and no evidence for transformation was found after several months of cultivation. Conditional immortalization allows diverse studies including those on cellular activities without the influence of the immortalizing gene(s), senescence as well as secondary effects from T-antigen expression.
Collapse
Affiliation(s)
- Tobias May
- Department of Gene Regulation and Differentiation, GBF-German Research Center for Biotechnology, Mascheroder Weg 1, D-38124 Braunschweig, Germany
| | | | | |
Collapse
|
44
|
MacLennan NK, James SJ, Melnyk S, Piroozi A, Jernigan S, Hsu JL, Janke SM, Pham TD, Lane RH. Uteroplacental insufficiency alters DNA methylation, one-carbon metabolism, and histone acetylation in IUGR rats. Physiol Genomics 2004; 18:43-50. [PMID: 15084713 DOI: 10.1152/physiolgenomics.00042.2004] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Uteroplacental insufficiency leads to intrauterine growth retardation (IUGR) and increases the risk of insulin resistance and hypertriglyceridemia in both humans and rats. Postnatal changes in hepatic gene expression characterize the postnatal IUGR rat, despite the transient nature of the initial in utero insult. Phenomena such as DNA methylation and histone acetylation can induce a relatively static reprogramming of gene transcription by altering chromatin infrastructure. We therefore hypothesized that uteroplacental insufficiency persistently affects DNA methylation and histone acetylation in the IUGR rat liver. IUGR rat pups were created by inducing uteroplacental insufficiency through bilateral uterine artery ligation of the pregnant dam on day 19 of gestation. The SssI methyltransferase assay and two-dimensional thin-layer chromatography demonstrated genome-wide DNA hypomethylation in postnatal IUGR liver. To investigate a possible mechanism for this hypomethylation, levels of hepatic metabolites and enzyme mRNAs involved in one-carbon metabolism were measured using HPLC with coulometric electrochemical detection and real-time RT-PCR, respectively. Uteroplacental insufficiency increased IUGR levels of S-adenosylhomocysteine, homocysteine, and methionine in association with decreased mRNA levels of methionine adenosyltransferase and cystathionine-beta-synthase. Western blotting further demonstrated that increased quantities of acetylated histone H3 also characterized the IUGR liver. Increased hepatic levels of S-adenosylhomocysteine can promote DNA hypomethylation, which is often associated with histone hyperacetylation. We speculate that the altered intrauterine milieu associated with uteroplacental insufficiency affects hepatic one-carbon metabolism and subsequent DNA methylation, which thereby alters chromatin dynamics and leads to persistent changes in hepatic gene expression.
Collapse
Affiliation(s)
- Nicole K MacLennan
- David Geffen School of Medicine, UCLA, Department of Pediatrics, Division of Neonatology and Developmental Biology, Mattel Children's Hospital, UCLA, Los Angeles, California, 90095-1752, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Casillas MA, Lopatina N, Andrews LG, Tollefsbol TO. Transcriptional control of the DNA methyltransferases is altered in aging and neoplastically-transformed human fibroblasts. Mol Cell Biochem 2004; 252:33-43. [PMID: 14577574 DOI: 10.1023/a:1025548623524] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Although it has been known for quite some time that genomic methylation is significantly altered in aging and neoplastic tissues and cells, the underlying mechanisms responsible for these alterations are not yet known. Since DNA methylation affects many different cellular processes including, most significantly, gene expression, elucidation of the basis for aberrations in DNA methylation in aging and cancer is of high priority. To address this problem, we sought to analyze changes in gene expression, protein production and enzyme activity of the three major DNA methyltransferases (Dnmtl, 3a, and 3b) in aging and neoplastically-transformed WI-38 human fetal lung fibroblasts. We have found that the gene expression of each of the three Dnmts parallels changes in protein production and enzyme activity of the Dnmts not only in aging cells, but also in WI-38 fibroblasts induced to undergo neoplastic transformation using defined genetic elements. These findings strongly implicate change in gene expression as an underlying mechanism in the altered genomic methylation of these cells. Striking changes in the gene expression of the Dnmts were observed in aging cells with the mRNA of Dnmtl becoming reduced while the mRNA of Dnmt3b increased steadily in aging cells consistent with our observations in protein production and activity of these enzymes. Surprisingly, Dnmt3a actually decreased in gene expression in aging cells. We therefore propose that the transcriptional control of Dnmt1, the predominant maintenance methyltransferase, is significantly suppressed in aging cells and contributes to the reduced genomic methylation of these cells. The paradoxical sporadic gene hypermethylation in aging cells appears to be related to transcriptional up-regulation of the Dnmt3b gene. In addition, we sought to explore the coordinated changes in gene expression, protein production, and enzyme activity of these Dnmts in early cellular transformation. In these cells, the gene expression of all the three major Dnmts were up-regulated followed by marked increases in Dnmt protein and enzyme activity. These results therefore collectively indicate that changes in transcriptional control of the Dnmts are the likely cause for the known alterations in DNA methylation in aging cells and in cells undergoing tumorigenesis. They also show that changes in transcription of Dnmtl and Dnmt3b are probably most important in affecting the generalized hypomethylation and specific hypermethylation seen in aging cells while gene expression of all the Dnmts is significantly increased in cancer cells. These findings should have broad implications in elucidating the underlying causes of changes in DNA methylation in aging and tumorigenesis and point to variations in gene expression of the individual Dnmts as a likely mechanism involved in these processes.
Collapse
Affiliation(s)
- Mark A Casillas
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294-1170, USA
| | | | | | | |
Collapse
|
46
|
Shivapurkar N, Takahashi T, Reddy J, Zheng Y, Stastny V, Collins R, Toyooka S, Suzuki M, Parikh G, Asplund S, Kroft SH, Timmons C, McKenna RW, Feng Z, Gazdar AF. Presence of Simian Virus 40 DNA Sequences in Human Lymphoid and Hematopoietic Malignancies and Their Relationship to Aberrant Promoter Methylation of Multiple Genes. Cancer Res 2004; 64:3757-60. [PMID: 15172980 DOI: 10.1158/0008-5472.can-03-3307] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The simian polyoma virus SV40 has been detected in specific human tumors including non-Hodgkin's lymphomas, although a causative role for the virus has not been convincingly demonstrated. Aberrant methylation of CpG islands in promoter regions is a frequent method of silencing tumor suppressor genes (TSGs) in cancers and may be induced by oncogenic viruses. We investigated the relationship between the presence of SV40 or EBV DNA sequences and the methylation profiles for 10 TSGs in 90 cases of non-Hodgkin's lymphomas/leukemias and 56 control tissues. SV40 sequences were present in 33/90 (37%) non-Hodgkin's lymphomas/leukemias, and EBV was present in 11/42 (26%) of non-Hodgkin's lymphomas. We found a highly significant correlation between the presence of SV40 and methylation of seven genes (P values, 0.006 to <0.0001). In lymphomas, there was no relationship between EBV and methylation. Oncogenic viruses and methylation were rarely present in control tissues. We investigated methylation of the same 10 TSGs in peripheral blood mononuclear cells (PBMC) from a healthy volunteer infected with EBV or EBV and SV40. Promoter methylation of CDH1 and CDH13 were noted in dual SV40- and EBV-infected PBMC, and these two genes were also highly significantly correlated to the presence of SV40 sequences in tumors. SV40 infection also resulted in appearance of the lymphoma/leukemia-specific marker, methylated SHP1. Methylation was completely absent in uninfected and EBV-infected PBMC. Our results demonstrate that the presence of SV40 in hematological malignancies is associated with promoter methylation of TSGs and that in all probability, the virus plays a role in tumor pathogenesis.
Collapse
Affiliation(s)
- Narayan Shivapurkar
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8593, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Soejima K, Fang W, Rollins BJ. DNA methyltransferase 3b contributes to oncogenic transformation induced by SV40T antigen and activated Ras. Oncogene 2003; 22:4723-33. [PMID: 12879017 DOI: 10.1038/sj.onc.1206510] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2003] [Revised: 02/11/2003] [Accepted: 02/17/2003] [Indexed: 01/03/2023]
Abstract
Transcriptional silencing of tumor suppressor genes in association with DNA methylation contributes to malignant transformation. However, the specific DNA methyltransferases that initiate this process are unknown. Here we show that a de novo DNA methyltransferase, DNMT3b, substantially contributes to the oncogenic phenotype in a lung cancer model. Normal human bronchial epithelial (NHBE) cells expressing telomerase, SV40 large T antigen, and activated Ras were immortal, formed colonies in soft agar, and expressed DNMT3b. Antisense suppression of DNMT3b prevented soft agar growth. Furthermore, mouse embryo fibroblasts expressing T antigen and Ras formed soft agar colonies and large tumors, but fibroblasts from Dnmt3b(-/-) mice did not grow in soft agar and were much less tumorigenic in vivo. The tumor suppressor genes, FHIT, TSLC1, and RASSF1A were downregulated in transformed NHBE cells, and antisense DNMT3b treatment resulted in re-expression of FHIT and TSLC1. While expression of TSCL1 correlated with methylation of CpG dinucleotides in its promoter region, the expression of FHIT did not, suggesting that DNMT3b may silence genes by several mechanisms including direct DNA methylation or recruitment of proteins that modify chromatin. Regardless of mechanism, our data indicate that DNMT3b plays an important role in transformation.
Collapse
Affiliation(s)
- Kenzo Soejima
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | | |
Collapse
|
48
|
Shi W, Hoeflich A, Flaswinkel H, Stojkovic M, Wolf E, Zakhartchenko V. Induction of a senescent-like phenotype does not confer the ability of bovine immortal cells to support the development of nuclear transfer embryos. Biol Reprod 2003; 69:301-9. [PMID: 12646489 DOI: 10.1095/biolreprod.102.012112] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Previously, we reported that cloned embryos derived from an immortalized bovine mammary epithelial cell line (MECL) failed to develop beyond 12- to 16-cell stage. To analyze whether induction of a senescent-like phenotype in MECL can improve their ability to support the development after transfer into enucleated oocytes, we treated MECL with DNA methylation inhibitor 5-aza-2-deoxycytidine (Aza-C), histone deacetylase inhibitors trichostatin A (TSA), sodium butyrate (NaBu), or 5-bromodeoxyuridine and used those cells for nuclear transfer. Primary bovine fetal fibroblasts (BFF) were used as control. All agents were capable to induce features of senescence including reduced cell proliferation, enlarged cell size with a considerable proportion of cells stained positive for acidic senescence-associated beta-galactosidase and G1/S cell cycle boundary arrest in MECL. Aza-C treatment induced genome demethylation. Acetylation of H3 and H4 was increased after TSA treatment in both MECL and BFF, whereas no obvious changes in global H3 or H4 acetylation were detected after NaBu treatment. Nuclear transfer experiments following diverse treatments demonstrated that the induced senescent-like phenotype of MECL did not confer their ability to support embryonic development, although 7.3% of reconstructed embryos derived from NaBu-treated cells developed to morula stage. Intriguingly, a much higher proportion of cloned embryos developed to blastocysts when using NaBu-treated BFF, compared with using untreated BFF (59% versus 26%). Our results suggest that the developmental failure of donor nuclei from bovine immortal cells could not be reversed by induction of senescent-like phenotype. The beneficial effect of NaBu on the developmental potential of cloned embryos reconstructed from BFF merits further studies.
Collapse
Affiliation(s)
- Wei Shi
- Department of Molecular Animal Breeding and Biotechnology, University of Munich, D-85764 Munich, Germany
| | | | | | | | | | | |
Collapse
|
49
|
Young JI, Sedivy JM, Smith JR. Telomerase expression in normal human fibroblasts stabilizes DNA 5-methylcytosine transferase I. J Biol Chem 2003; 278:19904-8. [PMID: 12665523 DOI: 10.1074/jbc.m301685200] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The finite proliferative potential of normal human fibroblasts can be overcome, a process commonly called immortalization, by the introduction of the catalytic subunit of human telomerase. In contrast to malignant transformation, the pattern of gene expression remains largely unmodified in telomerase-induced immortalization. Here we show evidence that suggests that the maintenance of a "young" pattern of gene expression by telomerization is mediated, at least in part, by a novel function of human telomerase that involves regulation of DNA methyltransferase I gene expression.
Collapse
Affiliation(s)
- Juan I Young
- Roy M. and Phyllis Gough Huffington Center on Aging, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | |
Collapse
|
50
|
Milutinovic S, Zhuang Q, Niveleau A, Szyf M. Epigenomic stress response. Knockdown of DNA methyltransferase 1 triggers an intra-S-phase arrest of DNA replication and induction of stress response genes. J Biol Chem 2003; 278:14985-95. [PMID: 12576480 DOI: 10.1074/jbc.m213219200] [Citation(s) in RCA: 81] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The DNA methylation pattern is an important component of the epigenome that regulates and maintains gene expression programs. In this paper, we test the hypothesis that vertebrate cells possess mechanisms protecting them from epigenomic stress similar to DNA damage checkpoints. We show that knockdown of DNMT1 (DNA methyltransferase 1) by an antisense oligonucleotide triggers an intra-S-phase arrest of DNA replication that is not observed with control oligonucleotide. The cells are arrested at different positions throughout the S-phase of the cell cycle, suggesting that this response is not specific to distinct classes of origins of replication. The intra-S-phase arrest of DNA replication is proposed to protect the genome from extensive DNA demethylation that could come about by replication in the absence of DNMT1. This protective mechanism is not induced by 5-aza-2'-deoxycytidine, a nucleoside analog that inhibits DNA methylation by trapping DNMT1 in the progressing replication fork, but does not reduce de novo synthesis of DNMT1. Our data therefore suggest that the intra-S-phase arrest is triggered by a reduction in DNMT1 and not by demethylation of DNA. DNMT1 knockdown also leads to an induction of a set of genes that are implicated in genotoxic stress response such as NF-kappaB, JunB, ATF-3, and GADD45beta (growth arrest DNA damage 45beta gene). Based on these data, we suggest that this stress response mechanism evolved to guard against buildup of DNA methylation errors and to coordinate inheritance of genomic and epigenomic information.
Collapse
Affiliation(s)
- Snezana Milutinovic
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | | | | | | |
Collapse
|