51
|
Moggs J, Terranova R. Chromatin dynamics underlying latent responses to xenobiotics. Toxicol Res (Camb) 2018; 7:606-617. [PMID: 30090610 PMCID: PMC6062062 DOI: 10.1039/c7tx00317j] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/26/2018] [Indexed: 11/21/2022] Open
Abstract
Pleiotropic xenobiotics can trigger dynamic alterations in mammalian chromatin structure and function but many of these are likely non-adverse and simply reflect short-term changes in DNA transactions underlying normal homeostatic, adaptive and protective cellular responses. However, it is plausible that a subset of xenobiotic-induced perturbations of somatic tissue or germline epigenomes result in delayed-onset and long-lasting adverse effects, in particular if they occur during critical stages of growth and development. These could include reprogramming, dedifferentiation, uncontrolled growth, and cumulative toxicity effects through molecular memory of prior xenobiotic exposures or altered susceptibility to subsequent xenobiotic exposures. Here we discuss the current evidence for epigenetic mechanisms underlying latent responses to xenobiotics, and the potential for identifying molecular epigenetic changes that are prodromal to overt morphologic or functional toxicity phenotypes.
Collapse
Affiliation(s)
- Jonathan Moggs
- Preclinical Safety , Translational Medicine , Novartis Institutes for BioMedical Research , Basel , Switzerland
| | - Rémi Terranova
- Preclinical Safety , Translational Medicine , Novartis Institutes for BioMedical Research , Basel , Switzerland
| |
Collapse
|
52
|
Yamada T. Case examples of an evaluation of the human relevance of the pyrethroids/pyrethrins-induced liver tumours in rodents based on the mode of action. Toxicol Res (Camb) 2018; 7:681-696. [PMID: 30090614 PMCID: PMC6062351 DOI: 10.1039/c7tx00288b] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/10/2018] [Indexed: 01/01/2023] Open
Abstract
Rodent carcinogenicity studies are useful for screening for human carcinogens but they are not perfect. Some modes of action (MOAs) lead to cancers in both experimental rodents and humans, but others that lead to cancers in rodents do not do so in humans. Therefore, analysing the MOAs by which chemicals produce tumours in rodents and determining the relevance of such tumour data for human risk are critical. Recently, experimental data were obtained as case examples of an evaluation of the human relevance of pyrethroid (metofluthrin and momfluorothrin)- and pyrethrins-induced liver tumours in rats based on MOA. The MOA analysis, based on the International Programme on Chemical Safety (IPCS) framework, concluded that experimental data strongly support that the postulated MOA for metofluthrin-, momfluorothrin- and pyrethrins-produced rat hepatocellular tumours is mediated by constitutive androstane receptor (CAR) activation. Since metofluthrin and momfluorothrin are close structural analogues, reproducible outcomes for both chemicals provide confidence in the MOA findings. Furthermore, cultured human hepatocyte studies and humanized chimeric mouse liver studies demonstrated species difference between human hepatocytes (refractory to the mitogenic effects of these compounds) and rat hepatocytes (sensitive to their mitogenic effects). These data strongly support the hypothesis that the CAR-mediated MOA for liver tumorigenesis is of low carcinogenic risk for humans. In this research, in addition to cultured human hepatocyte studies, the usefulness of the humanized chimeric liver mouse models was clearly demonstrated. These data substantially influenced decisions in regulatory toxicology. In this review I comprehensively discuss the human relevance of the CAR-mediated MOA for rodent liver tumorigenesis based on published information, including our recent molecular research on CAR-mediated MOA.
Collapse
Affiliation(s)
- Tomoya Yamada
- Environmental Health Science Laboratory , Sumitomo Chemical Co. , Ltd , 1-98 , 3-Chome , Kasugade-Naka , Konohana-ku , Osaka 554-8558 , Japan . ; ; Tel: +81-66466-5322
| |
Collapse
|
53
|
Chen Y, Hong T, Wang S, Mo J, Tian T, Zhou X. Epigenetic modification of nucleic acids: from basic studies to medical applications. Chem Soc Rev 2018; 46:2844-2872. [PMID: 28352906 DOI: 10.1039/c6cs00599c] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The epigenetic modification of nucleic acids represents one of the most significant areas of study in the field of nucleic acids because it makes gene regulation more complex and heredity more complicated, thus indicating its profound impact on aspects of heredity, growth, and diseases. The recent characterization of epigenetic modifications of DNA and RNA using chemical labelling strategies has promoted the discovery of these modifications, and the newly developed single-base or single-cell resolution mapping strategies have enabled large-scale epigenetic studies in eukaryotes. Due to these technological breakthroughs, several new epigenetic marks have been discovered that have greatly extended the scope and impact of epigenetic modifications in nucleic acids over the past few years. Because epigenetics is reversible and susceptible to environmental factors, it could potentially be a promising direction for clinical medicine research. In this review, we have comprehensively discussed how these epigenetic marks are involved in disease, including the pathogenesis, prevention, diagnosis and treatment of disease. These findings have revealed that the epigenetic modification of nucleic acids has considerable significance in various areas from methodology to clinical medicine and even in biomedical applications.
Collapse
Affiliation(s)
- Yuqi Chen
- College of Chemistry and Molecular Sciences, Institute of Advanced Studies, Key Laboratory of Biomedical Polymers of Ministry of Education, Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Hubei, Wuhan 430072, P. R. China.
| | | | | | | | | | | |
Collapse
|
54
|
Meehan RR, Thomson JP, Lentini A, Nestor CE, Pennings S. DNA methylation as a genomic marker of exposure to chemical and environmental agents. Curr Opin Chem Biol 2018; 45:48-56. [PMID: 29505975 DOI: 10.1016/j.cbpa.2018.02.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 02/07/2018] [Accepted: 02/12/2018] [Indexed: 02/06/2023]
Abstract
Recent progress in interpreting comprehensive genetic and epigenetic profiles for human cellular states has contributed new insights into the developmental origins of disease, elucidated novel signalling pathways and enhanced drug discovery programs. A similar comprehensive approach to decoding the epigenetic readouts from chemical challenges in vivo would yield new paradigms for monitoring and assessing environmental exposure in model systems and humans.
Collapse
Affiliation(s)
- Richard R Meehan
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK.
| | - John P Thomson
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Antonio Lentini
- Department of Clinical and Experimental Medicine, Linköping University, Linköping SE 58183, Sweden
| | - Colm E Nestor
- Department of Clinical and Experimental Medicine, Linköping University, Linköping SE 58183, Sweden.
| | - Sari Pennings
- Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, EH16 4TJ, UK.
| |
Collapse
|
55
|
|
56
|
Thomson JP, Ottaviano R, Buesen R, Moggs JG, Schwarz M, Meehan RR. Defining baseline epigenetic landscapes in the rat liver. Epigenomics 2017; 9:1503-1527. [PMID: 29130343 PMCID: PMC5957268 DOI: 10.2217/epi-2017-0029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Aim Characterization of the hepatic epigenome following exposure to chemicals and therapeutic drugs provides novel insights into toxicological and pharmacological mechanisms, however appreciation of genome-wide inter- and intra-strain baseline epigenetic variation, particularly in under-characterized species such as the rat is limited. Material & methods To enhance the utility of epigenomic endpoints safety assessment, we map both DNA modifications (5-methyl-cytosine and 5-hydroxymethyl-cytosine) and enhancer related chromatin marks (H3K4me1 and H3K27ac) across multiple male and female rat livers for two important outbred laboratory rat strains (Sprague–Dawley and Wistar). Results & conclusion Integration of DNA modification, enhancer chromatin marks and gene expression profiles reveals clear gender-specific chromatin states at genes which exhibit gender-specific transcription. Taken together this work provides a valuable baseline liver epigenome resource for rat strains that are commonly used in chemical and pharmaceutical safety assessment.
Collapse
Affiliation(s)
- John P Thomson
- MRC Human Genetics Unit, Genome Regulation, Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Raffaele Ottaviano
- MRC Human Genetics Unit, Genome Regulation, Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| | - Roland Buesen
- BASF SE, Experimental Toxicology & Ecology, 67056 Ludwigshafen, Germany
| | - Jonathan G Moggs
- Preclinical Safety, Translational Medicine, Novartis Institutes for BioMedical Research, CH-4057 Basel, Switzerland
| | - Michael Schwarz
- Department of Toxicology, Institute of Experimental & Clinical Pharmacology & Toxicology, University of Tübingen, 72074 Tübingen, Germany
| | - Richard R Meehan
- MRC Human Genetics Unit, Genome Regulation, Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh EH4 2XU, UK
| |
Collapse
|
57
|
TET1 exerts its tumour suppressor function by regulating autophagy in glioma cells. Biosci Rep 2017; 37:BSR20160523. [PMID: 28341638 PMCID: PMC5672083 DOI: 10.1042/bsr20160523] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 02/24/2017] [Accepted: 03/24/2017] [Indexed: 12/19/2022] Open
Abstract
DNA methylation and demethylation play a critical role in the regulation of the molecular pathogenesis of gliomas. Tet methylcytosine dioxygenase 1 (TET1) catalyses the sequential oxidation of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine, (5hmC) leading to eventual DNA demethylation. It has been reported that TET1 is a tumour suppressor in several cancers. However, whether TET1 plays a role in glioma development is largely unclear. Different glioma specimens and corresponding normal controls were collected to analyse the expression of TET1. At the same time, TET1 of glioma U251 cells was knocked down or overexpressed to observe its effect on glioma cell proliferation and invasion as well as autophagy level. Here, we reported that the expression of TET1 in glioma tissue was significantly lower than the corresponding non-tumour normal tissues, and the concentration of TET1 is negatively correlated with the glioma WHO classification. When TET1 gene in glioma U251 cells was knocked down by CRISPR/Caspase-9 system, the proliferation and invasive ability of U251 increased remarkably. But when TET1 was overexpressed in U251 cells, the proliferation and invasion were impaired. Following the down-expression of TET1, the level of autophagy in U251 cells decreased accordingly.However, when TET1 was overexpressed in U251 cells, the level of autophagy incraesed. Furthermore, bafilomycin A1 (Baf-A1) but not 3-methyladenine (3-MA) could decrease the autophagy level of TET1−/− U251 cells as the wild-type controls. It suggests that the tumour suppressor effect of TET1 seems to be mediated by regulating the level of autophagy, and the regulation of TET1 on autophagy is at an early stage.
Collapse
|
58
|
Yadav S, DE Sarkar N, Kumari N, Krishnani N, Kumar A, Mittal B. Targeted Gene Sequencing of Gallbladder Carcinoma Identifies High-impact Somatic and Rare Germline Mutations. Cancer Genomics Proteomics 2017; 14:495-506. [PMID: 29109099 PMCID: PMC6070325 DOI: 10.21873/cgp.20059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 10/05/2017] [Accepted: 10/09/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gallbladder carcinoma (GBC) is a subtype of biliary tract malignancy with poor prognosis and high fatality rate. The present study was designed to uncover somatic and rare germline mutations in GBC to reveal the disease biology and understand the clinical importance of mutation profile in terms of prognostics and actionability. MATERIALS AND METHODS We performed ultra-deep sequencing across 409 cancer-related genes in 11 GBC patients of North-Indian descent. NGS data analysis was performed using Ion Reporter and several other publicly available resources and databases. RESULTS We identified 184 nonsynonymous somatic and 60 rare germline mutations in bona-fide cancer drivers such as SMAD family member 4 (SMAD4), lysine methyltransferase 2C (KMT2C), and tumor protein p53 (TP53). All the early-onset cases or hypermutated cases harbored mutation(s) in critical DNA-repair genes. Additionally, we detected 9 novel genes with high-impact somatic mutations in GBC. CONCLUSION Our results indicated the significance of inherited rare germline mutations in DNA-repair pathway genes in addition to acquired somatic mutations in GB carcinogenesis.
Collapse
Affiliation(s)
- Saurabh Yadav
- Department of Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Navonil DE Sarkar
- Human Biology Division, Fred Hutchinson Cancer Research Centre, Seattle, WA, U.S.A
| | - Niraj Kumari
- Department of Pathology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Narendra Krishnani
- Department of Pathology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Ashok Kumar
- Department of Surgical Gastroenterology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Balraj Mittal
- Department of Genetics, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| |
Collapse
|
59
|
Ohara A, Takahashi Y, Kondo M, Okuda Y, Takeda S, Kushida M, Kobayashi K, Sumida K, Yamada T. Candidate genes responsible for early key events of phenobarbital-promoted mouse hepatocellular tumorigenesis based on differentiation of regulating genes between wild type mice and humanized chimeric mice. Toxicol Res (Camb) 2017; 6:795-813. [PMID: 30090543 PMCID: PMC6062386 DOI: 10.1039/c7tx00163k] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/23/2017] [Indexed: 12/12/2022] Open
Abstract
Phenobarbital (PB) is a nongenotoxic hepatocellular carcinogen in rodents. PB induces hepatocellular tumors by activating the constitutive androstane receptor (CAR). Some previous research has suggested the possible involvement of epigenetic regulation in PB-promoted hepatocellular tumorigenesis, but the details of its molecular mechanism are not fully understood. In the present study, comprehensive analyses of DNA methylation, hydroxymethylation and gene expression using microarrays were performed in mouse hepatocellular adenomas induced by a single 90 mg kg-1 intraperitoneal injection dose of diethylnitrosamine (DEN) followed by 500 ppm PB in the diet for 27 weeks. DNA modification and expression of hundreds of genes are coordinately altered in PB-induced mouse hepatocellular adenomas. Of these, gene network analysis showed alterations of CAR signaling and tumor development-related genes. Pathway enrichment analysis revealed that differentially methylated or hydroxymethylated genes belong mainly to pathways involved in development, immune response and cancer cells in contrast to differentially expressed genes belonging primarily to the cell cycle. Furthermore, overlap was evaluated between the genes with altered expression levels with 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) alterations in mouse hepatocellular adenoma induced by DEN/PB and the genes with altered expression levels in the liver of CD-1 mice or humanized chimeric mice treated with PB for 7 days. With the integration of transcriptomic and epigenetic approaches, we detected candidate genes responsible for early key events of PB-promoted mouse hepatocellular tumorigenesis. Interestingly, these genes did not overlap with genes altered by the PB treatment of humanized chimeric mice, thus suggesting a species difference between the effects of PB in mouse and human hepatocytes.
Collapse
Affiliation(s)
- Ayako Ohara
- Environmental Health Science Laboratory , Sumitomo Chemical Co. , Ltd. , 1-98 , 3-Chome , Kasugade-Naka , Konohana-ku , Osaka 554-8558 , Japan . ; ; Tel: +81-66466-5322
| | - Yasuhiko Takahashi
- Environmental Health Science Laboratory , Sumitomo Chemical Co. , Ltd. , 1-98 , 3-Chome , Kasugade-Naka , Konohana-ku , Osaka 554-8558 , Japan . ; ; Tel: +81-66466-5322
| | - Miwa Kondo
- Environmental Health Science Laboratory , Sumitomo Chemical Co. , Ltd. , 1-98 , 3-Chome , Kasugade-Naka , Konohana-ku , Osaka 554-8558 , Japan . ; ; Tel: +81-66466-5322
| | - Yu Okuda
- Environmental Health Science Laboratory , Sumitomo Chemical Co. , Ltd. , 1-98 , 3-Chome , Kasugade-Naka , Konohana-ku , Osaka 554-8558 , Japan . ; ; Tel: +81-66466-5322
| | - Shuji Takeda
- Environmental Health Science Laboratory , Sumitomo Chemical Co. , Ltd. , 1-98 , 3-Chome , Kasugade-Naka , Konohana-ku , Osaka 554-8558 , Japan . ; ; Tel: +81-66466-5322
| | - Masahiko Kushida
- Environmental Health Science Laboratory , Sumitomo Chemical Co. , Ltd. , 1-98 , 3-Chome , Kasugade-Naka , Konohana-ku , Osaka 554-8558 , Japan . ; ; Tel: +81-66466-5322
| | - Kentaro Kobayashi
- Environmental Health Science Laboratory , Sumitomo Chemical Co. , Ltd. , 1-98 , 3-Chome , Kasugade-Naka , Konohana-ku , Osaka 554-8558 , Japan . ; ; Tel: +81-66466-5322
| | - Kayo Sumida
- Environmental Health Science Laboratory , Sumitomo Chemical Co. , Ltd. , 1-98 , 3-Chome , Kasugade-Naka , Konohana-ku , Osaka 554-8558 , Japan . ; ; Tel: +81-66466-5322
| | - Tomoya Yamada
- Environmental Health Science Laboratory , Sumitomo Chemical Co. , Ltd. , 1-98 , 3-Chome , Kasugade-Naka , Konohana-ku , Osaka 554-8558 , Japan . ; ; Tel: +81-66466-5322
| |
Collapse
|
60
|
Liebig M, Hassanzada A, Kämmerling M, Genz B, Vollmar B, Abshagen K. Microcirculatory disturbances and cellular changes during progression of hepatic steatosis to liver tumors. Exp Biol Med (Maywood) 2017; 243:1-12. [PMID: 29065724 DOI: 10.1177/1535370217738730] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease is closely associated with metabolic syndrome and comprises a pathological spectrum of liver disease ranging from steatosis to steatohepatitis and can progress to fibrosis/cirrhosis and hepatocellular carcinoma. In 2013, a mouse model was described that mimics non-alcoholic fatty liver disease progression from steatohepatitis to tumors in a short time span and with high incidence. As microcirculatory disturbances play a crucial role in liver disease, the suitability of the steatosis-inflammation-tumor model for microcirculatory studies was assessed. Herein, we present a comprehensive view on morphological, microvascular, cellular, and functional aspects of non-alcoholic fatty liver disease progression in the steatosis-inflammation-tumor model using intravital microscopy, biochemical, and histological techniques. Mice develop steatohepatitis, mild fibrosis, and liver tumors at ages of 6, 12, and 20 weeks, respectively. Non-alcoholic fatty liver disease progression was accompanied by several general aspects of disease severity like increasing liver/body weight index, non-alcoholic fatty liver disease activity score, and hepatocellular apoptosis. Intravital microscopic analysis revealed significant changes in hepatic microcirculation with increasing structural alterations, elevated leukocyte adherence, and impaired nutritive perfusion. Non-alcoholic fatty liver disease was further characterized by a lower sinusoidal density with a striking rise at 20 weeks. The characteristic microcirculatory changes make the model a convenient tool for analysis of microcirculation during progression from steatosis to liver tumor. Impact statement Significant alterations of microcirculation contribute to progression of NAFLD, a chronic liver disease with increasing medical and socio-economic impact. Characterization of microcirculation in a NAFLD model reflecting all relevant stages of disease progression was still missing. Thus, we evaluated microcirculatory and cellular changes in a steatosis-inflammation-tumor model using in vivo microscopy. Analyses revealed increasing structural alterations, elevated leukocyte-endothelial interaction, and impaired nutritive perfusion. Thus, this model is suitable for further studies investigating therapeutic approaches targeting these progressive microcirculatory disturbances.
Collapse
Affiliation(s)
- Marie Liebig
- 1 Institute for Experimental Surgery, University Medicine Rostock, Rostock 18057, Germany
| | - Alireza Hassanzada
- 1 Institute for Experimental Surgery, University Medicine Rostock, Rostock 18057, Germany
| | - Malte Kämmerling
- 1 Institute for Experimental Surgery, University Medicine Rostock, Rostock 18057, Germany
| | - Berit Genz
- 1 Institute for Experimental Surgery, University Medicine Rostock, Rostock 18057, Germany.,2 QIMR Berghofer Medical Research Institute, Brisbane QLD 4006, Australia
| | - Brigitte Vollmar
- 1 Institute for Experimental Surgery, University Medicine Rostock, Rostock 18057, Germany
| | - Kerstin Abshagen
- 1 Institute for Experimental Surgery, University Medicine Rostock, Rostock 18057, Germany
| |
Collapse
|
61
|
Chen H, Cai W, Chu ESH, Tang J, Wong CC, Wong SH, Sun W, Liang Q, Fang J, Sun Z, Yu J. Hepatic cyclooxygenase-2 overexpression induced spontaneous hepatocellular carcinoma formation in mice. Oncogene 2017; 36:4415-4426. [PMID: 28346420 PMCID: PMC5543258 DOI: 10.1038/onc.2017.73] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 02/07/2017] [Accepted: 02/07/2017] [Indexed: 12/12/2022]
Abstract
Cyclooxygenase (COX)-2 is upregulated in hepatocellular carcinoma (HCC). However, the direct causative effect of COX-2 in spontaneous HCC formation remains unknown. We thus investigate the role and molecular pathogenesis of COX-2 in HCC by using liver-specific COX-2 transgenic (TG) mice. We found spontaneous HCC formation with elevated inflammatory infiltrates and neovessels in male TG mice (3/21, 14.3%), but not in any of male WT mice (0/19). Reduced representation bisulfite sequencing (RRBS) and gene expression microarrays were performed in the HCC tumor and non-HCC liver tissues to investigate the molecular mechanisms of COX-2-driven HCC. By RRBS, DNA promoter hypermethylation was identified in HCC from TG mice. Induction of promoter hypermethylation was associated with reduced tet methylcytosine dioxygenase 1 (TET1) expression by COX-2. TET1 could catalyze the conversion of 5-methylcytosine into 5-hydroxymethylcytosine (5hmC) and prevents DNA hypermethylation. In keeping with this, loss of 5hmC was demonstrated in COX-2-induced HCC. Consistently, COX-2 overexpression in human HCC cell lines could reduce both TET1 expression and 5hmc levels. Integrative analyses of DNA methylation and gene expression profiles further identified significantly downregulated genes including LTBP1, ADCY5 and PRKCZ by promoter methylation in COX-2-induced HCC. Reduced expression of LTBP1, ADCY5 and PRKCZ by promoter hypermethylation was further validated in human HCCs. Bio-functional investigation revealed that LTBP1 inhibited cell proliferation in HCC cell lines, suggesting its potential role as a tumor suppressor in HCC. Gene expression microarrays revealed that signaling cascades (AKT (protein kinase B), STK33 (Serine/Threonine kinase 33) and MTOR (mechanistic target of rapamycin) pathways) were enriched in COX-2-induced HCC. In conclusion, this study demonstrated for the first time that enhanced COX-2 expression in hepatocytes is sufficient to induce HCC through inducing promoter hypermethylation by reducing TET1, silencing tumor-suppressive genes and activating key oncogenic pathways. Inhibition of COX-2 represents a mechanism-based target for HCC prevention.
Collapse
Affiliation(s)
- H Chen
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - W Cai
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - E S H Chu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - J Tang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - C-C Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - S H Wong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - W Sun
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Q Liang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - J Fang
- State Key Laboratory for Oncogenes and Related Genes, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Z Sun
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - J Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
62
|
MicroRNA-34a Encapsulated in Hyaluronic Acid Nanoparticles Induces Epigenetic Changes with Altered Mitochondrial Bioenergetics and Apoptosis in Non-Small-Cell Lung Cancer Cells. Sci Rep 2017. [PMID: 28623259 PMCID: PMC5473901 DOI: 10.1038/s41598-017-02816-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Therapies targeting epigenetic changes for cancer treatment are in Phase I/II trials; however, all of these target only nuclear DNA. Emerging evidence suggests presence of methylation marks on mitochondrial DNA (mtDNA); but their contribution in cancer is unidentified. Expression of genes encoded on mtDNA are altered in cancer cells, along with increased glycolytic flux. Such glycolytic flux and elevated reactive oxygen species is supported by increased antioxidant; glutathione. MicroRNA-34a can translocate to mitochondria, mediate downstream apoptotic effects of tumor suppressor P53, and inhibit the antioxidant response element Nrf-2, resulting in depleted glutathione levels. Based on such strong rationale, we encapsulated microRNA-34a in our well-established Hyaluronic-Acid nanoparticles and delivered to cisplatin-sensitive and cisplatin-resistant A549-lung adenocarcinoma cells. Successful delivery and uptake in cells resulted in altered ATP levels, decreased glycolytic flux, Nrf-2 and glutathione levels, ultimately resulting in caspase-3 activation and apoptosis. Most important were the concurrent underlying molecular changes in epigenetic status of D-loop on the mtDNA and transcription of mtDNA-encoded genes. Although preliminary, we provide a novel therapeutic approach in form of altered mitochondrial bioenergetics and redox status of cancer cells with underlying changes in epigenetic status of mtDNA that can subsequently results in induction of cancer cell apoptosis.
Collapse
|
63
|
Abstract
Melanoma is a malignant tumor of melanocytes and is considered to be the most aggressive cancer among all skin diseases. The pathogenesis of melanoma has not been well documented, which may restrict the research and development of biomarkers and therapies. To date, several genetic and epigenetic factors have been identified as contributing to the development and progression of melanoma. Besides the findings on genetic susceptibilities, the recent progress in epigenetic studies has revealed that loss of the DNA hydroxymethylation mark, 5-hydroxymethylcytosine (5-hmC), along with high levels of DNA methylation at promoter regions of several tumor suppressor genes in melanoma, may serve as biomarkers for melanoma. Moreover, 5-Aza-2′-deoxycytidine, an epigenetic modifier causing DNA demethylation, and ten-eleven translocation family dioxygenase (TET), which catalyzes the generation of 5-hmC, demonstrate therapeutic potential in melanoma treatment. In this review, we will summarize the latest progress in research on DNA methylation/hydroxymethylation in melanoma, and we will discuss and provide insight for epigenetic biomarkers and therapies for melanoma. Particularly, we will discuss the role of DNA hydroxymethylation in melanoma infiltrating immune cells, which may also serve as a potential target for melanoma treatment.
Collapse
|
64
|
Wang Y, Hu H, Wang Q, Li Z, Zhu Y, Zhang W, Wang Y, Jiang H, Cheng J. The level and clinical significance of 5-hydroxymethylcytosine in oral squamous cell carcinoma: An immunohistochemical study in 95 patients. Pathol Res Pract 2017; 213:969-974. [PMID: 28554766 DOI: 10.1016/j.prp.2017.04.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 03/26/2017] [Accepted: 04/14/2017] [Indexed: 12/22/2022]
Abstract
Accumulating evidence has revealed that aberrant abundance of 5-hydroxymethylcytosine (5hmC) is critically involved in tumorigenesis. The aim of the present study was to investigate the level of 5hmC in primary oral squamous cell carcinoma (OSCC) and determine its clinical significance as well as prognostic value in predicting patients' outcomes. The expression levels of 5hmC in 95 human OSCC samples and 24 normal oral mucosa were evaluated by immunohistochemical staining. Moreover, the associations between the expression status of 5hmC and several clinicopathological parameters as well as patients' survival were further statistically assessed. Our immunohistochemical results revealed that 5hmC was significantly downregulated in a significant fraction of OSCC as compared their normal counterparts. However, elevated 5hmC level was found to be significantly associated with pathological grade and cervical node metastasis with P-values of 0.0239 and 0.0041, respectively. Results from Kaplan-Meier cumulative survival analyses indicated that high expression of 5hmC in OSCC was significantly associated with decreased overall survival, disease-free and disease-specific survival as compared to those with low 5hmC (Log-rank, P=0.0210, 0.0313, 0.0415, respectively). Furthermore, the univariate and multivariate survival analyses further identified the expression status of 5hmC as an independent prognostic factor affecting patients' survival. Taken together, our results reveal a significant decrease of 5hmC level in a large subset of OSCC. However, high level of 5hmC associates with tumor aggressive features and unfavorable prognosis in a fraction of OSCC patients.
Collapse
Affiliation(s)
- Yi Wang
- Jiangsu Key Institute of Stomatology, Nanjing Medical University, Nanjing, 210029, PR China; Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital, Nanjing Medical University, Nanjing, 210029, PR China
| | - Huijun Hu
- Jiangsu Key Institute of Stomatology, Nanjing Medical University, Nanjing, 210029, PR China
| | - Qiong Wang
- Jiangsu Key Institute of Stomatology, Nanjing Medical University, Nanjing, 210029, PR China; Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital, Nanjing Medical University, Nanjing, 210029, PR China
| | - Zhongwu Li
- Jiangsu Key Institute of Stomatology, Nanjing Medical University, Nanjing, 210029, PR China
| | - Yumin Zhu
- Jiangsu Key Institute of Stomatology, Nanjing Medical University, Nanjing, 210029, PR China
| | - Wei Zhang
- Department of Oral Pathology, Affiliated Stomatological Hospital, Nanjing Medical University, Nanjing, 210029, PR China
| | - Yanling Wang
- Jiangsu Key Institute of Stomatology, Nanjing Medical University, Nanjing, 210029, PR China
| | - Hongbing Jiang
- Jiangsu Key Institute of Stomatology, Nanjing Medical University, Nanjing, 210029, PR China.
| | - Jie Cheng
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital, Nanjing Medical University, Nanjing, 210029, PR China.
| |
Collapse
|
65
|
Terranova R, Vitobello A, Del Rio Espinola A, Wolf CR, Schwarz M, Thomson J, Meehan R, Moggs J. Progress in identifying epigenetic mechanisms of xenobiotic-induced non-genotoxic carcinogenesis. CURRENT OPINION IN TOXICOLOGY 2017. [DOI: 10.1016/j.cotox.2017.06.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
66
|
Thomson JP, Meehan RR. The application of genome-wide 5-hydroxymethylcytosine studies in cancer research. Epigenomics 2016; 9:77-91. [PMID: 27936926 DOI: 10.2217/epi-2016-0122] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Early detection and characterization of molecular events associated with tumorgenesis remain high priorities. Genome-wide epigenetic assays are promising diagnostic tools, as aberrant epigenetic events are frequent and often cancer specific. The deposition and analysis of multiple patient-derived cancer epigenomic profiles contributes to our appreciation of the underlying biology; aiding the detection of novel identifiers for cancer subtypes. Modifying enzymes and co-factors regulating these epigenetic marks are frequently mutated in cancers, and as epigenetic modifications themselves are reversible, this makes their study very attractive with respect to pharmaceutical intervention. Here we focus on the novel modified base, 5-hydoxymethylcytosine, and discuss how genome-wide 5-hydoxymethylcytosine profiling expedites our molecular understanding of cancer, serves as a lineage tracer, classifies the mode of action of potentially carcinogenic agents and clarifies the roles of potential novel cancer drug targets; thus assisting the development of new diagnostic/prognostic tools.
Collapse
Affiliation(s)
- John P Thomson
- MRC Human Genetics Unit, Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| | - Richard R Meehan
- MRC Human Genetics Unit, Institute of Genetics & Molecular Medicine, University of Edinburgh, Crewe Road, Edinburgh, EH4 2XU, UK
| |
Collapse
|
67
|
Zeng W, Zhu XN, Luo W, Chen QY, Zhang Y, Tan SK. Clinical significance of expression of CMTM7 in hepatocellular carcinoma. Shijie Huaren Xiaohua Zazhi 2016; 24:4568-4575. [DOI: 10.11569/wcjd.v24.i34.4568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To detect the expression of CMTM7 in hepatocellular carcinoma (HCC) and to analyze its clinical significance.
METHODS Tissue microarray and immunohistochemistry method were used to detect 75 pairs of HCC tissues and tumor-adjacent normal tissues. The relationship of CMTM7 expression with clinical pathological features and overall survival of postoperative patients was analyzed statistically.
RESULTS The expression rate of CMTM7 in HCC tissues (20.00%, 15/75) was significantly lower than that in paired para-carcinoma normal tissues (81.33%, 61/75; P < 0.05). The expression of CMTM7 in HCC had no obvious correlation with gender, age, smoking history, drinking history, family history of hepatitis B virus infection or tumor, tumor number, pathological classification, or clinical stage. However, CMTM7 expression had a significant correlation with liver cirrhosis, distant metastasis, tumor size, TNM stage, and α-fetoprotein levels (P < 0.05). The survival time of postoperative HCC patients in the CMTM7 negative group was obviously lower than that of patients in the CMTM7 positive group, suggesting that CMTM7 expression may be related to the prognosis of HCC patients.
CONCLUSION The down-regulated expression of CMTM7 may be closely related with the occurrence and development of HCC.
Collapse
|
68
|
Zhou D, Hlady RA, Schafer MJ, White TA, Liu C, Choi JH, Miller JD, Roberts LR, LeBrasseur NK, Robertson KD. High fat diet and exercise lead to a disrupted and pathogenic DNA methylome in mouse liver. Epigenetics 2016; 12:55-69. [PMID: 27858497 DOI: 10.1080/15592294.2016.1261239] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
High-fat diet consumption and sedentary lifestyle elevates risk for obesity, non-alcoholic fatty liver disease, and cancer. Exercise training conveys health benefits in populations with or without these chronic conditions. Diet and exercise regulate gene expression by mediating epigenetic mechanisms in many tissues; however, such effects are poorly documented in the liver, a central metabolic organ. To dissect the consequences of diet and exercise on the liver epigenome, we measured DNA methylation, using reduced representation bisulfite sequencing, and transcription, using RNA-seq, in mice maintained on a fast food diet with sedentary lifestyle or exercise, compared with control diet with and without exercise. Our analyses reveal that genome-wide differential DNA methylation and expression of gene clusters are induced by diet and/or exercise. A combination of fast food and exercise triggers extensive gene alterations, with enrichment of carbohydrate/lipid metabolic pathways and muscle developmental processes. Through evaluation of putative protective effects of exercise on diet-induced DNA methylation, we show that hypermethylation is effectively prevented, especially at promoters and enhancers, whereas hypomethylation is only partially attenuated. We assessed diet-induced DNA methylation changes associated with liver cancer-related epigenetic modifications and identified significant increases at liver-specific enhancers in fast food groups, suggesting partial loss of liver cell identity. Hypermethylation at a subset of gene promoters was associated with inhibition of tissue development and promotion of carcinogenic processes. Our study demonstrates extensive reprogramming of the epigenome by diet and exercise, emphasizing the functional relevance of epigenetic mechanisms as an interface between lifestyle modifications and phenotypic alterations.
Collapse
Affiliation(s)
- Dan Zhou
- a Department of Molecular Pharmacology and Experimental Therapeutics , Mayo Clinic , Rochester , MN , USA
| | - Ryan A Hlady
- a Department of Molecular Pharmacology and Experimental Therapeutics , Mayo Clinic , Rochester , MN , USA
| | - Marissa J Schafer
- b Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester , MN , USA.,c Department of Physical Medicine & Rehabilitation , Mayo Clinic , Rochester , MN , USA
| | - Thomas A White
- b Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester , MN , USA
| | - Chen Liu
- d Department of Pathology and Laboratory Medicine , Rutgers University , NJ , USA
| | - Jeong-Hyeon Choi
- e Department of Applied Research , Marine Biodiversity Institute of Korea , Korea
| | - Jordan D Miller
- b Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester , MN , USA.,f Department of Surgery , Mayo Clinic , Rochester , MN , USA
| | - Lewis R Roberts
- g Mayo Clinic Cancer Center , Mayo Clinic , Rochester , MN , USA.,h Division of Gastroenterology and Hepatology, Mayo Clinic , Rochester , MN , USA
| | - Nathan K LeBrasseur
- b Robert and Arlene Kogod Center on Aging, Mayo Clinic , Rochester , MN , USA.,c Department of Physical Medicine & Rehabilitation , Mayo Clinic , Rochester , MN , USA
| | - Keith D Robertson
- a Department of Molecular Pharmacology and Experimental Therapeutics , Mayo Clinic , Rochester , MN , USA.,g Mayo Clinic Cancer Center , Mayo Clinic , Rochester , MN , USA.,i Center for Individualized Medicine, Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
69
|
Zheng L, Zhai Y, Li N, Ma F, Zhu H, Du X, Li G, Hua J. The Modification of Tet1 in Male Germline Stem Cells and Interact with PCNA, HDAC1 to promote their Self-renewal and Proliferation. Sci Rep 2016; 6:37414. [PMID: 27857213 PMCID: PMC5114665 DOI: 10.1038/srep37414] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 10/25/2016] [Indexed: 12/14/2022] Open
Abstract
Epigenetic modification plays key roles in spermatogenesis, especially DNA methylation dynamic is important in sustaining normal spermatogenesis. Ten-eleven translocation 1 (Tet1) is not only a key demethylase, which works in specific gene regions, but also crosstalks with partners to regulate epigenetic progress as protein complexes. Dairy goat is an important livestock in China, while the unstable culture system in vitro inhibits optimization of new dairy goat species. The study of epigenetic modification in male germline stem cells (mGSCs) is beneficial to the optimization of adult stem cell culture system in vitro, and the improvement of sperm quality and breeding of selected livestock. In our study, we not only analyzed the morphology, gene expression, DNA methylation and histone methylation dynamic in mouse Tet1 (mTet1) modified mGSCs, we also analyzed the stemness ability by in vivo transplantation and explored the functional mechanism of Tet1 in dairy goat mGSCs. The results showed mTet1 modified mGSCs had better self-renewal and proliferation ability than wild-type mGSCs, mTet1 could also up-regulate JMJD3 to decrease H3K27me3, which also showed to suppress the MEK-ERK pathway. Furthermore, Co-IP analysis demonstrated that TET1 interact with PCNA and HDAC1 by forming protein complexes to comprehensively regulate dairy goat mGSCs and spermatogenesis.
Collapse
Affiliation(s)
- Liming Zheng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Yuanxin Zhai
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Na Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Fanglin Ma
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Haijing Zhu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Xiaomin Du
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Guangpeng Li
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University, Hohhot, 010021, China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| |
Collapse
|
70
|
Derisking Drug-Induced Carcinogenicity for Novel Therapeutics. Trends Cancer 2016; 2:398-408. [DOI: 10.1016/j.trecan.2016.07.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/07/2016] [Accepted: 07/08/2016] [Indexed: 12/21/2022]
|