1
|
Zhou X, Zhou X, Li J, He Y, Qiu S, Xu Y, Liu Z, Yao Y, Liu J, Wen Y, Xie S, Chen J, Liu L, Ou Z, Cai C, Lin J, Lei B, Zou F. Bclaf1 mediates super-enhancer-driven activation of POLR2A to enhance chromatin accessibility in nitrosamine-induced esophageal carcinogenesis. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138218. [PMID: 40220379 DOI: 10.1016/j.jhazmat.2025.138218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/30/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Gene-environment interactions are pivotal contributors to nitrosamine-induced esophageal carcinogenesis. While genetic mechanisms in esophageal carcinoma (ESCA) are well-defined, epigenetic drivers remain elusive. This study identifies a novel mechanism of epigenetic regulation centered on B-cell lymphoma-2-associated transcription factor 1 (Bclaf1) in nitrosamine-induced (Methylnitronitrosoguanidine, MNNG) esophageal carcinogenesis. In nitrosamine-induced malignant transformation cells (MNNG-M), Bclaf1 expression is progressively increased with malignancy, and elevated Bclaf1 levels are correlated with poor prognosis in ESCA patients. Functionally, Bclaf1 significantly promotes the abnormal proliferation of MNNG-M and ESCA cells in vitro and in vivo. Mechanistically, transposase-accessible chromatin sequencing (ATAC-seq) results suggest that Bclaf1 silencing markedly reduces chromatin accessibility, thereby impairing the synthesis of newly transcribed RNA. Bclaf1 activates RNA polymerase II subunit POLR2A to promote chromatin accessibility through distinct transcriptional and splicing mechanisms. More specifically, cleavage under targets and tagmentation (CUT&Tag) assays revealed Bclaf1/P300/H3K27ac co-recruitment at the POLR2A promoter, driving transcription via the E2/E3 elements of the POLR2A super-enhancer. Additionally, RNA-binding protein immunoprecipitation (RIP) assays demonstrated that the Bclaf1 cofactor, small nuclear ribonucleoprotein polypeptide A (SNRPA), interacts with pre-POLR2A to regulate its splicing. Collectively, our study reveals that Bclaf1 facilitates nitrosamine-induced ESCA by controlling POLR2A transcriptional and splicing activities, providing novel insight for early detection and intervention.
Collapse
Affiliation(s)
- Xiangjun Zhou
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Xueqiong Zhou
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China.
| | - Jun Li
- Department of thoracic surgery, The third affiliated hospital of Southern Medical University, Guangzhou, Guangdong 510630, China
| | - Yingzheng He
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Shizhen Qiu
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Ye Xu
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, China
| | - Zeyu Liu
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Yina Yao
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Jia Liu
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Ying Wen
- Guangzhou Women and Children's Medical Centre, Guangzhou Medical University Institute of Pediatrics, 9 Jinsui Road, Guangzhou 510623, China
| | - Sitong Xie
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Jialong Chen
- Department of Preventive Medicine, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Linhua Liu
- Department of Preventive Medicine, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Zejin Ou
- Key Laboratory of Occupational Environment and Health, Guangzhou Twelfth People's Hospital, Guangzhou, 510620, China
| | - Chunqing Cai
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Junyuan Lin
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China
| | - Bingxi Lei
- Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Fei Zou
- Department of Occupational Health and Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, China.
| |
Collapse
|
2
|
Kim JY, Yang AY, Kim K, Kwon HH, Leem J, Kim YA. Pharmacological inhibition of p300 ameliorates steatosis, inflammation, and fibrosis in mice with non-alcoholic steatohepatitis. Heliyon 2024; 10:e30908. [PMID: 38774067 PMCID: PMC11107220 DOI: 10.1016/j.heliyon.2024.e30908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 05/24/2024] Open
Abstract
The histone acetyltransferase p300 plays a pivotal role in regulating gene expression and cellular phenotype through epigenetic mechanisms. It significantly influences lipid metabolism, which is a key factor in the pathogenesis of non-alcoholic steatohepatitis (NASH), by modulating the transcription of genes involved in lipid synthesis and accumulation. This study aimed to investigate the protective potential of inhibiting p300 in NASH. Male C57BL/6J mice were subjected to a methionine- and choline-deficient (MCD) diet for 4 weeks to induce NASH, and during this period, the p300 inhibitor C646 (10 mg/kg) was administered three times a week. C646 treatment reduced the elevation of p300 expression and histone H3 acetylation, leading to a decrease in liver injury markers in the serum and an improvement in the histological abnormalities observed in MCD diet-fed mice. C646 also reduced lipid accumulation by modulating de novo lipogenesis and suppressed inflammation, including cytokine overproduction and macrophage infiltration. Furthermore, C646 mitigated liver fibrosis and myofibroblast accumulation. This protective effect was achieved through the inhibition of apoptosis by reducing p53 and Bax expression and the suppression of ferroptosis by decreasing lipid peroxidation while enhancing antioxidant defenses. Additionally, C646 alleviated endoplasmic reticulum stress, as evidenced by the downregulation of unfolded protein response signaling molecules. These results highlight the potential of p300 as a therapeutic target for NASH.
Collapse
Affiliation(s)
- Jung-Yeon Kim
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Ah Young Yang
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Kiryeong Kim
- Department of Internal Medicine, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Hyun Hee Kwon
- Department of Internal Medicine, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| | - Yun-A Kim
- Department of Family Medicine, School of Medicine, Daegu Catholic University, Daegu, Republic of Korea
| |
Collapse
|
3
|
Karabegović I, Maas SCE, Shuai Y, Ikram MA, Stricker B, Aerts J, Brusselle G, Lahousse L, Voortman T, Ghanbari M. Smoking-related dysregulation of plasma circulating microRNAs: the Rotterdam study. Hum Genomics 2023; 17:61. [PMID: 37430296 DOI: 10.1186/s40246-023-00504-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 06/26/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are post-transcriptional regulators of gene expression. Differential miRNA expression, which is widely shown to be associated with the pathogenesis of various diseases, can be influenced by lifestyle factors, including smoking. This study aimed to investigate the plasma miRNA signature of smoking habits, the potential effect of smoking cessation on miRNA levels, and relate the findings with lung cancer incidence. RESULTS A targeted RNA-sequencing approach measured plasma miRNA levels in 2686 participants from the population-based Rotterdam study cohort. The association between cigarette smoking (current versus never) and 591 well-expressed miRNAs was assessed via adjusted linear regression models, identifying 41 smoking-associated miRNAs that passed the Bonferroni-corrected threshold (P < 0.05/591 = 8.46 × 10-5). Moreover, we found 42 miRNAs with a significant association (P < 8.46 × 10-5) between current (reference group) and former smokers. Then, we used adjusted linear regression models to explore the effect of smoking cessation time on miRNA expression levels. The expression levels of two miRNAs were significantly different within 5 years of cessation (P < 0.05/41 = 1.22 × 10-3) from current smokers, while for cessation time between 5 and 15 years we found 19 miRNAs to be significantly different from current smokers, and finally, 38 miRNAs were significantly different after more than 15 years of cessation time (P < 1.22 × 10-3). These results imply the reversibility of the smoking effect on plasma levels of at least 38 out of the 41 smoking-miRNAs following smoking cessation. Next, we found 8 out of the 41 smoking-related miRNAs to be nominally associated (P < 0.05) with the incidence of lung cancer. CONCLUSIONS This study demonstrates smoking-related dysregulation of plasma miRNAs, which might have a potential for reversibility when comparing different smoking cessation groups. The identified miRNAs are involved in several cancer-related pathways and include 8 miRNAs associated with lung cancer incidence. Our results may lay the groundwork for further investigation of miRNAs as potential mechanism linking smoking, gene expression and cancer.
Collapse
Affiliation(s)
- Irma Karabegović
- Department of Epidemiology, Erasmus MC University Medical Center, Dr Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Silvana C E Maas
- Department of Epidemiology, Erasmus MC University Medical Center, Dr Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- Vall d'Hebron Institute of Oncology (VHIO), 08035, Barcelona, Spain
| | - Yu Shuai
- Department of Epidemiology, Erasmus MC University Medical Center, Dr Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Dr Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Bruno Stricker
- Department of Epidemiology, Erasmus MC University Medical Center, Dr Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Joachim Aerts
- Department of Pulmonary Medicine, Erasmus MC University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Guy Brusselle
- Department of Respiratory Medicine, Ghent University Hospital, 9000, Ghent, Belgium
| | - Lies Lahousse
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, 9000, Ghent, Belgium
| | - Trudy Voortman
- Department of Epidemiology, Erasmus MC University Medical Center, Dr Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
- Division of Human Nutrition and Health, Wageningen University and Research, 6708 PB, Wageningen, The Netherlands
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC University Medical Center, Dr Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
4
|
Ortega MA, Álvarez-Mon MA, García-Montero C, Fraile-Martínez Ó, Monserrat J, Martinez-Rozas L, Rodríguez-Jiménez R, Álvarez-Mon M, Lahera G. Microbiota-gut-brain axis mechanisms in the complex network of bipolar disorders: potential clinical implications and translational opportunities. Mol Psychiatry 2023; 28:2645-2673. [PMID: 36707651 PMCID: PMC10615769 DOI: 10.1038/s41380-023-01964-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 01/02/2023] [Accepted: 01/13/2023] [Indexed: 01/28/2023]
Abstract
Bipolar disorders (BD) represent a severe leading disabling mental condition worldwide characterized by episodic and often progressive mood fluctuations with manic and depressive stages. The biological mechanisms underlying the pathophysiology of BD remain incompletely understood, but it seems that there is a complex picture of genetic and environmental factors implicated. Nowadays, gut microbiota is in the spotlight of new research related to this kind of psychiatric disorder, as it can be consistently related to several pathophysiological events observed in BD. In the context of the so-called microbiota-gut-brain (MGB) axis, it is shown to have a strong influence on host neuromodulation and endocrine functions (i.e., controlling the synthesis of neurotransmitters like serotonin or mediating the activation of the hypothalamic-pituitary-adrenal axis), as well as in modulation of host immune responses, critically regulating intestinal, systemic and brain inflammation (neuroinflammation). The present review aims to elucidate pathophysiological mechanisms derived from the MGB axis disruption and possible therapeutic approaches mainly focusing on gut microbiota in the complex network of BD. Understanding the mechanisms of gut microbiota and its bidirectional communication with the immune and other systems can shed light on the discovery of new therapies for improving the clinical management of these patients. Besides, the effect of psychiatric drugs on gut microbiota currently used in BD patients, together with new therapeutical approaches targeting this ecosystem (dietary patterns, probiotics, prebiotics, and other novelties) will also be contemplated.
Collapse
Affiliation(s)
- Miguel A Ortega
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain.
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain.
| | - Miguel Angel Álvarez-Mon
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Psychiatry and Mental Health, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Óscar Fraile-Martínez
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Lucia Martinez-Rozas
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Roberto Rodríguez-Jiménez
- Department of Legal Medicine and Psychiatry, Complutense University, Madrid, Spain
- Institute for Health Research 12 de Octubre Hospital, (Imas 12)/CIBERSAM (Biomedical Research Networking Centre in Mental Health), Madrid, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine, University Hospital Príncipe de Asturias (CIBEREHD), Alcalá de Henares, Spain
- Psychiatry Service, Center for Biomedical Research in the Mental Health Network, University Hospital Príncipe de Asturias, Alcalá de Henares, Spain
| | - Guillermo Lahera
- Department of Medicine and Medical Specialities, University of Alcala, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Psychiatry Service, Center for Biomedical Research in the Mental Health Network, University Hospital Príncipe de Asturias, Alcalá de Henares, Spain
| |
Collapse
|
5
|
Zhu J, Li G, Huang Q, Wen J, Deng Y, Jiang J. TET3-mediated DNA demethylation and chromatin remodeling regulate T-2 toxin-induced human CYP1A1 expression and cytotoxicity in HepG2 cells. Biochem Pharmacol 2023; 211:115506. [PMID: 36948362 DOI: 10.1016/j.bcp.2023.115506] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/24/2023]
Abstract
T-2 toxin is a hazardous environmental pollutant that poses a risk to both farm animals and humans. Our previous research has reported that T-2 toxin highly induced the expression of human cytochrome P450 1A1 (CYP1A1), which may be a representative inducible marker of T-2 toxin and mediate the toxicity of T-2 toxin. In this study, we found that T-2 toxin decreased the DNA methylation levels of the CpG islands on the CYP1A1 promoter by inducing the expression of eleven translocation family protein 3 (TET3) and facilitating its binding to the promoter. These DNA methylation changes then generated an activated chromatin structure on the CYP1A1 promoter by releasing the repressor complex methyl-binding protein 2 (MeCP2) and histone deacetylase 2 (HDAC2), increasing the active histone modification markers, including H3K4ac, H3K9ac and H3K14ac, and facilitating RNA pol II and NRF1/Sp1 recruitment, which ultimately led to the transcriptional activation of CYP1A1. Interestingly, TET3-mediated CYP1A1 induction enhanced the cytotoxicity of T-2 toxin through inhibiting cell proliferation. Our results demonstrate that T-2 toxin-induced CYP1A1 expression is detrimental to cells and clearly show how T-2 toxin inhibits cell proliferation through regulating CYP1A1 expression from an epigenetic perspective. The findings broaden our current knowledge of the epigenetic mechanisms regulating environmental factors-induced CYP1A1 expression and cytotoxicity. TET3 may serve as a potential new target for toxicogenic detoxification.
Collapse
Affiliation(s)
- Jiahui Zhu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, Guangdong, P. R. China; Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Guihong Li
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, Guangdong, P. R. China; Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Qiang Huang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, Guangdong, P. R. China; Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Jikai Wen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, Guangdong, P. R. China; Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China
| | - Yiqun Deng
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, Guangdong, P. R. China; Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China.
| | - Jun Jiang
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, Guangdong, P. R. China; Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou 510642, Guangdong, P. R. China.
| |
Collapse
|
6
|
Yazar V, Dawson VL, Dawson TM, Kang SU. DNA Methylation Signature of Aging: Potential Impact on the Pathogenesis of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2023; 13:145-164. [PMID: 36710687 PMCID: PMC10041453 DOI: 10.3233/jpd-223517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Regulation of gene expression by epigenetic modifications means lasting and heritable changes in the function of genes without alterations in the DNA sequence. Of all epigenetic mechanisms identified thus far, DNA methylation has been of particular interest in both aging and age-related disease research over the last decade given the consistency of site-specific DNA methylation changes during aging that can predict future health and lifespan. An increasing line of evidence has implied the dynamic nature of DNA (de)methylation events that occur throughout the lifespan has a role in the pathophysiology of aging and age-associated neurodegenerative conditions, including Parkinson's disease (PD). In this regard, PD methylome shows, to some extent, similar genome-wide changes observed in the methylome of healthy individuals of matching age. In this review, we start by providing a brief overview of studies outlining global patterns of DNA methylation, then its mechanisms and regulation, within the context of aging and PD. Considering diverging lines of evidence from different experimental and animal models of neurodegeneration and how they combine to shape our current understanding of tissue-specific changes in DNA methylome in health and disease, we report a high-level comparison of the genomic methylation landscapes of brain, with an emphasis on dopaminergic neurons in PD and in natural aging. We believe this will be particularly useful for systematically dissecting overlapping genome-wide alterations in DNA methylation during PD and healthy aging, and for improving our knowledge of PD-specific changes in methylation patterns independent of aging process.
Collapse
Affiliation(s)
- Volkan Yazar
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA, USA
| | - Sung-Ung Kang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
7
|
Mourtzi N, Sertedaki A, Charmandari E. Glucocorticoid Signaling and Epigenetic Alterations in Stress-Related Disorders. Int J Mol Sci 2021; 22:5964. [PMID: 34073101 PMCID: PMC8198182 DOI: 10.3390/ijms22115964] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/26/2021] [Accepted: 05/29/2021] [Indexed: 12/31/2022] Open
Abstract
Stress is defined as a state of threatened or perceived as threatened homeostasis. The well-tuned coordination of the stress response system is necessary for an organism to respond to external or internal stressors and re-establish homeostasis. Glucocorticoid hormones are the main effectors of stress response and aberrant glucocorticoid signaling has been associated with an increased risk for psychiatric and mood disorders, including schizophrenia, post-traumatic stress disorder and depression. Emerging evidence suggests that life-stress experiences can alter the epigenetic landscape and impact the function of genes involved in the regulation of stress response. More importantly, epigenetic changes induced by stressors persist over time, leading to increased susceptibility for a number of stress-related disorders. In this review, we discuss the role of glucocorticoids in the regulation of stress response, the mechanism through which stressful experiences can become biologically embedded through epigenetic alterations, and we underline potential associations between epigenetic changes and the development of stress-related disorders.
Collapse
Affiliation(s)
- Niki Mourtzi
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (N.M.); (A.S.)
| | - Amalia Sertedaki
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (N.M.); (A.S.)
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens Medical School, 11527 Athens, Greece; (N.M.); (A.S.)
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
8
|
Kadiyala P, Carney SV, Gauss JC, Garcia-Fabiani MB, Haase S, Alghamri MS, Núñez FJ, Liu Y, Yu M, Taher A, Nunez FM, Li D, Edwards MB, Kleer CG, Appelman H, Sun Y, Zhao L, Moon JJ, Schwendeman A, Lowenstein PR, Castro MG. Inhibition of 2-hydroxyglutarate elicits metabolic reprogramming and mutant IDH1 glioma immunity in mice. J Clin Invest 2021; 131:139542. [PMID: 33332283 DOI: 10.1172/jci139542] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 12/09/2020] [Indexed: 02/06/2023] Open
Abstract
Mutant isocitrate dehydrogenase 1 (IDH1-R132H; mIDH1) is a hallmark of adult gliomas. Lower grade mIDH1 gliomas are classified into 2 molecular subgroups: 1p/19q codeletion/TERT-promoter mutations or inactivating mutations in α-thalassemia/mental retardation syndrome X-linked (ATRX) and TP53. This work focuses on glioma subtypes harboring mIDH1, TP53, and ATRX inactivation. IDH1-R132H is a gain-of-function mutation that converts α-ketoglutarate into 2-hydroxyglutarate (D-2HG). The role of D-2HG within the tumor microenvironment of mIDH1/mATRX/mTP53 gliomas remains unexplored. Inhibition of D-2HG, when used as monotherapy or in combination with radiation and temozolomide (IR/TMZ), led to increased median survival (MS) of mIDH1 glioma-bearing mice. Also, D-2HG inhibition elicited anti-mIDH1 glioma immunological memory. In response to D-2HG inhibition, PD-L1 expression levels on mIDH1-glioma cells increased to similar levels as observed in WT-IDH gliomas. Thus, we combined D-2HG inhibition/IR/TMZ with anti-PDL1 immune checkpoint blockade and observed complete tumor regression in 60% of mIDH1 glioma-bearing mice. This combination strategy reduced T cell exhaustion and favored the generation of memory CD8+ T cells. Our findings demonstrate that metabolic reprogramming elicits anti-mIDH1 glioma immunity, leading to increased MS and immunological memory. Our preclinical data support the testing of IDH-R132H inhibitors in combination with IR/TMZ and anti-PDL1 as targeted therapy for mIDH1/mATRX/mTP53 glioma patients.
Collapse
Affiliation(s)
- Padma Kadiyala
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Stephen V Carney
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Jessica C Gauss
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Maria B Garcia-Fabiani
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Santiago Haase
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Mahmoud S Alghamri
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Felipe J Núñez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yayuan Liu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Minzhi Yu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Ayman Taher
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Fernando M Nunez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Dan Li
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA
| | - Marta B Edwards
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Celina G Kleer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Henry Appelman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Yilun Sun
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Biostatistics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Lili Zhao
- Department of Biostatistics, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA.,Biointerfaces Institute, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
9
|
Wang M, Tan G, Eljaszewicz A, Meng Y, Wawrzyniak P, Acharya S, Altunbulakli C, Westermann P, Dreher A, Yan L, Wang C, Akdis M, Zhang L, Nadeau KC, Akdis CA. Laundry detergents and detergent residue after rinsing directly disrupt tight junction barrier integrity in human bronchial epithelial cells. J Allergy Clin Immunol 2018; 143:1892-1903. [PMID: 30500342 DOI: 10.1016/j.jaci.2018.11.016] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 10/31/2018] [Accepted: 11/09/2018] [Indexed: 11/30/2022]
Abstract
BACKGROUND Defects in the epithelial barrier have recently been associated with asthma and other allergies. The influence of laundry detergents on human bronchial epithelial cells (HBECs) and their barrier function remain unknown. OBJECTIVE We investigated the effects of laundry detergents on cytotoxicity, barrier function, the transcriptome, and the epigenome in HBECs. METHODS Air-liquid interface cultures of primary HBECs from healthy control subjects, patients with asthma, and patients with chronic obstructive pulmonary disease were exposed to laundry detergents and detergent residue after rinsing. Cytotoxicity and epithelial barrier function were evaluated. RNA sequencing, Assay for Transposase Accessible Chromatin with high-throughput sequencing, and DNA methylation arrays were used for checking the transcriptome and epigenome. RESULTS Laundry detergents and rinse residue showed dose-dependent toxic effects on HBECs, with irregular cell shape and leakage of lactate dehydrogenase after 24 hours of exposure. A disrupted epithelial barrier function was found with decreased transepithelial electrical resistance, increased paracellular flux, and stratified tight junction (TJ) immunostaining in HBECs exposed to laundry detergent at 1:25,000 dilutions or rinse residue at further 1:10 dilutions. RNA sequencing analysis showed that lipid metabolism, apoptosis progress, and epithelially derived alarmin-related gene expression were upregulated, whereas cell adhesion-related gene expression was downregulated by laundry detergent at 1:50,000 dilutions after 24 hours of exposure without substantially affecting chromatin accessibility and DNA methylation. CONCLUSION Our data demonstrate that laundry detergents, even at a very high dilution, and rinse residue show significant cell-toxic and directly disruptive effects on the TJ barrier integrity of HBECs without affecting the epigenome and TJ gene expression.
Collapse
Affiliation(s)
- Ming Wang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, and the Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland; Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, and the Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Ge Tan
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, and the Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland; Functional Genomics Center Zurich, ETH Zurich/University of Zurich, Zurich, Switzerland
| | - Andrzej Eljaszewicz
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, and the Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland; Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, Bialystok, Poland
| | - Yifan Meng
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, and the Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland; Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, and the Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Paulina Wawrzyniak
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, and the Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Swati Acharya
- Sean N Parker Center for Allergy and Asthma Research at Stanford University, Department of Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Can Altunbulakli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, and the Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Patrick Westermann
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, and the Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Anita Dreher
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, and the Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | | | - Chengshuo Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, and the Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, and the Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Luo Zhang
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, and the Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Kari C Nadeau
- Sean N Parker Center for Allergy and Asthma Research at Stanford University, Department of Medicine, Stanford University School of Medicine, Stanford, Calif
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, and the Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland.
| |
Collapse
|
10
|
Wang Y, Yan L, Zhang Z, Prado E, Fu L, Xu X, Du L. Epigenetic Regulation and Its Therapeutic Potential in Pulmonary Hypertension. Front Pharmacol 2018; 9:241. [PMID: 29615911 PMCID: PMC5870037 DOI: 10.3389/fphar.2018.00241] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 03/05/2018] [Indexed: 12/21/2022] Open
Abstract
Recent advances in epigenetics have made a tremendous impact on our knowledge of biological phenomena and the environmental stressors on complex diseases. Understanding the mechanism of epigenetic reprogramming during the occurrence of pulmonary hypertension (PH) is important for advanced studies and clinical therapy. In this article, we review the discovery of novel epigenetic mechanisms associated with PH including DNA methylation, histone modification, and noncoding RNA interference. In addition, we highlight the role of epigenetic mechanisms in adult PAH resulting from undesirable perinatal environments-Extrauterine growth restriction (EUGR) and Intrauterine growth retardation (IUGR). Lastly, we give a comprehensive summary for the remaining challenges and discuss future methods of epigenetic targeted therapy for pulmonary hypertension.
Collapse
Affiliation(s)
- Yu Wang
- Department of Pediatrics, Children's Hospital of Zhejiang University, Hangzhou, China
| | - Lingling Yan
- Department of Pediatrics, Children's Hospital of Zhejiang University, Hangzhou, China
| | - Ziming Zhang
- Department of Pediatrics, Children's Hospital of Zhejiang University, Hangzhou, China
| | - Eric Prado
- Loma Linda University School of Medicine, Loma Linda, CA, United States
| | - Linchen Fu
- Department of Pediatrics, Children's Hospital of Zhejiang University, Hangzhou, China
| | - Xuefeng Xu
- Department of Pediatrics, Children's Hospital of Zhejiang University, Hangzhou, China
| | - Lizhong Du
- Department of Pediatrics, Children's Hospital of Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Sar D, Kim B, Ostadhossein F, Misra SK, Pan D. Revisiting Polyarenes and Related Molecules: An Update of Synthetic Approaches and Structure-Activity-Mechanistic Correlation for Carcinogenesis. CHEM REC 2018; 18:619-658. [DOI: 10.1002/tcr.201700110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 02/05/2018] [Indexed: 01/07/2023]
Affiliation(s)
- Dinabandhu Sar
- Department of Bioengineering; University of Illinois at Urbana-Champaign; Biomedical Research Center, Office 3304; 3rd Floor, Mills Breast Cancer Institute, Carle Foundation Hospital; 502 N. Busey Urbana IL 61801 USA
- Mills Breast Cancer Institute and Carle Foundation Hospital; 502 North Busey Urbana, Illinois 61801 USA
| | | | - Fatemeh Ostadhossein
- Department of Bioengineering; University of Illinois at Urbana-Champaign; Biomedical Research Center, Office 3304; 3rd Floor, Mills Breast Cancer Institute, Carle Foundation Hospital; 502 N. Busey Urbana IL 61801 USA
- Mills Breast Cancer Institute and Carle Foundation Hospital; 502 North Busey Urbana, Illinois 61801 USA
| | - Santosh K. Misra
- Department of Bioengineering; University of Illinois at Urbana-Champaign; Biomedical Research Center, Office 3304; 3rd Floor, Mills Breast Cancer Institute, Carle Foundation Hospital; 502 N. Busey Urbana IL 61801 USA
- Mills Breast Cancer Institute and Carle Foundation Hospital; 502 North Busey Urbana, Illinois 61801 USA
| | - Dipanjan Pan
- Department of Bioengineering; University of Illinois at Urbana-Champaign; Biomedical Research Center, Office 3304; 3rd Floor, Mills Breast Cancer Institute, Carle Foundation Hospital; 502 N. Busey Urbana IL 61801 USA
- Mills Breast Cancer Institute and Carle Foundation Hospital; 502 North Busey Urbana, Illinois 61801 USA
- Department of Materials Science and Engineering; University of Illinois at Urbana-Champaign, Urbana, Illinois; 61801 USA
- Beckman Institute; University of Illinois at Urbana-Champaign; Urbana, Illinois 61801 USA
| |
Collapse
|
12
|
Öner D, Ghosh M, Bové H, Moisse M, Boeckx B, Duca RC, Poels K, Luyts K, Putzeys E, Van Landuydt K, Vanoirbeek JA, Ameloot M, Lambrechts D, Godderis L, Hoet PH. Differences in MWCNT- and SWCNT-induced DNA methylation alterations in association with the nuclear deposition. Part Fibre Toxicol 2018; 15:11. [PMID: 29426343 PMCID: PMC5807760 DOI: 10.1186/s12989-018-0244-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 01/18/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Subtle DNA methylation alterations mediated by carbon nanotubes (CNTs) exposure might contribute to pathogenesis and disease susceptibility. It is known that both multi-walled carbon nanotubes (MWCNTs) and single-walled carbon nanotubes (SWCNTs) interact with nucleus. Such, nuclear-CNT interaction may affect the DNA methylation effects. In order to understand the epigenetic toxicity, in particular DNA methylation alterations, of SWCNTs and short MWCNTs, we performed global/genome-wide, gene-specific DNA methylation and RNA-expression analyses after exposing human bronchial epithelial cells (16HBE14o- cell line). In addition, the presence of CNTs on/in the cell nucleus was evaluated in a label-free way using femtosecond pulsed laser microscopy. RESULTS Generally, a higher number of SWCNTs, compared to MWCNTs, was deposited at both the cellular and nuclear level after exposure. Nonetheless, both CNT types were in physical contact with the nuclei. While particle type dependency was noticed for the identified genome-wide and gene-specific alterations, no global DNA methylation alteration on 5-methylcytosine (5-mC) sites was observed for both CNTs. After exposure to MWCNTs, 2398 genes were hypomethylated (at gene promoters), and after exposure to SWCNTs, 589 CpG sites (located on 501 genes) were either hypo- (N = 493 CpG sites) or hypermethylated (N = 96 CpG sites). Cells exposed to MWCNTs exhibited a better correlation between gene promoter methylation and gene expression alterations. Differentially methylated and expressed genes induced changes (MWCNTs > SWCNTs) at different cellular pathways, such as p53 signalling, DNA damage repair and cell cycle. On the other hand, SWCNT exposure showed hypermethylation on functionally important genes, such as SKI proto-oncogene (SKI), glutathione S-transferase pi 1 (GTSP1) and shroom family member 2 (SHROOM2) and neurofibromatosis type I (NF1), which the latter is both hypermethylated and downregulated. CONCLUSION After exposure to both types of CNTs, epigenetic alterations may contribute to toxic or repair response. Moreover, our results suggest that the observed differences in the epigenetic response depend on particle type and differential CNT-nucleus interactions.
Collapse
Affiliation(s)
- Deniz Öner
- Laboratory of Toxicology, Unit of Environment and Health, Department of Public Health and Primary Care, KU Leuven, 3000, Leuven, Belgium
| | - Manosij Ghosh
- Laboratory of Toxicology, Unit of Environment and Health, Department of Public Health and Primary Care, KU Leuven, 3000, Leuven, Belgium
| | - Hannelore Bové
- Centre for Surface Chemistry and Catalysis, KU Leuven, Celestijnenlaan 200F, 3001, Leuven, Belgium
- Biomedical Research Institute, Agoralaan Building C, Hasselt University, 3590, Diepenbeek, Belgium
| | - Matthieu Moisse
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium
- Laboratory for Translational Genetics, VIB Centre for Cancer Biology, VIB, 3000, Leuven, Belgium
| | - Bram Boeckx
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium
- Laboratory for Translational Genetics, VIB Centre for Cancer Biology, VIB, 3000, Leuven, Belgium
| | - Radu C Duca
- Laboratory for Occupational and Environmental Hygiene, Unit of Environment and Health, Department of Public Health and Primary Care, KU Leuven, 3000, Leuven, Belgium
| | - Katrien Poels
- Laboratory for Occupational and Environmental Hygiene, Unit of Environment and Health, Department of Public Health and Primary Care, KU Leuven, 3000, Leuven, Belgium
| | - Katrien Luyts
- Laboratory of Toxicology, Unit of Environment and Health, Department of Public Health and Primary Care, KU Leuven, 3000, Leuven, Belgium
| | - Eveline Putzeys
- Laboratory of Toxicology, Unit of Environment and Health, Department of Public Health and Primary Care, KU Leuven, 3000, Leuven, Belgium
- Department of Oral Health Sciences, Unit of Biomaterials (BIOMAT), KU Leuven, 3000, Leuven, Belgium
| | - Kirsten Van Landuydt
- Department of Oral Health Sciences, Unit of Biomaterials (BIOMAT), KU Leuven, 3000, Leuven, Belgium
| | - Jeroen Aj Vanoirbeek
- Laboratory of Toxicology, Unit of Environment and Health, Department of Public Health and Primary Care, KU Leuven, 3000, Leuven, Belgium
- Laboratory for Occupational and Environmental Hygiene, Unit of Environment and Health, Department of Public Health and Primary Care, KU Leuven, 3000, Leuven, Belgium
| | - Marcel Ameloot
- Biomedical Research Institute, Agoralaan Building C, Hasselt University, 3590, Diepenbeek, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, 3000, Leuven, Belgium
- Laboratory for Translational Genetics, VIB Centre for Cancer Biology, VIB, 3000, Leuven, Belgium
| | - Lode Godderis
- Laboratory for Occupational and Environmental Hygiene, Unit of Environment and Health, Department of Public Health and Primary Care, KU Leuven, 3000, Leuven, Belgium
- External Service for Prevention and Protection at Work, IDEWE, B-3001, Leuven, Belgium
| | - Peter Hm Hoet
- Laboratory of Toxicology, Unit of Environment and Health, Department of Public Health and Primary Care, KU Leuven, 3000, Leuven, Belgium.
| |
Collapse
|
13
|
Ershov NI, Bondar NP, Lepeshko AA, Reshetnikov VV, Ryabushkina JA, Merkulova TI. Consequences of early life stress on genomic landscape of H3K4me3 in prefrontal cortex of adult mice. BMC Genomics 2018; 19:93. [PMID: 29504911 PMCID: PMC5836825 DOI: 10.1186/s12864-018-4479-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Maternal separation models in rodents are widely used to establish molecular mechanisms underlying prolonged effects of early life adversity on neurobiological and behavioral outcomes in adulthood. However, global epigenetic signatures following early life stress in these models remain unclear. Results In this study, we carried out a ChIP-seq analysis of H3K4 trimethylation profile in the prefrontal cortex of adult male mice with a history of early life stress. Two types of stress were used: prolonged separation of pups from their mothers (for 3 h once a day, maternal separation, MS) and brief separation (for 15 min once a day, handling, HD). Adult offspring in the MS group demonstrated reduced locomotor activity in the open field test accompanied by reduced exploratory activity, while the HD group showed decreased anxiety-like behavior only. In a group of maternal separation, we have found a small number (45) of slightly up-regulated peaks, corresponding to promoters of 70 genes, while no changes were observed in a group of handling. Among the genes whose promoters have differential enrichment of H3K4me3, the most relevant ones participate in gene expression regulation, modulation of chromatin structure and mRNA processing. For two genes, Ddias and Pip4k2a, increased H3K4me3 levels were associated with the increased mRNA expression in MS group. Conclusion The distribution of H3K4me3 in prefrontal cortex showed relatively low variability across all individuals, and only some subtle changes were revealed in mice with a history of early life stress. It is possible that the observed long-lasting behavioral alterations induced by maternal separation are mediated by other epigenetic mechanisms, or other brain structures are responsible for these effects. Electronic supplementary material The online version of this article (10.1186/s12864-018-4479-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nikita I Ershov
- Laboratory of Gene Expression Regulation, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Prospect Lavrentyeva, 630090, Novosibirsk, Russia
| | - Natalya P Bondar
- Laboratory of Gene Expression Regulation, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Prospect Lavrentyeva, 630090, Novosibirsk, Russia. .,Novosibirsk National Research State University, 2 Pirogov Street, 630090, Novosibirsk, Russia.
| | - Arina A Lepeshko
- Laboratory of Gene Expression Regulation, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Prospect Lavrentyeva, 630090, Novosibirsk, Russia.,Novosibirsk National Research State University, 2 Pirogov Street, 630090, Novosibirsk, Russia
| | - Vasiliy V Reshetnikov
- Laboratory of Gene Expression Regulation, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Prospect Lavrentyeva, 630090, Novosibirsk, Russia
| | - Julia A Ryabushkina
- Novosibirsk National Research State University, 2 Pirogov Street, 630090, Novosibirsk, Russia
| | - Tatiana I Merkulova
- Laboratory of Gene Expression Regulation, Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Prospect Lavrentyeva, 630090, Novosibirsk, Russia.,Novosibirsk National Research State University, 2 Pirogov Street, 630090, Novosibirsk, Russia
| |
Collapse
|
14
|
Ateia IM, Sutthiboonyapan P, Kamarajan P, Jin T, Godovikova V, Kapila YL, Fenno JC. Treponema denticola increases MMP-2 expression and activation in the periodontium via reversible DNA and histone modifications. Cell Microbiol 2018; 20. [PMID: 29205773 DOI: 10.1111/cmi.12815] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/03/2017] [Accepted: 11/28/2017] [Indexed: 12/21/2022]
Abstract
Host-derived matrix metalloproteinases (MMPs) and bacterial proteases mediate destruction of extracellular matrices and supporting alveolar bone in periodontitis. The Treponema denticola dentilisin protease induces MMP-2 expression and activation in periodontal ligament (PDL) cells, and dentilisin-mediated activation of pro-MMP-2 is required for cellular fibronectin degradation. Here, we report that T. denticola regulates MMP-2 expression through epigenetic modifications in the periodontium. PDL cells were treated with epigenetic enzyme inhibitors before or after T. denticola challenge. Fibronectin fragmentation, MMP-2 expression, and activation were assessed by immunoblot, zymography, and qRT-PCR, respectively. Chromatin modification enzyme expression in T. denticola-challenged PDL cells and periodontal tissues were evaluated using gene arrays. Several classes of epigenetic enzymes showed significant alterations in transcription in diseased tissue and T. denticola-challenged PDL cells. T. denticola-mediated MMP-2 expression and activation were significantly reduced in PDL cells treated with inhibitors of aurora kinases and histone deacetylases. In contrast, DNA methyltransferase inhibitors had little effect, and inhibitors of histone acetyltransferases, methyltransferases, and demethylases exacerbated T. denticola-mediated MMP-2 expression and activation. Chronic epigenetic changes in periodontal tissues mediated by T. denticola or other oral microbes may contribute to the limited success of conventional treatment of chronic periodontitis and may be amenable to therapeutic reversal.
Collapse
Affiliation(s)
- Islam M Ateia
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Department of Periodontics and Oral Medicine, University of Mansoura Faculty of Dentistry, Mansoura, Egypt
| | - Pimchanok Sutthiboonyapan
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Department of Periodontology, Chulalongkorn University Faculty of Dentistry, Bangkok, Thailand
| | - Pachiyappan Kamarajan
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Department of Orofacial Sciences, University of California San Francisco School of Dentistry, San Francisco, CA, USA
| | - Taocong Jin
- Office of Research, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Valentina Godovikova
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - Yvonne L Kapila
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI, USA.,Department of Orofacial Sciences, University of California San Francisco School of Dentistry, San Francisco, CA, USA
| | - J Christopher Fenno
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| |
Collapse
|
15
|
Nilendu P, Sharma NK. Epigenomic Hard Drive Imprinting: A Hidden Code Beyond the Biological Death of Cancer Patients. J Cancer Prev 2017; 22:211-218. [PMID: 29302578 PMCID: PMC5751838 DOI: 10.15430/jcp.2017.22.4.211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/07/2017] [Accepted: 12/11/2017] [Indexed: 12/26/2022] Open
Abstract
Several genetic and epigenetic theories have been suggested to explain the intricacies of life and death. However, several questions remain unsettled regarding cellular death events, particularly of living tissue in the case of cancer patients, such as the fate and adaptation of cancer cells after biological death. It is possible that cancer cells can display the intent to communicate with the external environment after biological death by means of molecular, genetic, and epigenetic pathways. Whether these cancer cells contain special information in the form of coding that may help them survive beyond the biological death of cancer patients is unknown. To understand these queries in the cancer field, we hypothesize the epigenomic hard drive (EHD) as a cellular component to record and store global epigenetic events in cancerous and non-cancerous tissues of cancer patients. This mini-review presents the novel concept of EHD that is reinforced with the existing knowledge of genetic and epigenetic events in cancer. Further, we summarize the EHD understanding that may impart much potential and interest for basic and clinical scientists to unravel mechanisms of carcinogenesis, therapeutic markers, and differential drug responses.
Collapse
Affiliation(s)
- Pritish Nilendu
- Cancer and Translational Research Lab, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Maharashtra, India
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth, Maharashtra, India
| |
Collapse
|
16
|
Koschmann C, Nunez FJ, Mendez F, Brosnan-Cashman JA, Meeker AK, Lowenstein PR, Castro MG. Mutated Chromatin Regulatory Factors as Tumor Drivers in Cancer. Cancer Res 2017; 77:227-233. [PMID: 28062403 DOI: 10.1158/0008-5472.can-16-2301] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/04/2016] [Accepted: 10/08/2016] [Indexed: 11/16/2022]
Abstract
Genes encoding proteins that regulate chromatin structure and DNA modifications [i.e., chromatin regulatory factors (CRF)] and genes encoding histone proteins harbor recurrent mutations in most human cancers. These mutations lead to modifications in tumor chromatin and DNA structure and an altered epigenetic state that contribute to tumorigenesis. Mutated CRFs have now been identified in most types of cancer and are increasingly regarded as novel therapeutic targets. In this review, we discuss DNA alterations in CRFs and how these influence tumor chromatin structure and function, which in turn leads to tumorigenesis. We also discuss the clinical implications and review concepts of targeted treatments for these mutations. Continued research on CRF mutations will be critical for our future understanding of cancer biology and the development and implementation of novel cancer therapies. Cancer Res; 77(2); 227-33. ©2017 AACR.
Collapse
Affiliation(s)
- Carl Koschmann
- Department of Pediatrics, Division of Pediatric Hematology-Oncology, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan
| | - Felipe J Nunez
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Flor Mendez
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | | | - Alan K Meeker
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland.,Department of Urology, Johns Hopkins University, Baltimore, Michigan
| | - Pedro R Lowenstein
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan.,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Maria G Castro
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, Michigan. .,Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
17
|
Ereful NC, Liu LY, Tsai E, Kao SM, Dixit S, Mauleon R, Malabanan K, Thomson M, Laurena A, Lee D, Mackay I, Greenland A, Powell W, Leung H. Analysis of Allelic Imbalance in Rice Hybrids Under Water Stress and Association of Asymmetrically Expressed Genes with Drought-Response QTLs. RICE (NEW YORK, N.Y.) 2016; 9:50. [PMID: 27671164 PMCID: PMC5037104 DOI: 10.1186/s12284-016-0123-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 09/18/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND Information on the effect of stress on the allele-specific expression (ASE) profile of rice hybrids is limited. More so, the association of allelically imbalanced genes to important traits is yet to be understood. Here we assessed allelic imbalance (AI) in the heterozygote state of rice under non- and water-stress treatments and determined association of asymmetrically expressed genes with grain yield (GY) under drought stress by in-silico co-localization analysis and selective genotyping. The genotypes IR64, Apo and their F1 hybrid (IR64 × Apo) were grown under normal and water-limiting conditions. We sequenced the total RNA transcripts for all genotypes then reconstructed the two chromosomes in the heterozygote. RESULTS We are able to estimate the transcript abundance of and the differential expression (DE) between the two parent-specific alleles in the rice hybrids. The magnitude and direction of AI are classified into two categories: (1) symmetrical or biallelic and (2) asymmetrical. The latter can be further classified as either IR64- or Apo-favoring gene. Analysis showed that in the hybrids grown under non-stress conditions, 179 and 183 favor Apo- and IR64-specific alleles, respectively. Hence, the number of IR64- and Apo-favoring genes is relatively equal. Under water-stress conditions, 179 and 255 favor Apo- and IR64-specific alleles, respectively, indicating that the number of allelically imbalanced genes is skewed towards IR64. This is nearly 40-60 % preference for Apo and IR64 alleles, respectively, to the hybrid transcriptome. We also observed genes which exhibit allele preference switching when exposed to water-stress conditions. Results of in-silico co-localization procedure and selective genotyping of Apo/IR64 F3:5 progenies revealed significant association of several asymmetrically expressed genes with GY under drought stress conditions. CONCLUSION Our data suggest that water stress skews AI on a genome-wide scale towards the IR64 allele, the cross-specific maternal allele. Several asymmetrically expressed genes are strongly associated with GY under drought stress which may shed hints that genes associated with important traits are allelically imbalanced. Our approach of integrating hybrid expression analysis and QTL mapping analysis may be an efficient strategy for shortlisting candidate genes for gene discovery.
Collapse
Affiliation(s)
- Nelzo C. Ereful
- Genetics and Biotechnology Division, International Rice Research Institute (IRRI), Los Baños, Laguna Philippines
- The John Bingham Laboratory, National Institute of Agricultural Botany (NIAB), Huntingdon Road, Cambridge, CB3 0LE UK
| | - Li-Yu Liu
- Department of Agronomy, National Taiwan University (NTU), Taipei City, 100 Taiwan
| | - Eric Tsai
- Department of Agronomy, National Taiwan University (NTU), Taipei City, 100 Taiwan
| | - Shu-Min Kao
- Department of Agronomy, National Taiwan University (NTU), Taipei City, 100 Taiwan
| | - Shalabh Dixit
- Genetics and Biotechnology Division, International Rice Research Institute (IRRI), Los Baños, Laguna Philippines
| | - Ramil Mauleon
- Genetics and Biotechnology Division, International Rice Research Institute (IRRI), Los Baños, Laguna Philippines
| | - Katrina Malabanan
- Genetics and Biotechnology Division, International Rice Research Institute (IRRI), Los Baños, Laguna Philippines
- Crop Science Cluster, College of Agriculture, University of the Philippines, Los Baños, Laguna 4031 Philippines
| | - Michael Thomson
- Genetics and Biotechnology Division, International Rice Research Institute (IRRI), Los Baños, Laguna Philippines
- Texas A &M, Department of Soil and Crop Sciences 2474 TAMU, College Station, TX 77843-2474 USA
| | - Antonio Laurena
- Institute of Plant Breeding, University of the Philippines, Los Baños, Laguna Philippines
| | - David Lee
- The John Bingham Laboratory, National Institute of Agricultural Botany (NIAB), Huntingdon Road, Cambridge, CB3 0LE UK
| | - Ian Mackay
- The John Bingham Laboratory, National Institute of Agricultural Botany (NIAB), Huntingdon Road, Cambridge, CB3 0LE UK
| | - Andy Greenland
- The John Bingham Laboratory, National Institute of Agricultural Botany (NIAB), Huntingdon Road, Cambridge, CB3 0LE UK
| | - Wayne Powell
- SRUC, Peter Wilson Building, West Mains Road, Edinburgh, EH9 3JG UK
| | - Hei Leung
- Genetics and Biotechnology Division, International Rice Research Institute (IRRI), Los Baños, Laguna Philippines
| |
Collapse
|