1
|
Fujiki J, Maeda N, Yamaguchi K, Ohtsuki Y, Iwano H. DNA methylation of Ad4BP/SF-1 suppresses Cyp11a1 and StAR transcripts in C2C12 myoblasts. Mol Cell Endocrinol 2024; 593:112336. [PMID: 39094930 DOI: 10.1016/j.mce.2024.112336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/04/2024]
Abstract
Steroidogenesis occurs locally in peripheral tissues and via adrenal and gonadal glands' biosynthesis. The C2C12 mouse myoblast cell line and rat skeletal muscles harbor a local steroidogenesis pathway for glucocorticoids, and corticosterone is biosynthesized from skeletal muscle cells. However, Cyp11a1 and StAR protein expressions are not observed in C2C12 cells or rat muscular tissues. In this context, this study investigated the relationship between DNA methylation and key steroidogenic genes. Bioinformatics analysis of methylated DNA immune precipitation showed that C2C12 myoblasts and myotubes did not have remarkable DNA methylated regions in the gene-body of Cyp11a1. However, a highly methylated region in the CpG island was detected in the intronic enhancer of Ad4BP/SF-1, known as the transcriptional factor for steroidogenic genes. After C2C12 myoblasts treatment with 5-aza-2-deoxycytidine, the gene expressions of Ad4BP/SF-1, Cyp11a1, and StAR were significantly time- and concentration-dependent upregulated. To clarify the contribution of Ad4BP/SF-1 on Cyp11a1 and StAR transcripts, we silenced Ad4BP/SF-1 during the 5-aza-2-deoxycytidine treatment in C2C12 myoblasts, resulting in significant suppression of both Cyp11a1 and StAR. Additionally, pregnenolone levels in the supernatants of C2C12 cells were enhanced by 5-aza-2-deoxycytidine treatment, whereas pregnenolone production by C2C12 myoblasts was significantly suppressed by Ad4BP/SF-1 knockdown. These results indicate that DNA methylation of Ad4BP/SF-1 might be involved in the downregulation of steroidogenic genes, such as Cyp11a1 and StAR in C2C12 myoblasts.
Collapse
Affiliation(s)
- Jumpei Fujiki
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Naoyuki Maeda
- Laboratory of Meat Science, Department of Food Science and Human Wellness, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan.
| | - Kosuke Yamaguchi
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Yuya Ohtsuki
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| | - Hidetomo Iwano
- Laboratory of Veterinary Biochemistry, Department of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido, Japan
| |
Collapse
|
2
|
Kober P, Rymuza J, Baluszek S, Maksymowicz M, Nyc A, Mossakowska BJ, Zieliński G, Kunicki J, Bujko M. DNA Methylation Pattern in Somatotroph Pituitary Neuroendocrine Tumors. Neuroendocrinology 2023; 114:51-63. [PMID: 37699356 DOI: 10.1159/000533692] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/14/2023] [Indexed: 09/14/2023]
Abstract
INTRODUCTION Growth hormone secretion by sporadic somatotroph neuroendocrine pituitary tumors (PitNETs) is a major cause of acromegaly. These tumors are relatively heterogenous in terms of histopathological and molecular features. Our previous transcriptomic profiling of somatotroph tumors revealed three distinct molecular subtypes. This study aimed to investigate the difference in DNA methylation patterns in subtypes of somatotroph PitNETs and its role in distinctive gene expression. METHODS Genome-wide DNA methylation was investigated in 48 somatotroph PitNETs with EPIC microarrays. Gene expression was assessed with RNAseq. Bisulfite pyrosequencing and qRT-PCR were used for verifying the results of DNA methylation and gene expression. RESULTS Clustering tumor samples based on methylation data reflected the transcriptome-related classification. Subtype 1 tumors are densely granulated without GNAS mutation, characterized by high expression of NR5A1 (SF-1) and GIPR. The expression of both genes is correlated with specific methylation of the gene body and promoter. This subtype has a lower methylation level of 5' gene regions and CpG islands than the remaining tumors. Subtype 2 PitNETs are densely granulated and frequently GNAS-mutated, while those in subtype 3 are mainly sparsely granulated. Methylation/expression analysis indicates that ∼50% genes located in differentially methylated regions are those differentially expressed between tumor subtypes. Correlation analysis revealed DNA methylation-controlled genes, including CDKN1B, CCND2, EBF3, CDH4, CDH12, MGMT, STAT5A, PLXND1, PTPRE, and MMP16, and genes encoding ion channels and semaphorins. CONCLUSION DNA methylation profiling confirmed the existence of three molecular subtypes of somatotroph PitNETs. High expression of NR5A1 and GIPR in subtype 1 tumors is correlated with specific methylation of both genes.
Collapse
Affiliation(s)
- Paulina Kober
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Julia Rymuza
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Szymon Baluszek
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Maria Maksymowicz
- Department of Cancer Pathomorphology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Aleksandra Nyc
- Department of Cancer Pathomorphology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Beata J Mossakowska
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Grzegorz Zieliński
- Department of Neurosurgery, Military Institute of Medicine, Warsaw, Poland
| | - Jacek Kunicki
- Department of Neurosurgery, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Mateusz Bujko
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| |
Collapse
|
3
|
Szukiewicz D. Aberrant epigenetic regulation of estrogen and progesterone signaling at the level of endometrial/endometriotic tissue in the pathomechanism of endometriosis. VITAMINS AND HORMONES 2023; 122:193-235. [PMID: 36863794 DOI: 10.1016/bs.vh.2022.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Endometriosis is a term referring to a condition whereby the endometrial tissue is found outside the uterine cavity. This progressive and debilitating condition affects up to 15% of women of reproductive age. Due to the fact that endometriosis cells may express estrogen receptors (ERα, Erβ, GPER) and progesterone (P4) receptors (PR-A, PR-B), their growth, cyclic proliferation, and breakdown are similar to the processes occurring in the endometrium. The underlying etiology and pathogenesis of endometriosis are still not fully explained. The retrograde transport of viable menstrual endometrial cells with the retained ability to attach within the pelvic cavity, proliferate, differentiate and invade into the surrounding tissue explains the most widely accepted implantation theory. Endometrial stromal cells (EnSCs) with clonogenic potential constitute the most abundant population of cells within endometrium that resemble the properties of mesenchymal stem cells (MSCs). Accordingly, formation of the endometriotic foci in endometriosis may be due to a kind of EnSCs dysfunction. Increasing evidence indicates the underestimated role of epigenetic mechanisms in the pathogenesis of endometriosis. Hormone-mediated epigenetic modifications of the genome in EnSCs or even MSCs were attributed an important role in the etiopathogenesis of endometriosis. The roles of excess estrogen exposure and P4 resistance were also found to be crucial in the development of epigenetic homeostasis failure. Therefore, the aim of this review was to consolidate the current knowledge regarding the epigenetic background of EnSCs and MSCs and the changed properties due to estrogen/P4 imbalances in the context of the etiopathogenesis of endometriosis.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
4
|
Abstract
The world of long non-coding RNAs (lncRNAs) has opened up massive new prospects in understanding the regulation of gene expression. Not only are there seemingly almost infinite numbers of lncRNAs in the mammalian cell, but they have highly diverse mechanisms of action. In the nucleus, some are chromatin-associated, transcribed from transcriptional enhancers (eRNAs) and/or direct changes in the epigenetic landscape with profound effects on gene expression. The pituitary gonadotrope is responsible for activation of reproduction through production and secretion of appropriate levels of the gonadotropic hormones. As such, it exemplifies a cell whose function is defined through changes in developmental and temporal patterns of gene expression, including those that are hormonally induced. Roles for diverse distal regulatory elements and eRNAs in gonadotrope biology have only just begun to emerge. Here, we will present an overview of the different kinds of lncRNAs that alter gene expression, and what is known about their roles in regulating some of the key gonadotrope genes. We will also review various screens that have detected differentially expressed pituitary lncRNAs associated with changes in reproductive state and those whose expression is found to play a role in gonadotrope-derived nonfunctioning pituitary adenomas. We hope to shed light on this exciting new field, emphasize the open questions, and encourage research to illuminate the roles of lncRNAs in various endocrine systems.
Collapse
Affiliation(s)
- Tal Refael
- Faculty of Biology, Technion Israel Institute of Technology, Haifa 32000, Israel
| | - Philippa Melamed
- Faculty of Biology, Technion Israel Institute of Technology, Haifa 32000, Israel
- Correspondence: Philippa Melamed, PhD, Faculty of Biology, Technion - Israel Institute of Technology, Haifa 32000, Israel.
| |
Collapse
|
5
|
Nam AR, Lee KH, Hwang HJ, Schabort JJ, An JH, Won SH, Cho JY. Alternative methylation of intron motifs is associated with cancer-related gene expression in both canine mammary tumor and human breast cancer. Clin Epigenetics 2020; 12:110. [PMID: 32693820 PMCID: PMC7374976 DOI: 10.1186/s13148-020-00888-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/23/2020] [Indexed: 12/15/2022] Open
Abstract
Background Canine mammary tumor (CMT) has long been considered as a good animal model for human breast cancer (HBC) due to their pathological and biological similarities. However, only a few aspects of the epigenome have been explored in both HBC and CMT. Moreover, DNA methylation studies have mainly been limited to the promoter regions of genes. Results Genome-wide methylation analysis was performed in CMT and adjacent normal tissues and focused on the intron regions as potential targets for epigenetic regulation. As expected, many tumor suppressors and oncogenes were identified. Of note, most cancer-associated biological processes were enriched in differentially methylated genes (DMGs) that included intron DMRs (differentially methylated regions). Interestingly, two PAX motifs, PAX5 (tumor suppressive) and PAX6 (oncogenic), were frequently found in hyper- and hypomethylated intron DMRs, respectively. Hypermethylation at the PAX5 motifs in the intron regions of CDH5 and LRIG1 genes were found to be anti-correlated with gene expression, while CDH2 and ADAM19 genes harboring hypomethylated PAX6 motifs in their intron region were upregulated. These results were validated from the specimens originally MBD-sequenced as well as additional clinical samples. We also comparatively investigated the intron methylation and downstream gene expression of these genes using human breast invasive carcinoma (BRCA) datasets in TCGA (The Cancer Genome Atlas) public database. Regional alteration of methylation was conserved in the corresponding intron regions and, consequently, gene expression was also altered in HBC. Conclusions This study provides good evidence for the conservation of epigenetic regulation in CMT and HBC, and suggests that intronic methylation can be an important factor in better understanding gene regulation in both CMT and HBC.
Collapse
Affiliation(s)
- A-Reum Nam
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Gwanak-ro1, Gwanak-gu, Seoul, Korea
| | - Kang-Hoon Lee
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Gwanak-ro1, Gwanak-gu, Seoul, Korea
| | - Hyeon-Ji Hwang
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Gwanak-ro1, Gwanak-gu, Seoul, Korea
| | - Johannes J Schabort
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Gwanak-ro1, Gwanak-gu, Seoul, Korea
| | - Jae-Hoon An
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Korea
| | - Sung-Ho Won
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Korea
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Gwanak-ro1, Gwanak-gu, Seoul, Korea.
| |
Collapse
|
6
|
Boni JL, Kahanovitch U, Nwaobi SE, Floyd CL, Olsen ML. DNA methylation: A mechanism for sustained alteration of KIR4.1 expression following central nervous system insult. Glia 2020; 68:1495-1512. [PMID: 32068308 PMCID: PMC8665281 DOI: 10.1002/glia.23797] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/29/2020] [Accepted: 01/31/2020] [Indexed: 12/22/2022]
Abstract
Kir4.1, a glial-specific inwardly rectifying potassium channel, is implicated in astrocytic maintenance of K+ homeostasis. Underscoring the role of Kir4.1 in central nervous system (CNS) functioning, genetic mutations in KCNJ10, the gene which encodes Kir4.1, causes seizures, ataxia and developmental disability in humans. Kir4.1 protein and mRNA loss are consistently observed in CNS injury and neurological diseases linked to hyperexcitability and neuronal dysfunction, leading to the notion that Kir4.1 represents an attractive therapeutic target. Despite this, little is understood regarding the mechanisms that underpin this downregulation. Previous work by our lab revealed that DNA hypomethylation of the Kcnj10 gene functions to regulate mRNA levels during astrocyte maturation whereas hypermethylation in vitro led to decreased promoter activity. In the present study, we utilized two vastly different injury models with known acute and chronic loss of Kir4.1 protein and mRNA to evaluate the methylation status of Kcnj10 as a candidate molecular mechanism for reduced transcription and subsequent protein loss. Examining whole hippocampal tissue and isolated astrocytes, in a lithium-pilocarpine model of epilepsy, we consistently identified hypermethylation of CpG island two, which resides in the large intronic region spanning the Kcnj10 gene. Strikingly similar results were observed using the second injury paradigm, a fifth cervical (C5) vertebral hemi-contusion model of spinal cord injury. Our previous work indicates the same gene region is significantly hypomethylated when transcription increases during astrocyte maturation. Our results suggest that DNA methylation can bidirectionally modulate Kcnj10 transcription and may represent a targetable molecular mechanism for the restoring astroglial Kir4.1 expression following CNS insult.
Collapse
Affiliation(s)
- Jessica L Boni
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
- School of Neuroscience, Virginia Polytechnic and State University, Blacksburg, Virginia
| | - Uri Kahanovitch
- School of Neuroscience, Virginia Polytechnic and State University, Blacksburg, Virginia
| | - Sinifunanya E Nwaobi
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
- Division of Pediatric Neurology, UCLA Mattel Children's Hospital, University of California Los Angeles, Los Angeles, California
| | - Candace L Floyd
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Physical Medicine and Rehabilitation, University of Utah Health, Salt Lake City, Utah
| | - Michelle L Olsen
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
- School of Neuroscience, Virginia Polytechnic and State University, Blacksburg, Virginia
| |
Collapse
|
7
|
Jorgensen BG, Ro S. Role of DNA Methylation in the Development and Differentiation of Intestinal Epithelial Cells and Smooth Muscle Cells. J Neurogastroenterol Motil 2019; 25:377-386. [PMID: 31327220 PMCID: PMC6657918 DOI: 10.5056/jnm19077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 05/27/2019] [Indexed: 12/15/2022] Open
Abstract
The mammalian intestine contains many different cell types but is comprised of 2 main cell types: epithelial cells and smooth muscle cells. Recent in vivo and in vitro evidence has revealed that various alterations to the DNA methylation apparatus within both of these cell types can result in a variety of cellular phenotypes including modified differentiation status, apoptosis, and uncontrolled growth. Methyl groups added to cytosines in regulatory genomic regions typically act to repress associated gene transcription. Aberrant DNA methylation patterns are often found in cells with abnormal growth/differentiation patterns, including those cells involved in burdensome intestinal pathologies including inflammatory bowel diseases and intestinal pseudo-obstructions. The altered methylation patterns being observed in various cell cultures and DNA methyltransferase knockout models indicate an influential connection between DNA methylation and gastrointestinal cells’ development and their response to environmental signaling. As these modified DNA methylation levels are found in a number of pathological gastrointestinal conditions, further investigations into uncovering the causative nature, and controlled regulation, of this epigenetic modification is of great interest.
Collapse
Affiliation(s)
- Brian G Jorgensen
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, USA
| | - Seungil Ro
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, USA
| |
Collapse
|
8
|
Maternal and Post-weaning High-Fat Diets Produce Distinct DNA Methylation Patterns in Hepatic Metabolic Pathways within Specific Genomic Contexts. Int J Mol Sci 2019; 20:ijms20133229. [PMID: 31262088 PMCID: PMC6651091 DOI: 10.3390/ijms20133229] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/24/2019] [Accepted: 06/26/2019] [Indexed: 02/07/2023] Open
Abstract
Calorie-dense high-fat diets (HF) are associated with detrimental health outcomes, including obesity, cardiovascular disease, and diabetes. Both pre- and post-natal HF diets have been hypothesized to negatively impact long-term metabolic health via epigenetic mechanisms. To understand how the timing of HF diet intake impacts DNA methylation and metabolism, male Sprague–Dawley rats were exposed to either maternal HF (MHF) or post-weaning HF diet (PHF). At post-natal week 12, PHF rats had similar body weights but greater hepatic lipid accumulation compared to the MHF rats. Genome-wide DNA methylation was evaluated, and analysis revealed 1744 differentially methylation regions (DMRs) between the groups with the majority of the DMR located outside of gene-coding regions. Within differentially methylated genes (DMGs), intragenic DNA methylation closer to the transcription start site was associated with lower gene expression, whereas DNA methylation further downstream was positively correlated with gene expression. The insulin and phosphatidylinositol (PI) signaling pathways were enriched with 25 DMRs that were associated with 20 DMGs, including PI3 kinase (Pi3k), pyruvate kinase (Pklr), and phosphodiesterase 3 (Pde3). Together, these results suggest that the timing of HF diet intake determines DNA methylation and gene expression patterns in hepatic metabolic pathways that target specific genomic contexts.
Collapse
|
9
|
Anastasiadi D, Esteve-Codina A, Piferrer F. Consistent inverse correlation between DNA methylation of the first intron and gene expression across tissues and species. Epigenetics Chromatin 2018; 11:37. [PMID: 29958539 PMCID: PMC6025724 DOI: 10.1186/s13072-018-0205-1] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/19/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND DNA methylation is one of the main epigenetic mechanisms for the regulation of gene expression in eukaryotes. In the standard model, methylation in gene promoters has received the most attention since it is generally associated with transcriptional silencing. Nevertheless, recent studies in human tissues reveal that methylation of the region downstream of the transcription start site is highly informative of gene expression. Also, in some cell types and specific genes it has been found that methylation of the first intron, a gene feature typically rich in enhancers, is linked with gene expression. However, a genome-wide, tissue-independent, systematic comparative analysis of the relationship between DNA methylation in the first intron and gene expression across vertebrates has not been explored yet. RESULTS The most important findings of this study are: (1) using different tissues from a modern fish, we show a clear genome-wide, tissue-independent quasi-linear inverse relationship between DNA methylation of the first intron and gene expression. (2) This relationship is conserved across vertebrates, since it is also present in the genomes of a model pufferfish, a model frog and different human tissues. Among the gene features, tissues and species interrogated, the first intron's negative correlation with the gene expression was most consistent. (3) We identified more tissue-specific differentially methylated regions (tDMRs) in the first intron than in any other gene feature. These tDMRs have positive or negative correlation with gene expression, indicative of distinct mechanisms of tissue-specific regulation. (4) Lastly, we identified CpGs in transcription factor binding motifs, enriched in the first intron, the methylation of which tended to increase with the distance from the first exon-first intron boundary, with a concomitant decrease in gene expression. CONCLUSIONS Our integrative analysis clearly reveals the important and conserved role of the methylation level of the first intron and its inverse association with gene expression regardless of tissue and species. These findings not only contribute to our basic understanding of the epigenetic regulation of gene expression but also identify the first intron as an informative gene feature regarding the relationship between DNA methylation and gene expression where future studies should be focused.
Collapse
Affiliation(s)
- Dafni Anastasiadi
- Institute of Marine Sciences (ICM-CSIC), Passeig Marítim de la Barceloneta, 37-49, 08003, Barcelona, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Center for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 4, 08028, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Francesc Piferrer
- Institute of Marine Sciences (ICM-CSIC), Passeig Marítim de la Barceloneta, 37-49, 08003, Barcelona, Spain.
| |
Collapse
|
10
|
Takechi T, Hirota T, Sakai T, Maeda N, Kobayashi D, Ieiri I. Interindividual Differences in the Expression of ATP-Binding Cassette and Solute Carrier Family Transporters in Human Skin: DNA Methylation Regulates Transcriptional Activity of the Human ABCC3 Gene. Drug Metab Dispos 2018; 46:628-635. [PMID: 29437875 DOI: 10.1124/dmd.117.079061] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 01/30/2018] [Indexed: 01/01/2023] Open
Abstract
The identification of drug transporters expressed in human skin and interindividual differences in gene expression is important for understanding the role of drug transporters in human skin. In the present study, we evaluated the expression of ATP-binding cassette (ABC) and solute carrier (SLC) transporters using human skin tissues. In skin samples, ABCC3 was expressed at the highest levels, followed by SLCO3A1, SLC22A3, SLC16A7, ABCA2, ABCC1, and SLCO2B1. Among the quantitated transporters, ABCC3 accounted for 20.0% of the total mean transporter mRNA content. The expression of ABCC3 mRNA showed large interindividual variability (9.5-fold). None of the single nucleotide polymorphisms tested (-1767G>A, -1328G>A, -1213C>G, -897delC, -260T>A, and -211C>T) in the promoter region of the ABCC3 gene showed a significant change in ABCC3 mRNA levels. ABCC3 expression levels negatively correlated with the methylation status of the CpG island (CGI) located approximately 10 kilobase pairs upstream of ABCC3 (Rs: -0.323, P < 0.05). The reporter gene assay revealed a significant increase in transcriptional activity in the presence of CGI. ABCC3 mRNA was upregulated in HaCaT cells by the demethylating agent 5-aza-2'-deoxycytidine. Furthermore, the deletion of the region surrounding CGI using the clustered regularly interspaced short palindromic repeat/Cas9 system resulted in significantly lower ABCC3 mRNA levels than those in control clones in HaCaT cells. Herein, we demonstrated large interindividual differences in the expression of drug transporters in human skin. CGI may function as an enhancer of the transcription of ABCC3, and methylation levels in CGI contribute to the variability of ABCC3 expression in human skin.
Collapse
Affiliation(s)
- Tomoki Takechi
- Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (T.T., T.H., T.S., N.M., I.I.); Drug Development Research Laboratories, Kyoto R&D Center, Maruho Co., Ltd., Kyoto, Japan (T.T.); and Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (D.K.)
| | - Takeshi Hirota
- Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (T.T., T.H., T.S., N.M., I.I.); Drug Development Research Laboratories, Kyoto R&D Center, Maruho Co., Ltd., Kyoto, Japan (T.T.); and Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (D.K.)
| | - Tatsuya Sakai
- Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (T.T., T.H., T.S., N.M., I.I.); Drug Development Research Laboratories, Kyoto R&D Center, Maruho Co., Ltd., Kyoto, Japan (T.T.); and Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (D.K.)
| | - Natsumi Maeda
- Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (T.T., T.H., T.S., N.M., I.I.); Drug Development Research Laboratories, Kyoto R&D Center, Maruho Co., Ltd., Kyoto, Japan (T.T.); and Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (D.K.)
| | - Daisuke Kobayashi
- Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (T.T., T.H., T.S., N.M., I.I.); Drug Development Research Laboratories, Kyoto R&D Center, Maruho Co., Ltd., Kyoto, Japan (T.T.); and Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (D.K.)
| | - Ichiro Ieiri
- Department of Clinical Pharmacokinetics, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (T.T., T.H., T.S., N.M., I.I.); Drug Development Research Laboratories, Kyoto R&D Center, Maruho Co., Ltd., Kyoto, Japan (T.T.); and Department of Clinical Pharmacy and Pharmaceutical Care, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan (D.K.)
| |
Collapse
|
11
|
Wang W, Sun Y, Guo Y, Cai P, Li Y, Liu J, Cai G, Kiyoshi A, Zhang W. Continuous soy isoflavones exposure from weaning to maturity induces downregulation of ovarian steroidogenic factor 1 gene expression and corresponding changes in DNA methylation pattern. Toxicol Lett 2017; 281:175-183. [PMID: 28964809 DOI: 10.1016/j.toxlet.2017.09.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/19/2017] [Accepted: 09/26/2017] [Indexed: 11/26/2022]
Abstract
Female Wistar rats were treated with orally administered soy isoflavones at concentrations of 0, 25, 50, or 100mg/kg body weight from weaning until sexual maturity (3 mo.), and ovarian steroidogenesis was evaluated. After soy isoflavones were administered, a significant (P<0.05) decrease (44%) in the serum estrodial levels of the high-dose (HD) group were observed. Cultured granulosa cells from the middle- (MD) and HD groups showed significantly (P<0.05) reduced (31%, 45%, respectively) in vitro estradiol secretion, and those from the HD group showed significantly (P<0.05) reduced progesterone (25%) secretion. Compared with the control group, the mRNA expression of the steroidogenic acute regulatory protein (Star), cytochromeP450 cholesterol side chain cleavage (Cyp11a1 and Cyp19a1), and hydroxysteroid dehydrogenase 3b (Hsd3b) genes also decreased. Real-time quantitative PCR and Western blotting revealed a significant (P<0.05) decrease in key transcription factor steroidogenic factor-1 (SF-1) expression in the HD group. The detection of DNA methylation using bisulfitesequencing PCR (BSP) suggested a significantly (P<0.05) increased total methylation rate in the proximal SF-1 promoter in the HD group. Further studies showed that treatment with soy isoflavones can significantly (P<0.05) increase the mRNA expression of DNA methyltransferase (DNMT) 1 and DNMT3a. This study proved that soy isoflavone administration from weaning until sexual maturity could inhibit ovarian steroidogenesis, suggesting that SF-1 might play an important role in this effect. In addition, DNA methylation might play a role in the downregulation of SF-1 gene expression induced by soy isoflavones.
Collapse
Affiliation(s)
- Wenxiang Wang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China; Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China.
| | - Yan Sun
- Center for Reproductive Medicine, Teaching Hospital of Fujian Medical University, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
| | - Yiwei Guo
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Ping Cai
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Yuchen Li
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Jin Liu
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Guoxi Cai
- Nagasaki Prefectural Institute of Environmental Research and Public Health, Nagasaki, Japan
| | - Aoyagi Kiyoshi
- Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Wenchang Zhang
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
12
|
An epigenetic switch repressing Tet1 in gonadotropes activates the reproductive axis. Proc Natl Acad Sci U S A 2017; 114:10131-10136. [PMID: 28855337 DOI: 10.1073/pnas.1704393114] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The TET enzymes catalyze conversion of 5-methyl cytosine (5mC) to 5-hydroxymethyl cytosine (5hmC) and play important roles during development. TET1 has been particularly well-studied in pluripotent stem cells, but Tet1-KO mice are viable, and the most marked defect is abnormal ovarian follicle development, resulting in impaired fertility. We hypothesized that TET1 might play a role in the central control of reproduction by regulating expression of the gonadotropin hormones, which are responsible for follicle development and maturation and ovarian function. We find that all three TET enzymes are expressed in gonadotrope-precursor cells, but Tet1 mRNA levels decrease markedly with completion of cell differentiation, corresponding with an increase in expression of the luteinizing hormone gene, Lhb We demonstrate that poorly differentiated gonadotropes express a TET1 isoform lacking the N-terminal CXXC-domain, which represses Lhb gene expression directly and does not catalyze 5hmC at the gene promoter. We show that this isoform is also expressed in other differentiated tissues, and that it is regulated by an alternative promoter whose activity is repressed by the liganded estrogen and androgen receptors, and by the hypothalamic gonadotropin-releasing hormone through activation of PKA. Its expression is also regulated by DNA methylation, including at an upstream enhancer that is protected by TET2, to allow Tet1 expression. The down-regulation of TET1 relieves its repression of the methylated Lhb gene promoter, which is then hydroxymethylated and activated by TET2 for full reproductive competence.
Collapse
|
13
|
Lyu Y, Lou J, Yang Y, Feng J, Hao Y, Huang S, Yin L, Xu J, Huang D, Ma B, Zou D, Wang Y, Zhang Y, Zhang B, Chen P, Yu K, Lam EWF, Wang X, Liu Q, Yan J, Jin B. Dysfunction of the WT1-MEG3 signaling promotes AML leukemogenesis via p53-dependent and -independent pathways. Leukemia 2017; 31:2543-2551. [PMID: 28400619 PMCID: PMC5729340 DOI: 10.1038/leu.2017.116] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 03/16/2017] [Accepted: 04/04/2017] [Indexed: 12/14/2022]
Abstract
Long non-coding RNAs (lncRNAs) play a pivotal role in tumorigenesis, exemplified by the recent finding that lncRNA maternally expressed gene 3 (MEG3) inhibits tumor growth in a p53-dependent manner. Acute myeloid leukemia (AML) is the most common malignant myeloid disorder in adults, and TP53 mutations or loss are frequently detected in patients with therapy-related AML or AML with complex karyotype. Here, we reveal that MEG3 is significantly downregulated in AML and suppresses leukemogenesis not only in a p53-dependent, but also a p53-independent manner. In addition, MEG3 is proven to be transcriptionally activated by Wilms’ tumor 1 (WT1), dysregulation of which by epigenetic silencing or mutations is causally involved in AML. Therefore MEG3 is identified as a novel target of the WT1 molecule. Ten–eleven translocation-2 (TET2) mutations frequently occur in AML and significantly promote leukemogenesis of this disorder. In our study, TET2, acting as a cofactor of WT1, increases MEG3 expression. Taken together, our work demonstrates that TET2 dysregulated WT1-MEG3 axis significantly promotes AML leukemogenesis, paving a new avenue for diagnosis and treatment of AML patients.
Collapse
Affiliation(s)
- Y Lyu
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China.,Department of Hematology, the Second Affiliated Hospital, Institute of Hematopoeitic Stem Cell Transplantation of Dalian Medical University, Liaoning Hematopoeitic Stem Cell Transplantation Medical Center, Dalian Key Laboratory of Hematology, Dalian Medical University, Dalian, China
| | - J Lou
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China.,Department of Neurosurgery, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Y Yang
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China.,Department of Hematology, the Second Affiliated Hospital, Institute of Hematopoeitic Stem Cell Transplantation of Dalian Medical University, Liaoning Hematopoeitic Stem Cell Transplantation Medical Center, Dalian Key Laboratory of Hematology, Dalian Medical University, Dalian, China
| | - J Feng
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Y Hao
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - S Huang
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - L Yin
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China.,Department of Hematology, the Second Affiliated Hospital, Institute of Hematopoeitic Stem Cell Transplantation of Dalian Medical University, Liaoning Hematopoeitic Stem Cell Transplantation Medical Center, Dalian Key Laboratory of Hematology, Dalian Medical University, Dalian, China
| | - J Xu
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - D Huang
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China.,Department of Hematology, the Second Affiliated Hospital, Institute of Hematopoeitic Stem Cell Transplantation of Dalian Medical University, Liaoning Hematopoeitic Stem Cell Transplantation Medical Center, Dalian Key Laboratory of Hematology, Dalian Medical University, Dalian, China
| | - B Ma
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China.,Department of Neurosurgery, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - D Zou
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Y Wang
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China.,Department of Hematology, the Second Affiliated Hospital, Institute of Hematopoeitic Stem Cell Transplantation of Dalian Medical University, Liaoning Hematopoeitic Stem Cell Transplantation Medical Center, Dalian Key Laboratory of Hematology, Dalian Medical University, Dalian, China
| | - Y Zhang
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - B Zhang
- Department of Neurosurgery, the Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - P Chen
- Department of Obstetrics and Gynecology, the Second Xiangya Hospital, Central South University, Changsha, China
| | - K Yu
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, USA
| | - E W-F Lam
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - X Wang
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - Q Liu
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| | - J Yan
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China.,Department of Hematology, the Second Affiliated Hospital, Institute of Hematopoeitic Stem Cell Transplantation of Dalian Medical University, Liaoning Hematopoeitic Stem Cell Transplantation Medical Center, Dalian Key Laboratory of Hematology, Dalian Medical University, Dalian, China
| | - B Jin
- Department of Hematology, the Second Affiliated Hospital, Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, China
| |
Collapse
|
14
|
Rahamim-Ben Navi L, Tsukerman A, Feldman A, Melamed P, Tomić M, Stojilkovic SS, Boehm U, Seger R, Naor Z. GnRH Induces ERK-Dependent Bleb Formation in Gonadotrope Cells, Involving Recruitment of Members of a GnRH Receptor-Associated Signalosome to the Blebs. Front Endocrinol (Lausanne) 2017; 8:113. [PMID: 28626446 PMCID: PMC5454083 DOI: 10.3389/fendo.2017.00113] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
We have previously described a signaling complex (signalosome) associated with the GnRH receptor (GnRHR). We now report that GnRH induces bleb formation in the gonadotrope-derived LβT2 cells. The blebs appear within ~2 min at a turnover rate of ~2-3 blebs/min and last for at least 90 min. Formation of the blebs requires active ERK1/2 and RhoA-ROCK but not active c-Src. Although the following ligands stimulate ERK1/2 in LβT2 cells: EGF > GnRH > PMA > cyclic adenosine monophosphate (cAMP), they produced little or no effect on bleb formation as compared to the robust effect of GnRH (GnRH > PMA > cAMP > EGF), indicating that ERK1/2 is required but not sufficient for bleb formation possibly due to compartmentalization. Members of the above mentioned signalosome are recruited to the blebs, some during bleb formation (GnRHR, c-Src, ERK1/2, focal adhesion kinase, paxillin, and tubulin), and some during bleb retraction (vinculin), while F-actin decorates the blebs during retraction. Fluorescence intensity measurements for the above proteins across the cells showed higher intensity in the blebs vs. intracellular area. Moreover, GnRH induces blebs in primary cultures of rat pituitary cells and isolated mouse gonadotropes in an ERK1/2-dependent manner. The novel signalosome-bleb pathway suggests that as with the signalosome, the blebs are apparently involved in cell migration. Hence, we have extended the potential candidates which are involved in the blebs life cycle in general and for the GnRHR in particular.
Collapse
Affiliation(s)
- Liat Rahamim-Ben Navi
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv, Israel
| | - Anna Tsukerman
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Alona Feldman
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Philippa Melamed
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Melanija Tomić
- National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, United States
| | - Stanko S. Stojilkovic
- National Institute of Child Health and Human Development, National Institute of Health, Bethesda, MD, United States
| | - Ulrich Boehm
- Department of Pharmacology and Toxicology, University of Saarland School of Medicine, Homburg, Germany
| | - Rony Seger
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Zvi Naor
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv, Israel
- *Correspondence: Zvi Naor,
| |
Collapse
|
15
|
Weinhouse C, Bergin IL, Harris C, Dolinoy DC. Stat3 is a candidate epigenetic biomarker of perinatal Bisphenol A exposure associated with murine hepatic tumors with implications for human health. Epigenetics 2016; 10:1099-110. [PMID: 26542749 DOI: 10.1080/15592294.2015.1107694] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Bisphenol A (BPA) is an endocrine disrupting chemical (EDC) that has been implicated as a potential carcinogen and epigenotoxicant. We have previously reported dose-dependent incidence of hepatic tumors in 10-month-old isogenic mice perinatally exposed to BPA. Here, we evaluated DNA methylation at 3 candidate genes (Esr1, Il-6st, and Stat3) in liver tissue of BPA-exposed mice euthanized at 2 time points: post-natal day 22 (PND22; n = 147) or 10-months of age (n = 78, including n = 18 with hepatic tumors). Additionally, DNA methylation profiles were analyzed at human homologs of murine candidate genes in human fetal liver samples (n = 50) with known liver tissue BPA levels. Candidate genes were chosen based on reported expression changes in both rodent and human hepatocellular carcinoma (HCC). Regions for bisulfite sequencing were chosen by mining whole genome next generation sequencing methylation datasets of both mice and human liver samples with known perinatal BPA exposures. One of 3 candidate genes, Stat3, displayed dose-dependent DNA methylation changes in both 10-month mice with liver tumors as compared to those without liver tumors and 3-week sibling mice from the same exposure study, implicating Stat3 as a potential epigenetic biomarker of both early life BPA exposure and adult disease in mice. DNA methylation profiles within STAT3 varied with liver tissue BPA level in human fetal liver samples as well, suggesting STAT3 may be a translationally relevant candidate biomarker. These data implicate Stat3 as a potential early life biomarker of adult murine liver tumor risk following early BPA exposure with early evidence of relevance to human health.
Collapse
Affiliation(s)
- Caren Weinhouse
- a Department of Environmental Health Sciences ; University of Michigan ; Ann Arbor , Michigan , USA
| | - Ingrid L Bergin
- b Unit for Laboratory Animal Medicine; University of Michigan ; Ann Arbor , Michigan , USA
| | - Craig Harris
- a Department of Environmental Health Sciences ; University of Michigan ; Ann Arbor , Michigan , USA
| | - Dana C Dolinoy
- a Department of Environmental Health Sciences ; University of Michigan ; Ann Arbor , Michigan , USA.,c Department of Nutritional Sciences ; University of Michigan ; Ann Arbor , Michigan , USA
| |
Collapse
|
16
|
Laverrière JN, L'Hôte D, Tabouy L, Schang AL, Quérat B, Cohen-Tannoudji J. Epigenetic regulation of alternative promoters and enhancers in progenitor, immature, and mature gonadotrope cell lines. Mol Cell Endocrinol 2016; 434:250-65. [PMID: 27402603 DOI: 10.1016/j.mce.2016.07.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 07/05/2016] [Accepted: 07/05/2016] [Indexed: 11/25/2022]
Abstract
Gonadotrope cell identity genes emerge in a stepwise process during mouse pituitary development. Cga, encoding for the α-subunit of TSH, LH, and FSH, is initially detected at E11.5 followed by Gnrhr and steroidogenic factor Sf1 at E13.5, specifying cells engaged in a gonadotrope cell fate. Lhb and Fshb appear at E16.5 and 17.5, respectively, typifying differentiated gonadotrope cells. Using the αT1-1, αT3-1 and LβT2 cell lines recapitulating these stages of gonadotrope differentiation, DNA methylation at Gnrhr and Sf1 was investigated. Regulatory regions were found hypermethylated in progenitor αT1-1 cells and hypomethylated in differentiated LβT2 cells. Abundance of RNA polymerase II together with active histone modifications including H3K4me1, H3K4me3, and H3K27ac were strictly correlated with DNA hypomethylation. Analyses of epigenomic modifications and chromatin accessibility were further extended to Isl1, Lhx3, Gata2, and Pitx2, highlighting alternative usages of specific regulatory gene domains in progenitor αT1-1, immature αT3-1, and mature LβT2 gonadotrope cells.
Collapse
Affiliation(s)
- Jean-Noël Laverrière
- Univ Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative (BFA), F-75013, Paris, France; CNRS UMR 8251, F-75013, Paris, France; Physiologie de l'axe gonadotrope INSERM U1133, F-75013, Paris, France.
| | - David L'Hôte
- Univ Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative (BFA), F-75013, Paris, France; CNRS UMR 8251, F-75013, Paris, France; Physiologie de l'axe gonadotrope INSERM U1133, F-75013, Paris, France
| | - Laure Tabouy
- Univ Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative (BFA), F-75013, Paris, France; CNRS UMR 8251, F-75013, Paris, France; Physiologie de l'axe gonadotrope INSERM U1133, F-75013, Paris, France
| | - Anne-Laure Schang
- Univ Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative (BFA), F-75013, Paris, France; CNRS UMR 8251, F-75013, Paris, France; Physiologie de l'axe gonadotrope INSERM U1133, F-75013, Paris, France
| | - Bruno Quérat
- Univ Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative (BFA), F-75013, Paris, France; CNRS UMR 8251, F-75013, Paris, France; Physiologie de l'axe gonadotrope INSERM U1133, F-75013, Paris, France
| | - Joëlle Cohen-Tannoudji
- Univ Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative (BFA), F-75013, Paris, France; CNRS UMR 8251, F-75013, Paris, France; Physiologie de l'axe gonadotrope INSERM U1133, F-75013, Paris, France
| |
Collapse
|
17
|
KOUKOURA OURANIA, SIFAKIS STAVROS, SPANDIDOS DEMETRIOSA. DNA methylation in endometriosis (Review). Mol Med Rep 2016; 13:2939-48. [PMID: 26934855 PMCID: PMC4805102 DOI: 10.3892/mmr.2016.4925] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/22/2016] [Indexed: 12/01/2022] Open
Abstract
Endometriosis is defined by the presence and growth of functional endometrial tissue, outside the uterine cavity, primarily in the ovaries, pelvic peritoneum and rectovaginal septum. Although it is a benign disease, it presents with malignant characteristics, such as invasion to surrounding tissues, metastasis to distant locations and recurrence following treatment. Accumulating evidence suggests that various epigenetic aberrations may play an essential role in the pathogenesis of endometriosis. Aberrant DNA methylation represents a possible mechanism repsonsible for this disease, linking gene expression alterations observed in endometriosis with hormonal and environmental factors. Several lines of evidence indicate that endometriosis may partially be due to selective epigenetic deregulations influenced by extrinsic factors. Previous studies have shed light into the epigenetic component of endometriosis, reporting variations in the epigenetic patterns of genes known to be involved in the aberrant hormonal, immunologic and inflammatory status of endometriosis. Although recent studies, utilizing advanced molecular techniques, have allowed us to further elucidate the possible association of DNA methylation with altered gene expression, whether these molecular changes represent the cause or merely the consequence of the disease is a question which remains to be answered. This review provides an overview of the current literature on the role of DNA methylation in the pathophysiology and malignant evolution of endometriosis. We also provide insight into the mechanisms through which DNA methylation-modifying agents may be the next step in the research of the pharmaceutical treatment of endometriosis.
Collapse
Affiliation(s)
- OURANIA KOUKOURA
- Department of Obstetrics and Gynecology, University Hospital of Larissa, Larissa 41500, Greece
| | - STAVROS SIFAKIS
- Department of Obstetrics and Gynecology, University Hospital of Heraklion, Heraklion 71003, Greece
| | - DEMETRIOS A. SPANDIDOS
- Laboratory of Clinical Virology, University of Crete Medical School, Heraklion 71409, Greece
| |
Collapse
|
18
|
Hu Z, Li J, Kuang Z, Wang M, Azhar S, Guo Z. Cell-Specific Polymorphism and Hormonal Regulation of DNA Methylation in Scavenger Receptor Class B, Type I. DNA Cell Biol 2016; 35:280-9. [PMID: 26981684 DOI: 10.1089/dna.2015.3185] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The scavenger receptor class B, type I (SR-BI), is a cell-surface glycoprotein that mediates selective uptake of high density lipoprotein (HDL)-derived cholesteryl ester. SR-BI plays an important role in cellular delivery of cholesterol. Both human and rodent SR-BI are expressed most abundantly in the liver parenchymal cells and steroidogenic cells of the adrenal gland and gonads, where the selective pathway exhibits its highest activity. In steroidogenic cells, the expression of SR-BI is regulated by trophic hormones (adrenocorticotropic hormone or gonadotropins luteinizing hormone or follicle-stimulating hormone) in concert with the regulation of steroid hormone production. DNA methylation has been implicated in a large number of biological processes mainly by regulating gene expression. The SR-BI promoter contains one CpG island (CGI) in its promoter and seven CGIs in its intronic regions. Here, we studied the DNA methylation status of SR-BI gene and provide evidence that the DNA methylation is cell specific in this gene promoter as well as in intronic regions. The DNA methylation in the SR-BI promoter is subject to N(6), 2'-O-dibutyryladenosine3':5'-cyclic monophosphate regulation in mouse adrenal Y1 cells and mouse Leydig tumor cells (MLTCs). The seven intron CGIs are methylated differentially in Y1 cells, MLTCs, ovarian granulosa cells, and mouse liver hepa 1-6 cells. Our experiments raised the possibility that DNA methylation participates in hormonal regulation of SR-BI expression in a tissue-specific manner. We further suggest that the cell-specific DNA methylation in SR-BI intronic regions may be associated with specific biological function(s) of these regions, including regulation of gene expression.
Collapse
Affiliation(s)
- Zhigang Hu
- 1 Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University , Nanjing, China
| | - Jiaxin Li
- 1 Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University , Nanjing, China
| | - Zhihui Kuang
- 1 Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University , Nanjing, China
| | - Meina Wang
- 1 Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University , Nanjing, China
| | - Salman Azhar
- 2 Geriatric Research, Education and Clinical Center, VA Palo Alto Health Care System, and Stanford University School of Medicine , Palo Alto, California
| | - Zhigang Guo
- 1 Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University , Nanjing, China
| |
Collapse
|
19
|
Qiao S, Nordström K, Muijs L, Gasparoni G, Tierling S, Krause E, Walter J, Boehm U. Molecular Plasticity of Male and Female Murine Gonadotropes Revealed by mRNA Sequencing. Endocrinology 2016; 157:1082-93. [PMID: 26677881 DOI: 10.1210/en.2015-1836] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Gonadotropes in the anterior pituitary gland are of particular importance within the hypothalamic-pituitary-gonadal axis because they provide a means of communication and thus a functional link between the brain and the gonads. Recent results indicate that female gonadotropes may be organized in the form of a network that shows plasticity and adapts to the altered endocrine conditions of different physiological states. However, little is known about functional changes on the molecular level within gonadotropes during these different conditions. In this study we capitalize on a binary genetic strategy in order to fluorescently label murine gonadotrope cells. Using this mouse model allows to produce an enriched gonadotrope population using fluorescence activated cell sorting to perform mRNA sequencing. By using this strategy, we analyze and compare the expression profile of murine gonadotropes in different genders and developmental and hormonal stages. We find that gonadotropes taken from juvenile males and females, from cycling females at diestrus and at proestrus, from lactating females, and from adult males each have unique gene expression patterns with approximately 100 to approximately 500 genes expressed only in one particular stage. We also demonstrate extensive gene-expression profile changes with up to approximately 2200 differentially expressed genes when comparing female and male development, juveniles and adults, and cycling females. Differentially expressed genes were significantly enriched in the GnRH signaling, calcium signaling, and MAPK signaling pathways by Kyoto Encyclopedia of Genes and Genomes analysis. Our data provide an unprecedented molecular view of the primary gonadotropes and reveal a high degree of molecular plasticity within the gonadotrope population.
Collapse
Affiliation(s)
- Sen Qiao
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| | - Karl Nordström
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| | - Leon Muijs
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| | - Gilles Gasparoni
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| | - Sascha Tierling
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| | - Elmar Krause
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| | - Jörn Walter
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| | - Ulrich Boehm
- Department of Pharmacology and Toxicology (S.Q., L.M., U.B.) and Center for Integrative Physiology and Molecular Medicine (E.K.), University of Saarland School of Medicine, Kirrberger Straße D-66421 Homburg, Germany; and Department of Genetics (K.N., G.G., S.T., J.W.), University of Saarland, D-66123 Saarbrücken, Germany
| |
Collapse
|
20
|
Verhulst EC, Mateman AC, Zwier MV, Caro SP, Verhoeven KJF, van Oers K. Evidence from pyrosequencing indicates that natural variation in animal personality is associated with DRD4 DNA methylation. Mol Ecol 2016; 25:1801-11. [PMID: 26678756 DOI: 10.1111/mec.13519] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 12/08/2015] [Accepted: 12/14/2015] [Indexed: 12/19/2022]
Abstract
Personality traits are heritable and respond to natural selection, but are at the same time influenced by the ontogenetic environment. Epigenetic effects, such as DNA methylation, have been proposed as a key mechanism to control personality variation. However, to date little is known about the contribution of epigenetic effects to natural variation in behaviour. Here, we show that great tit (Parus major) lines artificially selected for divergent exploratory behaviour for four generations differ in their DNA methylation levels at the dopamine receptor D4 (DRD4) gene. This D4 receptor is statistically associated with personality traits in both humans and nonhuman animals, including the great tit. Previous work in this songbird failed to detect functional genetic polymorphisms within DRD4 that could account for the gene-trait association. However, our observation supports the idea that DRD4 is functionally involved in exploratory behaviour but that its effects are mediated by DNA methylation. While the exact mechanism underlying the transgenerational consistency of DRD4 methylation remains to be elucidated, this study shows that epigenetic mechanisms are involved in shaping natural variation in personality traits. We outline how this first finding provides a basis for investigating the epigenetic contribution to personality traits in natural systems and its subsequent role for understanding the ecology and evolution of behavioural consistency.
Collapse
Affiliation(s)
- Eveline C Verhulst
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Droevendaalsesteeg 10, 6708 PB, Wageningen, The Netherlands.,Department of Terrestrial Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Droevendaalsesteeg 10, 6708 PB, Wageningen, The Netherlands
| | - A Christa Mateman
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Droevendaalsesteeg 10, 6708 PB, Wageningen, The Netherlands
| | - Mathijs V Zwier
- Center for Liver, Digestive and Metabolic Diseases, Department of Pediatrics, University Medical Center Groningen, University of Groningen, P.O. Box 196, 9700 AD, Groningen, The Netherlands
| | - Samuel P Caro
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Droevendaalsesteeg 10, 6708 PB, Wageningen, The Netherlands
| | - Koen J F Verhoeven
- Department of Terrestrial Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Droevendaalsesteeg 10, 6708 PB, Wageningen, The Netherlands
| | - Kees van Oers
- Department of Animal Ecology, Netherlands Institute of Ecology (NIOO-KNAW), Droevendaalsesteeg 10, 6708 PB, Wageningen, The Netherlands
| |
Collapse
|
21
|
Developmental programming of energy balance regulation: is physical activity more ‘programmable’ than food intake? Proc Nutr Soc 2015; 75:73-77. [DOI: 10.1017/s0029665115004127] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Extensive human and animal model data show that environmental influences during critical periods of prenatal and early postnatal development can cause persistent alterations in energy balance regulation. Although a potentially important factor in the worldwide obesity epidemic, the fundamental mechanisms underlying such developmental programming of energy balance are poorly understood, limiting our ability to intervene. Most studies of developmental programming of energy balance have focused on persistent alterations in the regulation of energy intake; energy expenditure has been relatively underemphasised. In particular, very few studies have evaluated developmental programming of physical activity. The aim of this review is to summarise recent evidence that early environment may have a profound impact on establishment of individual propensity for physical activity. Recently, we characterised two different mouse models of developmental programming of obesity; one models fetal growth restriction followed by catch-up growth, and the other models early postnatal overnutrition. In both studies, we observed alterations in body-weight regulation that persisted to adulthood, but no group differences in food intake. Rather, in both cases, programming of energy balance appeared to be due to persistent alterations in energy expenditure and spontaneous physical activity (SPA). These effects were stronger in female offspring. We are currently exploring the hypothesis that developmental programming of SPA occurs via induced sex-specific alterations in epigenetic regulation in the hypothalamus and other regions of the central nervous system. We will summarise the current progress towards testing this hypothesis. Early environmental influences on establishment of physical activity are likely an important factor in developmental programming of energy balance. Understanding the fundamental underlying mechanisms in appropriate animal models will help determine whether early life interventions may be a practical approach to promote physical activity in man.
Collapse
|
22
|
Dyson MT, Kakinuma T, Pavone ME, Monsivais D, Navarro A, Malpani SS, Ono M, Bulun SE. Aberrant expression and localization of deoxyribonucleic acid methyltransferase 3B in endometriotic stromal cells. Fertil Steril 2015; 104:953-963.e2. [PMID: 26239024 PMCID: PMC4603532 DOI: 10.1016/j.fertnstert.2015.06.046] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 06/30/2015] [Accepted: 06/30/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To define the expression and function of DNA methyltransferases (DNMTs) in response to decidualizing stimuli in endometriotic cells compared with healthy endometrial stroma. DESIGN Basic science. SETTING University research center. PATIENT(S) Premenopausal women with or without endometriosis. INTERVENTION(S) Primary cultures of stromal cells from healthy endometrium (E-IUM) or endometriomas (E-OSIS) were subjected to in vitro decidualization (IVD) using 1 μM medroxyprogesterone acetate, 35 nM 17β-estradiol, and 0.05 mM 8-Br-cAMP. MAIN OUTCOME MEASURE(S) Expression of DNMT1, DNMT3A, and DNMT3B in E-IUM and E-OSIS were assessed by quantitative real-time polymerase chain reaction and immunoblotting. Recruitment of DNMT3B to the promoters of steroidogenic factor 1 (SF-1) and estrogen receptor α (ESR1) was examined by chromatin immunoprecipitation. RESULT(S) IVD treatment reduced DNMT3B messenger RNA (74%) and protein levels (81%) only in E-IUM; DNMT1 and DNMT3A were unchanged in both cell types. Significantly more DNMT3B bound to the SF-1 promoter in E-IUM compared with E-OSIS, and IVD treatment reduced binding in E-IUM to levels similar to those in E-OSIS. Enrichment of DNMT3B across 3 ESR1 promoters was reduced in E-IUM after IVD, although the more-distal promoter showed increased DNMT3B enrichment in E-OSIS after IVD. CONCLUSION(S) The inability to downregulate DNMT3B expression in E-OSIS may contribute to an aberrant epigenetic fingerprint that misdirects gene expression in endometriosis and contributes to its altered response to steroid hormones.
Collapse
Affiliation(s)
- Matthew T Dyson
- Division of Reproductive Biology Research, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | - Toshiyuki Kakinuma
- Division of Reproductive Biology Research, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Mary Ellen Pavone
- Division of Reproductive Biology Research, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Diana Monsivais
- Division of Reproductive Biology Research, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Antonia Navarro
- Division of Reproductive Biology Research, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Saurabh S Malpani
- Division of Reproductive Biology Research, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Masanori Ono
- Division of Reproductive Biology Research, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Serdar E Bulun
- Division of Reproductive Biology Research, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
23
|
Reizel Y, Spiro A, Sabag O, Skversky Y, Hecht M, Keshet I, Berman BP, Cedar H. Gender-specific postnatal demethylation and establishment of epigenetic memory. Genes Dev 2015; 29:923-33. [PMID: 25934504 PMCID: PMC4421981 DOI: 10.1101/gad.259309.115] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
DNA methylation patterns are set up in a relatively fixed programmed manner during normal embryonic development and are then stably maintained. Using genome-wide analysis, we discovered a postnatal pathway involving gender-specific demethylation that occurs exclusively in the male liver. This demodification is programmed to take place at tissue-specific enhancer sequences, and our data show that the methylation state at these loci is associated with and appears to play a role in the transcriptional regulation of nearby genes. This process is mediated by the secretion of testosterone at the time of sexual maturity, but the resulting methylation profile is stable and therefore can serve as an epigenetic memory even in the absence of this inducer. These findings add a new dimension to our understanding of the role of DNA methylation in vivo and provide the foundations for deciphering how environment can impact on the epigenetic regulation of genes in general.
Collapse
Affiliation(s)
- Yitzhak Reizel
- Department of Developmental Biology and Cancer Research, Hebrew University Medical School, Jerusalem 91120, Israel
| | - Adam Spiro
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ofra Sabag
- Department of Developmental Biology and Cancer Research, Hebrew University Medical School, Jerusalem 91120, Israel
| | - Yael Skversky
- Department of Developmental Biology and Cancer Research, Hebrew University Medical School, Jerusalem 91120, Israel
| | - Merav Hecht
- Department of Developmental Biology and Cancer Research, Hebrew University Medical School, Jerusalem 91120, Israel
| | - Ilana Keshet
- Department of Developmental Biology and Cancer Research, Hebrew University Medical School, Jerusalem 91120, Israel
| | - Benjamin P Berman
- Bioinformatics and Computational Biology Research Center, Cedars-Sinai Medical Center, Los Angeles, California 90048, USA
| | - Howard Cedar
- Department of Developmental Biology and Cancer Research, Hebrew University Medical School, Jerusalem 91120, Israel;
| |
Collapse
|
24
|
Schillebeeckx M, Pihlajoki M, Gretzinger E, Yang W, Thol F, Hiller T, Löbs AK, Röhrig T, Schrade A, Cochran R, Jay PY, Heikinheimo M, Mitra RD, Wilson DB. Novel markers of gonadectomy-induced adrenocortical neoplasia in the mouse and ferret. Mol Cell Endocrinol 2015; 399:122-30. [PMID: 25289806 PMCID: PMC4262703 DOI: 10.1016/j.mce.2014.09.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 07/18/2014] [Accepted: 09/29/2014] [Indexed: 12/25/2022]
Abstract
Gonadectomy (GDX) induces sex steroid-producing adrenocortical tumors in certain mouse strains and in the domestic ferret. Transcriptome analysis and DNA methylation mapping were used to identify novel genetic and epigenetic markers of GDX-induced adrenocortical neoplasia in female DBA/2J mice. Markers were validated using a combination of laser capture microdissection, quantitative RT-PCR, in situ hybridization, and immunohistochemistry. Microarray expression profiling of whole adrenal mRNA from ovariectomized vs. intact mice demonstrated selective upregulation of gonadal-like genes including Spinlw1 and Insl3 in GDX-induced adrenocortical tumors of the mouse. A complementary candidate gene approach identified Foxl2 as another gonadal-like marker expressed in GDX-induced neoplasms of the mouse and ferret. That both "male-specific" (Spinlw1) and "female-specific" (Foxl2) markers were identified is noteworthy and implies that the neoplasms exhibit mixed characteristics of male and female gonadal somatic cells. Genome-wide methylation analysis showed that two genes with hypomethylated promoters, Igfbp6 and Foxs1, are upregulated in GDX-induced adrenocortical neoplasms. These new genetic and epigenetic markers may prove useful for studies of steroidogenic cell development and for diagnostic testing.
Collapse
Affiliation(s)
- Maximiliaan Schillebeeckx
- Department of Genetics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - Marjut Pihlajoki
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki 00290, Finland
| | - Elisabeth Gretzinger
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Hochschule Mannheim, University of Applied Sciences, Mannheim 68163, Germany
| | - Wei Yang
- Department of Genetics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - Franziska Thol
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Hochschule Mannheim, University of Applied Sciences, Mannheim 68163, Germany
| | - Theresa Hiller
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Hochschule Mannheim, University of Applied Sciences, Mannheim 68163, Germany
| | - Ann-Kathrin Löbs
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Hochschule Mannheim, University of Applied Sciences, Mannheim 68163, Germany
| | - Theresa Röhrig
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Hochschule Mannheim, University of Applied Sciences, Mannheim 68163, Germany
| | - Anja Schrade
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki 00290, Finland
| | - Rebecca Cochran
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - Patrick Y Jay
- Department of Genetics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - Markku Heikinheimo
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Helsinki 00290, Finland
| | - Robi D Mitra
- Department of Genetics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA
| | - David B Wilson
- Department of Pediatrics, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis Children's Hospital, St. Louis, MO 63110, USA.
| |
Collapse
|
25
|
Diaz-Rodriguez E, Garcia-Rendueles AR, Ibáñez-Costa A, Gutierrez-Pascual E, Garcia-Lavandeira M, Leal A, Japon MA, Soto A, Venegas E, Tinahones FJ, Garcia-Arnes JA, Benito P, Angeles Galvez M, Jimenez-Reina L, Bernabeu I, Dieguez C, Luque RM, Castaño JP, Alvarez CV. Somatotropinomas, but not nonfunctioning pituitary adenomas, maintain a functional apoptotic RET/Pit1/ARF/p53 pathway that is blocked by excess GDNF. Endocrinology 2014; 155:4329-40. [PMID: 25137025 DOI: 10.1210/en.2014-1034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Acromegaly is caused by somatotroph cell adenomas (somatotropinomas [ACROs]), which secrete GH. Human and rodent somatotroph cells express the RET receptor. In rodents, when normal somatotrophs are deprived of the RET ligand, GDNF (Glial Cell Derived Neurotrophic Factor), RET is processed intracellularly to induce overexpression of Pit1 [Transcription factor (gene : POUF1) essential for transcription of Pituitary hormones GH, PRL and TSHb], which in turn leads to p19Arf/p53-dependent apoptosis. Our purpose was to ascertain whether human ACROs maintain the RET/Pit1/p14ARF/p53/apoptosis pathway, relative to nonfunctioning pituitary adenomas (NFPAs). Apoptosis in the absence and presence of GDNF was studied in primary cultures of 8 ACROs and 3 NFPAs. Parallel protein extracts were analyzed for expression of RET, Pit1, p19Arf, p53, and phospho-Akt. When GDNF deprived, ACRO cells, but not NFPAs, presented marked level of apoptosis that was prevented in the presence of GDNF. Apoptosis was accompanied by RET processing, Pit1 accumulation, and p14ARF and p53 induction. GDNF prevented all these effects via activation of phospho-AKT. Overexpression of human Pit1 (hPit1) directly induced p19Arf/p53 and apoptosis in a pituitary cell line. Using in silico studies, 2 CCAAT/enhancer binding protein alpha (cEBPα) consensus-binding sites were found to be 100% conserved in mouse, rat, and hPit1 promoters. Deletion of 1 cEBPα site prevented the RET-induced increase in hPit1 promoter expression. TaqMan qRT-PCR (real time RT-PCR) for RET, Pit1, Arf, TP53, GDNF, steroidogenic factor 1, and GH was performed in RNA from whole ACRO and NFPA tumors. ACRO but not NFPA adenomas express RET and Pit1. GDNF expression in the tumors was positively correlated with RET and negatively correlated with p53. In conclusion, ACROs maintain an active RET/Pit1/p14Arf/p53/apoptosis pathway that is inhibited by GDNF. Disruption of GDNF's survival function might constitute a new therapeutic route in acromegaly.
Collapse
Affiliation(s)
- Esther Diaz-Rodriguez
- Centre for Investigations in Medicine of the USC (E.D.-R., A.R.G.-G., M.G.-L., C.D., C.V.A.), University of Santiago de Compostela, Santiago de Compostela, Spain 15782; Department of Endocrinology (I.B.), University Hospital (University Hospital of Santiago de Compostela), Instituto de Investigación Sanitaria, Santiago de Compostela, Spain 15706; Departments of Cell Biology, Physiology, and Immunology (A.I.-C., E.G.-P., R.M.L., J.P.C.), and Morphological Sciences (L.J.-R.), University of Cordoba, and Reina Sofia University Hospital (P.B., M.A.G.), Maimonides Institute for Research in Biomedicine of Cordoba, Córdoba, Spain 14014; Departments of Endocrinology and Pathology (A.L., M.A.J., A.S., E.V.), Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, University of Sevilla, Sevilla, Spain 41013; Department of Endocrinology (F.J.T.), Hospital Virgen de la Victoria, and Department of Endocrinology (J.A.G.-A.), Hospital Carlos Haya, Malaga, Spain 29010; and CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn) 15706, spain (A.I.-C., F.J.T., P.B., I.B., C.D., R.M.L., J.P.C., C.V.A.), Spain 15706
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Chandra S, Baribault C, Lacey M, Ehrlich M. Myogenic differential methylation: diverse associations with chromatin structure. BIOLOGY 2014; 3:426-51. [PMID: 24949935 PMCID: PMC4085616 DOI: 10.3390/biology3020426] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 05/21/2014] [Accepted: 05/21/2014] [Indexed: 11/16/2022]
Abstract
Employing a new algorithm for identifying differentially methylated regions (DMRs) from reduced representation bisulfite sequencing profiles, we identified 1972 hypermethylated and 3250 hypomethylated myogenic DMRs in a comparison of myoblasts (Mb) and myotubes (Mt) with 16 types of nonmuscle cell cultures. DMRs co-localized with a variety of chromatin structures, as deduced from ENCODE whole-genome profiles. Myogenic hypomethylation was highly associated with both weak and strong enhancer-type chromatin, while hypermethylation was infrequently associated with enhancer-type chromatin. Both myogenic hypermethylation and hypomethylation often overlapped weak transcription-type chromatin and Polycomb-repressed-type chromatin. For representative genes, we illustrate relationships between DNA methylation, the local chromatin state, DNaseI hypersensitivity, and gene expression. For example, MARVELD2 exhibited myogenic hypermethylation in transcription-type chromatin that overlapped a silenced promoter in Mb and Mt while TEAD4 had myogenic hypomethylation in intronic subregions displaying enhancer-type or transcription-type chromatin in these cells. For LSP1, alternative promoter usage and active promoter-type chromatin were linked to highly specific myogenic or lymphogenic hypomethylated DMRs. Lastly, despite its myogenesis-associated expression, TBX15 had multiple hypermethylated myogenic DMRs framing its promoter region. This could help explain why TBX15 was previously reported to be underexpressed and, unexpectedly, its promoter undermethylated in placentas exhibiting vascular intrauterine growth restriction.
Collapse
Affiliation(s)
- Sruti Chandra
- Center for Bioinformatics and Genomics, New Orleans, LA 70112, USA.
| | - Carl Baribault
- Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA 70112, USA.
| | - Michelle Lacey
- Tulane Cancer Center, Tulane University Health Sciences Center, New Orleans, LA 70112, USA.
| | - Melanie Ehrlich
- Center for Bioinformatics and Genomics, New Orleans, LA 70112, USA.
| |
Collapse
|
27
|
Yan YE, Liu L, Wang JF, Liu F, Li XH, Qin HQ, Wang H. Prenatal nicotinic exposure suppresses fetal adrenal steroidogenesis via steroidogenic factor 1 (SF-1) deacetylation. Toxicol Appl Pharmacol 2014; 277:231-41. [DOI: 10.1016/j.taap.2014.03.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 02/26/2014] [Accepted: 03/22/2014] [Indexed: 10/25/2022]
|
28
|
Waterland RA. Epigenetic mechanisms affecting regulation of energy balance: many questions, few answers. Annu Rev Nutr 2014; 34:337-55. [PMID: 24850387 DOI: 10.1146/annurev-nutr-071813-105315] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Extensive human and animal model data show that nutrition and other environmental influences during critical periods of embryonic, fetal, and early postnatal life can affect the development of body weight regulatory pathways, with permanent consequences for risk of obesity. Epigenetic processes are widely viewed as a leading mechanism to explain the lifelong persistence of such "developmental programming" of energy balance. Despite meaningful progress in recent years, however, significant research obstacles impede our ability to test this hypothesis. Accordingly, this review attempts to summarize progress toward answering the following outstanding questions: Is epigenetic dysregulation a major cause of human obesity? In what cells/tissues is epigenetic regulation most important for energy balance? Does developmental programming of human body weight regulation occur via epigenetic mechanisms? Do epigenetic mechanisms have a greater impact on food intake or energy expenditure? Does epigenetic inheritance contribute to transgenerational patterns of obesity? In each case, significant obstacles and suggested approaches to surmounting them are elaborated.
Collapse
Affiliation(s)
- Robert A Waterland
- Departments of Pediatrics and Molecular & Human Genetics, Baylor College of Medicine, USDA/ARS Children's Nutrition Research Center, Houston, Texas 77030;
| |
Collapse
|
29
|
Heymsfield SB, Avena NM, Baier L, Brantley P, Bray GA, Burnett LC, Butler MG, Driscoll DJ, Egli D, Elmquist J, Forster JL, Goldstone AP, Gourash LM, Greenway FL, Han JC, Kane JG, Leibel RL, Loos RJ, Scheimann AO, Roth CL, Seeley RJ, Sheffield V, Tauber M, Vaisse C, Wang L, Waterland RA, Wevrick R, Yanovski JA, Zinn AR. Hyperphagia: current concepts and future directions proceedings of the 2nd international conference on hyperphagia. Obesity (Silver Spring) 2014; 22 Suppl 1:S1-S17. [PMID: 24574081 PMCID: PMC4159941 DOI: 10.1002/oby.20646] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 10/11/2013] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Hyperphagia is a central feature of inherited disorders (e.g., Prader-Willi Syndrome) in which obesity is a primary phenotypic component. Hyperphagia may also contribute to obesity as observed in the general population, thus raising the potential importance of common underlying mechanisms and treatments. Substantial gaps in understanding the molecular basis of inherited hyperphagia syndromes are present as are a lack of mechanistic of mechanistic targets that can serve as a basis for pharmacologic and behavioral treatments. DESIGN AND METHODS International conference with 28 experts, including scientists and caregivers, providing presentations, panel discussions, and debates. RESULTS The reviewed collective research and clinical experience provides a critical body of new and novel information on hyperphagia at levels ranging from molecular to population. Gaps in understanding and tools needed for additional research were identified. CONCLUSIONS This report documents the full scope of important topics reviewed at a comprehensive international meeting devoted to the topic of hyperphagia and identifies key areas for future funding and research.
Collapse
Affiliation(s)
- Steven B. Heymsfield
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Nicole M. Avena
- Department of Psychiatry, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Leslie Baier
- Diabetes Molecular Genetics Section, Phoenix Epidemiology and Clinical Research Branch, NIDDK, NIH, Phoenix, Arizona, USA
| | - Phillip Brantley
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - George A. Bray
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Lisa C. Burnett
- College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | | | - Daniel J. Driscoll
- Division of Genetics and Metabolism, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, USA
| | - Dieter Egli
- College of Physicians and Surgeons, Columbia University, New York, New York, USA
- New York Stem Cell Foundation, New York, New York, USA
| | | | | | - Anthony P. Goldstone
- Metabolic & Molecular Imaging Group, MRC Clinical Sciences Centre, Imperial College London, UK
| | | | - Frank L. Greenway
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana, USA
| | - Joan C. Han
- Section on Growth and Obesity, National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - James G. Kane
- Prader-Willi Syndrome Association (USA), Sarasota, Florida, USA
| | - Rudolph L. Leibel
- College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Ruth J.F. Loos
- The Genetics of Obesity and Related Metabolic Traits Program, The Charles Bronfman Institute for Personalized Medicine, The Mindich Child Health and Development Institute, The Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ann O. Scheimann
- Division of Pediatric Gastroenterology, Nutrition and Hepatology at Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Christian L. Roth
- Center for Integrative Brain Research, Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Randy J. Seeley
- Center of Excellence in Obesity and Diabetes, University of Cincinnati, Cincinnati, Ohio, USA
| | - Val Sheffield
- Pediatrics and Medical Genetics, University of Iowa College of Medicine, Iowa City, Iowa, USA
| | - Maïthé Tauber
- Department of Endocrinology, Hôpital des Enfants and Paul Sabatier Université, Toulouse, France
| | - Christian Vaisse
- University of California, San Francisco, School of Medicine, San Francisco, California, USA
| | - Liheng Wang
- College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Robert A. Waterland
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics and Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Rachel Wevrick
- Department of Medical Genetics, University of Alberta, Edmonton, Canada
| | - Jack A. Yanovski
- Section on Growth and Obesity, National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Andrew R. Zinn
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
30
|
Abstract
Transient receptor potential (TRP) ion channels have been detected in neurons that are part of the neural network controlling reproductive physiology and behavior. In this chapter we will primarily take a look at the classical/canonical TRP (TRPC) channels but will also examine some other members of the TRP channel superfamily in reproductive (neuro)endocrinology. The referenced data suggest that different TRP proteins could play functional roles at different levels of the reproductive pathway. Still, our understanding of TRP channel involvement in (neuro)endocrinology is quite limited. Due to their mechanism of activation and complex regulation, these channels are however ideally suited to be part of the transduction machinery of hormone-secreting cells.
Collapse
Affiliation(s)
- Trese Leinders-Zufall
- Department of Physiology, University of Saarland School of Medicine, 66421, Homburg, Germany,
| | | |
Collapse
|
31
|
Fortin J, Boehm U, Weinstein MB, Graff JM, Bernard DJ. Follicle-stimulating hormone synthesis and fertility are intact in mice lacking SMAD3 DNA binding activity and SMAD2 in gonadotrope cells. FASEB J 2013; 28:1474-85. [PMID: 24308975 DOI: 10.1096/fj.13-237818] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The activin/inhibin system regulates follicle-stimulating hormone (FSH) synthesis and release by pituitary gonadotrope cells in mammals. In vitro cell line data suggest that activins stimulate FSH β-subunit (Fshb) transcription via complexes containing the receptor-regulated SMAD proteins SMAD2 and SMAD3. Here, we used a Cre-loxP approach to determine the necessity for SMAD2 and/or SMAD3 in FSH synthesis in vivo. Surprisingly, mice with conditional mutations in both Smad2 and Smad3 specifically in gonadotrope cells are fertile and produce FSH at quantitatively normal levels. Notably, however, we discovered that the recombined Smad3 allele produces a transcript that encodes the entirety of the SMAD3 C-terminal Mad homology 2 (MH2) domain. This protein behaves similarly to full-length SMAD3 in Fshb transcriptional assays. As the truncated protein lacks the N-terminal Mad homology 1 (MH1) domain, these results show that SMAD3 DNA-binding activity as well as SMAD2 are dispensable for normal FSH synthesis in vivo. Furthermore, the observation that deletion of proximal exons does not remove all SMAD3 function may facilitate interpretation of divergent phenotypes previously described in different Smad3 knockout mouse lines.
Collapse
Affiliation(s)
- Jérôme Fortin
- 1Department of Pharmacology and Therapeutics, McGill University, 3655 Promenade Sir William Osler, Rm. 1315, Montréal, QC, H3G 1Y6, Canada. J.F.,
| | | | | | | | | |
Collapse
|
32
|
Li G, Zhang W, Baker MS, Laritsky E, Mattan-Hung N, Yu D, Kunde-Ramamoorthy G, Simerly RB, Chen R, Shen L, Waterland RA. Major epigenetic development distinguishing neuronal and non-neuronal cells occurs postnatally in the murine hypothalamus. Hum Mol Genet 2013; 23:1579-90. [PMID: 24186871 DOI: 10.1093/hmg/ddt548] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Prenatal and early postnatal environment can persistently alter one's risk of obesity. Environmental effects on hypothalamic developmental epigenetics constitute a likely mechanism underlying such 'developmental programming' of energy balance regulation. To advance our understanding of these processes, it is essential to develop approaches to disentangle the cellular and regional heterogeneity of hypothalamic developmental epigenetics. We therefore performed genome-scale DNA methylation profiling in hypothalamic neurons and non-neuronal cells at postnatal day 0 (P0) and P21 and found, surprisingly, that most of the DNA methylation differences distinguishing these two cell types are established postnatally. In particular, neuron-specific increases in DNA methylation occurred extensively at genes involved in neuronal development. Quantitative bisulfite pyrosequencing verified our methylation profiling results in all 15 regions examined, and expression differences were associated with DNA methylation at several genes. We also identified extensive methylation differences between the arcuate (ARH) and paraventricular nucleus of the hypothalamus (PVH). Integrating these two data sets showed that genomic regions with PVH versus ARH differential methylation strongly overlap with those undergoing neuron-specific increases from P0 to P21, suggesting that these developmental changes occur preferentially in either the ARH or PVH. In particular, neuron-specific methylation increases at the 3' end of Shh localized to the ARH and were positively associated with gene expression. Our data indicate a key role for DNA methylation in establishing the gene expression potential of diverse hypothalamic cell types, and provide the novel insight that early postnatal life is a critical period for cell type-specific epigenetic development in the murine hypothalamus.
Collapse
Affiliation(s)
- Ge Li
- Department of Pediatrics, Baylor College of Medicine, USDA/ARS Children's Nutrition Research Center
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Webber BR, Iacovino M, Choi SH, Tolar J, Kyba M, Blazar BR. DNA methylation of Runx1 regulatory regions correlates with transition from primitive to definitive hematopoietic potential in vitro and in vivo. Blood 2013; 122:2978-86. [PMID: 24030384 PMCID: PMC3811172 DOI: 10.1182/blood-2013-03-489369] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 09/03/2013] [Indexed: 12/11/2022] Open
Abstract
The transcription factor Runx1 (AML1) is a central regulator of hematopoiesis and is required for the formation of definitive hematopoietic stem cells (HSCs). Runx1 is alternatively expressed from two promoters: the proximal (P2) prevails during primitive hematopoiesis, while the distal (P1) dominates in definitive HSCs. Although some transcription factor binding sites and cis-regulatory elements have been identified, a mechanistic explanation for the alternative promoter usage remains elusive. We investigated DNA methylation of known Runx1 cis-elements at stages of hematopoietic development in vivo and during differentiation of murine embryonic stem cells (ESCs) in vitro. In vivo, we find loss of methylation correlated with the primitive to definitive transition at the P1 promoter. In vitro, hypomethylation, acquisition of active chromatin modifications, and increased transcriptional activity at P1 are promoted by direct interaction with HOXB4, a transcription factor that confers definitive repopulation status on primitive hematopoietic progenitors. These data demonstrate a novel role for DNA methylation in the alternative promoter usage at the Runx1 locus and identify HOXB4 as a direct activator of the P1 promoter. This epigenetic signature should serve as a novel biomarker of HSC potential in vivo, and during ESC differentiation in vitro.
Collapse
Affiliation(s)
- Beau R Webber
- Division of Hematology-Oncology, Blood and Marrow Transplantation, and
| | | | | | | | | | | |
Collapse
|
34
|
Chanda S, Dasgupta UB, Mazumder DG, Saha J, Gupta B. Human GMDS gene fragment hypermethylation in chronic high level of arsenic exposure with and without arsenic induced cancer. SPRINGERPLUS 2013; 2:557. [PMID: 24255851 PMCID: PMC3825097 DOI: 10.1186/2193-1801-2-557] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 09/26/2013] [Indexed: 12/31/2022]
Abstract
Arsenic, though a poor mutagen, is an accepted environmental carcinogen. Perturbation of DNA methylation pattern leading to aberrant gene expression has been hypothesized as the mechanism for arsenic induced carcinogenesis. We had earlier demonstrated the hypermethylation of promoter region of p53 and p16 genes in persons exposed to different doses of arsenic. Till now no genomic hot spot has been identified which is frequently hypermethylated or hypomethylated in persons chronically exposed to environmental arsenic. In the present work, we have identified one hypermethylated sequence by methyl-sensitive arbitrarily primed polymerase chain reaction in the peripheral blood leukocyte DNA of chronically arsenic exposed persons with and without arsenic induced skin cancer. The sequence is from GMDS gene responsible for fucose metabolism. Southern hybridization of the sequence to the amplification products of methyl sensitive restriction enzyme digested genome of persons exposed to different doses of arsenic indicated that methylation increased in a dose dependent manner.
Collapse
Affiliation(s)
- Sarmishtha Chanda
- Department of Biophysics, Molecular biology & Genetics, University of Calcutta, Kolkata, West Bengal 700092 India ; Department of Physiology, Presidency University, Kolkata, West Bengal 700073 India
| | | | | | | | | |
Collapse
|
35
|
The CpG island in the murine foxl2 proximal promoter is differentially methylated in primary and immortalized cells. PLoS One 2013; 8:e76642. [PMID: 24098544 PMCID: PMC3788739 DOI: 10.1371/journal.pone.0076642] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 08/30/2013] [Indexed: 11/23/2022] Open
Abstract
Forkhead box L2 (Foxl2), a member of the forkhead transcription factor family, plays important roles in pituitary follicle-stimulating hormone synthesis and in ovarian maintenance and function. Mutations in the human FOXL2 gene cause eyelid malformations and premature ovarian failure. FOXL2/Foxl2 is expressed in pituitary gonadotrope and thyrotrope cells, the perioptic mesenchyme of the developing eyelid, and ovarian granulosa cells. The mechanisms governing this cell-restricted expression have not been described. We mapped the Foxl2 transcriptional start site in immortalized murine gonadotrope-like cells, LβT2, by 5’ rapid amplification of cDNA ends and then PCR amplified approximately 1 kb of 5’ flanking sequence from murine genomic DNA. When ligated into a reporter plasmid, the proximal promoter conferred luciferase activity in both homologous (LβT2) and, unexpectedly, heterologous (NIH3T3) cells. In silico analyses identified a CpG island in the proximal promoter and 5’ untranslated region, suggesting that Foxl2 transcription might be regulated epigenetically. Indeed, pyrosequencing and quantitative analysis of DNA methylation using real-time PCR revealed Foxl2 proximal promoter hypomethylation in homologous compared to some, though not all, heterologous cell lines. The promoter was also hypomethylated in purified murine gonadotropes. In vitro promoter methylation completely silenced reporter activity in heterologous and homologous cells. Collectively, the data suggest that differential proximal promoter DNA methylation may contribute to cell-specific Foxl2 expression in some cellular contexts. However, gonadotrope-specific expression of the gene cannot be explained by promoter hypomethylation alone.
Collapse
|
36
|
Savulescu D, Feng J, Ping YS, Mai O, Boehm U, He B, O'Malley BW, Melamed P. Gonadotropin-releasing hormone-regulated prohibitin mediates apoptosis of the gonadotrope cells. Mol Endocrinol 2013; 27:1856-70. [PMID: 24085822 DOI: 10.1210/me.2013-1210] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
GnRH regulates circulating levels of the gonadotropins but has also been implicated in establishing the gonadotrope cell population. Consistent with this, GnRH induces proliferation of partially differentiated gonadotropes, while reducing the numbers of fully differentiated cells. We have previously reported that the proapoptotic protein, prohibitin (PHB) is expressed more abundantly in gonadotrope-derived LβT2 cells than in partially differentiated αT3-1 gonadotrope precursor cells, suggesting a possible role for PHB in GnRH-induced apoptosis. We show here that PHB is required for GnRH-induced apoptosis in mature gonadotropes. PHB expression and activity are regulated by GnRH: its transcription is via c-Jun NH2-terminal kinase, whereas its nuclear export follows activation of ERK. Moreover, PHB levels are down-regulated by microRNA27, which is expressed at lower levels in mature gonadotropes, possibly explaining the switch to an apoptotic response with development. PHB is required for mitochondrial import of the proapoptotic BAX, whose expression is also induced by GnRH-activated c-Jun NH2-terminal kinase, as is expression of the BH3-only protein, HRK, and this too plays a role in GnRH-induced apoptosis. Finally, we show that gonadotrope-specific PHB-knockout mice display reproductive abnormalities, including a larger gonadotrope population, increased LH levels, reduced fertility, and altered gonad development. We thus demonstrate a role for PHB in GnRH-induced cell death in mature gonadotropes, which is crucial for the normal development and function of the reproductive axis.
Collapse
Affiliation(s)
- Dana Savulescu
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, 32000 Israel.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Intragenic DNA methylation in transcriptional regulation, normal differentiation and cancer. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2013; 1829:1161-74. [PMID: 23938249 DOI: 10.1016/j.bbagrm.2013.08.001] [Citation(s) in RCA: 157] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 08/02/2013] [Accepted: 08/05/2013] [Indexed: 02/06/2023]
Abstract
Ever since the discovery of DNA methylation at cytosine residues, the role of this so called fifth base has been extensively studied and debated. Until recently, the majority of DNA methylation studies focused on the analysis of CpG islands associated to promoter regions. However, with the upcoming possibilities to study DNA methylation in a genome-wide context, this epigenetic mark can now be studied in an unbiased manner. As a result, recent studies have shown that not only promoters but also intragenic and intergenic regions are widely modulated during physiological processes and disease. In particular, it is becoming increasingly clear that DNA methylation in the gene body is not just a passive witness of gene transcription but it seems to be actively involved in multiple gene regulation processes. In this review we discuss the potential role of intragenic DNA methylation in alternative promoter usage, regulation of short and long non-coding RNAs, alternative RNA processing, as well as enhancer activity. Furthermore, we summarize how the intragenic DNA methylome is modified both during normal cell differentiation and neoplastic transformation.
Collapse
|
38
|
Li G, Kohorst JJ, Zhang W, Laritsky E, Kunde-Ramamoorthy G, Baker MS, Fiorotto ML, Waterland RA. Early postnatal nutrition determines adult physical activity and energy expenditure in female mice. Diabetes 2013; 62:2773-83. [PMID: 23545705 PMCID: PMC3717861 DOI: 10.2337/db12-1306] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Decades of research in rodent models has shown that early postnatal overnutrition induces excess adiposity and other components of metabolic syndrome that persist into adulthood. The specific biologic mechanisms explaining the persistence of these effects, however, remain unknown. On postnatal day 1 (P1), mice were fostered in control (C) or small litters (SL). SL mice had increased body weight and adiposity at weaning (P21), which persisted to adulthood (P180). Detailed metabolic studies indicated that female adult SL mice have decreased physical activity and energy expenditure but not increased food intake. Genome-scale DNA methylation profiling identified extensive changes in hypothalamic DNA methylation during the suckling period, suggesting that it is a critical period for developmental epigenetics in the mouse hypothalamus. Indeed, SL mice exhibited subtle and sex-specific changes in hypothalamic DNA methylation that persisted from early life to adulthood, providing a potential mechanistic basis for the sustained physiological effects. Expression profiling in adult hypothalamus likewise provided evidence of widespread sex-specific alterations in gene expression. Together, our data indicate that early postnatal overnutrition leads to a reduction in spontaneous physical activity and energy expenditure in females and suggest that early postnatal life is a critical period during which nutrition can affect hypothalamic developmental epigenetics.
Collapse
Affiliation(s)
- Ge Li
- Department of Pediatrics, Baylor College of Medicine, United States Department of Agriculture/Agricultural Research Service Children’s Nutrition Research Center, Houston, Texas
| | - John J. Kohorst
- Department of Pediatrics, Baylor College of Medicine, United States Department of Agriculture/Agricultural Research Service Children’s Nutrition Research Center, Houston, Texas
| | - Wenjuan Zhang
- Department of Pediatrics, Baylor College of Medicine, United States Department of Agriculture/Agricultural Research Service Children’s Nutrition Research Center, Houston, Texas
| | - Eleonora Laritsky
- Department of Pediatrics, Baylor College of Medicine, United States Department of Agriculture/Agricultural Research Service Children’s Nutrition Research Center, Houston, Texas
| | - Govindarajan Kunde-Ramamoorthy
- Department of Pediatrics, Baylor College of Medicine, United States Department of Agriculture/Agricultural Research Service Children’s Nutrition Research Center, Houston, Texas
| | - Maria S. Baker
- Department of Pediatrics, Baylor College of Medicine, United States Department of Agriculture/Agricultural Research Service Children’s Nutrition Research Center, Houston, Texas
| | - Marta L. Fiorotto
- Department of Pediatrics, Baylor College of Medicine, United States Department of Agriculture/Agricultural Research Service Children’s Nutrition Research Center, Houston, Texas
| | - Robert A. Waterland
- Department of Pediatrics, Baylor College of Medicine, United States Department of Agriculture/Agricultural Research Service Children’s Nutrition Research Center, Houston, Texas
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, Texas
- Corresponding author: Robert A. Waterland,
| |
Collapse
|
39
|
Xue Q, Xu Y, Yang H, Zhang L, Shang J, Zeng C, Yin P, Bulun SE. Methylation of a novel CpG island of intron 1 is associated with steroidogenic factor 1 expression in endometriotic stromal cells. Reprod Sci 2013; 21:395-400. [PMID: 23899549 DOI: 10.1177/1933719113497283] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Steroidogenic factor 1 (SF-1), a transcriptional factor essential for estrogen biosynthesis, is undetectable in endometrial stromal cells and aberrantly expressed in endometriotic stromal cells. OBJECTIVE We tried to gain further insight into the mechanism for differential SF-1 expression in endometrial and endometriotic stromal cells. DESIGN We had previously identified a novel CpG island in SF-1, which is located in the downstream intron 1 region. Here, we evaluated the methylation status of this CpG island. PATIENTS We obtained the eutopic endometrium from disease-free participants (n = 8) and the walls of cystic endometriosis lesions of the ovaries from another group of participants (n = 8). None of the patients had received any preoperative hormonal therapy. INTERVENTIONS Stromal cells were isolated from these 2 types of tissues and subjected to DNA bisulfite treatment and sequence analysis. RESULTS The SF-1 messenger RNA (mRNA) levels in endometriotic stromal cells were significantly higher than those in endometrial stromal cells. Bisulfite sequencing showed strikingly increased methylation of a 1-kbp region around the previously identified CpG island in endometriotic cells compared with endometrial cells (P < .001). A strong correlation between SF-1 mRNA levels and percentage methylation of the intron 1 region of the SF-1 gene was observed in endometriotic cells (Spearman correlation coefficient, .96; P < .001). CONCLUSIONS Methylation of the intron 1 region of the SF-1 gene is associated with its expression in endometriotic cells. This CpG island therefore plays an important role in regulating SF-1 expression.
Collapse
Affiliation(s)
- Qing Xue
- 1Department of Obstetrics and Gynecology, First Hospital of Beijing University, Beijing, P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Hoivik EA, Witsoe SL, Bergheim IR, Xu Y, Jakobsson I, Tengholm A, Doskeland SO, Bakke M. DNA methylation of alternative promoters directs tissue specific expression of Epac2 isoforms. PLoS One 2013; 8:e67925. [PMID: 23861833 PMCID: PMC3701594 DOI: 10.1371/journal.pone.0067925] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 05/23/2013] [Indexed: 12/22/2022] Open
Abstract
Epac 1 and Epac 2 (Epac1/2; exchange factors directly activated by cAMP) are multidomain proteins that mediate cellular responses upon activation by the signaling molecule cAMP. Epac1 is ubiquitously expressed, whereas Epac2 exhibits a restricted expression pattern. The gene encoding Epac2 gives rise to at least three protein isoforms (Epac2A, Epac2B and Epac2C) that exhibit confined tissue and cell specific expression profiles. Here, we describe alternative promoter usage for the different isoforms of Epac2, and demonstrate that the activity of these promoters depend on the DNA methylation status. Bisulfite sequencing demonstrated that the level of methylation of the promoters in different tissues correlates with Epac2 isoform expression. The presented data indicate that the tissue-specific expression of the Epac2 isoforms is epigenetically regulated, and identify tissue-specific differentially methylated promoter regions within the Epac2 locus that are essential for its transcriptional control.
Collapse
Affiliation(s)
- Erling A. Hoivik
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Department of Clinical Medicine, Section for Gynecology and Obstetrics, University of Bergen, Bergen, Norway
| | | | | | - Yunjian Xu
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Ida Jakobsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Anders Tengholm
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | | | - Marit Bakke
- Department of Biomedicine, University of Bergen, Bergen, Norway
- * E-mail:
| |
Collapse
|
41
|
Hoivik EA, Bjanesoy TE, Bakke M. Epigenetic regulation of the gene encoding steroidogenic factor-1. Mol Cell Endocrinol 2013; 371:133-9. [PMID: 23313548 DOI: 10.1016/j.mce.2012.12.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 12/27/2012] [Accepted: 12/28/2012] [Indexed: 12/21/2022]
Abstract
The nuclear receptor steroidogenic factor 1 (SF-1) is expressed in a precise time and cell-specific pattern in the endocrine system. Three intronic enhancers and one upstream enhancer, which are required for controlling the restricted expression of SF-1, have been identified in the mouse gene encoding SF-1. In recent years, efforts from several laboratories have established that expression of SF-1 is controlled by DNA methylation. CpG-sites are found in the basal promoter as well as in the intronic enhancers, and the methylation status of these genomic regions nearly perfectly correlates with their transcriptional activity such that they are hypomethylated in tissues where they are active, and generally hypermethylated in tissues where they are not active. This review summarizes the present knowledge of how tissue differentially methylated regions control the transcriptional activity of the SF-1 gene, and how irregularities in the methylation pattern can contribute to disease development.
Collapse
Affiliation(s)
- Erling A Hoivik
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | | |
Collapse
|
42
|
Alim Z, Hartshorn C, Mai O, Stitt I, Clay C, Tobet S, Boehm U. Gonadotrope plasticity at cellular and population levels. Endocrinology 2012; 153:4729-39. [PMID: 22893721 PMCID: PMC3685717 DOI: 10.1210/en.2012-1360] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Hormone-secreting cells within the anterior pituitary gland may form organized and interdigitated networks that adapt to changing endocrine conditions in different physiological contexts. For gonadotropes, this might reflect a strategy to cope with acute changes throughout different female reproductive stages. The current study examined gonadotropes in female mice at characteristically different hormonal stages: prepubertal, postpubertal, and lactating. Gonadotrope plasticity was examined at the level of the whole population and single cells at different stages by imaging both fixed and live pituitary slices. The use of a model animal providing for the identification of selectively fluorescent gonadotropes allowed the particular advantage of defining cellular plasticity specifically for gonadotropes. In vivo analyses of gonadotropes relative to vasculature showed significantly different gonadotrope distributions across physiological states. Video microscopy studies using live slices ex vivo demonstrated pituitary cell plasticity in the form of movements and protrusions in response to GnRH. As positive feedback from rising estradiol levels is important for priming the anterior pituitary gland for the LH surge, experiments provide evidence of estradiol effects on GnRH signaling in gonadotropes. The experiments presented herein provide new insight into potential plasticity of gonadotropes within the anterior pituitary glands of female mice.
Collapse
Affiliation(s)
- Zahara Alim
- Institute for Neural Signal Transduction, Center for Molecular Neurobiology, 20251 Hamburg, Germany
| | | | | | | | | | | | | |
Collapse
|
43
|
Meldi KM, Gaconnet GA, Mayo KE. DNA methylation and histone modifications are associated with repression of the inhibin α promoter in the rat corpus luteum. Endocrinology 2012; 153:4905-17. [PMID: 22865368 PMCID: PMC3512026 DOI: 10.1210/en.2012-1292] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The transition from follicle to corpus luteum after ovulation is associated with profound morphological and functional changes and is accompanied by corresponding changes in gene expression. The gene encoding the α subunit of the dimeric reproductive hormone inhibin is maximally expressed in the granulosa cells of the preovulatory follicle, is rapidly repressed by the ovulatory LH surge, and is expressed at only very low levels in the corpus luteum. Although previous studies have identified transient repressors of inhibin α gene transcription, little is known about how this repression is maintained in the corpus luteum. This study examines the role of epigenetic changes, including DNA methylation and histone modification, in silencing of inhibin α gene expression. Bisulfite sequencing reveals that methylation of the inhibin α proximal promoter is low in preovulatory and ovulatory follicles but is elevated in the corpus luteum. Increased methylation during luteinization is observed within the cAMP response element in the promoter, and EMSA demonstrate that methylation of this site inhibits cAMP response element binding protein binding in vitro. Chromatin immunoprecipitation reveals that repressive histone marks H3K9 and H3K27 trimethylation are increased on the inhibin α promoter in primary luteal cells, whereas the activation mark H3K4 trimethylation is decreased. The changes in histone modification precede the alterations in DNA methylation, suggesting that they facilitate the recruitment of DNA methyltransferases. We show that the DNA methyltransferase DNMT3a is present in the ovary and in luteal cells when the inhibin α promoter becomes methylated and observe recruitment of DNMT3a to the inhibin promoter during luteinization.
Collapse
Affiliation(s)
- Kristen M Meldi
- Department of Molecular Biosciences, Center for Reproductive Science, Northwestern University, Evanston, Illinois 60208, USA
| | | | | |
Collapse
|
44
|
Abstract
Different cell types within a single organism are generally distinguished by strikingly different patterns of gene expression, which are dynamic throughout development and adult life. Distal enhancer elements are key drivers of spatiotemporal specificity in gene regulation. Often located tens of kilobases from their target promoters and functioning in an orientation-independent manner, the identification of bona fide enhancers has proved a formidable challenge. With the development of ChIP-seq, global cataloging of putative enhancers has become feasible. Here, we review the current understanding of the chromatin landscape at enhancers and how these chromatin features enable robust identification of tissue-specific enhancers.
Collapse
Affiliation(s)
- Gabriel E Zentner
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | |
Collapse
|
45
|
Current world literature. Curr Opin Endocrinol Diabetes Obes 2012; 19:233-47. [PMID: 22531108 DOI: 10.1097/med.0b013e3283542fb3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
46
|
Gardiner JR, Shima Y, Morohashi KI, Swain A. SF-1 expression during adrenal development and tumourigenesis. Mol Cell Endocrinol 2012; 351:12-8. [PMID: 22024498 DOI: 10.1016/j.mce.2011.10.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 10/05/2011] [Accepted: 10/07/2011] [Indexed: 02/01/2023]
Abstract
SF-1 is a master regulator of steroidogenesis whose expression is critical for normal adrenal and gonadal organogenesis. Strict maintenance of SF-1 levels is essential, and mutations causing under- or overexpression result in congenital adrenal and gonadal defects or hyperplasia, respectively. Data from transgenic mouse models points to a network of transcription factors responsible for stringent regulation of Sf-1 expression during development, which bind to intronic enhancer elements in addition to the basal promoter to specifically modulate transcription in each Sf-1-expressing tissue. Furthermore, analysis of the role of SF-1 in adrenal tumourigenesis implies that improper developmental regulation of Sf-1 expression may have postnatal consequences separate from the well-documented developmental defects.
Collapse
|
47
|
Roberson EDO, Liu Y, Ryan C, Joyce CE, Duan S, Cao L, Martin A, Liao W, Menter A, Bowcock AM. A subset of methylated CpG sites differentiate psoriatic from normal skin. J Invest Dermatol 2011; 132:583-92. [PMID: 22071477 PMCID: PMC3568942 DOI: 10.1038/jid.2011.348] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Psoriasis is a chronic inflammatory immune-mediated disorder affecting the skin and other organs including joints. Over 1,300 transcripts are altered in psoriatic involved skin compared to normal skin. However to our knowledge global epigenetic profiling of psoriatic skin is previously unreported. Here we describe a genome-wide study of altered CpG methylation in psoriatic skin. We determined the methylation levels at 27,578 CpG sites in skin samples from individuals with psoriasis (12 involved, 8 uninvolved) and 10 unaffected individuals. CpG methylation of involved skin differed from normal skin at 1,108 sites. Twelve mapped to the epidermal differentiation complex, upstream or within genes that are highly up-regulated in psoriasis. Hierarchical clustering of 50 of the top differentially methylated (DM) sites separated psoriatic from normal skin samples. CpG sites where methylation was correlated with gene expression are reported. Sites with inverse correlations between methylation and nearby gene expression include those of KYNU, OAS2, S100A12, and SERPINB3, whose strong transcriptional up-regulation are important discriminators of psoriasis. We observed intrinsic epigenetic differences in uninvolved skin. Pyrosequencing of bisulfite-treated DNA from skin biopsies at three DM loci confirmed earlier findings and revealed reversion of methylation levels towards the non-psoriatic state after one month of anti-TNF-α therapy.
Collapse
Affiliation(s)
- Elisha D O Roberson
- Department of Genetics, Washington University School of Medicine, St Louis, Missouri 63110, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Mello MPD, França ESDS, Fabbri HC, Maciel-Guerra AT, Guerra-Júnior G. Multifunctional role of steroidogenic factor 1 and disorders of sex development. ACTA ACUST UNITED AC 2011; 55:607-12. [DOI: 10.1590/s0004-27302011000800015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 10/19/2011] [Indexed: 02/02/2023]
Abstract
Disorders of sex development (DSD) involve several conditions that result from abnormalities during gonadal determination and differentiation. Some of these disorders may manifest at birth by ambiguous genitalia; others are diagnosed only at puberty, by the delayed onset of secondary sexual characteristics. Sex determination and differentiation in humans are processes that involve the interaction of several genes such as WT1, NR5A1, NR0B1, SOX9, among others, in the testicular pathway, and WNT4, DAX1, FOXL2 and RSPO1, in the ovarian pathway. One of the major proteins in mammalian gonadal differentiation is the steroidogenic nuclear receptor factor 1 (SF1). This review will cover some of the most recent data on SF1 functional roles and findings related to mutations in its coding gene, NR5A1.
Collapse
|