1
|
Ulrich A, Wu Y, Draisma H, Wharton J, Swietlik EM, Cebola I, Vasilaki E, Balkhiyarova Z, Jarvelin MR, Auvinen J, Herzig KH, Coghlan JG, Lordan J, Church C, Howard LS, Pepke-Zaba J, Toshner M, Wort SJ, Kiely DG, Condliffe R, Lawrie A, Gräf S, Morrell NW, Wilkins MR, Prokopenko I, Rhodes CJ. Blood DNA methylation profiling identifies cathepsin Z dysregulation in pulmonary arterial hypertension. Nat Commun 2024; 15:330. [PMID: 38184627 PMCID: PMC10771427 DOI: 10.1038/s41467-023-44683-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 12/28/2023] [Indexed: 01/08/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterised by pulmonary vascular remodelling causing premature death from right heart failure. Established DNA variants influence PAH risk, but susceptibility from epigenetic changes is unknown. We addressed this through epigenome-wide association study (EWAS), testing 865,848 CpG sites for association with PAH in 429 individuals with PAH and 1226 controls. Three loci, at Cathepsin Z (CTSZ, cg04917472), Conserved oligomeric Golgi complex 6 (COG6, cg27396197), and Zinc Finger Protein 678 (ZNF678, cg03144189), reached epigenome-wide significance (p < 10-7) and are hypermethylated in PAH, including in individuals with PAH at 1-year follow-up. Of 16 established PAH genes, only cg10976975 in BMP10 shows hypermethylation in PAH. Hypermethylation at CTSZ is associated with decreased blood cathepsin Z mRNA levels. Knockdown of CTSZ expression in human pulmonary artery endothelial cells increases caspase-3/7 activity (p < 10-4). DNA methylation profiles are altered in PAH, exemplified by the pulmonary endothelial function modifier CTSZ, encoding protease cathepsin Z.
Collapse
Affiliation(s)
- Anna Ulrich
- Department of Clinical and Experimental Medicine, University of Surrey, Surrey, UK
| | - Yukyee Wu
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Harmen Draisma
- Department of Clinical and Experimental Medicine, University of Surrey, Surrey, UK
- Section of Genetics & Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - John Wharton
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Emilia M Swietlik
- VPD Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Inês Cebola
- Section of Genetics & Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Eleni Vasilaki
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Zhanna Balkhiyarova
- Department of Clinical and Experimental Medicine, University of Surrey, Surrey, UK
- Section of Genetics & Genomics, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- People-Centred Artificial Intelligence Institute, University of Surrey, Guildford, UK
| | - Marjo-Riitta Jarvelin
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Unit of Primary Care, Oulu University Hospital, Oulu, Finland
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, London, UK
| | - Juha Auvinen
- Center for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Karl-Heinz Herzig
- Institute of Biomedicine, Medical Research Center Oulu, Oulu University and Oulu University Hospital, Oulu, Finland
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | - Colin Church
- Golden Jubilee National Hospital and University of Glasgow, Glasgow, UK
| | - Luke S Howard
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Mark Toshner
- VPD Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Stephen J Wort
- National Heart and Lung Institute, Imperial College London, London, UK
- National PH Service, Royal Brompton Hospital, London, UK
| | - David G Kiely
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield, UK
- NIHR Biomedical Research Centre Sheffield, Sheffield, UK
| | - Robin Condliffe
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, UK
- Sheffield Pulmonary Vascular Disease Unit, Royal Hallamshire Hospital, Sheffield, UK
| | - Allan Lawrie
- National Heart and Lung Institute, Imperial College London, London, UK
- Department of Infection, Immunity & Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Stefan Gräf
- VPD Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
- NIHR BioResource for Translational Research, Cambridge Biomedical Campus, Cambridge, UK
| | - Nicholas W Morrell
- VPD Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Martin R Wilkins
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Inga Prokopenko
- Department of Clinical and Experimental Medicine, University of Surrey, Surrey, UK
| | | |
Collapse
|
2
|
DNA hypomethylation during MSC chondrogenesis occurs predominantly at enhancer regions. Sci Rep 2020; 10:1169. [PMID: 31980739 PMCID: PMC6981252 DOI: 10.1038/s41598-020-58093-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 12/21/2019] [Indexed: 12/12/2022] Open
Abstract
Regulation of transcription occurs in a cell type specific manner orchestrated by epigenetic mechanisms including DNA methylation. Methylation changes may also play a key role in lineage specification during stem cell differentiation. To further our understanding of epigenetic regulation in chondrocytes we characterised the DNA methylation changes during chondrogenesis of mesenchymal stem cells (MSCs) by Infinium 450 K methylation array. Significant DNA hypomethylation was identified during chondrogenic differentiation including changes at many key cartilage gene loci. Integration with chondrogenesis gene expression data revealed an enrichment of significant CpGs in upregulated genes, while characterisation of significant CpG loci indicated their predominant localisation to enhancer regions. Comparison with methylation profiles of other tissues, including healthy and diseased adult cartilage, identified chondrocyte-specific regions of hypomethylation and the overlap with differentially methylated CpGs in osteoarthritis. Taken together we have associated DNA methylation levels with the chondrocyte phenotype. The consequences of which has potential to improve cartilage generation for tissue engineering purposes and also to provide context for observed methylation changes in cartilage diseases such as osteoarthritis.
Collapse
|
3
|
Zhu S, Qiu H, Bennett S, Kuek V, Rosen V, Xu H, Xu J. Chondromodulin-1 in health, osteoarthritis, cancer, and heart disease. Cell Mol Life Sci 2019; 76:4493-4502. [PMID: 31317206 PMCID: PMC6841647 DOI: 10.1007/s00018-019-03225-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 12/21/2022]
Abstract
The human chondromodulin-1 (Chm-1, Chm-I, CNMD, or Lect1) gene encodes a 334 amino acid type II transmembrane glycoprotein protein with characteristics of a furin cleavage site and a putative glycosylation site. Chm-1 is expressed most predominantly in healthy and developing avascular cartilage, and healthy cardiac valves. Chm-1 plays a vital role during endochondral ossification by the regulation of angiogenesis. The anti-angiogenic and chondrogenic properties of Chm-1 are attributed to its role in tissue development, homeostasis, repair and regeneration, and disease prevention. Chm-1 promotes chondrocyte differentiation, and is regulated by versatile transcription factors, such as Sox9, Sp3, YY1, p300, Pax1, and Nkx3.2. Decreased expression of Chm-1 is implicated in the onset and progression of osteoarthritis and infective endocarditis. Chm-1 appears to attenuate osteoarthritis progression by inhibiting catabolic activity, and to mediate anti-inflammatory effects. In this review, we present the molecular structure and expression profiling of Chm-1. In addition, we bring a summary to the potential role of Chm-1 in cartilage development and homeostasis, osteoarthritis onset and progression, and to the pathogenic role of Chm-1 in infective endocarditis and cancers. To date, knowledge of the Chm-1 receptor, cellular signalling, and the molecular mechanisms of Chm-1 is rudimentary. Advancing our understanding the role of Chm-1 and its mechanisms of action will pave the way for the development of Chm-1 as a therapeutic target for the treatment of diseases, such as osteoarthritis, infective endocarditis, and cancer, and for potential tissue regenerative bioengineering applications.
Collapse
Affiliation(s)
- Sipin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Molecular Laboratory and the Division of Regenerative Biology, School of Biomedical Sciences, M Block, QEII Medical Centre, The University of Western Australia (M504), 35 Stirling Hwy, Perth, WA, 6009, Australia
| | - Heng Qiu
- Molecular Laboratory and the Division of Regenerative Biology, School of Biomedical Sciences, M Block, QEII Medical Centre, The University of Western Australia (M504), 35 Stirling Hwy, Perth, WA, 6009, Australia
| | - Samuel Bennett
- Molecular Laboratory and the Division of Regenerative Biology, School of Biomedical Sciences, M Block, QEII Medical Centre, The University of Western Australia (M504), 35 Stirling Hwy, Perth, WA, 6009, Australia
| | - Vincent Kuek
- Molecular Laboratory and the Division of Regenerative Biology, School of Biomedical Sciences, M Block, QEII Medical Centre, The University of Western Australia (M504), 35 Stirling Hwy, Perth, WA, 6009, Australia
| | - Vicki Rosen
- Developmental Biology, Harvard School of Dental Medicine, Boston, MA, 02115, USA
| | - Huazi Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
- Molecular Laboratory and the Division of Regenerative Biology, School of Biomedical Sciences, M Block, QEII Medical Centre, The University of Western Australia (M504), 35 Stirling Hwy, Perth, WA, 6009, Australia.
| |
Collapse
|
4
|
von Heyking K, Calzada-Wack J, Göllner S, Neff F, Schmidt O, Hensel T, Schirmer D, Fasan A, Esposito I, Müller-Tidow C, Sorensen PH, Burdach S, Richter GHS. The endochondral bone protein CHM1 sustains an undifferentiated, invasive phenotype, promoting lung metastasis in Ewing sarcoma. Mol Oncol 2017; 11:1288-1301. [PMID: 28319320 PMCID: PMC5579336 DOI: 10.1002/1878-0261.12057] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/24/2017] [Accepted: 03/08/2017] [Indexed: 12/11/2022] Open
Abstract
Ewing sarcomas (ES) are highly malignant, osteolytic bone or soft tissue tumors, which are characterized by EWS–ETS translocations and early metastasis to lung and bone. In this study, we investigated the role of the BRICHOS chaperone domain‐containing endochondral bone protein chondromodulin I (CHM1) in ES pathogenesis. CHM1 is significantly overexpressed in ES, and chromosome immunoprecipitation (ChIP) data demonstrate CHM1 to be directly bound by an EWS–ETS translocation, EWS‐FLI1. Using RNA interference, we observed that CHM1 promoted chondrogenic differentiation capacity of ES cells but decreased the expression of osteolytic genes such as HIF1A,IL6,JAG1, and VEGF. This was in line with the induction of the number of tartrate‐resistant acid phosphatase (TRAP+)‐stained osteoclasts in an orthotopic model of local tumor growth after CHM1 knockdown, indicating that CHM1‐mediated inhibition of osteomimicry might play a role in homing, colonization, and invasion into bone tissues. We further demonstrate that CHM1 enhanced the invasive potential of ES cells in vitro. This invasiveness was in part mediated via CHM1‐regulated matrix metallopeptidase 9 expression and correlated with the observation that, in an xenograft mouse model, CHM1 was essential for the establishment of lung metastases. This finding is in line with the observed increase in CHM1 expression in patient specimens with ES lung metastases. Our results suggest that CHM1 seems to have pleiotropic functions in ES, which need to be further investigated, but appears to be essential for the invasive and metastatic capacities of ES.
Collapse
Affiliation(s)
- Kristina von Heyking
- Laboratory for Functional Genomics and Transplantation Biology, Children's Cancer Research Center and Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Comprehensive Cancer Center Munich (CCCM), German Translational Cancer Research Consortium (DKTK), Munich, Germany
| | - Julia Calzada-Wack
- Institute of Pathology, Helmholtz Zentrum München - German Research Centre for Environmental Health (GmbH), Neuherberg, Germany
| | - Stefanie Göllner
- Department of Medicine IV, Hematology and Oncology, University Hospital Halle, Germany
| | - Frauke Neff
- Institute of Pathology, Helmholtz Zentrum München - German Research Centre for Environmental Health (GmbH), Neuherberg, Germany
| | - Oxana Schmidt
- Laboratory for Functional Genomics and Transplantation Biology, Children's Cancer Research Center and Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Comprehensive Cancer Center Munich (CCCM), German Translational Cancer Research Consortium (DKTK), Munich, Germany
| | - Tim Hensel
- Laboratory for Functional Genomics and Transplantation Biology, Children's Cancer Research Center and Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Comprehensive Cancer Center Munich (CCCM), German Translational Cancer Research Consortium (DKTK), Munich, Germany
| | - David Schirmer
- Laboratory for Functional Genomics and Transplantation Biology, Children's Cancer Research Center and Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Comprehensive Cancer Center Munich (CCCM), German Translational Cancer Research Consortium (DKTK), Munich, Germany
| | - Annette Fasan
- Laboratory for Functional Genomics and Transplantation Biology, Children's Cancer Research Center and Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Comprehensive Cancer Center Munich (CCCM), German Translational Cancer Research Consortium (DKTK), Munich, Germany
| | | | - Carsten Müller-Tidow
- Department of Medicine IV, Hematology and Oncology, University Hospital Halle, Germany.,Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, Germany
| | - Poul H Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, Canada
| | - Stefan Burdach
- Laboratory for Functional Genomics and Transplantation Biology, Children's Cancer Research Center and Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Comprehensive Cancer Center Munich (CCCM), German Translational Cancer Research Consortium (DKTK), Munich, Germany
| | - Günther H S Richter
- Laboratory for Functional Genomics and Transplantation Biology, Children's Cancer Research Center and Department of Pediatrics, Klinikum rechts der Isar, Technische Universität München, Comprehensive Cancer Center Munich (CCCM), German Translational Cancer Research Consortium (DKTK), Munich, Germany
| |
Collapse
|
5
|
Mutant IDH1 Dysregulates the Differentiation of Mesenchymal Stem Cells in Association with Gene-Specific Histone Modifications to Cartilage- and Bone-Related Genes. PLoS One 2015; 10:e0131998. [PMID: 26161668 PMCID: PMC4498635 DOI: 10.1371/journal.pone.0131998] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 06/09/2015] [Indexed: 12/14/2022] Open
Abstract
Somatic mutations in the isocitrate dehydrogenase (IDH)1/2 genes endow encoding proteins with neomorphic activity to produce the potential oncometabolite, 2-hydroxyglutarate (2-HG), which induces the hypermethylation of histones and DNA. The incidence of IDH1/2 mutations in cartilaginous tumors was previously shown to be the highest among various types of tumors, except for those in the central nervous system. Mutations have been detected in both benign (enchondromas) and malignant (chondrosarcomas) types of cartilaginous tumors, whereas they have rarely been found in other mesenchymal tumors such as osteosarcomas. To address this unique tumor specificity, we herein examined the effects of IDH1 R132C, which is the most prevalent mutant in cartilaginous tumors, on the differentiation properties of human mesenchymal stem cells (hMSCs). The induction of the IDH1 R132C gene into MSCs markedly increased the amount of 2-HG and up-regulated global histone methylation. The induction of IDH1 R132C promoted the chondrogenic differentiation of hMSCs by enhancing the expression of SOX9 and COL2A1 genes in association with an increase in the active mark (H3K4me3), but disrupted cartilage matrix formation. On the other hand, IDH1 R132C inhibited expression of the ALPL gene in association with an increase in the repressive mark (H3K9me3), and subsequently inhibited the osteogenic properties of hMSCs and human osteosarcoma cells. Since osteogenic properties are an indispensable feature for the diagnosis of osteosarcoma, the inhibitory effects of IDH1 R132C on osteogenic properties may contribute to the lack of osteosarcomas with the IDH1 R132C mutation. These results suggested that IDH1 R132C contributed to the formation of cartilaginous tumors by dysregulating the chondrogenic and osteogenic differentiation of hMSCs via gene-specific histone modulation.
Collapse
|
6
|
Yokoyama K, Ikeya M, Umeda K, Oda H, Nodomi S, Nasu A, Matsumoto Y, Izawa K, Horigome K, Kusaka T, Tanaka T, Saito MK, Yasumi T, Nishikomori R, Ohara O, Nakayama N, Nakahata T, Heike T, Toguchida J. Enhanced chondrogenesis of induced pluripotent stem cells from patients with neonatal-onset multisystem inflammatory disease occurs via the caspase 1-independent cAMP/protein kinase A/CREB pathway. Arthritis Rheumatol 2015; 67:302-14. [PMID: 25302486 DOI: 10.1002/art.38912] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 10/07/2014] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Neonatal-onset multisystem inflammatory disease (NOMID) is a dominantly inherited autoinflammatory disease caused by NLRP3 mutations. NOMID pathophysiology is explained by the NLRP3 inflammasome, which produces interleukin-1β (IL-1β). However, epiphyseal overgrowth in NOMID is resistant to anti-IL-1 therapy and may therefore occur independently of the NLRP3 inflammasome. This study was undertaken to investigate the effect of mutated NLRP3 on chondrocytes using induced pluripotent stem cells (iPSCs) from patients with NOMID. METHODS We established isogenic iPSCs with wild-type or mutant NLRP3 from 2 NOMID patients with NLRP3 somatic mosaicism. The iPSCs were differentiated into chondrocytes in vitro and in vivo. The phenotypes of chondrocytes with wild-type and mutant NLRP3 were compared, particularly the size of the chondrocyte tissue produced. RESULTS Mutant iPSCs produced larger chondrocyte masses than wild-type iPSCs owing to glycosaminoglycan overproduction, which correlated with increased expression of the chondrocyte master regulator SOX9. In addition, in vivo transplantation of mutant cartilaginous pellets into immunodeficient mice caused disorganized endochondral ossification. Enhanced chondrogenesis was independent of caspase 1 and IL-1, and thus the NLRP3 inflammasome. Investigation of the human SOX9 promoter in chondroprogenitor cells revealed that the CREB/ATF-binding site was critical for SOX9 overexpression caused by mutated NLRP3. This was supported by increased levels of cAMP and phosphorylated CREB in mutant chondroprogenitor cells. CONCLUSION Our findings indicate that the intrinsic hyperplastic capacity of NOMID chondrocytes is dependent on the cAMP/PKA/CREB pathway, independent of the NLRP3 inflammasome.
Collapse
|
7
|
Zhou S, Zhang Z, Xu G. Notable epigenetic role of hyperhomocysteinemia in atherogenesis. Lipids Health Dis 2014; 13:134. [PMID: 25142226 PMCID: PMC4156629 DOI: 10.1186/1476-511x-13-134] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 08/15/2014] [Indexed: 12/21/2022] Open
Abstract
Atherosclerosis is associated with multiple genetic and modifiable risk factors. There is an increasing body of evidences to indicate that epigenetic mechanisms also play an essential role in atherogenesis by influencing gene expression. Homocysteine is a sulfur-containing amino acid formed during methionine metabolism. Elevated plasma level of homocysteine is generally termed as hyperhomocysteinemia. As a potential risk factor for cardiovascular diseases, hyperhomocysteinemia may initiate or motivate atherogenesis by modification of DNA methylation. The underlying epigenetic mechanism is still unclear with controversial findings. This review focuses on epigenetic involvement and mechanisms of hyperhomocysteinemia in atherogenesis. Considering the potential beneficial effects of anti-homocysteinemia treatments in preventing atherosclerosis, further studies on the role of hyperhomocysteinemia in atherogenesis are warranted.
Collapse
Affiliation(s)
- Shuyu Zhou
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Road, Nanjing, 210002 Jiangsu Province China
| | - Zhizhong Zhang
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Road, Nanjing, 210002 Jiangsu Province China
| | - Gelin Xu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Road, Nanjing, 210002 Jiangsu Province China
| |
Collapse
|
8
|
Reynard LN, Bui C, Syddall CM, Loughlin J. CpG methylation regulates allelic expression of GDF5 by modulating binding of SP1 and SP3 repressor proteins to the osteoarthritis susceptibility SNP rs143383. Hum Genet 2014; 133:1059-73. [PMID: 24861163 PMCID: PMC4099533 DOI: 10.1007/s00439-014-1447-z] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 05/12/2014] [Indexed: 01/25/2023]
Abstract
GDF5 encodes an extracellular signalling molecule that is essential for normal skeletal development. The rs144383 C to T SNP located in the 5'UTR of this gene is functional and has a pleiotropic effect on the musculoskeletal system, being a risk factor for knee-osteoarthritis (OA), congenital hip dysplasia, lumbar disc degeneration and Achilles tendon pathology. rs143383 exerts a joint-wide effect on GDF5 expression, with expression of the OA-associated T allele being significantly reduced relative to the C allele, termed allelic expression imbalance. We have previously reported that the GDF5 locus is subject to DNA methylation and that allelic imbalance of rs143383 is mediated by SP1, SP3 and DEAF1 transcriptional repressors. In this study, we have assayed GDF5 methylation in normal and osteoarthritic cartilage, and investigated the effect of methylation on the allelic imbalance of rs143383. We observed demethylation of the GDF5 5'UTR in OA knee cartilage relative to both OA (p = 0.009) and non-OA (p = 0.001) hip cartilage, with the most significant demethylation observed at the highly conserved +37 CpG site located 4 bp upstream of rs143383. Methylation modulates the level and direction of allelic imbalance of rs143383, with methylation of the +37 CpG dinucleotide within the SP1/SP3 binding site having an allele-specific effect on SP1 and SP3 binding. Furthermore, methylation attenuated the repressive effects of SP1, SP3 and DEAF1 on GDF5 promoter activity. This data suggest that the differential methylation of the +37 CpG site between osteoarthritic hip and knee cartilage may be responsible for the knee-specific effect of rs143383 on OA susceptibility.
Collapse
Affiliation(s)
- Louise N Reynard
- Musculoskeletal Research Group, Institute of Cellular Medicine, 4th Floor Catherine Cookson Building, The Medical School, Framlington Place, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK,
| | | | | | | |
Collapse
|
9
|
Kim GH, Ryan JJ, Archer SL. The role of redox signaling in epigenetics and cardiovascular disease. Antioxid Redox Signal 2013; 18:1920-36. [PMID: 23480168 PMCID: PMC3624767 DOI: 10.1089/ars.2012.4926] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 12/24/2012] [Accepted: 01/15/2013] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE The term epigenetics refers to the changes in the phenotype and gene expression that occur without alterations in the DNA sequence. There is a rapidly growing body of evidence that epigenetic modifications are involved in the pathological mechanisms of many cardiovascular diseases (CVDs), which intersect with many of the pathways involved in oxidative stress. RECENT ADVANCES Most studies relating epigenetics and human pathologies have focused on cancer. There has been a limited study of epigenetic mechanisms in CVDs. Although CVDs have multiple established genetic and environmental risk factors, these explain only a portion of the total CVD risk. The epigenetic perspective is beginning to shed new light on how the environment influences gene expression and disease susceptibility in CVDs. Known epigenetic changes contributing to CVD include hypomethylation in proliferating vascular smooth muscle cells in atherosclerosis, changes in estrogen receptor-α (ER-α) and ER-β methylation in vascular disease, decreased superoxide dismutase 2 expression in pulmonary hypertension (PH), as well as trimethylation of histones H3K4 and H3K9 in congestive heart failure. CRITICAL ISSUES In this review, we discuss the epigenetic modifications in CVDs, including atherosclerosis, congestive heart failure, hypertension, and PH, with a focus on altered redox signaling. FUTURE DIRECTIONS As advances in both the methodology and technology accelerate the study of epigenetic modifications, the critical role they play in CVD is beginning to emerge. A fundamental question in the field of epigenetics is to understand the biochemical mechanisms underlying reactive oxygen species-dependent regulation of epigenetic modification.
Collapse
Affiliation(s)
- Gene H Kim
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, Illinois 60637, USA.
| | | | | |
Collapse
|
10
|
Kasaai B, Gaumond MH, Moffatt P. Regulation of the bone-restricted IFITM-like (Bril) gene transcription by Sp and Gli family members and CpG methylation. J Biol Chem 2013; 288:13278-94. [PMID: 23530031 DOI: 10.1074/jbc.m113.457010] [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: 01/01/2023] Open
Abstract
BACKGROUND BRIL is a bone-specific membrane protein that is involved in osteogenesis imperfecta type V. RESULTS Bril transcription is activated by Sp1, Sp3, OSX, and GLI2 and by CpG demethylation. CONCLUSION Regulation of Bril involves trans-acting factors integrating at conserved promoter elements and epigenetic modifications. SIGNIFICANCE Identification of the mechanisms governing Bril transcription is important to understand its role in skeletal biology. Bril encodes a small membrane protein present in osteoblasts. In humans, a single recurrent mutation in the 5'-UTR of BRIL causes osteogenesis imperfecta type V. The exact function of BRIL and the mechanism by which it contributes to disease are still unknown. The goal of the current study was to characterize the mechanisms governing Bril transcription in humans, rats, and mice. In the three species, as detected by luciferase reporter assays in UMR106 cells, we found that most of the base-line regulatory activity was localized within ∼250 bp upstream of the coding ATG. Co-transfection experiments indicated that Sp1 and Sp3 were potent inducers of the promoter activity, through the binding of several GC-rich boxes. Osterix was a weak activator but acted cooperatively with Sp1 and GLI2 to synergistically induce the BRIL promoter. GLI2, a mediator of hedgehog signaling pathway, was also a potent activator of BRIL through a single GLI binding site. Correspondingly, agonists of the hedgehog pathway (purmorphamine and Indian hedgehog) in MC3T3 osteoblasts led to increased BRIL levels. The BRIL promoter activity was also found to be negatively modulated through two different mechanisms. First, the ZFP354C zinc finger protein repressed basal and Sp1-induced activity. Second, CpG methylation of the promoter region correlated with an inactive state and prevented Sp1 activation. The data provide the very first analyses of the cis- and trans-acting factors regulating Bril transcription. They revealed key roles for the Sp members and GLI2 that possibly cooperate to activate Bril when the promoter becomes demethylated.
Collapse
Affiliation(s)
- Bahar Kasaai
- Shriners Hospital for Children, Montreal, Quebec H3G 1A6, Canada
| | | | | |
Collapse
|
11
|
Abstract
Epigenetics refers to changes in phenotype and gene expression that occur without alterations in DNA sequence. Epigenetic modifications of the genome can be acquired de novo and are potentially heritable. This review focuses on the emerging recognition of a role for epigenetics in the development of pulmonary arterial hypertension (PAH). Lessons learned from the epigenetics in cancer and neurodevelopmental diseases, such as Prader-Willi syndrome, can be applied to PAH. These syndromes suggest that there is substantial genetic and epigenetic cross-talk such that a single phenotype can result from a genetic cause, an epigenetic cause, or a combined abnormality. There are three major mechanisms of epigenetic regulation, including methylation of CpG islands, mediated by DNA methyltransferases, modification of histone proteins, and microRNAs. There is substantial interaction between these epigenetic mechanisms. Recently, it was discovered that there may be an epigenetic component to PAH. In PAH there is downregulation of superoxide dismutase 2 (SOD2) and normoxic activation of hypoxia inducible factor (HIF-1α). This decrease in SOD2 results from methylation of CpG islands in SOD2 by lung DNA methyltransferases. The partial silencing of SOD2 alters redox signaling, activates HIF-1α) and leads to excessive cell proliferation. The same hyperproliferative epigenetic abnormality occurs in cancer. These epigenetic abnormalities can be therapeutically reversed. Epigenetic mechanisms may mediate gene-environment interactions in PAH and explain the great variability in susceptibility to stimuli such as anorexigens, virus, and shunts. Epigenetics may be relevant to the female predisposition to PAH and the incomplete penetrance of BMPR2 mutations in familial PAH.
Collapse
Affiliation(s)
- Gene H Kim
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | | | | | | |
Collapse
|
12
|
Abstract
Osteosarcoma is one of the most prevalent primary bone tumors. The pathogenesis and molecular development of this tumor remains elusive. The prognosis is unfavorable due to lack of effective treatment methods. Recent advances in the epigenetics have brought a profound impact on the understanding of molecular mechanisms that lead to osteosarcoma. In this review, we summarized the current literature on epigenetic changes that are thought to contribute to the carcinogenesis of osteosarcoma, and discussed the potential diagnostic and therapeutic applications as well as future areas of research.
Collapse
|
13
|
Affiliation(s)
- Diane E. Handy
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Rita Castro
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Metabolism & Genetics Group, Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon, Portugal
| | - Joseph Loscalzo
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
14
|
|
15
|
Aoyama T, Okamoto T, Fukiage K, Otsuka S, Furu M, Ito K, Jin Y, Ueda M, Nagayama S, Nakayama T, Nakamura T, Toguchida J. Histone modifiers, YY1 and p300, regulate the expression of cartilage-specific gene, chondromodulin-I, in mesenchymal stem cells. J Biol Chem 2010; 285:29842-50. [PMID: 20663886 DOI: 10.1074/jbc.m110.116319] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Elucidating the regulatory mechanism for tissue-specific gene expression is key to understanding the differentiation process. The chondromodulin-I gene (ChM-I) is a cartilage-specific gene, the expression of which is regulated by the transcription factor, Sp3. The binding of Sp3 to the core-promoter region is regulated by the methylation status of the Sp3-binding motif as we reported previously. In this study, we have investigated the molecular mechanisms of the down-regulation of ChM-I expression in mesenchymal stem cells (MSCs) and normal mesenchymal tissues other than cartilage. The core-promoter region of cells in bone and peripheral nerve tissues was hypermethylated, whereas the methylation status in cells of other tissues including MSCs did not differ from that in cells of cartilage, suggesting the presence of inhibitory mechanisms other than DNA methylation. We found that a transcriptional repressor, YY1, negatively regulated the expression of ChM-I by recruiting histone deacetylase and thus inducing the deacetylation of associated histones. As for a positive regulator, we found that a transcriptional co-activator, p300, bound to the core-promoter region with Sp3, inducing the acetylation of histone. Inhibition of YY1 in combination with forced expression of p300 and Sp3 restored the expression of ChM-I in cells with a hypomethylated promoter region, but not in cells with hypermethylation. These results suggested that the expression of tissue-specific genes is regulated in two steps; reversible down-regulation by transcriptional repressor complex and tight down-regulation via DNA methylation.
Collapse
Affiliation(s)
- Tomoki Aoyama
- Institute for Frontier Medical Sciences, Kyoto University,Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Zimmermann P, Boeuf S, Dickhut A, Boehmer S, Olek S, Richter W. Correlation of COL10A1 induction during chondrogenesis of mesenchymal stem cells with demethylation of two CpG sites in the COL10A1 promoter. ACTA ACUST UNITED AC 2010; 58:2743-53. [PMID: 18759285 DOI: 10.1002/art.23736] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Human articular chondrocytes do not express COL10A1 and do not undergo hypertrophy except in close vicinity to subchondral bone. In contrast, chondrocytes produced in vitro from mesenchymal stem cells (MSCs) show premature COL10A1 expression and cannot form stable ectopic cartilage transplants, which indicates that they may be phenotypically unstable and not suitable for treatment of articular cartilage lesions. CpG methylation established during natural development may play a role in suppression of COL10A1 expression and hypertrophy in human articular chondrocytes. This study was undertaken to compare gene methylation patterns and expression of COL10A1 and COL2A1 in chondrocyte and MSC populations, in order to determine whether failed genomic methylation patterns correlate with an unstable chondrocyte phenotype after chondrogenesis of MSCs. METHODS COL10A1 and COL2A1 regulatory gene regions were computationally searched for CpG-rich regions. CpG methylation of genomic DNA from human articular chondrocytes, MSCs, and MSC-derived chondrocytes was analyzed by Combined Bisulfite Restriction Analysis and by sequencing of polymerase chain reaction fragments amplified from bisulfite-treated genomic DNA. RESULTS The CpG island around the transcription start site of COL2A1 was unmethylated in all cell groups independent of COL2A1 expression, while 9 tested CpG sites in the sparse CpG promoter of COL10A1 were consistently methylated in human articular chondrocytes. Induction of COL10A1 expression during chondrogenesis of MSCs correlated with demethylation of 2 CpG sites in the COL10A1 promoter. CONCLUSION Our findings indicate that methylation-based COL10A1 gene silencing is established in cartilage tissue and human articular chondrocytes. Altered methylation levels at 2 CpG sites of COL10A1 in MSCs and their demethylation during chondrogenesis may facilitate induction of COL10A1 as observed during in vitro chondrogenesis of MSCs.
Collapse
Affiliation(s)
- Peter Zimmermann
- Orthopaedic University Hospital of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
17
|
Rapid quantification of DNA methylation by measuring relative peak heights in direct bisulfite-PCR sequencing traces. J Transl Med 2010; 90:282-90. [PMID: 20010852 DOI: 10.1038/labinvest.2009.132] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Various technologies are currently available to quantify DNA methylation. However, rapid and simple methods for determining the DNA methylation status of CpG sites in genes still remain elusive. In this report, we describe a novel method for the rapid quantification of CpG methylation on the basis of direct bisulfite-PCR sequencing method. According to the principles of bisulfite-PCR, converting unmethylated cytosines to thymine while leaving methylated cytosines unchanged, we regard the CpG site as a SNP and estimate the methylation status of cytosines in the given CG dinucleotides by measuring the ratio of the cytosine peak height to the sum of cytosine and thymine peak heights in automated DNA sequencing traces. Furthermore, we take several effective measures to break through the 'bottleneck' problems that render the routine bisulfite sequencing method unsuitable for quantitative methylation. In comparison with pyrosequencing and bisulfite-cloning sequencing, our method is confirmed to be a simple, high-throughput and cost-effective technology for determining the methylation status of specific genes. Accordingly, this novel method is anticipated to be an efficient and economical alternative tool for rapid quantification of methylation patterns in screening large numbers of clinical samples across multiple genes.
Collapse
|
18
|
Zhou H, Kepa JK, Siegel D, Miura S, Hiraki Y, Ross D. Benzene metabolite hydroquinone up-regulates chondromodulin-I and inhibits tube formation in human bone marrow endothelial cells. Mol Pharmacol 2009; 76:579-87. [PMID: 19525446 DOI: 10.1124/mol.109.057323] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Bone marrow is a major target of benzene toxicity, and NAD-(P)H:quinone oxidoreductase (NQO1), an enzyme protective against benzene toxicity, is present in human bone marrow endothelial cells, which form the hematopoietic stem cell vascular niche. In this study, we have employed a transformed human bone marrow endothelial cell (TrHBMEC) line to study the adverse effects induced by the benzene metabolite hydroquinone. Hydroquinone inhibited TrHBMEC tube formation at concentrations that were not overtly toxic, as demonstrated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide or sulforhodamine B analysis. Hydroquinone was found to up-regulate chondromodulin-I (ChM-I), a protein that promotes chondrocyte growth and inhibits endothelial cell growth and tube formation. Recombinant human ChM-I protein inhibited tube formation in TrHBMECs, suggesting that up-regulation of ChM-I may explain the ability of hydroquinone to inhibit TrHB-MEC tube formation. To explore this possibility further, anti-ChM-I small interfering RNA (siRNA) was used to deplete ChM-I mRNA and protein. Pretreatment with anti-ChM-I siRNA markedly abrogated hydroquinone-induced inhibition of tube formation in TrHBMECs. Overexpression of the protective enzyme NQO1 in TrHBMECs inhibited the up-regulation of ChM-I and abrogated the inhibition of tube formation induced by hydroquinone. In summary, hydroquinone treatment up-regulated ChM-I and inhibited tube formation in TrHBMECs; NQO1 inhibited hydroquinone-induced up-regulation of ChM-I in TrHB-MECs and protected cells from hydroquinone-induced inhibition of tube formation. This study demonstrates that ChM-I up-regulation is one of the underlying mechanisms of inhibition of tube formation and provides a mechanism that may contribute to benzene-induced toxicity at the level of bone marrow endothelium.
Collapse
Affiliation(s)
- Hongfei Zhou
- Department of Pharmaceutical Sciences, School of Pharmacy and Cancer Center, University of Colorado Denver, 12700 East 19th Avenue, Aurora, CO 80045, USA
| | | | | | | | | | | |
Collapse
|
19
|
Zhang W, Tong Q, Li S, Wang X, Wang Q. MG-132 Inhibits Telomerase Activity, Induces Apoptosis and G1Arrest Associated with Upregulated p27kip1 Expression and Downregulated Survivin Expression in Gastric Carcinoma Cells. Cancer Invest 2009; 26:1032-6. [DOI: 10.1080/07357900802104997] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
20
|
Regulation of natriuretic peptide receptor-A gene expression and stimulation of its guanylate cyclase activity by transcription factor Ets-1. Biosci Rep 2009; 29:57-70. [PMID: 18651838 DOI: 10.1042/bsr20080094] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
ANP (atrial natriuretic peptide) exerts its biological effects by binding to GC (guanylate cyclase)-A/NPR (natriuretic peptide receptor)-A, which generates the second messenger cGMP. The molecular mechanism mediating Npr1 (coding for GC-A/NPRA) gene regulation and expression is not well understood. The objective of the present study was to elucidate the mechanism by which Ets-1 [Ets (E twenty-six) transformation-specific sequence] contributes to the regulation of Npr1 gene transcription and expression. Chromatin immunoprecipitation and gel-shift assays confirmed the in vivo and in vitro binding of Ets-1 to the Npr1 promoter. Overexpression of Ets-1 enhanced significantly Npr1 mRNA levels, protein expression, GC activity and ANP-stimulated intracellular accumulation of cGMP in transfected cells. Depletion of endogenous Ets-1 by siRNA (small interfering RNA) dramatically decreased promoter activity by 80%. Moreover, methylation of the Npr1 promoter region (-356 to +55) reduced significantly the promoter activity and hypermethylation around the Ets-1 binding sites directly reduced Ets-1 binding to the Npr1 promoter. Collectively, the present study demonstrates that Npr1 gene transcription and GC activity of the receptor are critically controlled by Ets-1 in target cells.
Collapse
|
21
|
Aoyama T, Okamoto T, Kohno Y, Fukiage K, Otsuka S, Furu M, Ito K, Jin Y, Nagayama S, Nakayama T, Nakamura T, Toguchida J. Cell-specific epigenetic regulation of ChM-I gene expression: Crosstalk between DNA methylation and histone acetylation. Biochem Biophys Res Commun 2008; 365:124-30. [DOI: 10.1016/j.bbrc.2007.10.135] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2007] [Accepted: 10/24/2007] [Indexed: 10/22/2022]
|
22
|
Iliopoulos D, Malizos KN, Tsezou A. Epigenetic regulation of leptin affects MMP-13 expression in osteoarthritic chondrocytes: possible molecular target for osteoarthritis therapeutic intervention. Ann Rheum Dis 2007; 66:1616-21. [PMID: 17502362 PMCID: PMC2095321 DOI: 10.1136/ard.2007.069377] [Citation(s) in RCA: 136] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2007] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To investigate whether epigenetic mechanisms can regulate leptin's expression and affect its downstream targets as metalloproteinases 3,9,13 in osteoarthritic chondrocytes. METHODS DNA methylation in leptin promoter was measured by DNA bisulfite sequencing, and mRNA expression levels were measured by real-time quantitative PCR in osteoarthritic as well as in normal cartilage. Osteoarthritic articular cartilage samples were obtained from two distinct locations of the knee (n = 15); from the main defective area of maximum load (advanced osteoarthritis (OA)) and from adjacent macroscopically intact regions (minimal OA). Using small interference RNA, we tested if leptin downregulation would affect matrix metalloproteinase (MMP)-13 activity. We also evaluated the effect of the demethylating agent, 5'-Aza-2-deoxycytidine (AZA) and of the histone deacetylase inhibitor trichostatin A (TSA) on leptin expression in chondrocyte cultures. Furthermore, we performed chromatin immunoprecipitation in leptin's promoter area. RESULTS We found a correlation between leptin expression and DNA methylation and also that leptin controls MMP-13 activity in chondrocytes. Leptin's downregulation with small interference RNA inhibited MMP-13 expression dramatically. After 5-AZA application in normal chondrocytes, leptin's methylation was decreased, while its expression was upregulated, and MMP-13 was activated. Furthermore, TSA application in normal chondrocyte cultures increased leptin's expression. Also, chromatin immunoprecipitation in leptin's promoter after TSA treatment revealed that histone H3 lysines 9 and 14 were acetylated. CONCLUSION We found that epigenetic mechanisms regulate leptin's expression in chondrocytes affecting its downstream target MMP-13. Small interference RNA against leptin deactivated directly MMP-13, which was upregulated after leptin's epigenetic reactivation, raising the issue of leptin's therapeutic potential for osteoarthritis.
Collapse
Affiliation(s)
- D Iliopoulos
- University of Thessalia, Medical School, Department of Biology, 22 Papakyriazi str. 41 222 Larisa, Greece
| | | | | |
Collapse
|
23
|
Michelotti GA, Brinkley DM, Morris DP, Smith MP, Louie RJ, Schwinn DA. Epigenetic regulation of human alpha1d-adrenergic receptor gene expression: a role for DNA methylation in Sp1-dependent regulation. FASEB J 2007; 21:1979-93. [PMID: 17384146 PMCID: PMC2279228 DOI: 10.1096/fj.06-7118com] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A growing body of evidence implicates alpha1-adrenergic receptors (alpha1ARs) as potent regulators of growth pathways. The three alpha1AR subtypes (alpha1aAR, alpha1bAR, alpha1dAR) display highly restricted tissue expression that undergoes subtype switching with many pathological stimuli, the mechanistic basis of which remains unknown. To gain insight into transcriptional pathways governing cell-specific regulation of the human alpha1dAR subtype, we cloned and characterized the alpha1dAR promoter region in two human cellular models that display disparate levels of endogenous alpha1dAR expression (SK-N-MC and DU145). Results reveal that alpha1dAR basal expression is regulated by Sp1-dependent binding of two promoter-proximal GC boxes, the mutation of which attenuates alpha1dAR promoter activity 10-fold. Mechanistically, chromatin immunoprecipitation data demonstrate that Sp1 binding correlates with expression of the endogenous gene in vivo, correlating highly with alpha1dAR promoter methylation-dependent silencing of both episomally expressed reporter constructs and the endogenous gene. Further, analysis of methylation status of proximal GC boxes using sodium bisulfite sequencing reveals differential methylation of proximal GC boxes in the two cell lines examined. Together, the data support a mechanism of methylation-dependent disruption of Sp1 binding in a cell-specific manner resulting in repression of basal alpha1dAR expression.
Collapse
MESH Headings
- Azacitidine/analogs & derivatives
- Azacitidine/pharmacology
- Base Sequence
- Cell Line, Tumor
- Chromatin/chemistry
- DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors
- DNA Methylation
- Decitabine
- Gene Expression Regulation
- Gene Silencing
- Humans
- Immunoprecipitation
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Promoter Regions, Genetic/genetics
- Protein Binding
- RNA, Messenger/biosynthesis
- Receptors, Adrenergic, alpha-1/biosynthesis
- Receptors, Adrenergic, alpha-1/genetics
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Sequence Analysis, DNA
- Sp1 Transcription Factor/metabolism
- Sulfites/pharmacology
- Transcription, Genetic
Collapse
Affiliation(s)
- Gregory A Michelotti
- Department of Pharmacology/Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Wang X, Feng Y, Pan L, Wang Y, Xu X, Lu J, Huang B. The proximal GC-rich region of p16INK4a gene promoter plays a role in its transcriptional regulation. Mol Cell Biochem 2007; 301:259-66. [PMID: 17333389 DOI: 10.1007/s11010-007-9427-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2006] [Accepted: 02/02/2007] [Indexed: 12/19/2022]
Abstract
p16(INK4a) plays a key role in control of cell cycle progression by negatively regulating the CDK4/6 activity. This study establishes that the p16(INK4a) minimal promoter region required for the transcription factor Sp1 function is mapped at 62 bp upstream of the translation initiation codon. This region is GC-rich and shown to interact specifically with Sp1. siRNA-induced Sp1 silencing resulted in the inhibition of the p16(INK4a) minimal promoter activity. Additionally, by using a promoter sequence-directed siRNA method, we demonstrate that the histone H3 at the GC-rich region in the minimal promoter of p16(INK4a) is hypermethylated, with a concurrent reduction of both the activity of p16(INK4a) promoter and the level of endogenous p16(INK4a) mRNA. Moreover, we show that the specific mutation of the GC-rich region of the minimal promoter resulted in the complete loss of its regulatory activities. We conclude that the region spanning -62 to +1 bp of p16(INK4a) promoter plays a role in p16(INK4a) transcription regulation.
Collapse
Affiliation(s)
- Xiuli Wang
- Institute of Genetics and Cytology, Northeast Normal University, Changchun, PR China
| | | | | | | | | | | | | |
Collapse
|
25
|
Shima Y, Okamoto T, Aoyama T, Yasura K, Ishibe T, Nishijo K, Shibata KR, Kohno Y, Fukiage K, Otsuka S, Uejima D, Nakayama T, Nakamura T, Kiyono T, Toguchida J. In vitro transformation of mesenchymal stem cells by oncogenic H-rasVal12. Biochem Biophys Res Commun 2006; 353:60-6. [PMID: 17173860 DOI: 10.1016/j.bbrc.2006.11.137] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2006] [Accepted: 11/18/2006] [Indexed: 12/19/2022]
Abstract
Tissue stem cells may serve as progenitors for malignant tumors derived from the same tissue. Here, we report the establishment of immortalized human mesenchymal stem cells (ihMSC) and tested the feasibility of using ihMSC as presarcomatous cells. Immortalization was achieved by introducing the genes for human telomerase reverse transcriptase and Bmi1. ihMSC retained the potential for multi-directional differentiation of the original MSC. To transform ihMSC, we introduced an oncogenic H-ras(Val12) gene, and established the cell line ihMSC-ras. ihMSC-ras had the phenotype of fully transformed cells and retained adipogenic and chondrogenic, but not osteogenic, potential. Interestingly, ihMSC-ras demonstrated morphological features of autophagy, and inhibition of the ERK pathway suppressed the production of autophagosomes, indicating that ras/ERK signaling is responsible for the induction of autophagy. Thus ihMSC will serve as a material with which to analyze the tumorigenic and differentiation-modifying effects of candidate oncogenes involved in the development of sarcomas.
Collapse
Affiliation(s)
- Yasuko Shima
- Institute for Frontier Medical Sciences, Kyoto University, Shogoin, Kyoto 606-8507, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Wilson AS, Power BE, Molloy PL. DNA hypomethylation and human diseases. Biochim Biophys Acta Rev Cancer 2006; 1775:138-62. [PMID: 17045745 DOI: 10.1016/j.bbcan.2006.08.007] [Citation(s) in RCA: 327] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Revised: 08/24/2006] [Accepted: 08/27/2006] [Indexed: 12/14/2022]
Abstract
Changes in human DNA methylation patterns are an important feature of cancer development and progression and a potential role in other conditions such as atherosclerosis and autoimmune diseases (e.g., multiple sclerosis and lupus) is being recognised. The cancer genome is frequently characterised by hypermethylation of specific genes concurrently with an overall decrease in the level of 5 methyl cytosine. This hypomethylation of the genome largely affects the intergenic and intronic regions of the DNA, particularly repeat sequences and transposable elements, and is believed to result in chromosomal instability and increased mutation events. This review examines our understanding of the patterns of cancer-associated hypomethylation, and how recent advances in understanding of chromatin biology may help elucidate the mechanisms underlying repeat sequence demethylation. It also considers how global demethylation of repeat sequences including transposable elements and the site-specific hypomethylation of certain genes might contribute to the deleterious effects that ultimately result in the initiation and progression of cancer and other diseases. The use of hypomethylation of interspersed repeat sequences and genes as potential biomarkers in the early detection of tumors and their prognostic use in monitoring disease progression are also examined.
Collapse
Affiliation(s)
- Ann S Wilson
- Preventative Health National Research Flagship, North Ryde, NSW, Australia
| | | | | |
Collapse
|
27
|
Murayama A, Sakura K, Nakama M, Yasuzawa-Tanaka K, Fujita E, Tateishi Y, Wang Y, Ushijima T, Baba T, Shibuya K, Shibuya A, Kawabe YI, Yanagisawa J. A specific CpG site demethylation in the human interleukin 2 gene promoter is an epigenetic memory. EMBO J 2006; 25:1081-92. [PMID: 16498406 PMCID: PMC1409718 DOI: 10.1038/sj.emboj.7601012] [Citation(s) in RCA: 144] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2005] [Accepted: 01/27/2006] [Indexed: 02/05/2023] Open
Abstract
DNA demethylation plays a critical role in transcriptional regulation in differentiated somatic cells. However, there is no experimental evidence that CpG methylation in a small region of a genome restricts gene expression. Here, we show that the anti-CD3repsilon/CD28 antibody stimulation of human CD4+ T cells induces IL2 expression following epigenetic changes, including active demethylation of a specific CpG site, recruitment of Oct-1, and changes in histone modifications. When the stimulatory signal is withdrawn, Oct-1 remains on the enhancer region as a stable marker of the stimulation, causing the second induction to be faster and stronger. Our observations indicate that Oct-1-binding followed by CpG demethylation are key events in the epigenetic regulation of IL2 expression and may act as a memory of the regulatory event.
Collapse
Affiliation(s)
- Akiko Murayama
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba Science City, Ibaraki, Japan
| | - Kazuhisa Sakura
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba Science City, Ibaraki, Japan
| | - Mina Nakama
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba Science City, Ibaraki, Japan
| | - Kayoko Yasuzawa-Tanaka
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba Science City, Ibaraki, Japan
| | - Etsuko Fujita
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba Science City, Ibaraki, Japan
| | - Yukiyo Tateishi
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba Science City, Ibaraki, Japan
| | - Yinan Wang
- Department of Immunology, Institute of Basic Medical Sciences, Graduate School of Comprehensive Human Sciences, Center for TARA, University of Tsukuba, Ibaraki, Japan
| | - Toshikazu Ushijima
- Carcinogenesis Division, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Tadashi Baba
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba Science City, Ibaraki, Japan
| | - Kazuko Shibuya
- Department of Immunology, Institute of Basic Medical Sciences, Graduate School of Comprehensive Human Sciences, Center for TARA, University of Tsukuba, Ibaraki, Japan
| | - Akira Shibuya
- Department of Immunology, Institute of Basic Medical Sciences, Graduate School of Comprehensive Human Sciences, Center for TARA, University of Tsukuba, Ibaraki, Japan
| | - Yoh-ichi Kawabe
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba Science City, Ibaraki, Japan
| | - Junn Yanagisawa
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba Science City, Ibaraki, Japan
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tenno-dai, Tsukuba Science City, Ibaraki 305-8572, Japan. Tel.: +81 29 853 6632; Fax: 81 29 853 4605; E-mail:
| |
Collapse
|
28
|
Zhang Y, Fatima N, Dufau ML. Coordinated changes in DNA methylation and histone modifications regulate silencing/derepression of luteinizing hormone receptor gene transcription. Mol Cell Biol 2005; 25:7929-39. [PMID: 16135786 PMCID: PMC1234307 DOI: 10.1128/mcb.25.18.7929-7939.2005] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We have previously demonstrated that transcription of the luteinizing hormone receptor (LHR) gene is subject to repression by histone deacetylation at its promoter region, where a histone deacetylase (HDAC)/mSin3A complex is anchored at a proximal Sp1 site. The present studies have shown that epigenetic silencing and activation of the LHR gene is achieved through coordinated regulation at both the histone and DNA levels. The HDAC inhibitor trichostatin A (TSA) evoked robust but significantly lower activation of the LHR gene in JAR than in MCF-7 cells. This effect was localized to the 176-bp promoter region, which is highly methylated in JAR and lightly methylated in MCF-7 cells. Consequently, TSA and the DNA demethylating reagent 5-azacytidine (5-AzaC) caused marked synergistic activation of the LHR gene in JAR but not in MCF-7 cells. Multiple site-specific lysine acetylation of H3/H4 is associated with such LHR gene activation. Methylation or acetylation of H3 at K9 is present at the silenced and derepressed LHR promoter, respectively. While DNA methylation levels did not affect the histone code of the LHR gene promoter, demethylation of the promoter CpG sites was necessary for maximal stimulation of this gene. Mechanistically, the combined actions of TSA and 5-AzaC, but not either 5-AzaC or TSA alone, resulted in complete demethylation of the LHR gene promoter in JAR cells. Release of the repressive HDAC/mSin3A complex from the LHR gene promoter in both cell types required both TSA-induced changes of histone modifications and, concurrently, a demethylated promoter. Also, Dnmt1 was largely dissociated from the LHR gene promoter in the presence of TSA or TSA plus 5-AzaC, and binding of MBD2 in JAR cells was diminished upon conversion of the promoter to a demethylated state. Such changes induced a more permissive chromatin where recruitment of polymerase II and TFIIB to the promoter was significantly increased. The activated state of the LHR gene induced by TSA and 5-AzaC in JAR and MCF-7 cells was observed basally in LHR-expressing PLC cells, in which the promoter is unmethylated and associated with hyperacetylated histones. Consequently, PLC cells are unresponsive to drug treatment. These findings have elucidated a regulatory mechanism whereby concurrent dissociation of repressors and association of activators and basal transcriptional components, resulting from coordinated histone hyperacetylation and DNA demethylation, lead to derepression of the LHR gene expression.
Collapse
Affiliation(s)
- Ying Zhang
- ERRB, NICHD, National Institutes of Health, Bldg. 49, Rm. 6A-36, 49 Convent Dr., MSC 4510, Bethesda, MD 20892-4510, USA
| | | | | |
Collapse
|
29
|
Isomura H, Stinski MF, Kudoh A, Daikoku T, Shirata N, Tsurumi T. Two Sp1/Sp3 binding sites in the major immediate-early proximal enhancer of human cytomegalovirus have a significant role in viral replication. J Virol 2005; 79:9597-607. [PMID: 16014922 PMCID: PMC1181558 DOI: 10.1128/jvi.79.15.9597-9607.2005] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2005] [Accepted: 04/06/2005] [Indexed: 01/29/2023] Open
Abstract
We previously demonstrated that the major immediate early (MIE) proximal enhancer containing one GC box and the TATA box containing promoter are minimal elements required for transcription and viral replication in human fibroblast cells (H. Isomura, T. Tsurumi, M. F. Stinski, J. Virol. 78:12788-12799, 2004). After infection, the level of Sp1 increased while Sp3 remained constant. Here we report that either Sp1 or Sp3 transcription factors bind to the GC boxes located at approximately positions -55 and -75 relative to the transcription start site (+1). Both the Sp1 and Sp3 binding sites have a positive and synergistic effect on the human cytomegalovirus (HCMV) major immediate-early (MIE) promoter. There was little to no change in MIE transcription or viral replication for recombinant viruses with one or the other Sp1 or Sp3 binding site mutated. In contrast, mutation of both the Sp1 and Sp3 binding sites caused inefficient MIE transcription and viral replication. These data indicate that the Sp1 and Sp3 binding sites have a significant role in HCMV replication in human fibroblast cells.
Collapse
Affiliation(s)
- Hiroki Isomura
- Division of Virology, Aichi Cancer Center Research Institute, Chikusa-ku, Nagoya, Japan
| | | | | | | | | | | |
Collapse
|
30
|
Hiraki Y, Shukunami C. Angiogenesis inhibitors localized in hypovascular mesenchymal tissues: chondromodulin-I and tenomodulin. Connect Tissue Res 2005; 46:3-11. [PMID: 16019413 DOI: 10.1080/03008200590935547] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The majority of mesenchymal tissues obtain their nutrients via a well-developed network of capillaries. Cartilage, however, is normally devoid of capillary networks and, with the exception of endochondral bone formation, is resistant to vascular invasion from surrounding tissues. However, because of its avascular nature, cartilage is widely regarded as an enriched source of endogenous angiogenesis inhibitors, and many previous attempts have been made to identify these factors. We have identified chondromodulin-I (ChM-I) as an angiogenesis inhibitor derived from extracts of fetal epiphyseal cartilage, based upon its growth inhibitory activity in vascular endothelial cells in vitro. In the musculoskeletal system, ChM-I is specifically expressed in the avascular zones of cartilage. Upon functional expression of human ChM-I precursor cDNA, the purified recombinant protein was found to block the growth of solid tumors by inhibiting angiogenesis. Recently, we also cloned a cDNA that encodes a novel type II transmembrane glycoprotein containing a cysteine rich C-terminal domain homologous to ChM-I. We termed this glycoprotein "tenomodulin" (TeM) after tendons that were found to be the predominant expression sites in addition to other dense connective tissues including ligaments and cornea. Subsequently, by employing an adenovirus-mediated expression system, we demonstrated that the ChM-I-like domain of TeM is both antiangiogenic and antitumorigenic. In this article, we summarize the structural characteristics and biological activities of these two antiangiogenic molecules.
Collapse
Affiliation(s)
- Yuji Hiraki
- Department of Cellular Differentiation, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan.
| | | |
Collapse
|